ArticlePDF Available

Statin use and risk of breast cancer: a meta-analysis of observational studies

Authors:
  • National Institute of Pharmaceutical Education and Research (NIPER) Guwahati

Abstract and Figures

Emerging evidence suggests that statins' may decrease the risk of cancers. However, available evidence on breast cancer is conflicting. We, therefore, examined the association between statin use and risk of breast cancer by conducting a detailed meta-analysis of all observational studies published regarding this subject. PubMed database and bibliographies of retrieved articles were searched for epidemiological studies published up to January 2012, investigating the relationship between statin use and breast cancer. Before meta-analysis, the studies were evaluated for publication bias and heterogeneity. Combined relative risk (RR) and 95 % confidence interval (CI) were calculated using a random-effects model (DerSimonian and Laird method). Subgroup analyses, sensitivity analysis, and cumulative meta-analysis were also performed. A total of 24 (13 cohort and 11 case-control) studies involving more than 2.4 million participants, including 76,759 breast cancer cases contributed to this analysis. We found no evidence of publication bias and evidence of heterogeneity among the studies. Statin use and long-term statin use did not significantly affect breast cancer risk (RR = 0.99, 95 % CI = 0.94, 1.04 and RR = 1.03, 95 % CI = 0.96, 1.11, respectively). When the analysis was stratified into subgroups, there was no evidence that study design substantially influenced the effect estimate. Sensitivity analysis confirmed the stability of our results. Cumulative meta-analysis showed a change in trend of reporting risk of breast cancer from positive to negative in statin users between 1993 and 2011. Our meta-analysis findings do not support the hypothesis that statins' have a protective effect against breast cancer. More randomized clinical trials and observational studies are needed to confirm this association with underlying biological mechanisms in the future
Content may be subject to copyright.
EPIDEMIOLOGY
Statin use and risk of breast cancer: a meta-analysis
of observational studies
Krishna Undela Vallakatla Srikanth
Dipika Bansal
Received: 13 February 2012 / Accepted: 25 June 2012 / Published online: 18 July 2012
ÓSpringer Science+Business Media, LLC. 2012
Abstract Emerging evidence suggests that statins’ may
decrease the risk of cancers. However, available evidence
on breast cancer is conflicting. We, therefore, examined the
association between statin use and risk of breast cancer by
conducting a detailed meta-analysis of all observational
studies published regarding this subject. PubMed database
and bibliographies of retrieved articles were searched for
epidemiological studies published up to January 2012,
investigating the relationship between statin use and breast
cancer. Before meta-analysis, the studies were evaluated
for publication bias and heterogeneity. Combined relative
risk (RR) and 95 % confidence interval (CI) were calcu-
lated using a random-effects model (DerSimonian and
Laird method). Subgroup analyses, sensitivity analysis, and
cumulative meta-analysis were also performed. A total of
24 (13 cohort and 11 case–control) studies involving more
than 2.4 million participants, including 76,759 breast can-
cer cases contributed to this analysis. We found no evi-
dence of publication bias and evidence of heterogeneity
among the studies. Statin use and long-term statin use did
not significantly affect breast cancer risk (RR =0.99,
95 % CI =0.94, 1.04 and RR =1.03, 95 % CI =0.96,
1.11, respectively). When the analysis was stratified into
subgroups, there was no evidence that study design sub-
stantially influenced the effect estimate. Sensitivity analy-
sis confirmed the stability of our results. Cumulative meta-
analysis showed a change in trend of reporting risk of
breast cancer from positive to negative in statin users
between 1993 and 2011. Our meta-analysis findings do not
support the hypothesis that statins’ have a protective effect
against breast cancer. More randomized clinical trials and
observational studies are needed to confirm this association
with underlying biological mechanisms in the future.
Keywords Statin Breast cancer Meta-analysis
Introduction
Breast cancer is by far the most common cancer diagnosed in
women (ranking second in both sexes combined) and the
most common cause of death in women (ranking fifth in both
sexes combined) worldwide [1]. An estimated 1.38 million
women across the world were diagnosed with breast cancer
in 2008, accounting for nearly 23 % of the total cancer cases
and 14 % of the cancer deaths in women [2]. Breast cancer is
now also the leading cause of cancer death among females in
economically developing countries, a shift from the previous
decade during which the most common cause of cancer
death was cervical cancer [2].
Statins’ (3-hydroxy-3-methyl glutaryl-coenzyme A
reductase inhibitors), a therapeutic class of drugs that
reduce plasma cholesterol levels, are used to manage and
prevent coronary heart disease. As such, statins’ are among
the most commonly prescribed drugs worldwide [3].
Recent experimental [4,5] and clinical evidence [6,7]
suggests an additional chemopreventive potential of stat-
ins’. However, there is an inconsistency in the reporting
risk of breast cancer in statin users. Some randomized
clinical trials (RCTs) of statin use in coronary heart dis-
ease, [8,9] report some levels of incidence of breast can-
cer, but most results were ambiguous because of
inadequate power. Moreover, the results from observa-
tional studies published on the association between statin
K. Undela (&)V. Srikanth D. Bansal
Department of Pharmacy Practice, National Institute
of Pharmaceutical Education and Research, SAS Nagar,
Punjab, India
e-mail: krishna.niperian10@gmail.com
123
Breast Cancer Res Treat (2012) 135:261–269
DOI 10.1007/s10549-012-2154-x
use and risk of breast cancer were conflicting, while some
studies reported reduced risk [10,11], others described an
increased risk [1216] and one study have not identified
any effect [17].
This issue was discussed in a meta-analysis done by
Bonovas et al. [18] using seven RCTs and nine observa-
tional studies published between 1993 and 2005, and
concluded, for no association between statin use and breast
cancer. Recently published observational studies after 2005
have shown contrasting results, including decreased risk
[1923], increased risk [2428], and some also reported no
probable association [2934], which raised the interest of
adding new evidence to the previous analysis. In this meta-
analysis, we examined statin use in relation to breast can-
cer, using epidemiological studies published until January
2012. The strength of the present analysis lies in the
inclusion of 24 observational studies that reported data of
more than 2.4 million participants, including 76,759 breast
cancer cases. Inclusion of additional studies to the previous
analysis has enabled us to perform subgroup analyses,
analyzing various risk factors involved in breast cancer and
statin use. In this meta-analysis, we also analyzed the risk
of breast cancer in long-term statin users and the risk of
breast cancer recurrence in statin users, which are not
discussed in the previous meta-analysis.
Materials and methods
Search strategy
A comprehensive literature search was carried out in
PubMed database. Search terms included: ‘‘HMG-CoA
reductase inhibitor(s)’ or ‘statin(s)’ or ‘lipid-lowering
agent(s)’ and ‘cancer(s)’ or ‘neoplasm(s)’ or ‘‘malig-
nancy(ies)’ with limits; Date (January 1966 through Jan-
uary 2012), Humans and English. The title and abstract of
studies were reviewed to exclude irrelevant studies. The
full texts of remaining articles were read to extract infor-
mation on the topic of interest. Bibliographies and citation
sections of retrieved articles were also reviewed for addi-
tional pertinent studies. Abstracts of research presented at
related conferences [American Society of Clinical Oncol-
ogy (ASCO) and American Association for Cancer
Research (AACR)] were also searched.
Selection criteria
The studies considered in this meta-analysis were all
observational (cohort or case–control) studies that evalu-
ated exposure to statins’ and risk of breast cancer. We
included all articles irrespective of publication length; that
is we did not exclude articles published as short reports or
conference abstracts, even though the critical appraisal of
such publications is limited. Articles were excluded from
the analysis if they had insufficient published data for
determining an estimate of relative risk (RR) and confi-
dence interval (CI). When there were multiple publications
from the same population, only data from the most-recent
report were included in the meta-analysis and remaining
were excluded [35,36]. Studies reporting different mea-
sures of RR like risk ratio, rate ratio, hazard ratio (HR), and
odds ratio (OR) were included in the meta-analysis. In
practice, these measures of effect yield a similar estimate
of RR, since the absolute risk of breast cancer is low.
Data extraction and quality assessment
Two investigators (K.U. and V.S.) independently reviewed
the primary studies to assess the appropriateness for
inclusion in the present meta-analysis and data were
extracted. The following information was assayed from
each study: (i) first author’s last name, year of publication,
and country of the population studied; (ii) study design;
(iii) number of female subjects and number of breast cancer
cases; (iv) RR estimates and 95 % CIs; (v) definition of
statin exposure and breast cancer assessment; (vi) control
for potential confounding factors by matching or adjust-
ments, if applicable. We extracted the RR estimates that
reflected the greatest degree of control for potential con-
founding factors.
Statistical analysis
Publication bias was assessed using Begg and Mazumdar
adjusted rank correlation test and the Egger regression
asymmetry test [37,38]. To assess heterogeneity among
the studies, we used the Cochran Qand I
2
statistics; for the
Qstatistic, a Pvalue \0.10 was considered statistically
significant for heterogeneity; for I
2
, a value [50 % is
considered a measure of severe heterogeneity [39]. The
primary measure was combined RR of breast cancer from
individual studies, calculated using the random-effects
model (DerSimonian and Laird method), which accounts
for heterogeneity among the studies. Test for interaction
using summary estimates were performed using the method
described by Altman and Bland [40]. All analyses were
performed using STATA version 11.0 (StataCorp, College
Station, TX). All statistical tests were two-sided and
P\0.05 was considered statistically significant, unless
otherwise specified.
We first estimated the risk of breast cancer in statin
users compared to the non-users. To assess any link
between the (i) long-term statin use and breast cancer risk
and (ii) statin use and breast cancer recurrence risk, we
used the available data from previously collected studies
262 Breast Cancer Res Treat (2012) 135:261–269
123
and some new studies, which reported RR estimates for
these particular associations.
Prespecified subgroup analyses were performed accord-
ing to (i) study design (cohort and case–control), (ii) control
for confounding factors (nC8, nB7), and (iii) studies
before and after Bonovas et al. [18], to examine the impact of
these factors on the association. To test the robustness of
association, we performed a sensitivity analysis by exclud-
ing one study at a time. Cumulative meta-analysis was also
performed to identify the change in trend of reporting risk
over time. The present study was performed according to the
guidelines proposed by the Meta-analysis of Observational
Studies in Epidemiology group [41].
Results
Search results
A total of 1,547,148 citations were identified during the
initial search (Fig. 1). After review of the citations,
1,547,113 citations were found ineligible. Two studies [20,
22] published in an abstract form in ASCO were added
later to the remaining 35 studies. After detailed evaluation
of 37 potential studies, 13 studies were excluded for rea-
sons described in Fig. 1.
Study characteristics
Twenty four relevant studies (13 cohort and 11 case–con-
trol) published between 1993 and 2011were identified. A
total of 2,440,988 female subjects, including 76,759 breast
cancer cases were involved in these studies and followed
for 2–15 years.
Thirteen cohort studies of statin use and risk of breast
cancer were published between 1993 and 2011 which
included 2,042,439 participants and followed for
6–15 years. A total of 1,845 and 17,405 incidents of breast
cancer cases were reported among 251,860 statin users and
1,700,938 non-statin users, respectively. Of them, eight
studies were conducted in United States of America, four in
Europe and remaining one in USA, and Europe. Eight
studies reported RR and 5 reported HR.
Eleven case–control studies were published between
2000 and 2011. These studies included 398,549 partici-
pants and followed for 2–14 years. A total of 1,904 statin
users among 46,575 breast cancer cases, and 6,947 statin
users among 351,974 controls were reported. Of them,
eight studies were conducted in USA and three in Europe.
All case–control studies reported OR.
Of the total 24 studies, 14 are population based and 10
are hospital-based. All studies evaluated exposure to stat-
ins’ and the risk of breast cancer except for three studies
-2 studies reported on similar population
-9 outcome was not breast cancer incidence
-2 exposure was not statin use
13 studies excluded
2 abstract retrieved from ASCO
35 full text studies retrieved for detailed
evaluation
-147 not relevant based on abstract
-838,309 not relevant based on title reading
-24,409 editorials
-81,496 letters
-303,248 case reports
-88,014 clinical trials
-211,490 review articles
1,547,113 citations excluded
1,547,148 citations identified from PubMed
and reviewed
24 studies included in the meta-analysis
Fig. 1 Flowchart representing
the selection process ASCO
American society of clinical
oncology
Breast Cancer Res Treat (2012) 135:261–269 263
123
[11,19,23] that examined the use of all cholesterol-low-
ering drugs and one study [33] examined the use of only
lipophilic statins’ as a surrogate measure for statin use. All
studies were controlled for potential confounding factors
(at least for age) by matching or adjustments. The char-
acteristics of the selected studies are presented in Table 1.
Further, ten studies [17,19,21,2325,2931,34] were
reported RR estimates of the association between long-
term statin use and risk of breast cancer (Table 2), and two
studies [42,43] presented an examination of statin use in
relation to breast cancer recurrence (Table 3).
Main analysis
No publication bias was observed among studies using
Begg’s Pvalue (P=0.71); Egger’s (P=0.32) test and
the funnel plot had an expected funnel shape (Fig. 2).
Because of significant heterogeneity (P
heterogeneity
\0.001,
Table 1 Studies included in the meta-analysis
Author, year
a
(country)
b
Study period
(years)
All female
subjects
BC cases Description
of exposure/
reference
e
Study quality
Definition of
statin use
Number of variables
adjusted
f
Lovastatin study groups, 1993
(US, Canada & Finland) [12]
c
NR 241 3 a Self-reported 1
Blais et al. 2000 (Canada) [10]
d
6 (1988–1994) 715 NR b NR 1, 9, 10, 13, 22
Beck et al. 2003 (Canada) [13]
c
8 (1989–1997) 67,472 879 e Database 1
Cauley et al. 2003 (US) [11]
c
15 (1986–2001) 7,528 240 d Medical records 1–4
Graaf et al. 2004 (Netherlands) [14]
d
3 (1995–1998) 9,182 NR c NR 1–3,6–9,11–13
Kaye and Jick 2004 (UK) [15]
d
12 (1990–2002) 8,091 236 d Medical records 1, 14, 15
Boudreau et al. 2004 (US) [17]
d
2 (1997–1999) 1,982 231 g Medical records 1, 5
Friis et al. 2005 (Denmark) [16]
c
13 (1989–2002) 171,937 3,141 e Database 1, 5, 28, 29
Eliassen et al. 2005 (US) [19]
c
12 (1988–2000) 75,828 1,624 d Self-reported 1, 3, 14, 17, 21, 23–25
Kochhar et al. 2005 (US) [20]
d
6 (1998–2004) 40,421 4,771 d Database 1, 11, 15, 21
Cauley et al. 2006 (US) [21]
c
11 (1993–2004) 156,351 4,383 d Medical records 1, 2, 14, 15, 17, 21, 24, 26–28
Dumasia et al 2006 (US) [22]
d
10 (1995–2005) 1,042 NR d Self-reported NR
Boudreau et al. 2007 (US) [24]
c
14 (1990–2004) 92,888 2,707 g Database 1, 3, 9, 11, 14
Setoguchi et al. 2007 (US) [29]
c
9 (1994–2003) 19991 227 d Medical records 1–3, 11, 20, 17, 29, 30
Coogan et al. 2007 (US) [25]
d
14 (1991–2005) 2,355 69 c Self-reported 1, 3, 14, 18–21
Friedman et al. 2008 (US) [30]
c
9 (1994–2003) NR 1,706 e Medical records 35
Smeeth et al. 2008 (UK) [26]
c
11 (1995–2006) 364,854 3,204 d Medical records 1–11, 22, 31, 32, 37–40
Pocobelli et al. 2008 (US) [31]
d
6 (1995–2001) 8,620 607 g Self-reported 1, 3, 14, 17, 18, 25, 27, 31, 35
Eaton et al. 2009 (US) [27]
d
3 (2005–2008) 189 NR d Self-reported 1
Haukka et al. 2010 (Finland) [32]
c
9 (1996–2005) 6,046 583 d Database 1, 33
Hippisley et al. 2010
(England & Wales) [28]
c
6 (2002–2008) 1,014,197 9,823 d Medical records NR
Woditschka et al. 2010 (US) [33]
d
10 (1997–2007) 247,348 NR d Medical records 3, 36
Jacobs et al. 2011 (US) [23]
c
10 (1997–2007) 65,106 2,489 f Self-reported 1–3, 13–15, 20, 24, 27, 34
Vinogradova et al. 2011 (UK) [34]
d
10 (1998–2008) 78,604 7,708 e Medical records 1, 2, 14, 15, 30, 37–40
NR not reported, BC breast cancer
a
Publication year,
b
country of study conducted,
c
cohort studies,
d
case–control studies
e
a, systematic use of lovastatin versus SEER data; b, any use of statins versus use of bile acid-binding resins; c, regular use of statins versus no use of
statins; d, current use of statins versus no current use of statins; e, any use of statins versus no use of statins; f, current use of cholesterol-lowering
drugs versus never use of cholesterol-lowering drugs; g, ever use of statins versus no use of statins
f
1 age, 2 use of nonsteroidal anti-inflammatory drugs, 3 use of hormones, 4 use of cardiovascular drugs, 5 use of antihypertensive drugs, 6 use of
diuretics, 7 use of angiotensin-converting enzyme inhibitors, 8 use of calcium channel blockers, 9 use of other lipid-lowering therapy, 10 use of fibric
acids, 11 diabetes mellitus, 12 prior hospitalization, 13 comorbidity score, 14 body mass index, 15 smoking, 16 body weight, 17 family history of
breast cancer, 18 education, 19 religion, 20 race, 21 alcohol consumption, 22 previous neoplasms, 23 height, 24 physical activity, 25 menopausal
status, 26 hysterectomy, 27 mammogram, 28 percentage of calories from fat, 29 health service utilization, 30 arthritis, 31 calendar year, 32 propensity
score, 33 follow-up period, 34 history of elevated cholesterol, 35 state of residence, 36 use of oral contraceptives, 37 cardiovascular disease, 38
hypertension, 39 use of Cox2-inhibitors, 40 use of aspirin
264 Breast Cancer Res Treat (2012) 135:261–269
123
I
2
=57 %) was observed, a random-effects model was
chosen over a fixed-effects model and we found that statin
use did not significantly affect the risk breast cancer
(RR =0.99, 95 % CI =0.94, 1.04). Both multivariable
adjusted RR estimates with 95 % CIs of each study and
combined RR is shown in Fig. 3.
Subgroup analyses, sensitivity analysis, and cumulative
meta-analysis
We found no association between statin use and risk of
breast cancer among cohort studies (RR =1.01, 95 %
CI =0.98, 1.04) as well as case–control studies
(RR =0.95, 95 % CI =0.84, 1.08), presented in Table 4.
When we examined if thorough adjustment of potential
confounders could affect the combined RR and degree of
heterogeneity, it was observed that studies with higher
control for potential confounders (nC8) as well as studies
with lower control (nB7) presented no association
(RR =0.99, 95 % CI =0.94, 1.04 and RR =0.99, 95 %
CI =0.92, 1.06, respectively). The combined RR for the
studies published in the same time frame included in Bo-
novas et al. [18] was 1.06 (95 % CI =0.95, 1.18) and
studies published after Bonovas et al. was 0.98 (95 %
CI =0.93, 1.03) presented no association between statin
use and breast cancer. Test for interaction showed non-
significant results for subgroups of study design, adjust-
ment for confounders and time frame of Bonovas et al.
analysis; (P
interaction
=0.35, 1.00 and 0.20, respectively)
(Table 4).
To test the robustness of our findings, we also carried
out a sensitivity analysis. To do this, the overall homoge-
neity and effect size were calculated by removing one
study at a time. This analysis showed no significant vari-
ation in combined RR by excluding any of the study (RR
lies between 0.98 and 1.01), confirming the stability of
present results.
A cumulative meta-analysis of total 24 studies was
carried out to evaluate the cumulative effect estimate over
time. In 1993, Lovastatin study groups reported a signifi-
cant effect estimate of 1.15. By adding an effect estimate
from one study published in 2000 [10] to the earlier, RR
Table 2 Studies evaluating the association between long-term statin
use and risk of breast cancer
Study RR 95 % CI Breast
cancer
cases
Definition of
‘long-term’’
statin use
Boudreau et al. [17]
a
0.70 0.40–1.00 84 [5 years
Eliassen et al. [19]
b
0.93 0.60–1.44 21 [4 years
Cauley et al. [21]
b
0.94 0.75–1.18 108 C3 years
Boudreau et al. [24]
b
1.27 0.89–1.81 57 C5 years
Setoguchi et al. [29]
b
1.28 0.90–1.84 156 C3 years
Coogan et al. [25]
a
1.5 0.7–3.2 20 C5 years
Friedman et al. [30]
b
1.02 0.86–1.21 NR [5 years
Pocobelli et al. [31]
a
0.8 0.5–1.4 62 C10 years
Jacobs et al. [23]
b
1.11 0.98–1.25 429 C5 years
Vinogradova et al. [34]
a
0.85 0.67–1.08 1768 C6 years
RR Relative risk, CI confidence interval, NR not reported
a
Case–control studies
b
Cohort studies
Table 3 Studies evaluating the association between statin use and risk of breast cancer recurrence
Author, Year
a
(Country)
b
Study period
(years)
All female
subjects
BC recurrence
cases
RR (95 % CI)
Kwan et al., 2008 (Oakland) [42]
c
6 (2000–2006) 1811 210 0.67 (0.39, 1.13)
Chae et al. 2011 (US) [43]
c
9 (1999–2008) 703 149 0.43 (0.26, 0.70)
BC Breast cancer, RR relative risk, CI confidence interval
a
Publication year
b
Country of study conducted
c
Cohort studies
Standard error
0.0
0.2
0.4
0.6
2-2
0.8
-1 0 1
Relative risk (lo
g
arithmic scale)
Fig. 2 Funnel plot (publication bias assessment plot) of the relative
risk of developing breast cancer, by the standard error, for all studies.
Circles studies included in the meta-analysis. Relative risks are
displayed on a logarithmic scale. P=0.71 for the Begg’s test, and
P=0.32 for the Egger’s test
Breast Cancer Res Treat (2012) 135:261–269 265
123
reached to 0.78. By adding another effect estimate from the
study published in 2003 [13] to the previous cumulative
estimate, RR reached to 1.07. By adding another 21 studies
published between 2003 and 2011, the overall effect esti-
mate of 0.99 was obtained.
Results for long-term statin use
The calculated combined RR for breast cancer in long-term
statin use was found to be 1.03 (95 % CI =0.96, 1.11)
(Table 4). However, there was high evidence of no heter-
ogeneity among these studies (P
heterogeneity
=0.21,
I
2
=26 %). Stratification by study design showed that the
association was neutral in cohort studies (RR =1.07, 95 %
CI =0.99, 1.17) and non-significant inverse association in
case–control studies (RR =0.84, 95 % CI =0.70, 1.02)
with P
interaction
being 0.02.
Results for recurrence of breast cancer in statin users
The calculated combined RR of two studies for breast
cancer recurrence in statin users was found to be 0.53
(95 % CI =0.37, 0.76) with no heterogeneity among the
studies (P
heterogeneity
=0.23) (Table 4).
Discussion
In the past decade, the role of statins’ in the development of
breast cancer has been increasingly understood. With the
present updated combined analysis of 24 observational
studies currently available, it is obvious that there is no
reduction in breast cancer risk among statin users as
compared to non-users and this association remained stable
even after the sensitivity analysis. Our results do not sup-
port the hypothesis that long-term statin use may reduce the
risk of breast cancer incidence. Overall, when compared to
non-users of statins’, we found no significant difference in
breast cancer risk among ever users, current users and even
in those taking hydrophobic statins’. However, an inverse
association was observed that is a 47 % reduction in risk of
breast cancer recurrence in statin users compared to non-
users. There was significant heterogeneity among studies in
the overall analysis except among the studies adjusted for
Study, Year RR (95% CI)
0.01
Combined estimate (n = 21)
Vinogradova et al., 2011
0.1 0.2 0.5 1
Relative risk (95% confidence interval)
2
0.99 (0.94, 1.04)
1.00 (0.93, 1.08)
0.98 (0.88, 1.10)
1.02 (0.97, 1.08)
1.04 (0.98, 1.11)
1.01 (0.96, 1.06)
1.00 (0.80, 1.20)
1.17 (0.95, 1.43)
0.99 (0.92, 1.06)
1.20 (0.80, 1.80)
0.99 (0.74, 1.33)
1.07 (0.88, 1.29)
Beck et al., 2003 1.09 (0.93, 1.28)
Cauley et al., 2003 0.28 (0.09, 0.86)
1.07 (0.65, 1.74)Graaf et al., 2004
1.30 (1.00, 1.90)Kaye and Jick, 2004
1.00 (0.70, 1.20)Boudreau et al., 2004
1.02 (0.76, 1.36)Friis et al., 2005
Eliassen et al., 2005 0.91 (0.76, 1.08)
0.49 (0.38, 0.62)Kochher et al., 2005
Cauley et al., 2006 0.91 (0.80, 1.05)
0.78 (0.47, 1.31)Dumasia et al., 2006
Boudreau et al., 2007
Setoguchi et al., 2007
Coogan et al., 2007
Friedman et al., 2008
Smeeth et al., 2008
Pocobelli et al., 2008
Haukka et al., 2010
Hippisley et al., 2010
Woditschka et al., 2010
Jacobs et al., 2011
Fig. 3 Combined estimate of
relative risk (RR) and 95 %
confidence intervals (CIs) of
breast cancer associated with
statin use based on 24 studies
{in figure, studies like
Lovastatin study groups [12]
(RR =1.15, 95 % CI =0.37,
3.55), Blais et al. [10]
(RR =0.67, 95 % CI =0.33,
1.38) and Eaton et al. [27]
(RR =1.30, 95 % CI =0.70,
2.50) were excluded due to their
large CIs and no effect on the
final combined estimated RR}
comprising 265,482 statin users
and 76,759 incident breast
cancer cases. Squares indicate
RR in each study. The square
size is proportional to the
weight of the corresponding
study in the meta-analysis; the
length of horizontal lines
represents the 95 % CI. The
unshaded diamond indicates the
combined RR and 95 % CI
(random-effects model)
266 Breast Cancer Res Treat (2012) 135:261–269
123
nC8 confounders, subgroup of cohort, published before
Bonovas et al. [18] analysis as well as in studies repre-
senting long-term statin use and recurrence of breast
cancer.
In our subgroup analyses, the results were not substan-
tially affected by study design, confounder adjustment and
studies in the time frame of Bonovas et al. [18] and
afterward. Cohort and case–control studies alone showed
no association between statin use and risk of breast cancer.
There is no deviation in the association by remaining
subgroup analyses of studies like thorough adjustment
(nC8) of confounders, adjusted for B7 confounders,
covered in Bonovas et al. and after Bonovas et al. The test
of interaction was not statistically significant in any of
these subgroup analyses but was significant among sub-
groups representing long-term statin use. Cumulative meta-
analysis showed a change in trend of reporting risk of
breast cancer from positive to negative in statin users
between 1993 and 2011.
Although we found no association between statin use
and breast cancer risk, it remains plausible that statins’
reduce breast cancer risk. The mechanistic data are rela-
tively strong and suggest that statins’ inhibit cancer cell
growth and lead to apoptotic cell death through their
inhibition of the mevalonate pathway, although other
mechanisms have also been suggested [44]. Many products
of the mevalonate pathway are necessary for critical cel-
lular functions such as membrane integrity, cell signaling,
protein synthesis, and cell cycle progression. Disruption of
these processes in neoplastic cells by statins’ may result in
control of tumor initiation, growth, and metastasis [45].
In a review of rodent carcinogenicity tests [46], it was
reported that lipid-lowering drugs, including statins’, ini-
tiate or promote cancer in rats and mice. In contrast, sev-
eral recent laboratory studies indicated that statins’ may
have chemopreventive potential against cancer at various
sites, including breast. However, the inhibitory effect of
statins’ on breast cancer cells has thus far been tested only
in vitro and may behave differently in vivo. Specifically,
statins’ are selectively localized to the liver, and less than
5 % of a given dose reaches the systemic circulation.
Thereby, the usefulness of statins’ as chemopreventive
agents for breast cancer is doubted given their selective
hepatic uptake and low systemic availability [47,48].
The strength of the present analysis lies in inclusion of
24 observational studies, reporting data of more than 2.4
million participants, including 76,759 breast cancer cases.
Our meta-analysis has several limitations. First, we did
not search for unpublished studies or for original data.
Second, the included studies were different in terms of
Table 4 Overall effect estimates for breast cancer and statin use according to study characteristics
No. of studies Pooled estimate Tests of heterogeneity P
interaction
Tests of publication
bias
RR 95 % CI Qvalue (d.f.)
b
Pvalue I
2
(%)
c
Begg’s P
f
Egger’s P
f
All studies 24 0.99 0.94–1.04 52.83 (23) \0.001 57 0.71 0.32
Study design 0.35
Cohort 13 1.01 0.98–1.04 13.29 (12) 0.35
d
10
d
0.77 0.45
Case–control 11 0.95 0.84–1.08 39.29 (10) \0.001 75 0.36 0.54
Adjusted for confounders 1.00
nC8 confounders 8 0.99 0.94–1.04 5.10 (7) 0.65
d
0
d
0.55 0.77
nB7 confounders 16 0.99 0.92–1.06 46.99 (15) \0.001 68 0.35 0.37
Bonovas et al. [18] analysis 0.20
Before 8 1.06 0.95–1.18 8.86 (7) 0.26
d
21
d
0.40 0.15
After 16 0.98 0.93–1.03 43.04 (15) \0.001 65 0.76 0.32
Results for long-term statin use 10 1.03 0.96–1.11 12.14 (9) 0.21
d
26
d
0.02
e
0.29 0.56
Cohort studies 6 1.07 0.99–1.17 4.16 (5) 0.53
d
0
d
0.72 0.93
Case–control studies 4 0.84 0.70–1.02 2.89 (3) 0.41
d
0
d
0.75 0.63
Results for recurrence of BC 2 0.53 0.37–0.76
a
1.43 (1) 0.23
d
0–
RR Relative risk, CI confidence interval, d.f. degree of freedom
a
Statistically significant for inverse association between statin use and breast cancer recurrence
b
As tested by Cochran’s Qtest
c
As tested by I
2
statistic
d
Statistically significant for homogeneity (pooled effect estimates from ‘fixed-effects model’’, remaining from ‘random-effects model’’)
e
Test of interaction was statistically significant
f
Statistically significant for no publication bias
Breast Cancer Res Treat (2012) 135:261–269 267
123
study design and definitions of drug exposure. Finally, our
analysis was restricted to the articles in the English lan-
guage, which may have somewhat biased the results.
In conclusion, the findings of this meta-analysis, using
observational studies do not support the hypothesis that
statins’ use reduced the risk of breast cancer. However, we
cannot rule out a reduction in risk of breast cancer recur-
rence in statin users. More RCTs and observational studies
were needed to confirm this association with underlying
biological mechanisms in the future.
Acknowledgments The authors thank Dr. Dimple Kondal, Senior
scientist (Biostatistician), Centre for excellence, Public Health
Foundation, India, for helping with the data analysis and Ms. Sahaja
Banda for checking the accuracy of English usage.
Conflict of interest No potential conflicts of interest relevant to this
article were reported. No funding was provided for the analysis.
References
1. World Health Organization (2011) CancerStats Cancer world-
wide. http://info.cancerresearchuk.org/cancerstats/world/. Acces-
sed 7 February 2011
2. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D (2011)
Global cancer statistics. CA Cancer J Clin 61:69–90
3. Lamb E (2009) Top 200 drugs of 2008. http://www.pharmacy
times.com/issue/pharmacy/2009/2009-05/RxFocusTop200Drugs-
0509. Accessed 7 February 2011
4. Mandal CC, Ghosh-Choudhury N, Yoneda T, Choudhury GG
(2011) Simvastatin prevents skeletal metastasis of breast cancer
by an antagonistic interplay between p53 and CD44. J Biol Chem
286:11314–11327
5. Ghosh-Choudhury N, Mandal CC, Ghosh Choudhury G (2010)
Simvastatin induces derepression of PTEN expression via
NFkappaB to inhibit breast cancer cell growth. Cell Signal 22:
749–758
6. Jacobs EJ, Newton CC, Thun MJ, Gapstur SM (2011) Long-term
use of cholesterol-lowering drugs and cancer incidence in a large
United States cohort. Cancer Res 71:1763–1771
7. Vinogradova Y, Coupland C, Hippisley-Cox J (2011) Exposure to
statins’ and risk of common cancers: a series of nested case-
control studies. BMC Cancer 11:409
8. Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM,
Cobbe SM, Ford I, Gaw A, Hyland M, Jukema JW, Kamper AM,
Macfarlane PW, Meinders AE, Norrie J, Packard CJ, Perry IJ, Stott
DJ, Sweeney BJ, Twomey C, Westendorp RG (2002) Pravastatin
in elderly individuals at risk of vascular disease (PROSPER): a
randomised controlled trial. Lancet 360:1623–1630
9. Strandberg TE, Pyorala K, Cook TJ, Wilhelmsen L, Faergeman
O, Thorgeirsson G, Pedersen TR, Kjekshus J (2004) Mortality
and incidence of cancer during 10-year follow-up of the Scan-
dinavian Simvastatin Survival Study (4S). Lancet 364:771–777
10. Blais L, Desgagne
´A, LeLorier J (2000) 3-Hydroxy-3-methyl-
glutaryl coenzyme A reductase inhibitors and the risk of cancer: a
nested case-control study. Arch Intern Med 160:2363–2368
11. Cauley JA, Zmuda JM, Lui LY, Hillier TA, Ness RB, Stone KL,
Cummings SR, Bauer DC (2003) Lipid-lowering drug use and
breast cancer in older women: a prospective study. J Womens
Health (Larchmt) 12:749–756
12. Lovastatin Study Groups I through IV (1993) Lovastatin 5-year
safety and efficacy study. Arch Intern Med 153:1079–1087
13. Beck P, Wysowski DK, Downey W, Butler-Jones D (2003) Statin
use and the risk of breast cancer. J Clin Epidemiol 56:280–285
14. Graaf MR, Beiderbeck AB, Egberts AC, Richel DJ, Guchelaar HJ
(2004) The risk of cancer in users of statins’. J Clin Oncol
22:2388–2394
15. Kaye JA, Jick H (2004) Statin use and cancer risk in the general
practice research database. Br J Cancer 90:635–637
16. Friis S, Poulsen AH, Johnsen SP, McLaughlin JK, Fryzek JP,
Dalton SO, Sorensen HT, Olsen JH (2005) Cancer risk among
statin users: a population-based cohort study. Int J Cancer
114:643–647
17. Boudreau DM, Gardner JS, Malone KE, Heckbert SR, Blough
DK, Daling JR (2004) The association between 3-hydroxy-3-
methylglutaryl conenzyme A inhibitor use and breast carcinoma
risk among postmenopausal women: a case-control study. Cancer
100:2308–2316
18. Bonovas S, Filioussi K, Tsavaris N, Sitaras NM (2005) Use of
statins’ and breast cancer: a meta-analysis of seven randomized
clinical trials and nine observational studies. J Clin Oncol
23:8606–8612
19. Eliassen AH, Colditz GA, Rosner B, Willett WC, Hankinson SE
(2005) Serum lipids, lipid-lowering drugs, and the risk of breast
cancer. Arch Intern Med 165:2264–2271
20. Kochhar R, Khurana V, Bejjanki H, Caldito G, Fort C (2005)
Statins’ reduce breast cancer risk: a case control study in US
female veterans. J Clin Oncol 23(Suppl 1):16S, Abstract 514
21. Cauley JA, McTiernan A, Rodabough RJ, LaCroix A, Bauer DC,
Margolis KL, Paskett ED, Vitolins MZ, Furberg CD, Chlebowski
RT (2006) Statin use and breast cancer: prospective results from
the Women’s Health Initiative. J Natl Cancer Inst 98:700–707
22. Dumasia L, Lobocki C, Couturier A, Lebeis C, Derlichman A
(2006) Use of statins’ and breast carcinoma: a case–control study.
J Clin Oncol 24(Suppl 20):18S, Abstract 592
23. Jacobs EJ, Newton CC, Thun MJ, Gapstur SM (2011) Long-term
use of cholesterol-lowering drugs and cancer incidence in a large
United States cohort. Cancer Res 71:1763–1771
24. Boudreau DM, Yu O, Miglioretti DL, Buist DS, Heckbert SR,
Daling JR (2007) Statin use and breast cancer risk in a large
population-based setting. Cancer Epidemiol Biomarkers Prev
16:416–421
25. Coogan PF, Rosenberg L, Strom BL (2007) Statin use and the
risk of 10 cancers. Epidemiology 18:213–219
26. Smeeth L, Douglas I, Hall AJ, Hubbard R, Evans S (2008) Effect
of statins’ on a wide range of health outcomes: a cohort study
validated by comparison with randomized trials. Br J Clin
Pharmacol 67:99–109
27. Eaton M, Eklof J, Beal JR, Sahmoun AE (2009) Statins’ and
breast cancer in postmenopausal women without hormone ther-
apy. Anticancer Res 29:5143–5148
28. Hippisley-Cox J, Coupland C (2010) Unintended effects of stat-
ins’ in men and women in England and Wales: population based
cohort study using the QResearch database. BMJ 340:c2197
29. Setoguchi S, Glynn RJ, Avorn J, Mogun H, Schneeweiss S (2007)
Statins’ and the risk of lung, breast, and colorectal cancer in the
elderly. Circulation 115:27–33
30. Friedman GD, Flick ED, Udaltsova N, Chan J, Quesenberry CP,
Habel LA (2008) Screening statins’ for possible carcinogenic
risk: up to 9 years of follow-up of 361,859 recipients. Pharma-
coepidemiol Drug Saf 17:27–36
31. Pocobelli G, Newcomb PA, Trentham-Dietz A, Titus-Ernstoff L,
Hampton JM, Egan KM (2008) Statin use and risk of breast
cancer. Cancer 112:27–33
32. Haukka J, Sankila R, Klaukka T, Lonnqvist J, Niskanen L,
Tanskanen A, Wahlbeck K, Tiihonen J (2010) Incidence of
268 Breast Cancer Res Treat (2012) 135:261–269
123
cancer and statin usage record linkage study. Int J Cancer 126:
279–284
33. Woditschka S, Habel LA, Udaltsova N, Friedman GD, Sieh W
(2010) Lipophilic statin use and risk of breast cancer subtypes.
Cancer Epidemiol Biomarkers Prev 19:2479–2487
34. Vinogradova Y, Coupland C, Hippisley-Cox J (2011) Exposure to
statins’ and risk of common cancers: a series of nested case-
control studies. BMC Cancer 11:409
35. Coogan PF, Rosenberg L, Palmer JR, Strom BL, Zauber AG,
Shapiro S (2002) Statin use and the risk of breast and prostate
cancer. Epidemiology 13:262–267
36. Kaye JA, Meier CR, Walker AM, Jick H (2002) Statin use,
hyperlipidaemia, and the risk of breast cancer. Br J Cancer 86:
1436–1439
37. Begg CB, Mazumdar M (1994) Operating characteristics of a
rank correlation test for publication bias. Biometrics 50:
1088–1101
38. Egger M, Davey SG, Schneider M, Minder C (1997) Bias in
meta-analysis detected by a simple, graphical test. BMJ 315:
629–634
39. Higgins JP, Thompson SG, Deeks JJ, Altman DG (2003) Mea-
suring inconsistency in meta-analyses. BMJ 327:557–560
40. Altman DG, Bland JM (2003) Interaction revisited: the difference
between two estimates. BMJ 326:219
41. Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD,
Rennie D, Moher D, Becker BJ, Sipe TA, Thacker SB (2000)
Meta-analysis of observational studies in epidemiology: a pro-
posal for reporting. meta-analysis of observational studies in
epidemiology (MOOSE) group. JAMA 283:2008–2012
42. Kwan ML, Habel LA, Flick ED, Quesenberry CP, Caan B (2008)
Post-diagnosis statin use and breast cancer recurrence in a pro-
spective cohort study of early stage breast cancer survivors.
Breast Cancer Res Treat 109:573–579
43. Chae YK, Valsecchi ME, Kim J, Bianchi AL, Khemasuwan D,
Desai A, Tester W (2011) Reduced risk of breast cancer recur-
rence in patients using ACE inhibitors, ARBs, and/or statins’.
Cancer Invest 29:585–593
44. Wong WW, Dimitroulakos J, Minden MD, Penn LZ (2002)
HMG-CoA reductase inhibitors and the malignant cell: the statin
family of drugs as triggers of tumor-specific apoptosis. Leukemia
16:508–519
45. Chan KK, Oza AM, Siu LL (2003) The statins’ as anticancer
agents. Clin Cancer Res 9:10–19
46. Newman TB, Hulley SB (1996) Carcinogenicity of lipid-lowering
drugs. JAMA 275:55–60
47. Rogers MJ (2000) Statins’: lower lipids and better bones? Nat
Med 6:21–23
48. Hamelin BA, Turgeon J (1998) Hydrophilicity/lipophilicity: rel-
evance for the pharmacology and clinical effects of HMG-CoA
reductase inhibitors. Trends Pharmacol Sci 19:26–37
Breast Cancer Res Treat (2012) 135:261–269 269
123
... Bonovas, 2008 12 studies No significant relationship between statins and pancreatic cancer risk [129] Khurana, 2007 483,733 Protective against the development of pancreatic cancer [130] Lin, 2016 19,727 Prevent H. pylori-associated gastric cancer [105] Singh, 2013 11 studies Prevent gastric cancer risk in both Asian and Western population [131] Tsan, 2012 33,413 Reduce the risk for hepatocellular carcinoma in HBV-infected patients [132] Chen, 2015 2,053 Decrease hepatocellular carcinoma in diabetic patients [133] Zhang, 2013 13 studies No association between statin use and risk of bladder cancer [134] Peng, 2015 3,174 Reduce the risk of cholangiocarcinoma [108] Yi, 2014 20 studies Preventive effects against hematological malignancies [135] Pradelli, 2015 14 studies Negatively associated with all hematological malignancies [136] Wang, 2013 20 studies Nonsignificant association between statin users and lung cancer risk [137] Bansal, 2012 27 studies Reduce the risk of total and advanced prostate cancer [138] Jacobs, 2007 55,454 Reduce the risk of advanced prostate cancer [109] Undela, 2012 24 studies Do not support that statins have a protective effect against breast cancer [139] Lytras, 2014 40 studies Do not support that statin users reduce the risk of colorectal cancer [140] Setoguchi, 2007 24,439 No effect in the risk of colorectal, lung, or breast cancer in older patients [141] Kuoppala, 2008 42 studies No effect on the incidence of lung, breast, or prostate cancer Protect from stomach and liver cancer and from lymphoma Increase the incidence of both melanoma and nonmelanoma skin cancer [142] ...
... Although cholesterol-lowering drugs have also been shown to possess an important antitumor activity that reduces cell growth, proliferation , and migration through ERK-mediated and Akt-mediated signaling pathways and is capable of inducing apoptosis through extrinsic and intrinsic pathways using different cancer cells as models [43?45, 75, 78, 104, 118?123], it is still unclear whether statins are suitable to prevent the incidence of cancer. More than a hundred of epidemiological studies around the world have been performed to evaluate the effect of statin on the risk of cancer incidence [105, 108, 109,[126][127][128][129][130][131][132][133][134][135][136][137][138][139][140][141][142]. These studies have been focused on statin type, potency, lipophilic or hydrophobicity status, and duration of use. ...
Chapter
Full-text available
The accumulation of cholesterol in cancer cells and tumor tissues promotes cell growth, proliferation, and migration as well as tumor progression. Cholesterol synthesis is catalyzed by a series of enzymatic reactions. Regulation of these key enzymes can control cholesterol synthesis and modulate cellular cholesterol levels in the cells. Meanwhile, controlling cholesterol transportation, absorption, and depletion could also significantly reduce cellular cholesterol levels. The current evidence supports that cholesterol lowering agents, beyond the expected cholesterol-lowering properties, also display an important anticancer activity in reducing cancer cell growth, proliferation and migration, and inducing apoptosis in a variety of cancer cells. Understanding the mechanisms of cholesterol metabolism and cholesterol lowering could potentially benefit cancer patients in cancer prevention and treatment.
... The use of statins as chemopreventive agent is controversially discussed in the scientific literature: Some meta-analyses report the risk reduction potential of statins for different entities [402][403][404][405] whereas others observed no such effects [406][407][408][409]. Numerous further studies provide evidence pro chemopreventive effects of statins: Poynter and colleagues found a 47% reduced risk for colorectal cancer with statin use while Voorneveld et al. ...
Thesis
Full-text available
Das metastasierende kolorektale Karzinom stellt eine große Herausforderung in der Krebstherapie dar. Verlässliche und effiziente Biomarker zur Prognose des Krankheitsverlaufes oder der Therapieantwort (Prädiktion) sind rar. Metastasis-associated in colon cancer 1 (MACC1) ist ein prognostischer, prädiktiver und kausaler Biomarker für verschiedene Tumorentitäten. Durch die Induzierung von Zielgenen, wie z.B. MET, beeinflusst es Signalwege wie MEK/ERK und AKT/β-catenin und fördert so Zellproliferation und -motilität sowie Tumorprogression und Metastasierung in vivo. Diese Arbeit sollte neue Strategien erforschen diese Prozesse durch die Inhibition von MACC1 auf Transkriptions- und Signaltransduktionsebene zu unterbinden. Mit zwei verschiedenen Screeningmethoden konnten wir Statine als potente transkriptionelle Inhibitoren von MACC1 als auch phosphotyrosin (pY)-abhängige Interaktionen von MACC1 mit essentiellen Signalmolekülen identifizieren: SHP2, GRB2, SHC1, PLCG1 und STAT5B. Statine verringerten MACC1-spezifische Proliferation und Koloniebildung in vitro als auch Tumor Wachstum und Metastasierung in vivo bei Dosen äquivalent der humanen Standardtherapie zur Blutlipidsenkung. Mutation der pY-Bindungsstellen reduzierte die Aktivität des MACC1-induzierten ERK Signalwegs sowie Zellmigration und -proliferation. Anhand unserer Daten orchestriert MACC1, abhängig von MET und EGFR, neue SHP2/SRC/ERK und PKA/SRC/CREB Signalkaskaden zu einem malignen Phänotyp. Gezielte Intervention restringierte die MACC1-abhängige Koloniebildung, was neue therapeutische Interventionspunkte identifiziert und eine hervorragende Basis für Untersuchungen zur Kombinationstherapie darstellt. Die weitere Erforschung der spatiotemporalen Organisation des MACC1 Signalosoms und assoziierter Signalkaskaden soll das volle therapeutische Potential von MACC1 ausschöpfen. Wir empfehlen zudem Statine in der Krebstherapie bzw. -prävention, besonders bei MACC1-stratifzierten Patienten, anzuwenden.
... Some data support a beneficial role for their uses in breast cancer management, other studies are less promising and argue against their prescription in cancer treatment. [17][18][19] Moreover, all these studies were carried out using statins alone, its effectiveness in combination with TAM as neoadjuvant therapy in ER+ breast cancer has not yet been explored. Therefore, it is worthwhile examining whether SIM can potentiate the tumor response of TAM, the conventional breast cancer therapy or not. ...
Article
Full-text available
Purpose Tamoxifen (TAM) is a non-steroidal antiestrogen drug, used in the prevention and treatment of all stages of hormone-responsive breast cancer. Simvastatin (SIM) is a lipid-lowering agent and has been shown to inhibit cancer cell growth. The study aimed to investigate the effect of the combination of TAM and SIM in the treatment of estrogen receptor positive (ER+) breast cancer cell line, MCF-7, and in mice-bearing Ehrlich solid tumors. Methods MCF-7 cells were treated with different concentrations of TAM or/and SIM for 72 hours and the effects of the combination treatment on cytotoxicity, oxidative stress markers, apoptosis, angiogenesis, and metastasis were investigated using different techniques. In addition, tumor volume, oxidative markers, and inflammatory markers of the combined therapy were explored in mice bearing solid EAC tumors. Results The results showed that treatment of MCF-7 cells with the combination of 10 µM TAM, and 2 µM SIM significantly inhibited the increase in oxidative stress markers, LDH, and NF-kB induced by TAM. In addition, there was a significant decrease in the total apoptotic ratio, caspase-3 activity, and glucose uptake, while there was a non-significant change in Bax/bcl-2 ratio compared to the TAM-treated group. Using the isobologram equation, the drug interaction was antagonistic with combination index, CI=1.18. On the other hand, the combination regimen decreased VEGF, and matrix metalloproteinases, MMP 2&9 compared to TAM-treated cells. Additionally, in vivo, the combination regimen resulted in a non-significant decrease in the tumor volume, decreased oxidative markers, and the protein expression of TNF-α, and NF-κB compared to the TAM treated group. Conclusion Although the combination regimen of TAM and SIM showed an antagonistic drug interaction in MCF-7 breast cancer, it displayed favorable antiangiogenic, anti-metastatic, and anti-inflammatory effects.
... An inconsistency was indicated between relationship of statin use and incidence of breast cancer. Few studies indicated a decrease in risk of breast cancer, among statin users [38,[41][42][43][44] whereas other studies suggested its long-term use did not significantly affect risk of breast cancer [45,46] . For the investigation of the biological effects of statins in the prevention and modulation of breast cancer biomarkers several trials have been initiated [47,48] . ...
... [48][49][50] Some epidemiological studies previously indicated lower cancer incidence among statin users, 51 52 although the results have been inconsistent across studies. [53][54][55][56] Our recent update from the large, prospective Nurse's Health Study (NHS) showed no association between different statin exposures and risk of invasive breast cancer. 57 Results from a prominent meta-analysis were null. ...
Article
Full-text available
Statin drugs have been used for more than two decades to treat hypercholesterolemia and as cardio‐preventive drugs, resulting in a marked decrease in cardiovascular morbidity and mortality worldwide. Statins halt hepatic cholesterol biosynthesis by inhibiting the rate‐limiting enzyme in the mevalonate pathway, hydroxymethylglutaryl‐coenzyme A reductase (HMGCR). The mevalonate pathway regulates a host of biochemical processes in addition to cholesterol production. Attenuation of these pathways is likely responsible for the myriad benefits of statin therapy beyond cholesterol reduction—the so‐called pleiotropic effects of statins. Chief among these purported effects is anti‐cancer activity. A considerable body of preclinical, epidemiologic, and clinical evidence shows that statins impair proliferation of breast cancer cells and reduce the risk of breast cancer recurrence. Potential mechanisms for this effect have been explored in laboratory models, but remain poorly understood and require further investigation. The number of clinical trials assessing the putative clinical benefit of statins in breast cancer is increasing. Currently, a total of 30 breast cancer/statin trials are listed at the global trial identifier website clinicaltrials. gov.Given the compelling evidence from performed trials in a variety of clinical settings, there have been calls for a clinical trial of statins in the adjuvant breast cancer setting. It would be imperative for such a trial to incorporate tumor biomarkers predictive of statin response in its design and analysis plan. Ongoing translational clinical trials aimed at biomarker discovery will help identify, which breast cancer patients are most likely to benefit from adjuvant statin therapy, and will add valuable clinical knowledge to the field. This article is protected by copyright. All rights reserved.
... Consensus regarding the clinical effects of statins on breast cancer has not been reached, which has resulted in inconsistency in the relationship between statin use and the incidence of breast cancer. Many studies have demonstrated a decrease in the risk of a variety of cancers, including breast cancer, among statin users [70][71][72][73][74]. Conversely, several studies revealed that long-term use of statins did not significantly affect the risk of breast cancer [75][76][77] . However, through a systematic review and meta- analysis, Wu et al. found that although statin use may not influence the risk of breast cancer, it is associated with a decrease in mortality of breast cancer patients [78]. ...
Article
Full-text available
Triple-negative breast cancer (TNBC), which accounts for 15-20% of all breast cancers, does not express estrogen receptor (ER) or progesterone receptor (PR) and lacks human epidermal growth factor receptor 2 (HER2) overexpression or amplification. These tumors have a more aggressive phenotype and a poorer prognosis due to the high propensity for metastatic progression and absence of specific targeted treatments. Patients with TNBC do not benefit from hormonal or trastuzumab-based targeted therapies because of the loss of target receptors. Although these patients respond to chemotherapeutic agents such as taxanes and anthracyclines better than other subtypes of breast cancer, prognosis remains poor. A group of targeted therapies under investigation showed favorable results in TNBC, especially in cancers with BRCA mutation. The lipid-lowering statins (3-hydroxy-3-methyl-glutaryl coenzyme A reductase inhibitors), including lovastatin and simvastatin, have been shown to preferentially target TNBC compared with non-TNBC. These statins hold great promise for the management of TNBC. Only with the understanding of the molecular basis for the preference of statins for TNBC and more investigations in clinical trials can they be reformulated into a clinically approved drug against TNBC.
Article
Full-text available
Statins are reported to reduce the risk of cancer, but the results of various published studies have been contradictory. We carried out an umbrella review to provide an overview and understand the strength of evidence, extent of potential biases, and validity of claimed associations between the use of statins and cancer incidence. We comprehensively re-analyzed the data of meta-analyses of randomized controlled trials (RCTs) and observational studies on associations between statin use and cancer incidence. We also assessed the strength of evidence of the re-analyzed outcomes, which were determined from the criteria including statistical significance of the p-value of random-effects, as well as fixed-effects meta-analyses, small study effects, between-study heterogeneity, and a 95% prediction interval. Using a conventional method to assess the significance of meta-analysis (p-value < 0.05), statins had a statistically significant effect on reducing cancer incidence in 10 of 18 types of cancer. When we graded the level of evidence, no cancer type showed convincing evidence, and four cancers (esophageal cancer, hematological cancer, leukemia, and liver cancer) showed suggestive evidence of a preventive effect. There was weak evidence of an association with six cancers, and no significance for the remaining eight cancers. None of the meta-analyses of RCTs on the association of statin and cancer incidence showed a statistical significance. Although there was a preventive effect of statin on cancer incidence in 10 of the 18 cancer types, the evidence supporting the use of statins to reduce cancer incidence was low. Therefore, the associations between statin use and cancer incidence should be carefully considered by clinicians.
Article
In previous studies, we found modestly decreased and increased risks of second breast cancer events with the use of statins and antibiotics, respectively, after adjustment for surveillance mammography. We evaluated detection bias by comparing receipt of surveillance mammography among users of these 2 disparate classes of medication. Adult women diagnosed with early-stage breast cancer during 1990-2008 (n = 3,965) while enrolled in an integrated health-care plan (Group Health Cooperative; Washington State) were followed for up to 10 years in the Commonly Used Medications and Breast Cancer Outcomes (COMBO) Study. Categories of antibiotic use included infrequent (1-3 dispensings/12 months) and frequent (≥4 dispensings/12 months) use, and categories of statin use included less adherent (1 dispensing/6 months) and adherent (≥2 dispensings/6 months). We examined associations between medication use and surveillance mammography using multivariable generalized estimating equations and evaluated the impact of adjusting for surveillance within Cox proportional hazard models. Frequent antibiotic users were less likely to receive surveillance mammography (odds ratio (OR) = 0.90, 95% confidence interval (CI): 0.82, 0.99) than were nonusers; no association was found among infrequent users (OR = 0.96, 95% CI: 0.90, 1.03). Adherent statin use was associated with more surveillance compared with nonuse (OR = 1.11, 95% CI: 1.01, 1.25), but less adherent statin use was not (OR = 1.03, 95% CI: 0.81, 1.31). No difference in associations between medications of interest and second breast cancer events was observed when surveillance was removed from otherwise adjusted models. The influence of detection bias by medication use warrants further exploration.
Article
Full-text available
BACKGROUND: In the era of precision medicine, more attention is paid to the search for predictive markers of treatment efficacy and tolerability. Statins are one of the classes of drugs that could benefit from this approach because of their wide use and their incidence of adverse events. METHODS: Literature from PubMed databases and bibliography from retrieved publications have been analyzed according to terms such as statins, pharmacogenetics, epigenetics, toxicity and drug-drug interaction, among others. The search was performed until 1 October 2016 for articles published in English language. RESULTS: Several technical and methodological approaches have been adopted, including candidate gene and next generation sequencing (NGS) analyses, the latter being more robust and reliable. Among genes identified as possible predictive factors associated with statins toxicity, cytochrome P450 isoforms, transmembrane transporters and mitochondrial enzymes are the best characterized. Finally, the solute carrier organic anion transporter family member 1B1 (SLCO1B1) transporter seems to be the best target for future studies. Moreover, drug-drug interactions need to be considered for the best approach to personalized treatment. CONCLUSIONS: Pharmacogenetics of statins includes several possible genes and their polymorphisms, but muscular toxicities seem better related to SLCO1B1 variant alleles. Their analysis in the general population of patients taking statins could improve treatment adherence and efficacy; however, the cost-efficacy ratio should be carefully evaluated.
Article
Full-text available
Background Laboratory studies have demonstrated statin-induced apoptosis of cancer cells, including breast cancer cells, and evidence is accumulating on the mechanism of statin-induced apoptosis. However, despite numerous epidemiological studies, no consensus has been reached regarding the relationship between statin use and breast cancer risk. Methods This retrospective case–control study enrolled 4332 breast cancer patients and 21,660 age-matched controls registered in the National Health Insurance program of Taiwan, which covers approximately 99% of the population. The study cases were women for whom a diagnosis of breast cancer (ICD-9-CM code 174.X) had been recorded in LHID2005 between January 1, 2004 and December 31, 2010. A logistic regression model was adjusted for potential confounding factors, including the level of urbanization, and the Charlson Comorbidity Index was applied to assess potential comorbidities. We also considered possible bias caused by random urbanization, because nutrition and lifestyle factors are related to breast cancer incidence. ResultsOur results showed that lovastatin was associated with a lower risk of breast cancer (adjusted OR 0.596; 95% CI 0.497–0.714; p < 0.001), and atorvastatin exhibited a protective tendency against breast cancer (adjusted OR 0.887; 95% CI 0.776–1.013; p < 0.077). Conclusions Although no consensus has been established regarding the relationship between statin use and breast cancer risk, our study indicated that lovastatin is a potential chemopreventive agent against breast cancer. Further detailed research is warranted.
Article
Full-text available
Because of the pressure for timely, informed decisions in public health and clinical practice and the explosion of information in the scientific literature, research results must be synthesized. Meta-analyses are increasingly used to address this problem, and they often evaluate observational studies. A workshop was held in Atlanta, Ga, in April 1997, to examine the reporting of meta-analyses of observational studies and to make recommendations to aid authors, reviewers, editors, and readers. Twenty-seven participants were selected by a steering committee, based on expertise in clinical practice, trials, statistics, epidemiology, social sciences, and biomedical editing. Deliberations of the workshop were open to other interested scientists. Funding for this activity was provided by the Centers for Disease Control and Prevention. We conducted a systematic review of the published literature on the conduct and reporting of meta-analyses in observational studies using MEDLINE, Educational Research Information Center (ERIC), PsycLIT, and the Current Index to Statistics. We also examined reference lists of the 32 studies retrieved and contacted experts in the field. Participants were assigned to small-group discussions on the subjects of bias, searching and abstracting, heterogeneity, study categorization, and statistical methods. From the material presented at the workshop, the authors developed a checklist summarizing recommendations for reporting meta-analyses of observational studies. The checklist and supporting evidence were circulated to all conference attendees and additional experts. All suggestions for revisions were addressed. The proposed checklist contains specifications for reporting of meta-analyses of observational studies in epidemiology, including background, search strategy, methods, results, discussion, and conclusion. Use of the checklist should improve the usefulness of meta-analyses for authors, reviewers, editors, readers, and decision makers. An evaluation plan is suggested and research areas are explored.
Article
Objective: To review the findings and implications of studies of rodent carcinogenicity of lipid-lowering drugs. Data sources: Summaries of carcinogenicity studies published in the 1992 and 1994 Physicians' Desk Reference (PDR), additional information obtained from the US Food and Drug Administration, and published articles identified by computer searching, bibliographies, and consultation with experts. Study sample: We tabulated rodent carcinogenicity data from the 1994 PDR for all drugs listed as "hypolipidemics." For comparison, we selected a stratified random sample of antihypertensive drugs. We also reviewed methods and interpretation of carcinogenicity studies in rodents and results of clinical trials in humans. Data synthesis: All members of the two most popular classes of lipid-lowering drugs (the fibrates and the statins) cause cancer in rodents, in some cases at levels of animal exposure close to those prescribed to humans. In contrast, few of the antihypertensive drugs have been found to be carcinogenic in rodents. Evidence of carcinogenicity of lipid-lowering drugs from clinical trials in humans is inconclusive because of inconsistent results and insufficient duration of follow-up. Conclusions: Extrapolation of this evidence of carcinogenesis from rodents to humans is an uncertain process. Longer-term clinical trials and careful postmarketing surveillance during the next several decades are needed to determine whether cholesterol-lowering drugs cause cancer in humans. In the meantime, the results of experiments in animals and humans suggest that lipid-lowering drug treatment, especially with the fibrates and statins, should be avoided except in patients at high short-term risk of coronary heart disease.
Article
Summary Background Although statins reduce coronary and cerebrovascular morbidity and mortality in middle-aged individuals, their efficacy and safety in elderly people is not fully established. Our aim was to test the benefits of pravastatin treatment in an elderly cohort of men and women with, or at high risk of developing, cardiovascular disease and stroke. Methods We did a randomised controlled trial in which we assigned 5804 men (n=2804) and women (n=3000) aged 70-82 years with a history of, or risk factors for, vascular disease to pravastatin (40 mg per day; n=2891) or placebo (n=2913). Baseline cholesterol concentrations ranged from 4·0 mmol/L to 9·0 mmol/L. Follow-up was 3·2 years on average and our primary endpoint was a composite of coronary death, non-fatal myocardial infarction, and fatal or non-fatal stroke. Analysis was by intention-to-treat. Findings Pravastatin lowered LDL cholesterol concentrations by 34% and reduced the incidence of the primary endpoint to 408 events compared with 473 on placebo (hazard ratio 0·85, 95% CI 0·74-0·97, p=0·014). Coronary heart disease death and non-fatal myocardial infarction risk was also reduced (0·81, 0·69-0·94, p=0·006). Stroke risk was unaffected (1·03, 0·81-1·31, p=0·8), but the hazard ratio for transient ischaemic attack was 0·75 (0·55-1·00, p=0·051). New cancer diagnoses were more frequent on pravastatin than on placebo (1·25, 1·04-1·51, p=0·020).