ArticlePDF AvailableLiterature Review

Obesity and breast cancer: Status of leptin and adiponectin in pathological processes

Authors:
  • University of Minnesota, Hormel Institute

Abstract and Figures

It is well recognized that obesity increases the risk of various cancers, including breast malignancies in postmenopausal women. Furthermore, obesity may adversely affect tumor progression, metastasis, and overall prognosis in both pre- and postmenopausal women with breast cancer. However, the precise mechanism(s) through which obesity acts is/are still elusive and this relationship has been the subject of much investigation and speculation. Recently, adipose tissue and its associated cytokine-like proteins, adipokines, particularly leptin and adiponectin, have been investigated as mediators for the association of obesity with breast cancer. Higher circulating levels of leptin found in obese subjects could be a growth-enhancing factor as supported by in vitro and preclinical studies, whereas low adiponectin levels in obese women may be permissive for leptin's growth-promoting effects. These speculations are supported by in vitro studies which indicate that leptin promotes human breast cancer cell proliferation while adiponectin exhibits anti-proliferative actions. Further, estrogen and its receptors have a definite impact on the response of human breast cancer cell lines to leptin and adiponectin. More in-depth studies are needed to provide additional and precise links between the in vivo development of breast cancer and the balance of adiponectin and leptin.
Content may be subject to copyright.
NON-THEMATIC REVIEW
Obesity and breast cancer: status of leptin and adiponectin
in pathological processes
Michael E. Grossmann &Amitabha Ray &
Katai J. Nkhata &Dmitry A. Malakhov &
Olga P. Rogozina &Soner Dogan &Margot P. Cleary
Published online: 7 September 2010
#Springer Science+Business Media, LLC 2010
Abstract It is well recognized that obesity increases the
risk of various cancers, including breast malignancies in
postmenopausal women. Furthermore, obesity may adversely
affect tumor progression, metastasis, and overall prognosis in
both pre- and postmenopausal women with breast cancer.
However, the precise mechanism(s) through which obesity
acts is/are still elusive and this relationship has been the
subject of much investigation and speculation. Recently,
adipose tissue and its associated cytokine-like proteins,
adipokines, particularly leptin and adiponectin, have been
investigated as mediators for the association of obesity with
breast cancer. Higher circulating levels of leptin found in
obese subjects could be a growth-enhancing factor as
supported by in vitro and preclinical studies, whereas low
adiponectin levels in obese women may be permissive for
leptins growth-promoting effects. These speculations are
supported by in vitro studies which indicate that leptin
promotes human breast cancer cell proliferation while
adiponectin exhibits anti-proliferative actions. Further, estro-
gen and its receptors have a definite impact on the response
of human breast cancer cell lines to leptin and adiponectin.
More in-depth studies are needed to provide additional and
precise links between the in vivo development of breast
cancer and the balance of adiponectin and leptin.
Keywords Breast tumorigenesis .Cancer progression .
Adiposity .Leptin .Adiponectin .Caloric restriction
1 Introduction
A number of studies indicate that obesity, as reflected by
increased body mass index (BMI), is associated with
increased risk of more aggressive breast cancer as well as
reduced survival [14]. Obesity plays a complex role in
breast cancer and is associated with increased inflamma-
tion, angiogenesis, and alterations in serum levels of
potential growth regulators such as adiponectin, leptin,
and estrogen [5,6]. In women diagnosed with breast
cancer, the balance of adiponectin and leptin may be a key
factor both in disease development and progression.
Reduced levels of serum adiponectin have been reported
in breast cancer patients compared to healthy controls,
particularly in postmenopausal women [79]. On the other
hand, the role of serum leptin levels in breast cancer
appears to be more complex. Some studies have shown that
serum leptin levels are increased in women with breast
cancer but others have found leptin to be decreased or
unchanged [1014]. This may be due to confounding
factors such as menopausal status and disease stage. There
are several reports indicating that the presence of the leptin
receptor (Ob-R) in breast tumors and that high serum leptin
levels were associated with poor prognosis [15,16]possibly
in conjunction with hypoxia, and/or exposure to elevated
levels of insulin, insulin-like growth factor-I (IGF-I), and
estradiol. It may be that the levels of the receptors for
adiponectin and leptin as well as the balance of adiponectin
and leptin in the serum are critical factors in breast cancer
tumorigenesis.
Adiponectin and leptin are both secreted by adipose
tissue and appear to oppose each others actions. Adipo-
nectin, also known as adipocyte complement-related protein
of 30 kDa (Acrp30) [17], is measured in human serum in
the range of 220 μg/ml and is negatively correlated with
Michael E. Grossmann and Amitabha Ray contributed equally to this
review.
M. E. Grossmann :A. Ray :K. J. Nkhata :D. A. Malakhov :
O. P. Rogozina :S. Dogan :M. P. Cleary (*)
The Hormel Institute, University of Minnesota,
801-16th Avenue NE,
Austin, MN 55912, USA
e-mail: mpcleary@hi.umn.edu
Cancer Metastasis Rev (2010) 29:641653
DOI 10.1007/s10555-010-9252-1
body weight, BMI, body fat, and serum leptin in women [18].
On the other hand, leptin is positively correlated with body
weight, BMI, and total body fat. Increased serum levels of
adiponectin are associated with a number of positive health
factors such as decreased levels of triglycerides, increased
insulin sensitivity, increased high-density lipoprotein choles-
terol levels, and general anti-inflammatory and anti-vascular
effects [1921]. Conversely, increased serum leptin levels are
associated with negative health conditions such as type 2
diabetes, atherosclerosis, asthma, and other obesity-related
diseases [22]. Thus, adiponectin and leptin have global,
opposing effects on a number of different aspects of
pathophysiological conditions.
There are two different adiponectin receptors designated
as AdipoR1 and AdipoR2 [23]. Full-length adiponectin
binds with highest affinity to AdipoR2 [24] and cleaved
adiponectin, known as the globular form (gAcrp30) [25],
binds with highest affinity to AdipoR1 [23]. In vitro assays
have shown that a number of different breast cancer cell
lines express one or both of the adiponectin receptors and
undergo reduced growth and/or increased apoptosis in
response to the addition of adiponectin [2629]. Adipo-
nectin signaling through its receptors can block phosphor-
ylation of Akt and reduce the expression of cyclin D1. It
was also found that adiponectin can interact with growth
factors such as platelet-derived growth factor (type BB),
basic fibroblast growth factor, and heparin-binding epider-
mal growth factor-like growth factor and that in vitro, the
normal response to growth factors of breast cancer cells
could be blocked by adiponectin [30]. Adiponectin
inhibited angiogenesis as assessed by in vitro and in vivo
assays of non-breast cancer cell lines [31] further illustrat-
ing the broad range of anti-tumor growth effects of
adiponectin. In these studies, endothelial cell proliferation
and migration assays were performed for the in vitro
assessment of adiponectin action. The in vivo studies
included measurement of avascular zone formation using
a chick chorioallantoic membrane assay, immunohisto-
chemical evaluation in a mouse corneal angiogenesis assay
and evaluation of tissue sections from xenograft T241
fibrosarcomas which were treated daily with intratumoral
injections of adiponectin (50 μg) for 2 weeks. Thus, the
effects of adiponectin are mediated by receptors found in a
number of cell types including breast cancer.
Leptin was originally identified in 1994 as the product of
the obese gene [32]. Leptin is generally present in human
serum in the range of 550 ng/ml [22]. However, in obese
individuals, levels in excess of 100 ng/ml are common [33
35]. Leptin acts through receptors in the hypothalamus to
produce global effects on energy balance and body weight
[36]. In contrast to adiponectin, leptin has been implicated
as a growth-promoting factor for cancer including in vitro
proliferation of several human breast cancer cell lines [37
40]. Preclinical studies also support a role for leptin in
mammary tumor development as evidenced by the fact that
mice deficient in leptin, Lep
ob
Lep
ob
[41], or with nonfunc-
tioning leptin receptors, Lepr
db
Lepr
db
[42], did not develop
transgene-induced mammary tumors. The ability of leptin
to enhance breast cancer development may be related to
several different facets of tumor growth. For example,
leptin can initiate signaling pathways that are activated
during stimulation of estrogen receptor-α(ERα)by
estradiol. Leptin can also induce aromatase activity in
MCF-7 breast cancer cells enhancing in situ estradiol
production and promoting estrogen-dependent breast cancer
proliferation [43]. Furthermore, leptin is able to interfere
with the anti-cancer effects of the anti-estrogen ICI 182,780
[43], all of which illustrates the potential leptin has to
enhance breast cancer growth.
Disease characteristics that are associated with obesity
and/or breast cancer are also associated with the adiponec-
tin to leptin ratio. For example, the adiponectin to leptin
ratio is higher in individuals with a normal BMI as
compared to overweight or obese individuals [18,44].
Studies also suggest that the adiponectin to leptin ratio is a
better indicator of insulin resistance than the levels of
adiponectin or leptin alone or the homeostasis model
assessment ratio estimated from fasting plasma glucose
and insulin levels [45]. Furthermore, the estimation of the
ratio between these two adipokines can be used as a
valuable index in the evaluation of other obesity-related
complications such as metabolic syndrome [46]and
endothelial dysfunction [47]. In a recent review article,
the adiponectin to leptin ratio has been discussed in detail
in relation to the pathogenesis of breast cancer [48].
Interestingly, a study found that the ratio of adiponectin to
leptin may indicate the presence of aggressive breast cancer
independent of BMI [49]. Thus, the balance of adiponectin
to leptin may be indicative of, or influence, a number of
different disease processes. Here, we present an overview
of pertinent studies related to both adipokines, leptin and
adiponectin, conducted in recent years, including our own,
that may help shed light on the important issues related to
the link between obesity and postmenopausal breast cancer.
2 Clinical studies on obesity and disease progression
A number of studies have revealed an influence of obesity on
the clinical stages and histopathological grades of breast
carcinoma. These two important classification systems are
closely associated with phenomena like metastasis of cancer
cells, survival, and overall prognosis. In a recent study, Stark
et al. observed that obesity at the time of diagnosis of breast
cancer was associated with an 80% increased risk of more
advanced stages (III/IV) and poorly differentiated grade
642 Cancer Metastasis Rev (2010) 29:641653
compared with normal-weight women [50]. In a prospective
cohort study on invasive breast cancer consisting of 34%
obese patients, advanced grade and stage including lymph
node metastases were found more commonly among the
obese women [3]. Similarly, in a population-based follow-up
study on women younger than 45 years of age, the tumors of
the women in the highest quartile of BMI were more likely
to be high histologic grade, greater size and ER()[51].
Carmichael et al. noted that breast cancer in obese women
was associated with worse Nottingham prognostic index
(i.e., tumor grade, size, angiogenesis, and positive lymph
nodes) [52]. However, they did not observe any statistically
significant differences in overall and disease-free survival
between obese and non-obese group.
Several investigators documented a positive association
between obesity and advanced stage of breast cancer at
diagnosis [5357]. On the contrary, Chagpar et al. reported
that increased BMI did not lead to a worse stage at
presentation [58]. In an earlier study, Abe et al. found a
relatively advanced clinical stage, positive lymph node,
high vascular involvement, and low 5-year survival rate
among obese patients with primary breast cancer in
comparison with non-obese patients [59]. Similarly, a
recent report revealed higher prevalence of aggressive/
advanced stage malignancies in overweight/obese breast
cancer patients [60]. Interestingly, Healy et al. reported that
newly diagnosed postmenopausal breast cancer patients
with pathological stage (IIIV) were significantly more
likely to be obese, hyperglycemic, and hyperinsulinemic
with the majority exhibiting metabolic syndrome compared
with those with early stage of the disease [61]. Furthermore,
among obese patients, Litton et al. detected a significantly
higher number of hormone-negative cancers, stage III
tumors, and worse overall survival at a median follow-up
time of 4.1 years [62]. An analysis of nearly 2,000 women
diagnosed with invasive breast cancer indicated that
overweight women had significantly larger tumor size
compared with normal-weight patients [63]. Tumor size is
another important component to assess cancer stage and
Maehle et al. demonstrated a significant association
between body weight and tumor diameter in older patients
(more than 50 years of age) with hormone receptor-negative
breast cancer [64]. Several studies have also found a
positive relationship between body weight and lymph node
metastasis [2,3,6570]. Recent reports on the association
of obesity with cancer cell metastases, disease recurrence,
and patientssurvival have been summarized in Table 1[2,
7189]. Although the influence of obesity on estrogen
availability and hormonal effects on breast cancer prognosis
have been considered to be an important component of the
interrelationship of obesity and breast cancer [90], adipo-
kines also appear to play a significant role in the
progression of malignancies (Fig. 1)[16,9194].
3 Studies on breast cancer which considered both leptin
and adiponectin
A number of epidemiological studies have been carried out to
investigate the roles of adiponectin and leptin in breast cancer.
In an earlier study, Mantzoros et al. [8] found a significant
inverse association between serum adiponectin levels and
breast cancer risk among postmenopausal women, but there
appeared to be no association for leptin. A total of 174
women with newly diagnosed histologically confirmed
breast cancer and 167 women in the control group were
included; 125 cases and 123 controls belonged to the
postmenopausal group, and the remaining (i.e., 49 cases
and 44 controls) were premenopausal women. Another study
conducted in Taiwan reported decreased serum adiponectin
and increased leptin levels in breast cancer patients [49]. The
study analyzed various clinicopathological parameters along
with serum adiponectin and leptin concentrations in 100
breast cancer patients and 100 women in the control group.
The investigators did not find any significant differences
between the levels of leptin or adiponectin in patients within
the younger age group in comparison to older patients. In
addition, neither premenopausal nor postmenopausal patients
had any significant differences in leptin or adiponectin
levels. However, an interesting finding was that the serum
leptin to adiponectin ratio increased significantly in breast
cancer patients as compared with controls; and this ratio had
a positive and significant correlation with tumor size. In
another study, Hou et al. [95] analyzed blood samples of 80
breast cancer patients (43 premenopausal and 37 postmen-
opausal) and 50 healthy women (26 premenopausal and 24
postmenopausal). They found that serum adiponectin levels
were significantly lower whereas serum levels of leptin were
significantly higher in postmenopausal breast cancer patients
compared to controls. Furthermore, they observed that the
reduced levels of adiponectin and elevated leptin were
associated with lymph node metastasis. Interestingly, patients
with high histological grade tumors had lower serum
adiponectin in comparison to patients with low grade tumors
andleptinlevelspositivelycorrelatedwithtumorsize.Ina
recent study that included 561 breast cancer cases and an
equal number of matched controls from the participants of
The Northern Sweden Health and Disease Cohort, it was
reported that plasma adiponectin levels were negatively
correlated with leptin, and there was a trend of increased
risk for stage IIIV tumors with higher levels of leptin [96].
Surprisingly, leptin levels were inversely associated with
stage I breast cancer in this study. It is possible that the
associations of both leptin and adiponectin with breast cancer
differ depending on the pathological process or disease
course.
Analysis of human breast tumor tissues has also
suggested roles for leptin and adiponectin in breast cancer
Cancer Metastasis Rev (2010) 29:641653 643
development and progression. In a recent study, Jarde et al.
[97] analyzed the immunohistochemical expression of
adiponectin and leptin in 45 primary breast ductal cancers,
14 ductal carcinomas in situ, and 40 normal breast tissues
located adjacent to breast cancer. They found adiponectin
and leptin immunoreactivity in 75% and 80% of normal
breast tissue samples, respectively. Furthermore, leptin was
expressed in a similar manner in invasive ductal carcinoma
and in situ lesions; 80% of cases were positive in both
groups. In contrast, no tissue from in situ ductal carcinoma
exhibited adiponectin expression. However, 15% of cases
involving invasive cancer revealed immunoreactivity of
adiponectin. It was an interesting observation that these two
adipokines were inversely expressed in breast tissue. For
Table 1 Summary of studies that found a positive association of obesity with prognostic parameters such as distant metastasis, recurrence, and
survival in women with breast carcinoma
Investigators Study details Salient findings
Gillespie et al.
(2010) [71]
1,312 patients with stage IIII primary breast cancer/Michigan Obesity was associated with higher rates of tumor
neovascularization.
de Azambuja et
al. (2010) [72]
2,887 node-positive breast cancer patients/Belgium Obesity remained an independent prognostic factor for
shorter overall survival and disease-free survival.
Chen et al.
(2010) [73]
5,042 breast cancer patients with median follow-up of
46 months/Shanghai, China
Women with obesity at diagnosis and post-diagnostic
weight gain had higher mortality.
Majed et al.
(2009) [74]
Observational cohort of patients followed since a first
unilateral breast cancer without distant dissemination/France
Obese patients presented significantly higher risks of death
and metastasis recurrences.
Vona-Davis et
al. (2008) [75]
Retrospective study on 620 White patients/West Virginia Obesity was present in 50% cases with triple negative
tumors (ER,PR, HER2)
Caan et al.
(2008) [76]
Prospective cohort study of 1,692 breast cancer survivors,
7 years of follow-up/Northern California
Obesity before breast cancer diagnosis was associated with
increased risk of recurrence and poorer survival.
Dal Maso et al.
(2008) [77]
1,453 women with incidence of breast cancer/Italy Increased risk of death for breast cancer among obese
patients.
Kerlikowske et
al. (2008) [78]
287,115 postmenopausal women not using hormone therapy/
California
Higher rates of advanced disease, including large invasive
tumors, advanced-stage, and high nuclear grade
malignancies among overweight and obese subjects.
Dawood et al.
(2008) [79]
602 patients with locally advanced breast cancer/Texas Obese or overweight patients had a significantly worse
overall survival and recurrence-free survival and a higher
incidence of recurrence.
Majed et al.
(2008) [80]
Cohort of 14,709 patients/Paris, France Obesity appeared as a negative prognosis factor for several
events such as metastatic recurrence, shorter disease-free
interval and overall survival, and second primary cancer.
Demirkan et al.
(2007) [81]
266 patients/Turkey Postmenopausal obese patients had worse disease-free
survival and distant disease-free survival.
Abrahamson et
al. (2006) [82]
1,254 women diagnosed with invasive breast cancer/
metropolitan Atlanta and New Jersey
Increased mortality in women who were obese.
Dignam et al.
(2006) [83]
4,077 women with node-negative, ER() breast cancer/North
America
Obesity was associated with greater risk for second
cancers, contralateral breast cancer, and mortality.
Loi et al.
(2005) [2]
Population-based study of 1,360 women with breast cancer
before the age of 60 years/Australia
Obesity was associated with larger tumors, more involved
axillary nodes, increased breast cancer recurrence, and
death.
Whiteman et al.
(2005) [84]
3,924 women with incidence of breast cancer/U.S. Obese women were significantly more likely to die of
breast cancer than lean women.
Kroenke et al.
(2005) [85]
5,204 participants under NursesHealth Study diagnosed with
breast cancer/US
Pre-diagnosis obesity and weight gain were related to
higher rates of breast cancer recurrence and mortality.
Tillman et al.
(2005) [86]
139 Native American women with breast cancer/Phoenix,
Arizona
Obesity significantly correlated with larger tumor size and
more advanced stage.
Enger et al.
(2004) [87]
Retrospective cohort study of 1,376 patients followed up for a
median of 6.8 years/California
Women with higher body weight with early stage ER()
tumors had a nearly fivefold increased risk of dying
compared to lower weight and ER(+) tumors.
Dignam et al.
(2003) [88]
3,385 patients enrolled in National Surgical Adjuvant Breast
and Bowel Project (NSABP)/US
Obesity was associated with increased risk of contralateral
breast cancer.
Borugian et al.
(2003) [89]
603 patients with incident breast cancer/Vancouver,
British Columbia, Canada
Waist-to-hip ratio was directly related to breast cancer
mortality in postmenopausal women.
644 Cancer Metastasis Rev (2010) 29:641653
instance, myoepithelial cells of normal tissue adjacent to
breast cancer exhibited 65% positivity for adiponectin
while no cells in this group were positive for leptin
expression. In contrast, in ductal carcinoma in situ, high
levels of leptin expression and no adiponectin expression
were observed. This study has highlighted a fascinating role
of myoepithelial cells in the leptinadiponectin interaction;
the normal myoepithelial cells, which strongly expressed
adiponectin, might protect normal breast epithelial cells by
secreting adiponectin locally (in a paracrine manner).
Overall, the study has provided some evidence of antago-
nistic effects between leptin and adiponectin in breast
cancer development.
4In vitro studies focusing on both leptin
and adiponectin
In order to understand the relationship of adiponectin and
leptin in breast cancer development, our laboratory has
examined in vitro effects of both of these adipokines on
breast cancer cells. In our first study, a low physiological
level of adiponectin (1 μg/ml) and a high physiological level
of leptin (50 ng/ml) were used to reflect serum levels of
obese women. We examined how this adipokine combina-
tion affected cell signaling, apoptosis, and proliferation in
breast cancer cell lines with respect to two significant
characteristics, i.e., presence or absence of the estrogen
receptor (ER) and overexpression of HER2/neu. These two
biomarkers in breast tumor pathology are extremely impor-
tant factors in clinical outcome. Five breast cancer cell lines
[MCF-7 (ER+, HER2), T47-D (ER+, HER2), MDA-MB-
231 (ER,HER2), MDA-MB-361 (ER+, HER2+), and
SK-BR-3 (ER, HER2+)] were used; and results were
influenced by cell line [98]. Highlights of the findings
include that the adiponectin treatment increased the expres-
sion of both short and long isoforms of leptin receptors
(Ob-R and Ob-Rb) in T47-D cells; whereas in MDA-MB-
231 cells, protein expression of these receptors was reduced
by the addition of adiponectin. In general, adiponectin
decreased AdipoR1 expression in MCF-7, T47-D, MDA-
MB-231, and SK-BR-3 cells. In MCF-7 cells, adiponectin
stimulated leptin levels, although it suppressed adiponectin
expression. On the other hand, addition of leptin elevated
both leptin and adiponectin expression levels in MCF-7 cells.
Interestingly, simultaneous exposure to both leptin and
adiponectin caused overexpression of the progesterone
receptor-α(PRα) in MCF-7 cells, while adiponectin
inhibited PRα, but raised PRβexpression in T47-D cells.
Androgen receptor levels were increased by leptin in MCF-7
cells, and by adiponectin in T47-D cells. Two important
hormone-binding proteins, i.e., sex hormone-binding globu-
lin and insulin-like growth factor binding protein-3, were
increased in MCF-7 cells by adiponectin. It is worth
mentioning that both leptin and adiponectin may participate
in the maintenance of energy homoeostasis, which is
disturbed in obesity. These results were perhaps the
consequences of differential effects of these adipokines on
human breast cancer cell lines with diverse hormonal
sensitivities.
It has been reported that the ER-negative breast cancer
cell line, MDA-MB-231, is responsive to adiponectin as
Fig. 1 Possible role of
adipose tissue in breast
cancer progression. ER estrogen
receptor, PR progesterone
receptor, EGFR epidermal
growth factor receptor, SHBG
sex hormone-binding globulin,
IGF-I insulin-like growth
factor-I, upwards arrow
increase, downwards
arrow decrease
Cancer Metastasis Rev (2010) 29:641653 645
reflected by decreased proliferation [26]. To further
investigate the effects of adiponectin in relationship to the
presence or absence of estrogen receptor status, the ERα
gene was introduced into MDA-MB-231 cells by liposome-
mediated transfection. Several ER+ stably transfected
MDA-MB-231 cell lines exhibited higher levels of prolif-
eration compared to the original MDA-MB-231 cells
following addition of either estradiol or leptin. One of the
transfected cell lines designated as MDA-MB-231 ERα7
(MDA-ERα7) was selected for further study. This cell line
and the parental wild type MDA-MB-231 cells (MDA-wt)
expressed both adiponectin and leptin receptors. These two
cell lines were used to investigate the effects of full-length
adiponectin and globular adiponectin in the presence or
absence of leptin. The addition of 50 ng/ml of leptin
resulted in increased proliferation of both MDA-wt and
MDA-ERα7 cells, whereas a reduction in proliferation was
evident after addition of adiponectin and globular adipo-
nectin. Furthermore, cell proliferation assays were per-
formed using different ratios of adiponectin to leptin to
represent women with different body weight status, i.e.,
high adiponectin and low leptin levels for lean persons and
the reverse situation for the obese condition. Interestingly,
we found that an increasing adiponectin to leptin ratio
reduced cell proliferation, while a decreasing ratio failed to
influence the proliferation of both MDA-wt and MDA-
ERα7 cells [99].
Another study examined the effects of adiponectin to
leptin ratios on proliferation of human breast cancer cell
lines, MCF-7, T47-D, MDA-MB-231, MDA-MB-361, and
SK-BR-3 cells [100]. The cells were treated for 48 h with
10, 25, and 50 ng/ml of leptin and 100, 200, 500, 1,000,
and 5,000 ng/ml of adiponectin, respectively. Increasing the
adiponectin to leptin ratio resulted in reduced proliferation
of MCF-7 and T47-D cells. On the contrary, MDA-MB-
231, MDA-MB-361, and SK-BR-3 cells had an increase in
cell proliferation at lower adiponectin to leptin ratios. In
MCF-7 cells, adiponectin and leptin co-treatment enhanced
apoptosis; however, there were no significant differences in
cell cycle distribution between leptin-treated, adiponectin
and leptin co-treated and untreated cells. Overall, different
cell lines exhibited varied responses to different adiponectin
to leptin ratios. These studies indicate the complex
interaction of the characteristics of the breast cancer cell
line with respect to response to both leptin and adiponectin
as well as to the ratio of these two adipokines.
5 Preclinical studies of both leptin and adiponectin
in relation to mammary tumorigenesis
To evaluate the role of obesity and adipokine levels in the
development of breast cancer, several xenograft studies
were conducted using human breast cancer cells which had
been found to be responsive in vitro to leptin and/or
adiponectin. In one set of studies, CD-1 mice were made
obese by injecting them with goldthioglucose [101]. Serum
leptin levels were significantly higher in the obese mice
compared to lean ones while adiponectin levels were not
affected. The adiponectin to leptin ratio was significantly
reduced in the obese mice although it was not related to the
presence or absence of tumors formed following inocula-
tion of T47-D human breast cancer cells. This may be
partially attributable to the relatively low number of mice
with tumors. In an additional study, dietary-induced obesity
was used in conjunction with tumor formation from ER+
MCF-7 human breast cancer cells [102]. CD-1 ovariecto-
mized nude mice were fed a purified high-fat diet from
15 weeks of age (high-fat group, n= 46) while the control
mice, (low-fat group, n=7) were maintained on a purified
low-fat diet (AIN-93M) throughout the study. At 22
23 weeks of age, all mice were inoculated with MCF-7
cells, implanted with estradiol pellets, and followed for
tumor development. The study was terminated when the
mice reached 42 weeks of age. In contrast to other strains of
mice, high-fat CD-1 mice did not exhibit any significant
differences in body weight, fat pad weight, or serum
adiponectin levels compared to low-fat mice (Table 2).
Serum leptin and IGF-I levels were 65% and 72% higher in
the high-fat mice compared to low-fat mice, respectively.
Interestingly, western blot analysis of mammary fat pads
from low-fat mice exhibited significantly higher levels of
AdipoR2 compared to those from high-fat mice (Fig. 2).
Immunohistochemical staining of tumor tissue detected
expression of Ob-R in 21% of high-fat mice (Fig. 3and
Table 3). For the low-fat group, Ob-R expression was
detected in 75% of the tumors; additionally, expression of
leptin was detected in all tumors in this group. On the other
hand, both adiponectin receptors (AdipoR1 and AdipoR2)
were almost equally expressed in tumors from high-fat mice
(43% vs. 46%, respectively). For the low-fat mice,
AdipoR2 was expressed in all tumors, whereas 75% of
the tumors were positive for AdipoR1 expression. These
findings suggest that a high-fat diet can influence both
mammary tissue and tumor adipokine-related factors
independent of effects on body weight.
Another interesting aspect of this study was that of all the
MCF-7 tumors analyzed, only one of 32 had ERαidentifiable
by immunohistochemistry (Table 3). Thus, the originally
ER+ MCF-7 cells formed tumors which no longer appeared
to be estrogen-responsive. Recent studies have shown an
influence of estrogenic environment on the functions of both
leptin and adiponectin [101,103]. Therefore, in the above-
mentioned study, this phenomenon probably played a role on
the expression of leptin, Ob-R, and adiponectin receptors in
the developing mammary tumors.
646 Cancer Metastasis Rev (2010) 29:641653
In general, it is thought that postmenopausal obesity favors
the development of hormone-dependent breast cancer [104,
105]. On the other hand, obesity does not seem to be a risk
factor for the development of ER-negative breast cancer. In
support of this idea, a study using mice that overexpress
HER2/neu (MMTV-neu) showed no differences in tumor
incidence between low-fat and high-fat diet groups [106]. In
a recent study also using MMTV-neu mice [107], it was
reported that the time of onset of the first tumor and tumor
growth rates were not altered between high- and low-fat diet
groups, although the mice on a high-fat diet had an earlier
onset of second tumors along with a greater incidence of
multiple tumors. It is worth noting that dietary fat reduction
has been reported to improve the relapse-free survival in
women with hormone receptor-negative disease [108]. This
receptor-negative situation may be comparable with the
MCF-7 tumors in CD-1 mice [102] where the majority of
the tumors that arose had lost expression of ER. It may be
that MCF-7 breast cancer cells which express adiponectin,
leptin, and their associated receptors can provide a model for
the ER-negative breast cancer proliferative responses to
varying fat intake and adiposity.
It is speculated that besides estrogenic influences, adipo-
kines can stimulate breast cancer growth and metastasis in a
number of different ways [109]. Recent reports using an
adiponectin null mouse model of mammary cancer (MMTV-
PyV-mT/APN knockout mice) that resembles HER2/neu-
amplified human breast cancer indicated reduced vasculari-
zation resulting in nutrient deprivation of the tumors and
associated cell death; however, at the later stage, more
aggressive tumor behaviors were found in these mice [110,
111]. In contrast, an anti-angiogenic role of adiponectin in
cancer has been documented [31,112]. Accumulating
evidence also indicates a role for both adiponectin and leptin
with inflammation and oxidative stress [113117] which are
pathological processes associated with carcinogenesis [118].
In fact, pro-inflammatory pathways may play a pivotal role
in tumor development even in the absence of obesity. This is
supported by the fact that fatless A-Zip/F1 mice exhibit
accelerated skin papilloma formation [119]. Despite the lack
of fat tissue, A-Zip/F1 mice have close similarities with the
obese condition as they are hyperlipidemic and diabetic with
high blood levels of insulin, IGF-I, as well as pro-
inflammatory cytokines such as interleukin-6, IL-4, and
monocyte chemotactic protein-1. In addition, they have
elevated levels of vascular endothelial growth factor and
increased activity of several kinases including phosphatidy-
linositol 3-kinase, extracellular signal-regulated kinase, and
protein tyrosine kinase ErbB2 [119,120]. Many of these
proteins are linked with the signaling pathways of leptin.
Lack of adipose tissue associated with negligible adipokines
like an extremely low level of adiponectin may lead to
increased susceptibility to tumor growth in A-Zip/F1 mice
[121]. Remarkably, low levels of adiponectin most probably
are pathognomonic of obesity-related diseases. Therefore,
both obesity and the above-mentioned lipodystrophic condi-
tion generate a state of chronic inflammation, which is
perhaps coupled with oxidative stress. In a recent report on
breast cancer patients, leptin has been shown to be positively
correlated with oxidative stress parameters [122]. A study
from our laboratory on MCF-7 breast cancer cells docu-
mented a stimulating effect of leptin on the expression of
CYP1B1 [40], which is a phase I enzyme associated with
oxidativedamage[123] and metabolic activation of procar-
cinogens [124]. Unlike lipodystrophic conditions, obesity is
a common health problem connected with a derangement of
adipokines, particularly leptin and adiponectin, which possi-
bly creates an imbalanced state between various oxidants and
the bodys detoxifying system with one consequence being
the development of malignancies.
Table 2 Final body weights, mammary and visceral fat weights, serum adiponectin and leptin with their ratios, and IGF-I for CD-1 nude mice
inoculated with MCF-7 cells
Final body
wt. (g)
MFP
wt. (g)
Visceral fat
wt. (g)
Serum Acrp30
(μg/mL)
Serum leptin
(ng/mL)
Acrp30:leptin
ratio
Serum IGF-I
(ng/mL)
High-fat diet group (n=46) 31.58± 0.82 0.71 ± 0.08 1.24 ± 0.18 9.03± 0.46 8.63 ± 1.44 2.71± 0.55 446.9± 28.5a
Control, low-fat diet (n=7) 32.24 ±0.47 0.80±0.18 1.01 ± 0.27 8.70 ±1.24 5.22± 1.76 3.32 ±0.90 259±75.4b
ttest P=0.7432 P= 0.6719 P= 0.6229 P= 0.7974 P= 0.3626 P= 0.6678 P=0.0209
Values represent mean ± standard error of the mean. Values with lowercase letters indicate a significant difference after studentsttest
MFP mammary fat pad, Acrp30 adiponectin, IGF-I insulin-like growth factor-I
0.00
0.25
0.50
0.75
b
a
AdipoR2:beta actin rati o
(densit ometry units)
*
Fig. 2 Relative expression levels of AdipoR2 in mammary fat pads
between controls and animals on a high-fat diet by western blot
analyses. acontrols (low-fat diet group) and bhigh-fat diet group
studentsttest, *p<0.0001
Cancer Metastasis Rev (2010) 29:641653 647
6 Prevention of breast cancer through dietary alteration
of the leptinadiponectin ratio
If leptin and adiponectin play roles in breast cancer
development, being able to alter their circulating levels
may be an approach towards prevention. Interestingly,
several studies reported that leptin and/or adiponectin levels
can be modulated by dietary factors resulting in an
alteration in the adiponectin to leptin ratio. For instance, it
has been observed that bitter melon (Momordica charantia)
ameliorated fructose diet-induced hyperleptinemia and
reversed hypoadiponectinemia [125]. Similarly, dietary
conjugated linoleic acid supplementation has been shown
to reduce the levels of leptin and increase the mRNA
expression of adiponectin in the adipose tissue of obese rats
[126]. In general, leptin concentrations decreased and
adiponectin increased after weight loss [127]. In a study
conducted by Fenton et al. [128], calorie-restricted mice
displayed increased serum levels of adiponectin. In con-
trast, diet-induced obese (high calorie diet, fed ad libitum)
mice exhibited increased leptin levels [128]. In this
connection, our laboratory has been studying the impact
Fig. 3 Immunohistochemical expression of different parameters related to leptin and adiponectin in tumor tissues developed from MCF-7 cells in
CD-1 mice. aImmunohistochemically negative control. bExpression of leptin, cOb-R, dAdipoR1, eAdipoR2, and fERαexpression
Table 3 Overexpression of leptin and adiponectin receptors in tumors developed from MCF-7 human breast cancer cells in CD-1 mice
Groups Mice with tumor Tested Ob-R Leptin AdipoR1 AdipoR2 ERα
Controls, low-fat diet (n=7) 4 (tumor incidence= 57%) 4 3/4 (75.0%) 4/4 (100%) 3/4 (75.0%) 4/4 (100%) 0/4 (0%)
High-fat diet (n=46) 28 (tumor incidence= 61%) 28 6/28 (21.4%) 0/28 (0%) 12/28 (42.9%) 13/28 (46.4%) 1/28 (3.6%)
No tumor tissue showed overexpression of estrogen receptor β, sex hormone-binding globulin (SHBG), epidermal growth factor receptor
(EGFR), and p53
648 Cancer Metastasis Rev (2010) 29:641653
of caloric restriction in mammary tumorigenesis, particu-
larly the protective role of intermittent caloric restriction
[129132]. In a recently completed study, we found that
intermittent caloric-restricted mice had higher serum adi-
ponectin and lower serum leptin levels after 64 weeks of
caloric restriction. Moreover, the adiponectin to leptin ratio
was significantly higher in intermittent caloric-restricted
groups compared to the ad libitum-fed mice [133]. Howell
et al. [134] analyzed the influence of intermittent caloric
restriction versus chronic calorie restriction on biomarkers
of breast cancer risk and revealed that both approaches are
effective in improving various parameters including reduc-
tion of serum leptin levels. A recent study of obese women
who consumed a low-calorie diet showed an increase of
adiponectin concentrations and a decrease in the levels of
leptin [135]. Thus, preclinical as well as clinical studies
suggest that there are multiple mechanisms available to
influence the ratio of leptin to adiponectin.
7 Concluding remarks
It is clear that nutrition and overnutrition can have a profound
effect on breast cancer inhibition and growth [136]. A
comprehensive report by the World Cancer Research Fund/
American Institute for Cancer Research found that body
fatness, abdominal fatness, and adult weight gain all
represented a probable or convincing increased risk of
postmenopausal breast cancer [137]. Epidemiological studies
have found that the leptin to adiponectin ratio is influenced by
changes in body fatness. Increasing evidence suggests that it
is the ratio of adiponectin to leptin that is the key determinant
of the effects of these adipokines on breast cancer tumorigen-
esis. There appear to be multiple mechanisms of action
including direct cell proliferation, regulation of growth factors,
influences on angiogenesis, and alterations in oxidative stress,
and inflammation. Fortunately, there are also multiple
potential interventions including calorie restriction and alter-
ation of specific dietary factors. Continued investigations will
help clarify the role of body weight status and adipokines in
the development and/or prevention of breast cancer.
Acknowledgements We are thankful to Dr. Joseph P. Grande of the
Mayo Foundation, Rochester, Minnesota, for his pathological diagnosis
of the tumors. We also thank The Breast Cancer Research Foundation,
NCI grant CA101858 and the Hormel Foundation for the support.
References
1. Cleary, M. P., & Maihle, N. J. (1997). The role of body mass
index in the relative risk of developing premenopausal versus
postmenopausal breast cancer. Proceedings of the Society for
Experimental Biology and Medicine, 216,2843.
2. Loi, S., Milne, R. L., Friedlander, M. L., McCredie, M. R., Giles, G.
G., Hopper, J. L., et al. (2005). Obesity and outcomes in
premenopausal and postmenopausal breast cancer. Cancer Epide-
miology, Biomarkers & Prevention, 14,16861691.
3. Porter, G. A., Inglis, K. M., Wood, L. A., & Veugelers, P. J.
(2006). Effect of obesity on presentation of breast cancer. Annals
of Surgical Oncology, 13, 327332.
4. Sweeney, C., Blair, C. K., Anderson, K. E., Lazovich, D., &
Folsom, A. R. (2004). Risk factors for breast cancer in elderly
women. American Journal of Epidemiology, 160, 868875.
5. Fontana, L., Eagon, J. C., Trujillo, M. E., Scherer, P. E., & Klein,
S. (2007). Visceral fat adipokine secretion is associated with
systemic inflammation in obese humans. Diabetes, 56, 1010
1013.
6. McTiernan, A. (2005). Obesity and cancer: the risks, science,
and potential management strategies. Oncology (Williston Park),
19, 871881.
7. Bondanelli, M., Margutti, A., Ambrosio, M. R., Plaino, L.,
Cobellis, L., Petraglia, F., et al. (2001). Blood growth hormone-
binding protein levels in premenopausal and postmenopausal
women: roles of body weight and estrogen levels. The Journal of
Clinical Endocrinology and Metabolism, 86, 19731980.
8. Mantzoros, C., Petridou, E., Dessypris, N., Chavelas, C., Dalamaga,
M., Alexe, D. M., et al. (2004). Adiponectin and breast cancer risk.
The Journal of Clinical Endocrinology and Metabolism, 89,
11021107.
9. Miyoshi, Y., Funahashi, T., Kihara, S., Taguchi, T., Tamaki, Y.,
Matsuzawa, Y., et al. (2003). Association of serum adiponectin
levels with breast cancer risk. Clinical Cancer Research, 9, 5699
5704.
10. Coskun, U., Gunel, N., Toruner, F. B., Sancak, B., Onuk, E.,
Bayram, O., et al. (2003). Serum leptin, prolactin and vascular
endothelial growth factor (VEGF) levels in patients with breast
cancer. Neoplasma, 50,4146.
11. Stattin, P., Söderberg, S., Biessy, C., Lenner, P., Hallmans, G.,
Kaaks, R., et al. (2004). Plasma leptin and breast cancer risk: a
prospective study in northern Sweden. Breast Cancer Research
and Treatment, 86, 191196.
12. Woo, H. Y., Park, H., Ki, C. S., Park, Y. L., & Bae, W. G. (2006).
Relationships among serum leptin, leptin receptor gene poly-
morphisms, and breast cancer in Korea. Cancer Letters, 237,
137142.
13. Wu, M. H., Chou, Y. C., Chou, W. Y., Hsu, G. C., Chu, C. H.,
Yu, C. P., et al. (2009). Circulating levels of leptin, adiposity and
breast cancer risk. British Journal of Cancer, 100, 578582.
14. Hancke, K., Grubeck, D., Hauser, N., Kreienberg, R., & Weiss,
J. M. (2010). Adipocyte fatty acid-binding protein as a novel
prognostic factor in obese breast cancer patients. Breast Cancer
Research and Treatment, 119, 367377.
15. Garofalo, C., Koda, M., Cascio, S., Sulkowska, M., Kanczuga-
Koda, L., Golaszewska, J., et al. (2006). Increased expression of
leptin and the leptin receptor as a marker of breast cancer
progression: possible role of obesity-related stimuli. Clinical
Cancer Research, 12, 14471453.
16. Miyoshi, Y., Funahashi, T., Tanaka, S., Taguchi, T., Tamaki, Y.,
Shimomura, I., et al. (2006). High expression of leptin receptor
mRNA in breast cancer tissue predicts poor prognosis for
patients with high, but not low, serum leptin levels. International
Journal of Cancer, 118, 14141419.
17. Scherer, P. E., Williams, S., Fogliano, M., Baldini, G., & Lodish,
H. F. (1995). A novel serum protein similar to C1q, produced
exclusively in adipocytes. The Journal of Biological Chemistry,
270, 2674626749.
18.Ryan,A.S.,Berman,D.M.,Nicklas,B.J.,Sinha,M.,
Gingerich, R. L., Meneilly, G. S., et al. (2003). Plasma
adiponectin and leptin levels, body composition, and glucose
Cancer Metastasis Rev (2010) 29:641653 649
utilization in adult women with wide ranges of age and obesity.
Diabetes Care, 26, 23832388.
19. Hopkins, T. A., Ouchi, N., Shibata, R., & Walsh, K. (2007).
Adiponectin actions in the cardiovascular system. Cardiovascu-
lar Research, 74,1118.
20. Oh, D. K., Ciaraldi, T., & Henry, R. R. (2007). Adiponectin in
health and disease. Diabetes, Obesity & Metabolism, 9, 282289.
21. Ouchi, N., & Walsh, K. (2007). Adiponectin as an anti-
inflammatory factor. Clinica Chimica Acta, 380,2430.
22. Garofalo, C., & Surmacz, E. (2006). Leptin and cancer. Journal
of Cellular Physiology, 207,1222.
23. Yamauchi, T., Kamon, J., Ito, Y., Tsuchida, A., Yokomizo, T.,
Kita, S., et al. (2003). Cloning of adiponectin receptors that
mediate antidiabetic metabolic effects. Nature, 423, 762769.
24.Tsao,T.S.,Murrey,H.E.,Hug,C.,Lee,D.H.,&Lodish,H.F.
(2002). Oligomerization state-dependent activation of NF-kappa B
signaling pathway by adipocyte complement-related protein of 30
kDa (Acrp30). The Journal of Biological Chemistry, 277, 29359
29562.
25. Fruebis, J., Tsao, T. S., Javorschi, S., Ebbets-Reed, D., Erickson,
M. R., Yen, F. T., et al. (2001). Proteolytic cleavage product of
30-kDa adipocyte complement-related protein increases fatty
acid oxidation in muscle and causes weight loss in mice.
Proceedings of the National Academy of Sciences of the United
States of America, 98, 20052010.
26. Kang, J. H., Lee, Y. Y., Yu, B. Y., Yang, B. S., Cho, K. H., Yoon,
D. K., et al. (2005). Adiponectin induces growth arrest and
apoptosis of MDA-MB-231 breast cancer cell. Archives of
Pharmaceutical Research, 28, 12631269.
27. Wang, Y., Lam, J. B., Lam, K. S., Liu, J., Lam, M. C., Hoo, R.
L., et al. (2006). Adiponectin modulates the glycogen synthase
kinase-3beta/beta-catenin signaling pathway and attenuates
mammary tumorigenesis of MDA-MB-231 cells in nude mice.
Cancer Research, 66, 1146211470.
28. Arditi, J. D., Venihaki, M., Karalis, K. P., & Chrousos, G. P.
(2007). Antiproliferative effect of adiponectin on MCF7 breast
cancer cells: a potential hormonal link between obesity and
cancer. Hormone and Metabolic Research, 39,913.
29. Dieudonne, M. N., Bussiere, M., Dos Santos, E., Leneveu, M.
C., Giudicelli, Y., & Pecquery, R. (2006). Adiponectin mediates
antiproliferative and apoptotic responses in human MCF7 breast
cancer cells. Biochemical and Biophysical Research Communi-
cations, 345, 271279.
30. Wang, Y., Lam, K. S., Xu, J. Y., Lu, G., Xu, L. Y., Cooper, G. J., et
al. (2005). Adiponectin inhibits cell proliferation by interacting with
several growth factors in an oligomerization-dependent manner. The
Journal of Biological Chemistry, 280, 1834118347.
31. Brakenhielm, E., Veitonmaki, N., Cao, R., Kihara, S.,
Matsuzawa, Y., Zhivotovsky, B., et al. (2004). Adiponectin-
induced antiangiogenesis and antitumor activity involve
caspase-mediated endothelial cell apoptosis. Proceedings of
the National Academy of Sciences of the United States of
America, 101,24762481.
32. Zhang, Y., Proenca, R., Maffei, M., Barone, M., Leopold, L., &
Friedman, J. M. (1994). Positional cloning of the mouse obese
gene and its human homologue. Nature, 372, 425432.
33. Knerr, I., Herzog, D., Rauh, M., Rascher, W., & Horbach, T.
(2006). Leptin and ghrelin expression in adipose tissues and
serum levels in gastric banding patients. European Journal of
Clinical Investigation, 36, 389394.
34. Oksanen, L., Ohman, M., Heiman, M., Kainulainen, K., Kaprio, J.,
Mustajoki, P., et al. (1997). Markers for the gene ob and serum
leptin levels in human morbid obesity. Human Genetics, 99,559
564.
35. Ramos, A. P., de Abreu, M. R., Vendramini, R. C., Brunetti, I.
L., & Pepato, M. T. (2006). Decrease in circulating glucose,
insulin and leptin levels and improvement in insulin resistance at
1 and 3 months after gastric bypass. Obesity Surgery, 16, 1359
1364.
36. Klok, M. D., Jakobsdottir, S., & Drent, M. L. (2007). The role of
leptin and ghrelin in the regulation of food intake and body
weight in humans: a review. Obesity Reviews, 8,2134.
37. Dieudonne, M. N., Machinal-Quelin, F., Serazin-Leroy, V.,
Leneveu, M. C., Pecquery, R., & Giudicelli, Y. (2002). Leptin
mediates a proliferative response in human MCF7 breast cancer
cells. Biochemical and Biophysical Research Communications,
293, 622628.
38. Frankenberry, K. A., Skinner, H., Somasundar, P., McFadden, D.
W., & Vona-Davis, L. C. (2006). Leptin receptor expression and
cell signaling in breast cancer. International Journal of Oncol-
ogy, 28, 985993.
39. Hu, X., Juneja, S. C., Maihle, N. J., & Cleary, M. P. (2002).
Leptina growth factor in normal and malignant breast cells and
for normal mammary gland development. Journalofthe
National Cancer Institute, 94, 17041711.
40. Ray, A., Nkhata, K. J., & Cleary, M. P. (2007). Effects of leptin
on human breast cancer cell lines in relationship to estrogen
receptor and HER2 status. International Journal of Oncology,
30, 14991509.
41. Cleary, M. P., Phillips, F. C., Getzin, S. C., Jacobson, T. L.,
Jacobson, M. K., Christensen, T. A., et al. (2003). Genetically
obese MMTV-TGF-alpha/Lep(ob)Lep(ob) female mice do not
develop mammary tumors. Breast Cancer Research and Treat-
ment, 77, 205215.
42. Cleary, M. P., Juneja, S. C., Phillips, F. C., Hu, X., Grande, J. P.,
& Maihle, N. J. (2004). Leptin receptor-deficient MMTV-TGF-
alpha/Lepr(db)Lepr(db) female mice do not develop oncogene-
induced mammary tumors. Experimental Biology and Medicine
(Maywood), 229, 182193.
43. Garofalo, C., Sisci, D., & Surmacz, E. (2004). Leptin interferes
with the effects of the antiestrogen ICI 182, 780 in MCF-7 breast
cancer cells. Clinical Cancer Research, 10, 64666475.
44. Harvie, M., & Howell, A. (2006). Energy balance adiposity and
breast cancerenergy restriction strategies for breast cancer
prevention. Obesity Reviews, 7,3347.
45. Inoue, M., Maehata, E., Yano, M., Taniyama, M., & Suzuki, S.
(2005). Correlation between the adiponectinleptin ratio and
parameters of insulin resistance in patients with type 2 diabetes.
Metabolism, 54, 281286.
46. Zhuo, Q., Wang, Z., Fu, P., Piao, J., Tian, Y., Xu, J., et al. (2009).
Comparison of adiponectin, leptin and leptin to adiponectin ratio as
diagnostic marker for metabolic syndrome in older adults of
Chinese major cities. Diabetes Research and Clinical Practice,
84,2733.
47. Kiec-Klimczak, M., Malczewska-Malec, M., & Huszno, B.
(2008). Leptin to adiponectin ratio, as an index of insulin
resistance and atherosclerosis development. Przegla d Lekarski,
65, 844849.
48. Cleary, M. P., Ray, A., Rogozina, O. P., Dogan, S., &
Grossmann, M. E. (2009). Targeting the adiponectin:leptin ratio
for postmenopausal breast cancer prevention. Frontiers in
Bioscience, S1, 329357.
49. Chen, D. C., Chung, Y. F., Yeh, Y. T., Chaung, H. C., Kuo, F. C.,
Fu, O. Y., et al. (2006). Serum adiponectin and leptin levels in
Taiwanese breast cancer patients. Cancer Letters, 237, 109114.
50. Stark, A., Stahl, M.S., Kirchner, H.L., Krum, S., Prichard, J.,
Evans, J. (2010). Body mass index at the time of diagnosis and
the risk of advanced stages and poorly differentiated cancers of
the breast: findings from a case-series study. International
Journal of Obesity (London). PubMed ID: 20351736.
51. Daling, J. R., Malone, K. E., Doody, D. R., Johnson, L. G.,
Gralow, J. R., & Porter, P. L. (2001). Relation of body mass
650 Cancer Metastasis Rev (2010) 29:641653
index to tumor markers and survival among young women with
invasive ductal breast carcinoma. Cancer, 92, 720729.
52. Carmichael, A. R., Bendall, S., Lockerbie, L., Prescott, R. J., &
Bates, T. (2004). Does obesity compromise survival in women
with breast cancer? Breast, 13,9396.
53. Jones, B. A., Kasi, S. V., Curnen, M. G., Owens, P. H., &
Dubrow, R. (1997). Severe obesity as an explanatory factor for
the black/white difference in stage at diagnosis of breast cancer.
American Journal of Epidemiology, 146, 394404.
54. Moorman, P. G., Jones, B. A., Millikan, R. C., Hall, I. J., &
Newman, B. (2001). Race, anthropometric factors, and stage at
diagnosis of breast cancer. American Journal of Epidemiology,
153, 284291.
55. Cui, Y., Whiteman, M. K., Flaws, J. A., Langenberg, P.,
Tkaczuk, K. H., & Bush, T. L. (2002). Body mass and stage of
breast cancer at diagnosis. International Journal of Cancer, 98,
279283.
56. Cui, Y., Whiteman, M. K., Langenberg, P., Sexton, M., Tkaczuk,
K. H., Flaws, J. A., et al. (2002). Can obesity explain the racial
difference in stage of breast cancer at diagnosis between black
and white women? Journal of Womens Health & Gender-Based
Medicine, 11, 527536.
57. Deglise, C., Bouchardy, C., Burri, M., Usel, M., Neyroud-Caspar, I.,
Vlastos, G., et al. (2010). Impact of obesity on diagnosis and
treatment of breast cancer. Breast Cancer Research and Treatment,
120,185193.
58. Chagpar, A. B., McMasters, K. M., Saul, J., Nurko, J., Martin, R.
C., 2nd, Scoggins, C. R., et al. (2007). Body mass index
influences palpability but not stage of breast cancer at diagnosis.
The American Surgeon, 73, 555560.
59.Abe,R.,Kumagai,N.,Kimura,M.,Hirosaki,A.,&
Nakamura, T. (1976). Biological characteristics of breast
cancer in obesity. The Tohoku Journal of Experimental
Medicine, 120,351359.
60. Chaves, M. R., Tomé, C. B., Monteiro-Grillo, I., Camilo, M.,
Ravasco, P. (2010). The diversity of nutritional status in cancer:
New insights. Oncologist,15, 523530.
61. Healy, L. A., Ryan, A. M., Carroll, P., Ennis, D., Crowley, V.,
Boyle, T., et al. (2010). Metabolic syndrome, central obesity and
insulin resistance are associated with adverse pathological
features in postmenopausal breast cancer. Clinical Oncology
(Royal College of Radiologists), 22, 281288.
62. Litton, J. K., Gonzalez-Angulo, A. M., Warneke, C. L., Buzdar,
A. U., Kau, S. W., Bondy, M., et al. (2008). Relationship
between obesity and pathologic response to neoadjuvant chemo-
therapy among women with operable breast cancer. Journal of
Clinical Oncology, 26, 40724077.
63. McEligot, A. J., Im, T., Dillman, R. O., West, J. G., Salem, R.,
Haque, R., et al. (2008). Abstracting height and weight from
medical records, and breast cancer pathologic factors. Cancer
Causes & Control, 19, 12171226.
64. Maehle, B. O., Tretli, S., Skjaerven, R., & Thorsen, T. (2001).
Premorbid body weight and its relations to primary tumour
diameter in breast cancer patients; its dependence on estrogen
and progesterone receptor status. Breast Cancer Research and
Treatment, 68, 159169.
65. Kalish, L. A. (1984). Relationships of body size with breast
cancer. Journal of Clinical Oncology, 2, 287293.
66. Daniell, H. W. (1988). Increased lymph node metastases at
mastectomy for breast cancer associated with host obesity,
cigarette smoking, age, and large tumor size. Cancer, 62, 429
435.
67. Verreault, R., Brisson, J., Deschênes, L., & Naud, F. (1989).
Body weight and prognostic indicators in breast cancer.
Modifying effect of estrogen receptors. American Journal of
Epidemiology, 129, 260268.
68. Schapira, D. V., Kumar, N. B., Lyman, G. H., & Cox, C. E.
(1991). Obesity and body fat distribution and breast cancer
prognosis. Cancer, 67, 523528.
69. Honda, H., Ohi, Y., Umekita, Y., Takasaki, T., Kuriwaki, K.,
Ohyabu, I., et al. (1999). Obesity affects expression of
progesterone receptors and node metastasis of mammary
carcinomas in postmenopausal women without a family history.
Pathology International, 49, 198202.
70. Maehle, B. O., Tretli, S., & Thorsen, T. (2004). The associations
of obesity, lymph node status and prognosis in breast cancer
patients: dependence on estrogen and progesterone receptor
status. Acta Pathologica, Microbiologica, et Immunologica
Scandinavica, 112, 349357.
71. Gillespie, E. F., Sorbero, M. E., Hanauer, D. A., Sabel, M. S.,
Herrmann, E. J., Weiser, L. J., et al. (2010). Obesity and
angiolymphatic invasion in primary breast cancer. Annals of
Surgical Oncology, 17, 752759.
72. de Azambuja, E., McCaskill-Stevens, W., Francis, P., Quinaux, E.,
Crown, J. P., Vicente, M., et al. (2010). The effect of body mass
index on overall and disease-free survival in node-positive breast
cancer patients treated with docetaxel and doxorubicin-containing
adjuvant chemotherapy: the experience of the BIG 02-98 trial.
Breast Cancer Research and Treatment, 119,145153.
73. Chen, X., Lu, W., Zheng, W., Gu, K., Chen, Z., Zheng, Y., Shu,
X. O. (2010). Obesity and weight change in relation to breast
cancer survival. Breast Cancer Research and Treatment,122,
823833.
74. Majed, B., Moreau, T., Senouci, K., Sigal, B., Salmon, R. J.,
Fourquet, A., et al. (2009). Stoutness and prognosis of female
non-metastatic breast cancer: results from a French observational
cohort study. Bulletin du Cancer, 96, 531541.
75. Vona-Davis, L., Rose, D. P., Hazard, H., Howard-McNatt, M.,
Adkins, F., Partin, J., et al. (2008). Triple-negative breast cancer
and obesity in a rural Appalachian population. Cancer Epidemi-
ology, Biomarkers & Prevention, 17, 33193324.
76. Caan, B. J., Kwan, M. L., Hartzell, G., Castillo, A., Slattery, M.
L., Sternfeld, B., et al. (2008). Pre-diagnosis body mass index,
post-diagnosis weight change, and prognosis among women with
early stage breast cancer. Cancer Causes & Control, 19, 1319
1328.
77. Dal Maso, L., Zucchetto, A., Talamini, R., Serraino, D., Stocco,
C. F., Vercelli, M., et al. (2008). Effect of obesity and other
lifestyle factors on mortality in women with breast cancer.
International Journal of Cancer, 123, 21882194.
78. Kerlikowske, K., Walker, R., Miglioretti, D. L., Desai, A.,
Ballard-Barbash, R., & Buist, D. S. (2008). Obesity, mammog-
raphy use and accuracy, and advanced breast cancer risk. Journal
of the National Cancer Institute, 100, 17241733.
79. Dawood, S., Broglio, K., Gonzalez-Angulo, A. M., Kau, S. W.,
Islam, R., Hortobagyi, G. N., et al. (2008). Prognostic value of
body mass index in locally advanced breast cancer. Clinical
Cancer Research, 14, 17181725.
80. Majed, B., Moreau, T., Senouci, K., Salmon, R. J., Fourquet, A.,
& Asselain, B. (2008). Is obesity an independent prognosis
factor in woman breast cancer? Breast Cancer Research and
Treatment, 111, 329342.
81. Demirkan, B., Alacacioglu, A., & Yilmaz, U. (2007). Relation of
body mass index (BMI) to disease free (DFS) and distant disease
free survivals (DDFS) among Turkish women with operable
breast carcinoma. Japanese Journal of Clinical Oncology, 37,
256265.
82. Abrahamson, P. E., Gammon, M. D., Lund, M. J., Flagg, E. W.,
Porter, P. L., Stevens, J., et al. (2006). General and abdominal
obesity and survival among young women with breast cancer.
Cancer Epidemiology, Biomarkers & Prevention, 15, 1871
1877.
Cancer Metastasis Rev (2010) 29:641653 651
83. Dignam, J. J., Wieand, K., Johnson, K. A., Raich, P., Anderson,
S. J., Somkin, C., et al. (2006). Effects of obesity and race on
prognosis in lymph node-negative, estrogen receptor-negative
breast cancer. Breast Cancer Research and Treatment, 97, 245
254.
84. Whiteman, M. K., Hillis, S. D., Curtis, K. M., McDonald, J. A.,
Wingo, P. A., & Marchbanks, P. A. (2005). Body mass and
mortality after breast cancer diagnosis. Cancer Epidemiology,
Biomarkers & Prevention, 14, 20092014.
85. Kroenke, C. H., Chen, W. Y., Rosner, B., & Holmes, M. D.
(2005). Weight, weight gain, and survival after breast cancer
diagnosis. Journal of Clinical Oncology, 23, 13701378.
86. Tillman, L., Myers, S., Pockaj, B., Perry, C., Bay, R. C., & Al-
kasspooles, M. (2005). Breast cancer in Native American women
treated at an urban-based Indian health referral center 1982
2003. American Journal of Surgery, 190, 895902.
87. Enger, S. M., Greif, J. M., Polikoff, J., & Press, M. (2004). Body
weight correlates with mortality in early-stage breast cancer.
Archives of Surgery, 139, 954958.
88. Dignam, J. J., Wieand, K., Johnson, K. A., Fisher, B., Xu, L., &
Mamounas, E. P. (2003). Obesity, tamoxifen use, and outcomes
in women with estrogen receptor-positive early-stage breast
cancer. Journal of the National Cancer Institute, 95, 14671476.
89. Borugian, M. J., Sheps, S. B., Kim-Sing, C., Olivotto, I. A., Van
Patten, C., Dunn, B. P., et al. (2003). Waist-to-hip ratio and
breast cancer mortality. American Journal of Epidemiology, 158,
963968.
90. McTiernan, A., Rajan, K. B., Tworoger, S. S., Irwin, M.,
Bernstein, L., Baumgartner, R., et al. (2003). Adiposity and sex
hormones in postmenopausal breast cancer survivors. Journal of
Clinical Oncology, 21, 19611966.
91. Ishikawa, M., Kitayama, J., & Nagawa, H. (2004). Enhanced
expression of leptin and leptin receptor (OB-R) in human breast
cancer. Clinical Cancer Research, 10, 43254331.
92. Liu, C. L., Chang, Y. C., Cheng, S. P., Chern, S. R., Yang, T. L.,
Lee, J. J., et al. (2007). The roles of serum leptin concentration
and polymorphism in leptin receptor gene at codon 109 in breast
cancer. Oncology, 72,7581.
93. Macciò, A., Madeddu, C., Gramignano, G., Mulas, C., Floris, C.,
Massa, D., et al. (2010). Correlation of body mass index and
leptin with tumor size and stage of disease in hormone-
dependent postmenopausal breast cancer: preliminary results
and therapeutic implications. Journal of Molecular Medicine,88,
677686.
94. Ray, A., & Cleary, M. P. (2010). Leptin as a potential therapeutic
target for breast cancer prevention and treatment. Expert Opinion
on Therapeutic Targets, 14, 443451.
95. Hou, W. K., Xu, Y. X., Yu, T., Zhang, L., Zhang, W. W., Fu, C.
L., et al. (2007). Adipocytokines and breast cancer risk. Chinese
Medical Journal, 120, 15921596.
96. Cust, A. E., Stocks, T., Lukanova, A., Lundin, E., Hallmans, G.,
Kaaks, R., et al. (2009). The influence of over-weight and insulin
resistance on breast cancer risk and tumour stage at diagnosis: a
prospective study. Breast Cancer Research and Treatment, 113,
567576.
97. Jarde, T., Caldefie-Chezet, F., Damez, M., Mishellany, F.,
Perrone, D., Penault-Llorca, F., et al. (2008). Adiponectin and
leptin expression in primary ductal breast cancer and in adjacent
healthy epithelial and myoepithelial tissue. Histopathology, 53,
484487.
98. Grossmann, M. E., Ray, A., Dogan, S., Mizuno, N. K.,
Cleary, M. P. (2007). Balance of adiponectin and leptin in
relationship to breast cancer cell growth and signaling.
Advances in Breast Cancer Research: Genetics, Biology, and
Clinical Applications. San Diego, California.October1720,
2007; Abstract #B50.
99. Grossmann, M. E., Ray, A., Dogan, S., Mizuno, N. K., & Cleary,
M. P. (2008). Balance of adiponectin and leptin modulates breast
cancer cell growth. Cell Research, 18, 11541156.
100. Nkhata, K. J., Ray, A., Schuster, T. F., Grossmann, M. E., &
Cleary, M. P. (2009). Effects of adiponectin and leptin co-
treatment on human breast cancer cell growth. Oncology
Reports, 21, 16111619.
101. Nkhata, K. J., Ray, A., Dogan, S., Grande, J. P., & Cleary, M. P.
(2009). Mammary tumor development from T47-D human breast
cancer cells in obese ovariectomized mice with and without
estradiol supplements. Breast Cancer Research and Treatment,
114 ,7183.
102. Ray, A., Nkhata, K. J., Malakhov, D. A., Grossmann, M. E.,
Grande, J. P., Cleary, M. P. (2008). Characteristics of MCF-7
breast cancer cell line tumors in response to obesity and diet.
Third Frontiers in Cancer Research and 8th International Skin
Carcinogenesis Conferences. Austin, Minnesota. October 47,
2008; p.172/Abstract #36.
103. Pfeiler, G. H., Buechler, C., Neumeier, M., Schaffler, A.,
Schmitz, G., Ortmann, O., et al. (2008). Adiponectin effects on
human breast cancer cells are dependent on 17-beta estradiol.
Oncology Reports, 19, 787793.
104. Althuis, M. D., Fergenbaum, J. H., Garcia-Closas, M., Brinton,
L. A., Madigan, M. P., & Sherman, M. E. (2004). Etiology of
hormone receptor-defined breast cancer: a systematic review of
the literature. Cancer Epidemiology, Biomarkers & Prevention,
13, 15581568.
105. Suzuki, R., Rylander-Rudqvist, T., Ye, W., Saji, S., & Wolk, A.
(2006). Body weight and postmenopausal breast cancer risk
defined by estrogen and progesterone receptor status among
Swedish women: a prospective cohort study. International
Journal of Cancer, 119, 16831689.
106. Cleary, M. P., Grande, J. P., Juneja, S. C., & Maihle, N. J.
(2004). Diet-induced obesity and mammary tumor development
in MMTV-neu female mice. Nutrition and Cancer, 50, 174180.
107. Khalid, S., Hwang, D., Babichev, Y., Kolli, R., Altamentova, S.,
Koren, S., et al. (2010). Evidence for a tumor promoting effect of
high-fat diet independent of insulin resistance in HER2/Neu
mammary carcinogenesis. Breast Cancer Research and Treat-
ment,122, 647659.
108. Blackburn, G. L., & Wang, K. A. (2007). Dietary fat reduction
and breast cancer outcome: results from the Womens Interven-
tion Nutrition Study (WINS). The American Journal of Clinical
Nutrition, 86, s878s881.
109. Rose, D. P., & Vona-Davis, L. (2009). Influence of obesity on
breast cancer receptor status and prognosis. Expert Review of
Anticancer Therapy, 9, 10911101.
110. Denzel, M. S., Hebbard, L. W., Shostak, G., Shapiro, L., Cardiff,
R. D., & Ranscht, B. (2009). Adiponectin deficiency limits
tumor vascularization in the MMTV-PyV-mT mouse model of
mammary cancer. Clinical Cancer Research, 15, 32563264.
111. Landskroner-Eiger, S., Qian, B., Muise, E. S., Nawrocki, A. R.,
Berger, J. P., Fine, E. J., et al. (2009). Proangiogenic contribution
of adiponectin toward mammary tumor growth in vivo.Clinical
Cancer Research, 15, 32653276.
112. Barresi, V., Tuccari, G., & Barresi, G. (2009). Adiponectin
immunohistochemical expression in colorectal cancer and its
correlation with histological grade and tumour microvessel
density. Pathology, 41, 533538.
113. Soares, A. F., Guichardant, M., Cozzone, D., Bernoud-Hubac,
N., Bouzaïdi-Tiali, N., Lagarde, M., et al. (2005). Effects of
oxidative stress on adiponectin secretion and lactate production
in 3T3-L1 adipocytes. Free Radical Biology & Medicine, 38,
882889.
114. Swarbrick, M. M., & Havel, P. J. (2008). Physiological,
pharmacological, and nutritional regulation of circulating adipo-
652 Cancer Metastasis Rev (2010) 29:641653
nectin concentrations in humans. Metabolic Syndrome and
Related Disorders, 6,87102.
115. Goldstein, B. J., Scalia, R. G., & Ma, X. L. (2009). Protective
vascular and myocardial effects of adiponectin. Nature Clinical
Practice. Cardiovascular Medicine, 6,2735.
116. Dubey, L., & Hesong, Z. (2006). Role of leptin in atherogenesis.
Experimental and Clinical Cardiology, 11, 269275.
117. Wu, B., Fukuo, K., Suzuki, K., Yoshino, G., & Kazumi, T.
(2009). Relationships of systemic oxidative stress to body fat
distribution, adipokines and inflammatory markers in healthy
middle-aged women. Endocrine Journal, 56, 773782.
118. Federico, A., Morgillo, F., Tuccillo, C., Ciardiello, F., & Loguercio,
C. (2007). Chronic inflammation and oxidative stress in human
carcinogenesis. International Journal of Cancer, 121,23812386.
119. Hursting, S. D., Nunez, N. P., Varticovski, L., & Vinson, C.
(2007). The obesitycancer link: lessons learned from a fatless
mouse. Cancer Research, 67, 23912393.
120. Nunez, N. P., Oh, W. J., Rozenberg, J., Perella, C., Anver, M.,
Barrett, J. C., et al. (2006). Accelerated tumor formation in a
fatless mouse with type 2 diabetes and inflammation. Cancer
Research, 66, 54695576.
121. Ablamunits, V., Cohen, Y., Brazee, I. B., Gaetz, H. P., Vinson, C., &
Klebanov, S. (2006). Susceptibility to induced and spontaneous
carcinogenesis is increased in fatless A-ZIP/F-1 but not in obese ob/
ob mice. Cancer Research, 66,88978902.
122. Badid, N., Baba Ahmed, F. Z., Merzouk, H., Belbraouet, S.,
Mokhtari, N., Merzouk, S. A., et al. (2010). Oxidant/antioxidant
status, lipids and hormonal profile in overweight women with
breast cancer. Pathology & Oncology Research,16, 159167.
123. Tsuchiya, Y., Nakajima, M., & Yokoi, T. (2005). Cytochrome
P450-mediated metabolism of estrogens and its regulation in
human. Cancer Letters, 227,115124.
124. Bozina, N., Bradamante, V., & Lovrić, M. (2009). Genetic
polymorphism of metabolic enzymes P450 (CYP) as a suscep-
tibility factor for drug response, toxicity, and cancer risk. Arhiv
za Higijenu Rada i Toksikologiju, 60, 217242.
125. Shih, C. C., Lin, C. H., Lin, W. L., & Wu, J. B. (2009).
Momordica charantia extract on insulin resistance and the
skeletal muscle GLUT4 protein in fructose-fed rats. Journal of
Ethnopharmacology, 123,8290.
126. Zhou, X. R., Sun, C. H., Liu, J. R., & Zhao, D. (2008). Dietary
conjugated linoleic acid increases PPAR gamma gene expression
in adipose tissue of obese rat, and improves insulin resistance.
Growth Hormone & IGF Research, 18, 361368.
127. Kotidis, E. V., Koliakos, G. G., Baltzopoulos, V. G., Ioannidis,
K. N., Yovos, J. G., & Papavramidis, S. T. (2006). Serum
ghrelin, leptin and adiponectin levels before and after weight
loss: comparison of three methods of treatmenta prospective
study. Obesity Surgery, 16, 14251432.
128. Fenton, J. I., Nuñez, N. P., Yakar, S., Perkins, S. N., Hord,
N. G., & Hursting, S. D. (2009). Diet-induced adiposity alters
the serum profile of inflammation in C57BL/6N mice as
measured by antibody array. Diabetes, Obesity & Metabolism,
11,343354.
129. Cleary, M. P., Jacobson, M. K., Phillips, F. C., Getzin, S. C.,
Grande, J. P., & Maihle, N. J. (2002). Weight-cycling decreases
incidence and increases latency of mammary tumors to a greater
extent than does chronic caloric restriction in mouse mammary
tumor virus-transforming growth factor-alpha female mice.
Cancer Epidemiology, Biomarkers & Prevention, 11, 836843.
130. Pape-Ansorge, K. A., Grande, J. P., Christensen, T. A., Maihle, N.
J., & Cleary, M. P. (2002). Effect of moderate caloric restriction and/
or weight cycling on mammary tumor incidence and latency in
MMTV-Neu female mice. Nutrition and Cancer, 44,162168.
131. Cleary, M. P., Hu, X., Grossmann, M. E., Juneja, S. C., Dogan,
S., Grande, J. P., et al. (2007). Prevention of mammary
tumorigenesis by intermittent caloric restriction: does caloric
intake during refeeding modulate the response? Experimental
Biology and Medicine (Maywood), 232,7080.
132. Rogozina, O. P., Bonorden, M. J., Grande, J. P., & Cleary, M. P.
(2009). Serum insulin-like growth factor-I and mammary tumor
development in ad libitum-fed, chronic calorie-restricted, and
intermittent calorie-restricted MMTV-TGF-alpha mice. Cancer
Prevention Research (Phila Pa), 2, 712719.
133. Dogan, S., Rogozina, O. P., Loshkin, A., Grande, J. P., & Cleary,
M. P. (2010). Comparison of chronic vs intermittent calorie
restriction on mammary tumor incidence and serum adiponectin
and leptin levels in MMTV-TGF-a mice at different ages.
Oncology Letters, 1, 167176.
134. Howell, A., Chapman, M., & Harvie, M. (2009). Energy
restriction for breast cancer prevention. Recent Results in Cancer
Research, 181,97111.
135. Varady, K. A., Tussing, L., Bhutani, S., & Braunschweig, C. L.
(2009). Degree of weight loss required to improve adipokine
concentrations and decrease fat cell size in severely obese
women. Metabolism, 58, 10961101.
136. Glade, M. J. (1999). Food, nutrition, and the prevention of
cancer: a global perspective. American Institute for Cancer
Research/World Cancer Research Fund, American Institute for
Cancer Research, 1997. Nutrition, 15, 523526.
137. World Cancer Research Fund/American Institute for Cancer
Research. (2007). Food, nutrition, physical activity, and the
prevention of cancer: a global perspective. Washington, DC:
AICR, 2007.
Cancer Metastasis Rev (2010) 29:641653 653
... [16,19] Leptin is an adiponectin antagonist, which has pro-inflammatory properties. [20] In addition, it is involved in the regulation of food intake and fat metabolism. Short-term fluctuations in the blood content of leptin occur in the human body depending on the energy potential: leptin levels decrease after physical activity or fasting and increase after food intake. ...
... [23] In addition, leptin is involved in the regulation of proliferation, activation of T lymphocytes, and lipid metabolism. A high level of this adipocytokine is associated with the development of adverse cardiovascular events, such as myocardial infarction, acute cerebrovascular accident, arterial hypertension, etc. [19,20,23] Despite the proven epidemiological relationship between breast cancer and obesity, the pathogenetic mechanisms underlying this relationship (in particular, the adipocytokine profile) are still not fully understood. ...
Article
Full-text available
Context Breast cancer is the most common diagnosis established in women with malignant tumors. Aims The purpose is to investigate the blood contents of adiponectin and leptin in women with breast cancer and obesity. Settings and Design A total of 140 women aged 40–50 were examined. Materials and Methods Group 1 included 70 women from classes 1 or 2 obesity. Group 2 included 70 women with stage 1 or 2 breast cancer and classes 1 or 2 obesity. The control group included 30 apparently healthy women, with mean age of 42.5 ± 2.5 years. Statistical Analysis Used Statistical processing of the results obtained was performed using Statistica. Results Groups 1 and 2 were statistically significantly different from each other across all parameters, except for leptin resistance. In group 2, the course of breast cancer with concomitant obesity is characterized by disrupted adipocytokine homeostasis, which manifests as a 1.94-fold decrease in the blood content of adiponectin ( P < 0.05), a 4.14-fold increase in the blood content of leptin ( P < 0.05), and an 8.00-fold increase in the leptin/adiponectin ratio ( P < 0.05). Poorly differentiated breast tumors exhibit a more pronounced imbalance in the blood levels of adipocytokines. Thus, the serum content of leptin in women with poorly differentiated tumors (G3) was 1.79 times ( P < 0.05) higher than in women with moderately differentiated tumors (G2). Conclusions The course of breast cancer with concomitant obesity is characterized by disrupted adipocytokine homeostasis and decreased adiponectin concentration in the blood.
... The mechanisms and causal pathways of obesity affecting the onset of breast cancer are complex. For example, increased estrogens, insulin resistance, mammary fat inflammation, increased aromatase expression, and elevated leptin levels are all thought to play a role in the pathogenesis of obesity-associated breast cancer [32][33][34][35][36][37] . The specific mechanism of the interaction between BMI and a family history of cancer has not yet been reported. ...
Article
Full-text available
Both body mass index (BMI) and family history of cancer are established risk factors for female breast cancer. However, few studies explored the potential interaction between both factors. We assessed the association of BMI and its interaction with family cancer history on the risk of female breast cancer in Shanghai, China. Based on a population-based prospective cohort study started from 2008 to 2012 with 15,055 Chinese female participants in Minhang district, Shanghai. Cox regression models were used to estimate the association of BMI and its interaction with a family history of cancer on breast cancer risk. The additive interaction was evaluated by the relative excess risk due to interaction (RERI) and the attributable proportion due to interaction (AP), and the multiplicative interaction was assessed by the product term (BMI* family history of cancer) in the Cox regression model. Compared with BMI of < 24 kg/m² and no family history of cancer, women with BMI of ≥ 24 kg/m² and a family history of cancer had a higher risk for breast cancer with HR 2.06 (95% CI 1.39, 3.06). There was an additive interaction between BMI and family history of cancer on breast cancer incidence, with the RERI being 0.29 (95% CI 0.08, 0.51) and the AP being 0.37 (95% CI 0.08, 0.66). The coexistence of obesity and cancer family history may exacerbate breast cancer incidence risk, highlighting the importance of weight management in women with a family history of cancer.
... Leptin is primarily produced by secretion from white adipocytes (28). It has been previously reported that obesity is not only related to metabolic diseases but is also closely associated with the development of breast cancer (29). ...
Article
Full-text available
Breast cancer is a common tumor encountered in women, and triple-negative breast cancer (TNBC) has an extremely poor prognosis. The effect of leptin on the docetaxel sensitivity of MDA-MB-231 TNBC cells has not been investigated. The present study aimed to clarify the effect of leptin and M2 tumor-associated macrophages (TAMs) on the chemosensitivity of TNBC cell lines and its possible mechanisms. In the present study, the apoptosis of the MDA-MB-231 cell line was detected at 0, 24, 48 and 72 h using a Cell Counting Kit-8 assay to determine the appropriate concentration of docetaxel as well as the IC50 value. After determining the effect of leptin on TAMs, the conditioned medium with an appropriate concentration of docetaxel was collected to treat the breast cancer cells, and flow cytometry was used to detect the cell cycle distribution and apoptosis in different treatment groups. Interleukin 8 (IL-8) expression was detected using ELISA and western blot assay. The IL-8 antibody was used to neutralize IL-8, and invasion and scratch assays were used to detect changes in invasion and migration of breast cancer cells. Statistical analysis was performed using GraphPad Prism 9.0 and SPSS 22.0. It was revealed that the apoptotic rate of MDA-MB-231 cells in the leptin-treated TAMs group was lower than that in other groups. The expression of IL-8 was notably elevated in the group treated with leptin-activated TAMs compared with that in the other groups. The neutralization of IL-8 resulted in a significant reduction in the invasive migration of MDA-MB-231 cells compared with that in the non-neutralized group.
... APN receptors have been identified in breast cancer cells, and in vitro studies have demonstrated that APN exhibits anti-proliferative and pro-apoptotic effects on breast cancer cells (68). Indeed, epidemiological studies have reported a significant inverse association between APN and breast cancer risk (69,70). The link between APN and breast cancer exhibits a stronger correlation among postmenopausal women, although there have been some conflicting findings published. ...
Article
Full-text available
Obesity, a complex and multifactorial disease influenced by genetic, environmental, and psychological factors, has reached epidemic proportions globally, posing a significant health challenge. In addition to its established association with cardiovascular disease and type II diabetes, obesity has been implicated as a risk factor for various cancers. However, the precise biological mechanisms linking obesity and cancer remain largely understood. Adipose tissue, an active endocrine organ, produces numerous hormones and bioactive molecules known as adipokines, which play a crucial role in metabolism, immune responses, and systemic inflammation. Notably, adiponectin (APN), the principal adipocyte secretory protein, exhibits reduced expression levels in obesity. In this scoping review, we explore and discuss the role of APN in influencing cancer in common malignancies, including lung, breast, colorectal, prostate, gastric, and endometrial cancers. Our review aims to emphasize the critical significance of investigating this field, as it holds great potential for the development of innovative treatment strategies that specifically target obesity-related malignancies. Furthermore, the implementation of more rigorous and comprehensive prevention and treatment policies for obesity is imperative in order to effectively mitigate the risk of associated diseases, such as cancer.
... In breast cancer, a strong link exists between obesity, leptin, and tumor development. High circulating amounts of leptin in patients with obesity promote breast tumor development, whereas adiponectin has antiproliferative properties (21). The interaction between sex hormones, adipokines, and cancer progression requires further research. ...
Article
The overall goal of the annual Transdisciplinary Research in Energetics and Cancer (TREC) Training Workshop is to provide transdisciplinary training for scientists in energetics and cancer and clinical care. The 2022 Workshop included 27 early-to-mid career investigators (trainees) pursuing diverse TREC research areas in basic, clinical, and population sciences. The 2022 trainees participated in a gallery walk, an interactive qualitative program evaluation method, to summarize key takeaways related to program objectives. Writing groups were formed and collaborated on this summary of the 5 key takeaways from the TREC Workshop. The 2022 TREC Workshop provided a targeted and unique networking opportunity that facilitated meaningful collaborative work addressing research and clinical needs in energetics and cancer. This report summarizes the 2022 TREC Workshop's key takeaways and future directions for innovative transdisciplinary energetics and cancer research.
Article
Objective: To investigate the associations between breast cancer staging and body mass index (BMI), menopausal status, and family history of breast cancer, as well as to assess the expression of tumor markers in women with breast cancer in Brunei Darussalam. Methods: A retrospective cross-sectional study was carried out at The Brunei Cancer Centre. Cases of women with breast cancer between 2000 and 2013 were retrieved from the medical records section and entered into a clinical proforma. These were reviewed and analyzed using appropriate statistical methods. Results: Overall, the mean age at diagnosis was 53.6 years ± 9.83. The postmenopausal women had a significantly higher risk of being diagnosed with late-onset breast cancer than premenopausal women (P = 0.022). The expressions of PR (P = 0.034), Her2 (P = 0.046), and CatD (P = 0.004) were significantly different in breast cancer staging. Other factors did not have a statistically-significant association with breast cancer staging. Conclusion: While most of the factors studied were not significantly different in breast cancer staging, our findings showed that it is recommended that high-risk women perform regular breast screening after the age of 40 years old. A future study highly appreciates comparing breast cancer risk factors among the Bruneian and Western populations.
Article
The relations of body weight, height, and Quetelet index to axillary node involvement at diagnosis, estrogen receptor status, and histologic features of the primary tumor were examined In 656 patients with a newly diagnosed infiltrating breast carcinoma first treated in Québec City from July 1982 to December 1984. Node Involvement increased with body weight and Quetelet index. This association was more regular and much stronger among patients with estrogen receptor-positive tumors than among those with estrogen receptor-negative breast cancers. Among patients with estrogen receptor-positive tumors, the percentage with involved nodes at diagnosis increased regularly from 32.9% among lean patients (Quetelet Index <21 kg/m² to 65.6% among obese women (Quetetot index > 27 kg/m² This trend was seen even after adjustment for age and tumor size. In contrast, among patients with estrogen receptor-negative breast cancers, the associations of weight and Quetelet index with node involvement were weak and Irregular. The modifying effect of estrogen receptor status on the relation of obesity to node involvement was apparent in pre- and post- menopausal women. Body weight and Quetelet index were not related to estrogen receptor status or to any of the measured histologic features of breast tumors including nuclear grade, histologic grade, tubule formation, mitotic activity, and size of nucleus of cancer cells. These findings suggest that the observed deleterious effect of obesity on breast cancer prognosis is unlikely to be an artifact of delayed diagnosis in overweight patients. It may be due to hormonal changes associated with increases in body weight
Book
This Report has a number of inter-related general purposes. One is to explore the extent to which food , nutrition , physical activity , and body composition modify the risk of cancer , and to specify which factors are most important. To the extent that environmental factors such as ...
Article
Adiponectin (also known as 30-kDa adipocyte complement-related protein; Acrp30) is a hormone secreted by adipocytes that acts as an antidiabetic and anti-atherogenic adipokine. Levels of adiponectin in the blood are decreased under conditions of obesity, insulin resistance and type 2 diabetes. Administration of adiponectin causes glucose-lowering effects and ameliorates insulin resistance in mice. Conversely, adiponectin-deficient mice exhibit insulin resistance and diabetes. This insulin-sensitizing effect of adiponectin seems to be mediated by an increase in fatty-acid oxidation through activation of AMP kinase and PPAR-alpha. Here we report the cloning of complementary DNAs encoding adiponectin receptors 1 and 2 (AdipoR1 and AdipoR2) by expression cloning. AdipoR1 is abundantly expressed in skeletal muscle, whereas AdipoR2 is predominantly expressed in the liver. These two adiponectin receptors are predicted to contain seven transmembrane domains, but to be structurally and functionally distinct from G-protein-coupled receptors. Expression of AdipoR1/R2 or suppression of AdipoR1/R2 expression by small-interfering RNA supports our conclusion that they serve as receptors for globular and full-length adiponectin, and that they mediate increased AMP kinase and PPAR-alpha ligand activities, as well as fatty-acid oxidation and glucose uptake by adiponectin.
Article
Background: Obesity is associated with both increased breast cancer risk and poorer prognosis after disease onset. However, little is known about the effect of obesity on treatment efficacy. We evaluated the association of obesity with outcomes and with tamoxifen efficacy in women with early-stage, hormone-responsive breast cancer participating in a multicenter cancer cooperative group clinical trial. Methods: The cohort consisted of 3385 women enrolled in National Surgical Adjuvant Breast and Bowel Project (NSABP) protocol B-14, a randomized, placebo-controlled trial evaluating tamoxifen for lymph node–negative, estrogen receptor (ER)–positive breast cancer. Hazards of breast cancer recurrence, contralateral breast tumors, other new primary cancers, and several mortality endpoints were evaluated in relation to body mass index (BMI), using statistical modeling to adjust for other prognostic factors. Median follow-up time was 166 months. All statistical tests were two-sided. Results: The hazard of breast cancer recurrence was the same among obese (BMI ⩾30.0 kg/m2) women as compared with underweight and normal-weight women (BMI <25.0; hazard ratio [HR] = 0.98, 95% confidence interval [CI] = 0.80 to 1.18). Contralateral breast cancer hazard was higher in obese women than in underweight/normal-weight women (HR = 1.58, 95% CI = 1.10 to 2.25), as was the risk of other primary cancers (HR = 1.62, 95% CI = 1.16 to 2.24). Compared with normal-weight women, obese women had greater all-cause mortality (HR = 1.31, 95% CI = 1.12 to 1.54) and greater risk of deaths due to causes unrelated to breast cancer (HR = 1.49, 95% CI = 1.15 to 1.92). Breast cancer mortality was not statistically significantly increased for obese women (HR = 1.20, 95% CI = 0.97 to 1.49). Tamoxifen reduced breast cancer recurrence and mortality, regardless of BMI. Conclusions: For women with lymph node–negative, ER-positive breast cancer, obesity was not associated with a material increase in recurrence risk or a change in tamoxifen efficacy. However, because obesity was associated with increased risks of contralateral breast cancer, of other primary cancers, and of overall mortality, it may influence long-term outcomes for breast cancer survivors.
Article
Background: Obesity is a risk factor for breast cancer in postmenopausal women. As body weight and fat mass increase, circulating leptin increases. Leptin is an adipocyte-derived cytokine that acts through the long form of its receptor, termed OB-Rb. To investigate whether leptin is associated with breast cancer, we determined the expression of OB-Rb in human breast epithelial HBL100 cells and human breast carcinoma-derived T-47D cells, determined whether leptin influenced the proliferation of these cells, and evaluated the structure of mammary tissue in genetically obese leptin-deficient LepobLepob and leptin receptor-deficient LeprdbLeprdb mice. Methods: Cell numbers and cell colony formation by HBL100 and T-47D cells were determined by anchorage-dependent and anchorage-independent growth assays. OB-Rb expression was examined by reverse transcription–polymerase chain reaction and immunoblot analyses. Expression of leptin signaling pathway components was evaluated with immunoblot and electrophoretic mobility shift assays. Mammary gland development in lean and obese mice was investigated in whole-mount studies. All statistical tests were two-sided. Results: Leptin enhanced anchorage-dependent proliferation by 138% (95% confidence interval [CI] = 108% to 169%) in T-47D cells and 50% (95% CI = 38% to 60%) in HBL100 cells. In both cell lines, OB-Rb was expressed, and leptin increased the expression of phosphorylated signal transducers and activators of transcription 3 (STAT3), phosphorylated extracellular signal-regulated kinase (ERK), and transcript activator protein 1 (AP-1). However, leptin increased anchorage-independent cell growth only in the breast cancer cell line (by 81% [95% CI = 62% to 101%] compared with untreated cells). Obese LepobLepob and LeprdbLeprdb mice had minimal epithelial development in the mature mammary gland compared with their lean counterparts. Conclusions: Leptin appears to be able to control the proliferation of both normal and malignant breast epithelial cells. Consequently, the leptin pathway should be further studied as a target for interventions to treat or prevent breast cancer.
Article
High insulin levels have been associated with increased risk of breast cancer and poorer survival after a breast cancer diagnosis. Waist-to-hip ratio (WHR) is a marker for insulin resistance and hyperinsulinemia. In this study, the authors tested the hypothesis that elevated WHR is directly related to breast cancer mortality. For identification of modifiable factors affecting survival, data were collected on 603 patients with incident breast cancer who visited the Vancouver Cancer Centre of the British Columbia Cancer Agency (Vancouver, British Columbia, Canada) in 1991–1992, including body measurements and information on demographic, medical, reproductive, and dietary factors. These patients were followed for up to 10 years. Cox proportional hazards regression models were used to relate the variables to breast cancer mortality (n = 112). After adjustment for age, body mass index, family history, estrogen receptor (ER) status, tumor stage at diagnosis, and systemic treatment (chemotherapy or tamoxifen), WHR was directly related to breast cancer mortality in postmenopausal women (for highest quartile vs. lowest, relative risk = 3.3, 95% confidence interval: 1.1, 10.4) but not in premenopausal women (relative risk = 1.2, 95% confidence interval: 0.4, 3.4). Stratification according to ER status showed that the increased mortality was restricted to ER-positive postmenopausal women. Elevated WHR was confirmed as a predictor of breast cancer mortality, with menopausal status and ER status at diagnosis found to be important modifiers of that relation. body constitution; breast neoplasms; insulin resistance; mortality; receptors, estrogen