You are on page 1of 7

Available online at www.sciencedirect.

com

ScienceDirect

Inflammation, dysregulated metabolism and aromatase


in obesity and breast cancer
Heba Zahid1,2,4, Evan R Simpson3,4 and Kristy A Brown1,4

Obesity is associated with an increased risk of estrogen- Obesity affects the production of these factors and is
dependent breast cancer after menopause. Adipose tissue associated with a chronic state of low-grade inflammation,
undergoes important changes in obesity due to excess storage having profound effects on tumor development and pro-
of lipids, leading to adipocyte cell death and the recruitment of gression.
macrophages. The resultant state of chronic low-grade
inflammation is associated with the activation of NFkB According to the World Health Organization, 55% of
signaling and elevated levels of aromatase, the rate-limiting women worldwide are overweight or obese [2]. Higher
enzyme in estrogen biosynthesis. This occurs not only in the obesity rates are associated with an increased prevalence
visceral and subcutaneous fat, but also in the breast fat. The of obesity-associated diseases, such as cardiovascular dis-
regulation of aromatase in the breast adipose stromal cell in ease, diabetes and number of cancers, including breast
response to inflammatory mediators is under the control of cancer [3,4,5]. Breast cancer is the most diagnosed cancer
complex signaling pathways, including metabolic pathways in women in developed and developing countries. In the
involving LKB1/AMPK, p53, HIF1a and PKM2. Interventions United States, one in eight women will be diagnosed with
aimed at modifying weight, including diet and exercise, are breast cancer during their lifetime. Of these, two-thirds
associated with changes in adipose tissue inflammation and will develop estrogen receptor-positive (ER+) tumors
estrogen production that are likely to impact breast cancer risk. that depend on estrogens for growth [6]. The main source
This review will present an overview of these topics. of estrogen in premenopausal women is the ovaries, while
Addresses
in postmenopausal women, it is the adipose tissue [7].
1
Centre for Cancer Research, Hudson Institute of Medical Research,
Clayton, Victoria, Australia In postmenopausal women, obesity and weight gain have
2
Faculty of Applied Medical Science, Taibah University, Medina, Saudi been associated with significantly higher risk of ER+/
Arabia progesterone receptor-positive (PR+) breast cancer
3
Centre for Endocrinology and Metabolism, Hudson Institute for Medical
Research, Clayton, Victoria, Australia
(Table 1; [8–10]), attributable in part to increased local
4
Monash University, Clayton, Victoria, Australia estrogen production from the breast adipose tissue. In
ER+ breast cancer survivors, a 10% increase in body
Corresponding author: Brown, Kristy A (kristy.brown@hudson.org.au) weight is also associated with increased risk of recurrence
after 5 years [11]. Additionally, higher BMI is associated
Current Opinion in Pharmacology 2016, 31:90–96 with poor prognosis and worse overall survival and breast
cancer-specific survival in women with ER+/PR+/human
This review comes from a themed issue on Endocrine and metabolic
diseases epidermal growth factor receptor 2 (HER2) negative
breast cancers [12,13]. In premenopausal women, al-
Edited by Sebastiano Andò
though no significant effect of BMI on breast cancer risk
is observed [14], higher body mass index (BMI) is associ-
ated with larger tumors, higher incidence of lymph node
http://dx.doi.org/10.1016/j.coph.2016.11.003
metastases and higher histological grade [15,16]. In pre
and postmenopausal women, obesity has been found to
1471-4892/# 2016 Published by Elsevier Ltd.
be positively associated with breast cancer-specific mor-
tality [17,18] (Figure 1).

This review will discuss the role of obesity-associated


inflammation in driving the local expression of aromatase,
the rate-limiting enzyme in estrogen biosynthesis, via
Obesity and breast cancer
effects on metabolic pathways.
Adipose tissue is the largest endocrine organ in the body
and plays an important role in maintaining energy ho-
meostasis. Understanding the underlying biology of adi- Obesity and adipose tissue inflammation
pose tissue is important for understanding how obesity Adipose tissue is composed of multiple cell types, includ-
contributes to tumor development. It is well known that ing adipocytes and cells of the stromal vascular fraction
obesity is associated with excess storage of lipids [1]. In (e.g. preadipocytes, immune and endothelial cells). In
addition to lipid storage, adipose tissue also produces adipocytes, a positive energy balance leads to the storage
adipokines and steroid hormones, including estrogens. of excess calories as triglycerides, causing their expansion.

Current Opinion in Pharmacology 2016, 31:90–96 www.sciencedirect.com


Inflammation, dysregulated metabolism and aromatase Zahid, Simpson and Brown 91

Table 1

Effect of BMI on breast cancer risk in postmenopausal women.

Type of study Number of studies Years BMI (kg/m2) RR (95% CI) References
Meta-analysis 25 1997–2014 25 1.02 (0.98–1.06) [8]
30 1.12 (1.01–1.24)
35 1.26 (1.07–1.50)
Meta-analysis 11 1997–2011 Obese 1.25 (1.07–1.46) [9]
Meta-analysis 39 1980–2012 25–29.9 1.10 (1.06–1.13) [10]
30 1.18 (1.12–1.25)

With obesity, the unhealthy expansion of adipose tissue is inflammation, generate reactive oxygen species and re-
associated with endoplasmic reticulum stress, adipose lease cytokines such as tumor necrosis factor-a (TNFa)
tissue fibrosis and localized hypoxia [1]. This, in turn, and interleukin 6 (IL-6) [21]. Hypoxia inducible factor 1a
is associated with adipocyte cell death and initiation of an (HIF1a) is a key driver of this response, as loss of HIF1a
inflammatory response [19]. An increase in monocyte in adipocytes leads to the reduced expression of TNFa
chemoattractant protein-1 (MCP-1), also known as and MCP-1 mRNA, resulting in reduced macrophage
CCL2, in adipose tissue contributes to the infiltration infiltration and attenuation of the pro-inflammatory mi-
of macrophages [20] and their accumulation around dead lieu [22]. The adipokine leptin, which is elevated with
or dying adipocytes, leading to the formation of a distinct obesity, also stimulates the production of TNFa, IL-6
histological structure referred to as ‘crown like structure’ and other cytokines, and promotes monocyte phagocytic
(CLS). In contrast to lean adipose tissue, which predomi- function (reviewed in [23]). Studies of CLS have been
nantly contains alternatively activated M2 macrophages, undertaken in mouse models of diet-induced and genet-
obese adipose tissue is characterized by the infiltration of ic-induced obesity, as well as in humans, where increased
classically activated M1 macrophages which promote weight has been shown to be associated with a greater

Figure 1

Breast
Epithelial Adipose stromal cell
Cell PGE2

cytoplasm

E2 PKA
Adipose
Stromal Cell LKB1 Aha1
Arom CRTC
p p53
p HSP90
AMPK AMPK HIF1α
p
CRTC p53
Adipose Aha1
p
Tissue CLS-B CREB HSP90
+
nucleus
- p PKM2 CRTC p
PKM2

p53 HIF1α CREB

aromatase

Current Opinion in Pharmacology

Obesity, breast inflammation and the regulation of aromatase by metabolic pathways. Obesity is associated with increased breast white adipose
tissue inflammation, characterized by an increase in the presence of CLS-B. Inflammatory mediators, including PGE2, stimulate the expression of
aromatase in adipose stromal cells via effects on metabolic pathways involving LKB1/AMPK, CREB/CRTC, p53, HIF1a and PKM2. Aromatase
catalyzes the conversion of androgens into estrogens, known to drive the growth of obesity-related breast cancer cells. Large yellow cells
represent adipocytes; blue cells represent infiltrating macrophages forming CLS. CLS-B: crown-like structure of the breast; Arom: aromatase; E2:
estradiol; PGE2: prostaglandin E2; PKA: protein kinase A; LKB1: liver kinase B1; AMPK: AMP-activated protein kinase; CREB: cAMP response
element binding protein; CRTC: CREB-regulated transcription coactivator; HSP90: heat shock protein 90; Aha1: activator of heat shock 90 kDa
protein ATPase homolog 1; HIF1a: hypoxia inducible factor-1a; PKM2: protein kinase M2.

www.sciencedirect.com Current Opinion in Pharmacology 2016, 31:90–96


92 Endocrine and metabolic diseases

number of CLS/cm2 [24,25]. Interestingly, menopause is activation of aromatase expression. The tumor suppressor
also a determinant of breast white adipose tissue inflam- p53, also a recently established regulator of metabolism
mation [26]. The presence of CLS is associated with [35], was also found to inhibit the expression of aromatase
activation of nuclear factor kB (NF-kB) and increased by binding to a p53 response element on the aromatase
levels of TNFa, IL-1b, IL-6, and cyclooxygenase-2 promoter and be repressed by PGE2. On the other hand,
(COX-2)-derived prostaglandin E2 (PGE2) [27]. the oncogene HIF1a, also a driver of metabolic changes
in cancer cells, was found to be a potent stimulator of
Obesity, inflammation and breast cancer: aromatase expression in breast adipose stromal cells and
aromatase, the missing link required for the PGE2-mediated expression of aromatase
Obesity and inflammation have been associated with [36]. Recently, a link between p53, HIF1a and the
higher levels of aromatase in mammary adipose tissue metabolic regulator protein kinase M2 (PKM2) was also
in both mice and humans [24,25,27,28]. Interestingly, the demonstrated in stromal cells from Li–Fraumeni Syn-
presence of CLS in the mammary gland and breast (CLS- drome patients who have elevated aromatase expression
B) appears to be more strongly correlated with aromatase [37,38]. It was demonstrated that loss of p53 leads to the
expression and activity than weight or BMI, highlighting stabilization of HIF1a and PKM2 via heat shock protein
an important role for inflammatory mediators in driving 90 (Hsp90) dependent mechanisms, stimulates their in-
aromatase expression and/or potential limitations in using teraction with the aromatase promoter, and leads to an
BMI to define metabolic health. increase in aromatase expression and activity. Women
with Li–Fraumeni Syndrome tend to develop ER+ breast
The main site of aromatase expression in the breast fat is cancer and observed increases in breast aromatase may
the adipose stromal cell, a precursor to lipid-laden adi- account, at least in part, for why this occurs. Collectively,
pocytes and often referred to as preadipocyte or fibroblast. these data suggest that dysregulated metabolism not only
The stimulatory effect of inflammatory mediators on occurs in adipocytes and cancer cells, but also in adipose
aromatase transcript expression in isolated adipose stro- stromal cells in response to inflammatory mediators,
mal cells has been described in numerous studies. In thereby providing a molecular link between obesity
particular, cytokines, including TNFa, IL-6, IL-11, leu- and breast cancer.
kemia inhibitory factor (LIF) and oncostatin M, as well as
PGE2, have been shown to be potent drivers of aromatase Lifestyle interventions, inflammation,
expression in breast adipose stromal cells [29,30]. The aromatase and breast cancer risk
mechanism of aromatase transcriptional regulation by Considering the prevalence of obesity in today’s society
these factors is complex (reviewed in [31]) and con- and the now established link between obesity and breast
tinues to be the focus of investigation. For the purposes of cancer development, it is imperative that strategies are
this review, we will present recent discoveries relating to identified to reduce the health and economic burden
the role of metabolic pathways as regulators of aromatase predicted to occur as this population ages. As the majority
in the breast, as these pathways have been shown to be of postmenopausal breast cancers are dependent on estro-
dysregulated in both obesity and cancer. gens for growth, it is predicted that lowering systemic and
local estrogen levels will reduce the risk of breast cancer.
Dysregulated cell metabolism and aromatase A number of studies have been undertaken to examine
regulation the effect of lifestyle interventions, including moderate
Dysregulated cell metabolism has long been known to physical activity and controlled diet, on breast inflamma-
occur in obesity and is now an accepted Hallmark of tion, estrogen levels and breast cancer risk (Table 2).
cancer [32]. The role of metabolic pathways in regulating
aromatase expression in breast adipose stromal cells has Weight loss in postmenopausal women is associated with
recently been explored. Initial studies examined the role a reduced risk of breast cancer, while weight gain is
of the metabolic sensor, 50 AMP-activated protein kinase positively associated with risk in these women [39].
(AMPK), and its upstream kinase liver kinase B1 (LKB1). Reducing fat intake and altering diet, including increased
AMPK is well-characterized for its ability to sense consumption of fruits and vegetables, has been shown to
decreases in cellular ATP leading to stimulation of path- lower the risk of relapse in women after 5 years breast
ways of energy production and inhibition of pathways of cancer treatment [40,41]. Human caloric reduction and
energy utilization [33]. LKB1/AMPK were shown to exercise studies have also showed improvement in weight
inhibit aromatase expression in isolated adipose stromal loss, fat reduction, overall life quality and long-term
cells by inhibiting the nuclear entry of the CREB-coacti- prognosis [42,43].
vator (cAMP response element binding protein), CRTC2
(CREB regulated transcription cofactor 2) [34]. The In relation to effects of diet and exercise on estrogen
obesity-associated inflammatory factor, PGE2, sup- levels, studies have found that physical activity, alone and
pressed the expression and activity of LKB1/AMPK in combination with improved diet, leads to a decrease in
leading to the nuclear translocation of CRTC2 and circulating estrogens in healthy women, independent of

Current Opinion in Pharmacology 2016, 31:90–96 www.sciencedirect.com


Inflammation, dysregulated metabolism and aromatase Zahid, Simpson and Brown 93

Table 2

Effect of lifestyle and pharmacological interventions on breast cancer risk, inflammatory factors and estrogen

Intervention Outcome References


Lifestyle Diet Reduced risk of invasive breast cancer [40,41,44,47]
Lower risk of relapse
Decreased levels of E1, E2, free E2 and T
Increased levels of SHBG
Decreased levels of hsCRP, SAA and IL-6
Exercise Fat loss, improved quality of life [43–45,46,47]
Decreased levels of E1, E2, free E2, A4, DHEA-S and adiposity markers
Increased levels of SHBG
Combination of Weight loss, fat reduction [42,44,47]
diet and exercise
Lowered leptin, cholesterol, saturated fat, blood pressure, hsCRP, SAA and
IL-6 Decreased levels of E1, E2, free E2 and T Increased levels of SHBG
Pharmacological Metformin Reduced cancer risk and breast cancer mortality [49–56]
Reduced PKB/Akt and ERK1/2 phosphorylation, and insulin and insulin
receptor levels
Pioglitazone Improved HDL cholesterol, biomarkers of lipid metabolism, b-cell function, [64,65]
activity of the visceral adipose tissue, and chronic systemic inflammation
Decreased total adipose macrophage number, mast cells, M1 macrophages
Increases number of M2 macrophages

E1: estrone; E2: estradiol; T: testosterone; SHBG: sex hormone binding globulin; hsCRP: C-reactive protein; SAA: serum amyloid A; IL-6: interleukin-
6; A4: androstenedione; DHEA-S: dehydroepiandrosterone-sulfate; HDL: high density lipoprotein.

menopausal status [44,45,46]. Interestingly, these on tumor growth, although doses required to achieve
changes were dependent on loss of body fat, suggesting inhibitory effects on breast cancer cells in vitro suggest
that exercise reduces circulating estrogen levels originat- that effects observed clinically are likely to depend on
ing from the adipose tissue. Similarly, the effect of diet indirect effects of the drug. Metformin has also been
and exercise on adipose tissue inflammation has also been shown to inhibit adipose tissue inflammation in mouse
examined. Results suggest that diet alone or in combina- and rat models of diet-induced obesity [57–59], as well as
tion with exercise, leads to a reduction in systemic mar- in subcutaneous human adipose tissue [60]. Via effects on
kers of inflammation, including C-reactive protein and AMPK, metformin also inhibits aromatase transcript ex-
IL-6, while exercise alone did not [47]. These human pression in isolated breast adipose stromal cells [61,62].
studies suggest that chronic inflammation in obese wom- Despite being associated with weight gain, the peroxi-
en is reversible via combinations of controlled diet or/and some proliferator-activated receptor gamma (PPARg)
exercise. In mouse models, reducing caloric intake agonist and anti-diabetic drug pioglitazone, is associated
reverses elevations in adipose tissue inflammation and with a decrease in local and systemic inflammation in
aromatase associated with high fat feeding [48]. Specifi- humans when used alone or in combination with metfor-
cally, 30% caloric restriction is associated with more than min [63–65]. Interestingly, pioglitazone also causes a
75% reduction in mammary gland CLS, MCP-1 and decrease in the expression of aromatase in isolated human
aromatase. Additional studies are required to determine adipose stromal cells and mouse mammary gland [66].
whether observed decreases in inflammation in women
are also associated with a decrease in the local production Conclusions
of estrogens, and whether this will impact the develop- With a higher prevalence of obesity, it is expected that
ment and progression of breast cancers. the incidence of breast cancer will increase as the popu-
lation ages. Better understanding of the links, both phys-
Pharmacological interventions, inflammation, iological and molecular, will allow the identification of
aromatase and breast cancer new therapeutic strategies to break the linkage between
Weight-modulating drugs, in particular anti-diabetic obesity and breast cancer. There is a strong rationale for
drugs, have been examined for their effect on inflamma- targeting adipose tissue inflammation and the local pro-
tion, aromatase and breast cancer (Table 2). Several duction of estrogen in order to better treat and possibly
studies have described a beneficial effect of the anti- even prevent obesity-related postmenopausal breast can-
diabetic drug metformin on breast cancer risk, recurrence cers.
and cancer-associated mortality [49–56]. Multiple mech-
anisms have been proposed for the anti-cancer effects of Conflict of interest statement
metformin, including effects on insulin and direct effects Nothing declared.

www.sciencedirect.com Current Opinion in Pharmacology 2016, 31:90–96


94 Endocrine and metabolic diseases

15. Yanai A, Miyagawa Y, Murase K, Imamura M, Yagi T, Ichii S,


Acknowledgements Takatsuka Y, Ito T, Hirota S, Sasa M et al.: Influence of body mass
This work was supported by the Victorian Government Operational index on clinicopathological factors including estrogen
Infrastructure Support Program. KAB is supported by the Mavis Robertson receptor, progesterone receptor, and Ki67 expression levels in
Fellowship from the National Breast Cancer Foundation (ECF-16-004). breast cancers. Int J Clin Oncol 2014, 19:467-472.
16. Santa-Maria CA, Yan J, Xie X-J, Euhus DM: Aggressive estrogen-
References and recommended reading receptor-positive breast cancer arising in patients with
elevated body mass index. Int J Clin Oncol 2015, 20:317-323.
Papers of particular interest, published within the period of review,
have been highlighted as: 17. Protani M, Coory M, Martin JH: Effect of obesity on survival of
women with breast cancer: systematic review and meta-
 of special interest analysis. Breast Cancer Res Treat 2010, 123:627-635.
 of outstanding interest
18. Taghizadeh N, Boezen HM, Schouten JP, Schroder CP, Elisabeth
de Vries EG, Vonk JM: BMI and lifetime changes in BMI and
1. Rutkowski JM, Stern JH, Scherer PE: The cell biology of fat
cancer mortality risk. PLOS ONE 2015, 10:e0125261.
expansion. J Cell Biol 2015, 208:501-512.
19. Lee YS, Kim J-W, Osborne O, Oh DY, Sasik R, Schenk S, Chen A,
2. Obesity and Overweight. Available at: http://www.who.int/
 Chung H, Murphy A, Watkins SM et al.: Increased adipocyte O(2)
mediacentre/factsheets/fs311/en/.
consumption triggers HIF-1a causing inflammation and
3. Kelesidis I, Kelesidis T, Mantzoros CS: Adiponectin and cancer: a insulin resistance in obesity. Cell 2014, 157:1339-1352.
systematic review. Br J Cancer 2006, 94:1221-1225. In mouse models, high fat diet (HFD) leads to an increase in oxygen
consumption which triggers HIF-1a resulting in the induction of adipose
4. Keum N, Greenwood DC, Lee DH, Kim R, Aune D, Ju W: Adult tissue inflammation. Genetic deletion of adipocyte HIF-1a protects from
 weight gain and adiposity-related cancers: a dose–response HFD-induced adipose tissue inflammation.
meta-analysis of prospective observational studies. J Natl
Cancer Inst 2015:107. 20. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K-I, Kitazawa R,
The authors performed a meta-analysis on 50 observational studies Kitazawa S, Miyachi H, Maeda S, Egashira K et al.: MCP-1
investigating the relationship between adult weight gain and the risk of contributes to macrophage infiltration into adipose tissue,
obesity-related cancers, including breast cancer. For each 5 kg increase insulin resistance, and hepatic steatosis in obesity. J Clin
in adult weight, the relative risk was 1.11 (95% CI = 1.08–1.13) for Invest 2006, 116:1494.
postmenopausal breast cancer among no or low hormone replacement
therapy (HRT) users. Adult weight gain was not associated with 21. Lumeng CN, Bodzin JL, Saltiel AR: Obesity induces a phenotypic
increased breast cancer risk in premenopausal or postmenopausal switch in adipose tissue macrophage polarization. J Clin Invest
women on HRT. 2007, 117:175-184.

5. Chen GC, Chen SJ, Zhang R, Hidayat K, Qin JB, Zhang YS, Qin LQ: 22. Kihira Y, Miyake M, Hirata M, Hoshina Y, Kato K, Shirakawa H,
Central obesity and risks of pre- and postmenopausal breast Sakaue H, Yamano N, Izawa-Ishizawa Y, Ishizawa K et al.:
cancer: a dose–response meta-analysis of prospective Deletion of hypoxia-inducible factor-1a in adipocytes
studies. Obes Rev 2016. enhances glucagon-like peptide-1 secretion and reduces
adipose tissue inflammation. PLOS ONE 2014, 9:e93856.
6. Breast Cancer. Available at: http://www.cancer.org/acs/groups/
cid/documents/webcontent/003090-pdf.pdf. 23. Matarese G, Moschos S, Mantzoros CS: Leptin in immunology. J
Immunol (Baltim, MD: 1950) 2005, 174:3137-3142.
7. Brown KA, Simpson ER: Obesity and breast cancer:
mechanisms and therapeutic implications. Front Biosci 2012, 24. Subbaramaiah K, Howe LR, Bhardwaj P, Du B, Gravaghi C,
4:2515-2524. Yantiss RK, Zhou XK, Blaho VA, Hla T, Yang P et al.: Obesity is
associated with inflammation and elevated aromatase
8. Xia X, Chen W, Li J, Chen X, Rui R, Liu C, Sun Y, Liu L, Gong J, expression in the mouse mammary gland. Cancer Prev Res
Yuan P: Body mass index and risk of breast cancer: a nonlinear (Phila) 2011, 4:329-346.
dose-response meta-analysis of prospective studies. Sci Rep
2014, 4:7480. 25. Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK,
Du B, Brogi E, Crawford CB, Kopelovich L et al.: Inflammation and
9. Dobbins M, Decorby K, Choi BCK: The Association between increased aromatase expression occur in the breast tissue of
Obesity and Cancer Risk: A Meta-Analysis of Observational Studies obese women with breast cancer. Cancer Prev Res (Phila) 2011,
from 1985 to 2011. ISRN Preventive Medicine 2013. 2013:16. 4:1021-1029.
10. Munsell MF, Sprague BL, Berry DA, Chisholm G, Trentham- 26. Iyengar NM, Morris PG, Zhou XK, Gucalp A, Giri D, Harbus MD,
Dietz A: Body mass index and breast cancer risk according to  Falcone DJ, Krasne MD, Vahdat LT, Subbaramaiah K et al.:
postmenopausal estrogen-progestin use and hormone Menopause is a determinant of breast adipose inflammation.
receptor status. Epidemiol Rev 2014, 36:114-136. Cancer Prev Res (Phila) 2015, 8:349-358.
The authors demonstrated that white adipose tissue inflammation (WATi)
11. Nechuta S, Chen WY, Cai H, Poole EM, Kwan ML, Flatt SW, in 237 women is increased in postmenopausal women independent of
Patterson RE, Pierce JP, Caan BJ, Ou Shu X: A pooled analysis of BMI.
post-diagnosis lifestyle factors in association with late
estrogen-receptor-positive breast cancer prognosis. Int J 27. Subbaramaiah K, Morris PG, Zhou XK, Morrow M, Du B, Giri D,
Cancer 2016, 138:2088-2097. Kopelovich L, Hudis CA, Dannenberg AJ: Increased levels of
COX-2 and prostaglandin E2 contribute to elevated aromatase
12. Robinson PJ, Bell RJ, Davis SR: Obesity is associated with a expression in inflamed breast tissue of obese women. Cancer
 poorer prognosis in women with hormone receptor positive Discov 2012, 2:356-365.
breast cancer. Maturitas 2014, 79:279-286.
Five-year follow up with 1155 Australian women with ER+/PR+/HER2 28. Purohit A, Ghilchik MW, Duncan L, Wang DY, Singh A, Walker MM,
breast cancer revealed that obesity was associated with an independent Reed MJ: Aromatase activity and interleukin-6 production by
increased likelihood of breast cancer recurrence and metastasis. normal and malignant breast tissues. J Clin Endocrinol Metabol
1995, 80:3052-3058.
13. Jeon YW, Kang SH, Park MH, Lim W, Cho SH, Suh YJ:
Relationship between body mass index and the expression of 29. Crichton MB, Nichols JE, Zhao Y, Bulun SE, Simpson ER:
hormone receptors or human epidermal growth factor Expression of transcripts of interleukin-6 and related
receptor 2 with respect to breast cancer survival. BMC Cancer cytokines by human breast tumors, breast cancer cells, and
2015, 15:865. adipose stromal cells. Mol Cell Endocrinol 1996, 118:215-220.
14. Cheraghi Z, Poorolajal J, Hashem T, Esmailnasab N, Doosti Irani A: 30. Zhao Y, Agarwal VR, Mendelson CR, Simpson ER:
Effect of body mass index on breast cancer during Transcriptional regulation of CYP19 gene (aromatase)
premenopausal and postmenopausal periods: a meta- expression in adipose stromal cells in primary culture. J
analysis. PLoS ONE 2012, 7:e51446. Steroid Biochem Mol Biol 1997, 61:203-210.

Current Opinion in Pharmacology 2016, 31:90–96 www.sciencedirect.com


Inflammation, dysregulated metabolism and aromatase Zahid, Simpson and Brown 95

31. Brown KA, Simpson ER: Obesity and Breast Cancer: The Role of 46. Ennour-Idrissi K, Maunsell E, Diorio C: Effect of physical activity
 Dysregulated Estrogen Metabolism. New York: Springer; 2014.  on sex hormones in women: a systematic review and meta-
Comperhensive review of the relationship between obesity and breast analysis of randomized controlled trials. Breast Cancer Res
cancer. In particular, the role of dysregulated estrogen metabolism in the 2015, 17:1-11.
breast and molecular mechanisms involved. Data for circulating female sex hormones, including total and free estra-
diol, were extracted from 23 randomized controlled trials. Physical activity
32. Hanahan D, Weinberg RA: Hallmarks of cancer: the next significantly reduced total and free estradiol levels, independent of
generation. Cell 2011, 144:646-674. menopausal status. Physical activity also decreased free testosterone,
androstenedione, dehydroepiandrosterone-sulfate and adiposity mar-
33. Brown KA, Samarajeewa NU, Simpson ER: Endocrine-related kers. A significant increase in sex hormone-binding globulin (SHBG)
cancers and the role of AMPK. Mol Cell Endocrinol 2013, was observed.
366:170-179.
47. Imayama I, Ulrich CM, Alfano CM, Wang C, Xiao L, Wener MH,
34. Samarajeewa NU, Docanto MM, Simpson ER, Brown KA: CREB- Campbell KL, Duggan C, Foster-Schubert KE, Kong A et al.:
regulated transcription co-activator family stimulates Effects of a caloric restriction weight loss diet and exercise on
promoter II-driven aromatase expression in preadipocytes. inflammatory biomarkers in overweight/obese
Horm Cancer 2013, 4:233-241. postmenopausal women: a randomized controlled trial.
Cancer Res 2012, 72:2314-2326.
35. Wang X, Simpson ER, Brown KA: p53: protection against tumor
growth beyond effects on cell cycle and apoptosis. Cancer Res 48. Bhardwaj P, Du B, Zhou XK, Sue E, Harbus MD, Falcone DJ, Giri D,
2016, 76:1668. Hudis CA, Kopelovich L, Subbaramaiah K et al.: Caloric
restriction reverses obesity-induced mammary gland
36. Samarajeewa NU, Yang F, Docanto MM, Sakurai M,
inflammation in mice. Cancer Prev Res (Phila) 2013, 6:282-289.
McNamara KM, Sasano H, Fox SB, Simpson ER, Brown KA: HIF-
1alpha stimulates aromatase expression driven by 49. Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JM:
prostaglandin E2 in breast adipose stroma. Breast Cancer Res New users of metformin are at low risk of incident cancer: a
2013, 15:R30. cohort study among people with type 2 diabetes. Diabetes Care
2009, 32:1620-1625.
37. Subbaramaiah K, Brown KA, Zahid H, Balmus G, Weiss RS,
Herbert BS, Dannenberg AJ: Hsp90 and PKM2 drive the 50. Currie CJ, Poole CD, Gale EA: The influence of glucose-
expression of aromatase in Li–Fraumeni syndrome breast lowering therapies on cancer risk in type 2 diabetes.
adipose stromal cells. J Biol Chem 2016, 291:16011-16023. Diabetologia 2009, 52:1766-1777.
38. Wang X, Docanto MM, Sasano H, Cancer KCFCfRiFB, Lo C, 51. Bodmer M, Meier C, Krahenbuhl S, Jick SS, Meier CR: Long-term
 Simpson ER, Brown KA: Prostaglandin E2 inhibits p53 in human metformin use is associated with decreased risk of breast
breast adipose stromal cells: a novel mechanism for the cancer. Diabetes Care 2010, 33:1304-1308.
regulation of aromatase in obesity and breast cancer. Cancer
Res 2015. 52. Bosco JL, Antonsen S, Sorensen HT, Pedersen L, Lash TL:
Using human breast adipose stromal cells, the authors demonstrated that Metformin and incident breast cancer among diabetic women:
p53 is a repressor of aromatase expression and is negatively regulated by a population-based case-control study in Denmark. Cancer
PGE2. It therefore demonstrates a novel tumor-suppressor role for p53. In Epidemiol Biomark Prevent 2011, 20:101-111.
Li–Fraumeni patients who have a germline mutation in TP53, aromatase is
increased in ASCs, providing a rationale for the increased risk of estro- 53. Ruiter R, Visser LE, van Herk-Sukel MP, Coebergh JW, Haak HR,
gen-dependent breast tumors in these women. Geelhoed-Duijvestijn PH, Straus SM, Herings RM, Stricker BH:
Lower risk of cancer in patients on metformin in comparison
39. Eliassen AH, Colditz GA, Rosner B, Willett WC, Hankinson SE: with those on sulfonylurea derivatives: results from a large
Adult weight change and risk of postmenopausal breast population-based follow-up study. Diabetes Care 2012,
cancer. JAMA 2006, 296:193-201. 35:119-124.
40. Prentice RL, Caan B, Chlebowski RT, Patterson R, Kuller LH, 54. Chlebowski RT, McTiernan A, Wactawski-Wende J, Manson JE,
Ockene JK, Margolis KL, Limacher MC, Manson JE, Parker LM Aragaki AK, Rohan T, Ipp E, Kaklamani VG, Vitolins M, Wallace R
et al.: Low-fat dietary pattern and risk of invasive breast et al.: Diabetes, metformin, and breast cancer in
cancer: the Women’s Health Initiative Randomized Controlled postmenopausal women. J Clin Oncol 2012, 30:2844-2852.
Dietary Modification Trial. JAMA 2006, 295:629-642.
55. Vissers PA, Cardwell CR, van de Poll-Franse LV, Young IS,
41. Chlebowski RT, Blackburn GL, Thomson CA, Nixon DW, Pouwer F, Murray LJ: The association between glucose-
Shapiro A, Hoy MK, Goodman MT, Giuliano AE, Karanja N, lowering drug use and mortality among breast cancer patients
McAndrew P et al.: Dietary fat reduction and breast cancer with type 2 diabetes. Breast Cancer Res Treat 2015,
outcome: interim efficacy results from the Women’s 150:427-437.
Intervention Nutrition Study. J Natl Cancer Inst 2006,
98:1767-1776. 56. Dowling RJ, Niraula S, Chang MC, Done SJ, Ennis M,
McCready DR, Leong WL, Escallon JM, Reedijk M, Goodwin PJ
42. Scott E, Daley AJ, Doll H, Woodroofe N, Coleman RE, Mutrie N, et al.: Changes in insulin receptor signaling underlie
Crank H, Powers HJ, Saxton JM: Effects of an exercise and neoadjuvant metformin administration in breast cancer: a
hypocaloric healthy eating program on biomarkers associated prospective window of opportunity neoadjuvant study. Breast
with long-term prognosis after early-stage breast cancer: a Cancer Res 2015, 17:32.
randomized controlled trial. Cancer Causes Control 2013,
24:181-191. 57. Zhao W, Li A, Feng X, Hou T, Liu K, Liu B, Zhang N: Metformin and
resveratrol ameliorate muscle insulin resistance through
43. Swisher AK, Abraham J, Bonner D, Gilleland D, Hobbs G, Kurian S, preventing lipolysis and inflammation in hypoxic adipose
Yanosik MA, Vona-Davis L: Exercise and dietary advice tissue. Cell Signal 2016, 28:1401-1411.
intervention for survivors of triple-negative breast
cancer: effects on body fat, physical function, quality 58. Frendo-Cumbo S, MacPherson RE, Wright DC: Beneficial effects
of life, and adipokine profile. Support Care Cancer 2015, of combined resveratrol and metformin therapy in treating
23:2995-3003. diet-induced insulin resistance. Physiol Rep 2016, 4.
44. Campbell KL, Foster-Schubert KE, Alfano CM, Wang CC, 59. Lu CH, Hung YJ, Hsieh PS: Additional effect of metformin and
Wang CY, Duggan CR: Reduced-calorie dietary weight loss, celecoxib against lipid dysregulation and adipose tissue
exercise, and sex hormones in postmenopausal women: inflammation in high-fat fed rats with insulin resistance and
randomized controlled trial. J Clin Oncol 2012:30. fatty liver. Eur J Pharmacol 2016, 789:60-67.
45. McTiernan A, Tworoger SS, Ulrich CM, Yasui Y, Irwin ML, 60. Zulian A, Cancello R, Girola A, Gilardini L, Alberti L, Croci M,
Rajan KB: Effect of exercise on serum estrogens in Micheletto G, Danelli P, Invitti C: In vitro and in vivo effects of
postmenopausal women: a 12-month randomized clinical trial. metformin on human adipose tissue adiponectin. Obes Facts
Cancer Res 2004:64. 2011, 4:27-33.

www.sciencedirect.com Current Opinion in Pharmacology 2016, 31:90–96


96 Endocrine and metabolic diseases

61. Brown KA, Hunger NI, Docanto M, Simpson ER: Metformin pioglitazone/metformin fixed combination in comparison with
inhibits aromatase expression in human breast adipose a combination of metformin with glimepiride on diabetic
stromal cells via stimulation of AMP-activated protein kinase. dyslipidemia. Diabetes Technol Ther 2011, 13:637-643.
Breast Cancer Res Treat 2010, 123:591-596.
65. Spencer M, Yang L, Adu A, Finlin BS, Zhu B, Shipp LR, Rasouli N,
62. Samarajeewa NU, Ham S, Yang F, Simpson ER, Brown KA: Peterson CA, Kern PA: Pioglitazone treatment reduces adipose
Promoter-specific effects of metformin on aromatase tissue inflammation through reduction of mast cell and
transcript expression. Steroids 2011, 76:768-771. macrophage number and by improving vascularity. PLOS ONE
2014, 9:e102190.
63. Hartemann-Heurtier A, Halbron M, Golmard JL, Jacqueminet S,
Bastard JP, Rouault C, Ayed A, Pieroni L, Clement K, Grimaldi A: 66. Subbaramaiah K, Howe LR, Zhou XK, Yang P, Hudis CA,
Effects of bed-time insulin versus pioglitazone on abdominal Kopelovich L, Dannenberg AJ: Pioglitazone, a PPARgamma
fat accumulation, inflammation and gene expression in agonist, suppresses CYP19 transcription: evidence for
adipose tissue in patients with type 2 diabetes. Diabetes Res involvement of 15-hydroxyprostaglandin dehydrogenase and
Clin Pract 2009, 86:37-43. BRCA1. Cancer Prev Res (Phila) 2012, 5:1183-1194.
64. Pfutzner A, Schondorf T, Tschope D, Lobmann R, Merke J,
Muller J, Lehmann U, Fuchs W, Forst T: PIOfix-study: effects of

Current Opinion in Pharmacology 2016, 31:90–96 www.sciencedirect.com

You might also like