cytokine & growth factor reviews...role of adipokines and cytokines in obesity-associated breast...

11
Mini review Role of adipokines and cytokines in obesity-associated breast cancer: Therapeutic targets Sajid Khan, Samriddhi Shukla, Sonam Sinha, Syed Musthapa Meeran * Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India 1. Introduction Breast cancer is the most common type of cancer and also the leading cause of cancer-related deaths in women, worldwide. According to GLOBOCAN 2008, breast cancer accounted for 23% (1.38 million) of the total new cancer cases and 14% (458,000) of total cancer-related deaths in the year 2008 [1]. Several reproductive and lifestyle factors are associated with the development of breast cancer. Among the reproductive factors are long menstrual history, nulliparity, increased use of oral contraceptives, and giving birth to a child at later age [2]. Lifestyle factors including less physical activity, consumption of high calorie diets, cigarette smoking and alcohol consumption are strongly associated with increased risk of breast cancer development [3,4]. Obesity is an abnormal or excessive fat accumulation in adipose tissue which leads to impaired health [5]. According to World Health Organization (WHO), obesity is defined as having a body mass index (BMI) of equal to or higher than 30 kg/m 2 . It is the major cause of onset of a number of diseases such as type-2 diabetes, cardiovascular diseases (CVDs), infertility, and several types of cancers [6]. It is estimated that 25–30% of cancers at numerous sites such as esophagus, pancreas, colorectum, endo- metrium, kidney and postmenopausal breast are caused by obesity and physical inactivity [7,8]. Obesity increases the risk of breast cancer by 30% in postmenopausal women and accounts for 21% of all breast cancer deaths, worldwide [9–11]. Obesity is also associated with worse prognosis and poor treatment outcome in cancer [12,13]. In postmenopausal women, estrogen is a major risk factor for breast cancer development. In general, estrogen is synthesized by Cytokine & Growth Factor Reviews 24 (2013) 503–513 A R T I C L E I N F O Article history: Available online 21 October 2013 Keywords: Obesity Breast cancer Leptin Adiponectin Cytokines A B S T R A C T Obesity is the cause of a large proportion of breast cancer incidences and mortality in post-menopausal women. In obese people, elevated levels of various growth factors such as insulin and insulin-like growth factors (IGFs) are found. Elevated insulin level leads to increased secretion of estrogen by binding to the circulating sex hormone binding globulin (SHBG). The increased estrogen-mediated downstream signaling favors breast carcinogenesis. Obesity leads to altered expression profiles of various adipokines and cytokines including leptin, adiponectin, IL-6, TNF-a and IL-1b. The increased levels of leptin and decreased adiponectin secretion are directly associated with breast cancer development. Increased levels of pro-inflammatory cytokines within the tumor microenvironment promote tumor development. Efficacy of available breast cancer drugs against obesity-associated breast cancer is yet to be confirmed. In this review, we will discuss different adipokine- and cytokine-mediated molecular signaling pathways involved in obesity-associated breast cancer, available therapeutic strategies and potential therapeutic targets for obesity-associated breast cancer. ß 2013 Elsevier Ltd. All rights reserved. Abbreviations: IGFs, insulin-like growth factors; SHBG, sex hormone binding globulin; BMI, body mass index; IGFBPs, IGF binding proteins; IGF-1R, insulin- like growth factor-1 receptor; CLS, crown-like structure; NFkB, nuclear factor kappa-B; STAT3, signal transducer and activator of transcription-3; COX-2, cyclooxygenase-2; TNF-a, tumor necrosis factor-a; ILs, interleukins; IFNs, interferons; JNK, c-Jun N-terminal kinase; SOCS3, suppressor of cytokine signaling-3; JAK2, Janus kinase-2; MAPK, mitogen-activated protein kinase; VEGF, vascular endothelial growth factor; VEGF-R2, vascular endothelial growth factor receptor-2; ERa, estrogen receptor-a; ERK, extracellular signal-regulated kinase; HER-2, human epidermal growth factor receptor-2; hTERT, human telomerase reverse transcriptase; PARP, poly (ADP-ribose) polymerase; AMPK, AMP-activated protein kinase; LDL, low-density lipoprotein; PDGF-BB, platelet-derived growth factor subunit B homodimer; bFGF, basic fibroblast growth factor; HB-EGF, heparin- binding epidermal growth factor-like growth factor; GSK-3b, glycogen synthase kinase-3b; HIF-1a, hypoxia-inducible factor-1a; C/EBPa, CCAAT/enhancer binding protein-a; PFS, progression-free survival; CDK, cyclin-dependent kinase; PEG- LPrA2, pegylated leptin peptide receptor antagonist 2; mTOR, mammalian target of rapamycin; EGCG, epigallocatechin-3-gallate; NSAIDs, non-steroidal anti-inflam- matory drugs; PPARa, peroxisome proliferator-activated receptor-a. * Corresponding author at: Division of Endocrinology, CSIR-Central Drug Research Institute (CSIR-CDRI), Jankipuram Extn., Sector-10, Sitapur Road, Lucknow 226 031, India. Tel.: +91 522 2612411x4491; fax: +91 522 2623938. E-mail addresses: [email protected], [email protected] (S.M. Meeran). Contents lists available at ScienceDirect Cytokine & Growth Factor Reviews jo ur n al ho mep ag e: www .elsevier .c om /loc ate/c yto g fr 1359-6101/$ see front matter ß 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.cytogfr.2013.10.001

Upload: others

Post on 26-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Cytokine & Growth Factor Reviews 24 (2013) 503–513

Mini review

Role of adipokines and cytokines in obesity-associated breast cancer:Therapeutic targets

Sajid Khan, Samriddhi Shukla, Sonam Sinha, Syed Musthapa Meeran *

Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India

A R T I C L E I N F O

Article history:

Available online 21 October 2013

Keywords:

Obesity

Breast cancer

Leptin

Adiponectin

Cytokines

A B S T R A C T

Obesity is the cause of a large proportion of breast cancer incidences and mortality in post-menopausal

women. In obese people, elevated levels of various growth factors such as insulin and insulin-like growth

factors (IGFs) are found. Elevated insulin level leads to increased secretion of estrogen by binding to the

circulating sex hormone binding globulin (SHBG). The increased estrogen-mediated downstream

signaling favors breast carcinogenesis. Obesity leads to altered expression profiles of various adipokines

and cytokines including leptin, adiponectin, IL-6, TNF-a and IL-1b. The increased levels of leptin and

decreased adiponectin secretion are directly associated with breast cancer development. Increased

levels of pro-inflammatory cytokines within the tumor microenvironment promote tumor development.

Efficacy of available breast cancer drugs against obesity-associated breast cancer is yet to be confirmed.

In this review, we will discuss different adipokine- and cytokine-mediated molecular signaling pathways

involved in obesity-associated breast cancer, available therapeutic strategies and potential therapeutic

targets for obesity-associated breast cancer.

� 2013 Elsevier Ltd. All rights reserved.

Contents lists available at ScienceDirect

Cytokine & Growth Factor Reviews

jo ur n al ho mep ag e: www .e lsev ier . c om / loc ate /c yto g f r

1. Introduction

Breast cancer is the most common type of cancer and also theleading cause of cancer-related deaths in women, worldwide.

Abbreviations: IGFs, insulin-like growth factors; SHBG, sex hormone binding

globulin; BMI, body mass index; IGFBPs, IGF binding proteins; IGF-1R, insulin-

like growth factor-1 receptor; CLS, crown-like structure; NFkB, nuclear factor

kappa-B; STAT3, signal transducer and activator of transcription-3; COX-2,

cyclooxygenase-2; TNF-a, tumor necrosis factor-a; ILs, interleukins; IFNs,

interferons; JNK, c-Jun N-terminal kinase; SOCS3, suppressor of cytokine

signaling-3; JAK2, Janus kinase-2; MAPK, mitogen-activated protein kinase; VEGF,

vascular endothelial growth factor; VEGF-R2, vascular endothelial growth factor

receptor-2; ERa, estrogen receptor-a; ERK, extracellular signal-regulated kinase;

HER-2, human epidermal growth factor receptor-2; hTERT, human telomerase

reverse transcriptase; PARP, poly (ADP-ribose) polymerase; AMPK, AMP-activated

protein kinase; LDL, low-density lipoprotein; PDGF-BB, platelet-derived growth

factor subunit B homodimer; bFGF, basic fibroblast growth factor; HB-EGF, heparin-

binding epidermal growth factor-like growth factor; GSK-3b, glycogen synthase

kinase-3b; HIF-1a, hypoxia-inducible factor-1a; C/EBPa, CCAAT/enhancer binding

protein-a; PFS, progression-free survival; CDK, cyclin-dependent kinase; PEG-

LPrA2, pegylated leptin peptide receptor antagonist 2; mTOR, mammalian target of

rapamycin; EGCG, epigallocatechin-3-gallate; NSAIDs, non-steroidal anti-inflam-

matory drugs; PPARa, peroxisome proliferator-activated receptor-a.

* Corresponding author at: Division of Endocrinology, CSIR-Central Drug

Research Institute (CSIR-CDRI), Jankipuram Extn., Sector-10, Sitapur Road, Lucknow

226 031, India. Tel.: +91 522 2612411x4491; fax: +91 522 2623938.

E-mail addresses: [email protected], [email protected]

(S.M. Meeran).

1359-6101/$ – see front matter � 2013 Elsevier Ltd. All rights reserved.

http://dx.doi.org/10.1016/j.cytogfr.2013.10.001

According to GLOBOCAN 2008, breast cancer accounted for 23%(1.38 million) of the total new cancer cases and 14% (458,000) oftotal cancer-related deaths in the year 2008 [1]. Severalreproductive and lifestyle factors are associated with thedevelopment of breast cancer. Among the reproductive factorsare long menstrual history, nulliparity, increased use of oralcontraceptives, and giving birth to a child at later age [2]. Lifestylefactors including less physical activity, consumption of high caloriediets, cigarette smoking and alcohol consumption are stronglyassociated with increased risk of breast cancer development [3,4].

Obesity is an abnormal or excessive fat accumulation in adiposetissue which leads to impaired health [5]. According to WorldHealth Organization (WHO), obesity is defined as having a bodymass index (BMI) of equal to or higher than 30 kg/m2. It is themajor cause of onset of a number of diseases such as type-2diabetes, cardiovascular diseases (CVDs), infertility, and severaltypes of cancers [6]. It is estimated that 25–30% of cancers atnumerous sites such as esophagus, pancreas, colorectum, endo-metrium, kidney and postmenopausal breast are caused by obesityand physical inactivity [7,8]. Obesity increases the risk of breastcancer by 30% in postmenopausal women and accounts for 21% ofall breast cancer deaths, worldwide [9–11]. Obesity is alsoassociated with worse prognosis and poor treatment outcome incancer [12,13].

In postmenopausal women, estrogen is a major risk factor forbreast cancer development. In general, estrogen is synthesized by

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513504

sexual organs, whereas in obese postmenopausal women adiposetissue is the main source of estrogen synthesis [11]. Obesity leadsto altered expression of hormones, growth factors, inflammatorycytokines and adipokines which promote cancer cell survival,metastasis, angiogenesis, and decreased cancer cell apoptosis. Thedetailed molecular mechanisms through which obesity promotesbreast cancer development are discussed in the next section.

2. Factors involved in obesity-associated breast cancer andtheir mechanisms

2.1. Hormones and growth factors

Obesity is accompanied by increased estrogen synthesis fromadipose-tissue associated stromal cells and elevated levels ofinsulin and insulin-like growth factors (IGFs) [14–16]. This rise ininsulin level is found to be associated with the activation of IGFsystem and altered levels of IGF binding protein-1 (IGFBP1) and -2(IGFBP2) which leads to increased bioavailability of IGFs. IGFBPsstabilize and prolong the half life of IGFs and prevent their bindingto IGF receptors [17]. IGFBPs also influence the duration ofsignaling via the IGF receptor by slow release of IGF to its receptor.Further, IGFs lead to macrophage migration and invasion andincreased production of pro-inflammatory cytokines by macro-phages [18,19]. In chronic hyperinsulinemia, a decreased level ofcirculating sex-hormone binding globulin (SHBG) leads toincreased bio-available estrogen levels which promote mammarytumorigenesis [14].

Insulin, IGFs and insulin like growth factor-1 receptor (IGF-1R)are over expressed in several subtypes of breast cancer [20]. Thebinding of these ligands to IGF-1R leads to activation of its tyrosinekinase activity [21]. Activation of IGF-1R can promote cellmigration and redistribution of E-cadherin and, a- and b-cateninsfrom adherens junctions into the cytoplasm and promote breasttumorigenesis [22]. In addition, IGF-1R leads to activation of PI3K/Akt and Ras-raf-MAPK signaling which alter the expression ofgenes involved in cellular proliferation and survival [23]. Overall,this altered hormonal and growth factor profile is associated withincreased breast cancer risk.

2.2. Inflammation and inflammatory cytokines

Obesity causes subclinical inflammation in both visceral as wellas subcutaneous adipose tissue. This inflammation is characterizedby necrotic adipocytes surrounded by macrophages which arevisualized as crown-like structures (CLS) under light microscope[24–26]. This subclinical inflammation might increases the risk ofbreast cancer.

The adipose tissue-derived factors activate key inflammatorymolecules such as nuclear factor kappa-B (NFkB) and signaltransducer and activator of transcription-3 (STAT3). Activation ofNFkB in adipose tissue further induces the expression of severalpro-inflammatory mediators like cyclooxygenase-2 (COX-2),tumor necrosis factor-a (TNF-a), and interleukin-1b (IL-1b),which in turn induces aromatase expression and activity.Activation of these inflammatory mediators leads to alteredexpression of genes involved in breast carcinogenesis [27–29].

Cytokines, including TNF-a, interleukin-6 (IL-6) and interferons(IFNs) have been reported to be associated with breast cancerdevelopment as indicated by their presence within tumormicroenvironment and in the tumor metastatic sites [30]. TNF-a regulates IL-6 synthesis and the expression of aromatase inadipose tissue [31]. Conditional media from preadipocyte-derivedadipocytes was found to increase the proliferation of breastcancer cells, possibly due to the presence of IL-6 from adipocytes[32]. TNF-a treatment in adipocytes drastically decreases the

adiponectin expression and secretion through insulin-like growthfactor binding protein-3 (IGFBP-3) and c-Jun N-terminal kinase(JNK) cascades [33,34].

2.3. Adipokines

Adipokines are small peptide hormonal growth factors whichare secreted mainly by adipocytes from white adipose tissue. Theseare the major contributing factors for obesity associated breastcancer [35]. The two most important adipokines which areassociated with breast cancer development are leptin andadiponectin.

2.3.1. Leptin

Leptin, a multifunctional neuroendocrine peptide hormone,plays a key role in satiety, energy expenditure, food intake, andvarious reproductive processes [36,37]. Leptin is encoded by obese

(ob) gene which is located on chromosome 7, in humans [38]. Itconsists of 167 amino acids and has a molecular weight of 16 kDa[39]. The molecular actions of leptin are mediated through the cellsurface receptors which are members of cytokine family ofreceptors and are present in various tissues [40]. Six isoforms ofleptin receptors ranging from Ob-Ra to Ob-Rf have been identified.Ob-Rb is a long isoform of leptin receptor as it contains a longintracellular domain, comprising of approximately 306 aminoacids. The other isoforms (Ob-Ra, Ob-Rc, Ob-Rd and Ob-Rf) aremore abundant in peripheral tissues and contain a shortintracellular domain consisting of 23 amino acids [41]. Ob-Re isa soluble leptin receptor, and has been shown to control circulatingleptin levels [42].

Adipose tissue is the main source of leptin secretion. Inaddition, some amount of leptin is also secreted from normal andmalignant breast tissue, placenta, stomach and skeletal muscle.The level of leptin increases in proportion to BMI. Serum leptinlevels in obese individuals are higher due to its increased releasefrom adipocytes [43]. Leptin could increase or decrease the risk ofbreast cancer depending on the menopausal status. Plasma leptinlevel increases the risk of breast cancer in postmenopausal women,whereas its level is inversely related to breast cancer risk inpremenopausal women [44]. Leptin and its receptors are overexpressed in breast tumors and are associated with distantmetastasis [45,46]. Genetically obese leptin-deficient LepobLepob

and leptin receptor-deficient LeprdbLeprdb female mice do notdevelop mammary tumors which provide supporting evidencethat leptin and its receptor is involved in breast tumorigenesis[47,48].

The long form of leptin receptor which is mainly expressed inthe hypothalamus acts through the activation of JAK2/STAT3 andMAPK pathways, whereas short isoforms activate mainly MAPkinases and appear to be responsible for mitogenic activity [49].Through activation of JAK2/STAT3 pathway, leptin induces theexpression of c-MYC and consequently leads to increased cellsurvival and proliferation [50]. Another downstream target of JAK/STAT signaling, cyclin D1 which promotes G1 to S-phase transitionduring cell cycle progression is found to be increased due toincreased JAK/STAT signaling mediated by leptin [51]. Suppressorof cytokine signaling-3 (SOCS3) negatively regulates leptin-mediated activation of JAK2/STAT3 signaling by binding tophosphorylated JAK2 proteins [52,53]. Recently, SOCS3 has beenreported to down regulate the expression of anti-apoptotic proteinsurvivin through binding with long isoform of leptin receptor (Ob-Rb) and thus inhibiting leptin signaling through JAK/STAT pathway[54].

In a recent study, leptin has been shown to be involved in theregulation of endothelial cell proliferation and in the promotion ofangiogenesis [55]. Leptin increases endothelial COX-2 expression

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513 505

through p38 MAPK and PI3K/Akt activated mechanisms [56]. Inmouse mammary cancer cells, leptin may promote angiogenesisthrough vascular endothelial growth factor (VEGF) signaling, asleptin treatment results in increased expression of genes for VEGFand VEGF receptor-2 (VEGF-R2) in these cells [57].

Leptin promotes the synthesis of estrogens or converselyreduces follicular estradiol secretion and thus may influencebreast cancer risk [58,59]. Leptin exerts its effects by increasingcell viability and proliferation through crosstalk with estrogenreceptor-alpha (ERa). The expression of leptin receptor (Ob-R)and its signaling correlates with the presence of ERa in humanbreast cancer cell lines [60]. Leptin receptor expression was alsofound to be positively correlated with ERa expression in primarybreast carcinoma, which further illustrates the positive associa-tion between estrogen and leptin systems in the development ofbreast cancer in human. In a study on breast cancer patients, theserum leptin level was found to be higher in tamoxifen-treatedpatients as compared to controls [61,62]. The possible explana-tion for this is that the binding of tamoxifen reduces theexpression of ER which results in concomitant decrease in theleptin receptor expression and ultimately an increased serumleptin level.

Leptin induces the functional activation of estrogen receptors inMCF-7 cells via the extracellular signal-regulated kinase 1 and 2(ERK1/ERK2) signal transduction pathway [63]. The antiestrogeniceffects of ICI 182,780 on MCF-7 cells have been shown to be ceasedby simultaneous treatment with leptin [64]. This study furtherproved that leptin results in ER activation which neglects theeffects of antiestrogenic compound by an unknown mechanism. Ina preclinical tumor model for breast cancer, tamoxifen andletrozole did not affect leptin level in the serum [65]. In anotherstudy, treatment with exemestane caused 27% decrease in plasmaleptin levels following about 3 months of treatment in postmeno-pausal breast cancer patients [66]. This differential effect might bedue to menopausal status or time duration after which plasmaleptin level was measured. Leptin can also transactivate HER-2 inbreast cancer cells through both the HER-1 and JAK-2 pathways[67]. In a large number of breast tumors, leptin and its receptors arecoexpressed with HER-2, which supports the possibility ofintratumoral ob-R/HER-2 interactions. The circulating levels ofleptin are directly correlated with human telomerase reverse

transcriptase (hTERT) expression which is highly expressed inalmost all cancer types including breast cancer [68,69]. Thus, leptinis known to be a proliferative, self-renewal and survival factor inobesity-associated breast cancer.

Polymorphism of leptin (LEP) and leptin receptor (Ob-R) geneswas found to be associated with risk of developing breast cancer inobese women. LEP-2548G/A and LEPR Q223R polymorphisms maybe related to obesity as well as enhanced gene expression andincreased circulating levels of leptin [70–72]. The presence of LEP-

2548A/A and LEP-2548G/A in breast cancer cells was found to beassociated with high and intermediate leptin mRNA expression,respectively, while cells containing LEP-2548G/G expressed lowleptin mRNA levels. The presence of LEP-2548G/A facilitatesefficient recruitment of transcription factor specificity protein 1(Sp1) to leptin promoter DNA under insulin treatment, while Sp1loading on DNA containing LEP-2548G/G was not insulin-depen-dent. In contrast, the binding of nucleolin, a transcriptionalrepressor to LEP-2548G/A was downregulated in response toinsulin, while it was not regulated on LEP-2548G/G [73]. Thus, thepresence of LEP-2548G/A might enhance leptin expression in breastcancer cells via Sp1- and nucleolin-dependent mechanisms andthis enhanced leptin expression is associated with breast cancerrisk. In a case–control study, a modest increase in risk ofdeveloping breast cancer was found to be associated with LEP-

2548A/A genotype compared to the LEP-2548G/G genotype and the

association was stronger among the postmenopausal women whowere obese [74].

2.3.2. Adiponectin

Another adipokine involved in obesity-associated breast canceris adiponectin, which is mainly secreted by adipocytes and someamount is also secreted by other types of cells [75,76]. It has amolecular weight of 28–30 kDa. Several isoforms of adiponectinhave been identified. Amongst them, full length adiponectinknown as Acrp30 and globular adiponectin known as gAcrp30 areimportant because of their strong and identified affinity withadiponectin receptors [77]. There are mainly two adiponectinreceptors (AdipoRs), AdipoR1 and AdipoR2. Of these, AdipoR1 hashigh affinity for gAcrp30 and AdipoR2 is the intermediate affinityreceptor for gAcrp30 and Acrp30 [75,78]. The altered expression ofadiponectin receptors have been reported in most of the breastcancer cell lines such as MDA-MB-231, MCF-7, T47D and SK-BR-3.The variation is found in the expression levels of AdipoR1 andAdipoR2 in breast cancer cell lines. MDA-MB-231, T47D, and MCF-7 cells showed higher expression of AdipoR1 and lower expressionof AdipoR2, whereas MDA-MB-361 cells showed higher expressionof AdipoR2 [79–81].

Several retrospective and prospective case–control studies haveproved the association of low serum adiponectin levels withincreased risk of breast cancer. There is a controversy on whetherlow adiponectin level increases the risk of breast cancer amongboth premenopausal and postmenopausal women or it isassociated with increased risk only in postmenopausal women.Some studies have reported that there is a well defined inverseassociation between adiponectin and breast cancer risk in bothpre- and post-menopausal women. In contrast, some other studieshave reported that lower adiponectin levels are associated withbreast cancer risk only in postmenopausal women [82–87]. Thesedifferences might be due to female sex hormones especially theestrogens since serum adiponectin concentration is inverselycorrelated with serum estradiol concentration in postmenopausalwomen but not in premenopausal women [88].

Addition of full length adiponectin, i.e. Acrp30 to ER-positivecell lines (MCF-7, T47D, and MDA-MB-361) results in increasedapoptosis by increased poly (ADP-ribose) polymerase (PARP)cleavage. Increase in cleaved caspase-8 on adiponectin treatmentwas also found in MDA-MB-231 and MCF-7 cells leading toapoptosis. The treatment of MDA-ERa7 cell line (produced bytransfection of ERa gene into MDA-MB-231 cell line) withtruncated form of adiponectin, i.e. gAcrp30 reduces its growthprobably by decreasing phosphorylation of JNK2 [79]. This studyshows that ER expression is important for signaling throughadiponectin receptors and consequently cell growth inhibition byadiponectin treatment. Estrogen has been found to suppressadiponectin expression in 3T3-L1 adipocyte cell line [89]. Thissuggests that estrogen might be an important factor in obesity-induced breast carcinogenesis through decreased adiponectinexpression in adipocytes of adipose tissue.

The anti-proliferative and pro-apoptotic effects of adiponectinmight be mediated by its binding to AdipoRs, and concomitantactivation of AMP-activated protein kinase (AMPK), and theinactivation of p42/p44 MAPK [90]. Adiponectin mediated activa-tion of AMPK is involved in anti-breast cancer activities throughregulation of PI3K/Akt pathway [91]. In MDA-MB-231 cells,adiponectin-activated AMPK reduces cell invasion by inducingAKT dephosphorylation through protein phosphatase-2 activity[92]. Adiponectin may also mediate its effects by regulating theexpression of different tumor suppressor genes, oncogenes, pro-and anti-apoptotic genes, and cell cycle regulatory genes includingp53, Bax, Bcl-2, c-myc, cyclin D1, MAPK3 and ataxia telangiectasia

mutated (ATM) [80,90]. Adiponectin significantly inhibits the cell

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513506

proliferation induced by leptin, oxidized low-density lipoprotein(LDL) and several other growth factors like platelet-derived growthfactor subunit B homodimer (PDGF-BB), basic fibroblast growthfactor (bFGF), and heparin-binding epidermal growth factor-likegrowth factor (HB-EGF) [93–95]. Adiponectin inhibits the serum-induced phosphorylation of AKT and glycogen synthase kinase-3b(GSK-3b) and suppresses the intracellular accumulation of b-catenin in MDA-MB-231 and T47D breast cancer cell lines. Thisshows that GSK-3b/b-catenin signaling pathway is involved inadiponectin-mediated growth inhibition in breast cancer cell lines[95]. Thus, adiponectin acts as anti-proliferative, pro-apoptotic andinhibitor of angiogenesis in breast cancer. Fig. 1 summarizes themain signaling pathways mediated by estrogen, leptin andadiponectin which are involved in the process of breastcarcinogenesis.

2.4. Tumor microenvironment

The tumor microenvironment comprises of cells, solublefactors, signaling molecules, and extracellular matrix that canpromote tumorigenesis, and resist the tumor from host immunityand therapeutic response. Obesity reduces the oxygen level in thetumor microenvironment leading to hypoxic condition [97].Hypoxia in the peri-tumoral fat is reported to promote tumorsite hypoxia. The up regulation of hypoxia-inducible factor-a (HIF-1a) takes place in the hypoxic state which leads to alteredexpression of several genes involved in angiogenesis, cellproliferation and apoptosis which ultimately results in cellular

Fig. 1. Estrogen, leptin and adiponectin signaling in breast carcinogenesis. The signaling

leptin and adiponectin) is shown here. Estrogen binding to estrogen receptor-a (ERa) in

nucleus and binds to estrogen responsive elements (EREs) at the promoter region of some

adipose tissue is directly correlated with the breast cancer progression. Leptin binds to it

JAK2/STAT3 pathway, PI3K/AKT pathway, and MAPK pathway (here we shown only JAK2

binds to cytoplasmic domains of leptin receptor and causes their phosphorylation which

and the dimerized STAT3 binds at the promoter region of some tumor promoting genes (e

genes (e.g. survivin) and leads to their expression. Leptin has its actions not only through

increases the expression of aromatase and thus causes increased estrogen synthesis. In th

dependent pathway [96]. The low level of adiponectin results in abrogated adiponectin s

end result of all of these signaling pathways is increased cell proliferation and surviva

adaptation to low oxygen concentration [98]. In the adipose tissue,hypoxia induces the secretion of pro-angiogenic and inflammatorycytokines [99,100]. Hypoxia down regulates the expression ofCCAAT/enhancer binding protein-a (C/EBPa) in T-47D breastcancer cells through binding of HIF-1a to C/EBPa promoter site[101]. C/EBPa is a transcription factor which induces apoptosis,inhibits cell proliferation and involved in cellular differentiation[102].

3. Current clinical strategies and trials on obesity-associatedbreast cancer

3.1. Hormones- and growth factors-targeted

Despite the significant heterogeneity, several advances havebeen made over the past decade for the care of patients with breastcancer. Available treatments for breast cancer include chemother-apy, radiotherapy, hormonal and targeted therapy. Anthracyclines(e.g. doxorubicin) and taxanes (e.g. docetaxel) are two mostcommonly used chemotherapeutic agents for the treatment ofbreast cancer. These days, targeted and hormonal therapies areshowing promising results in the prognosis of breast cancer.Agents used in targeted therapy include inhibitors of receptortyrosine kinases (epidermal growth factor family inhibitors, e.g.gefitinib and erlotinib) and nonreceptor tyrosine kinases, inhibi-tors of intracellular signaling pathways such as PI3K/Akt and Ras-Raf MAPK, angiogenesis inhibitors, agents that interfere with DNArepair (e.g. PARP inhibitors), and hormonal antagonists. The use of

through three major players involved in obesity-associated breast cancer (estrogen,

the cytoplasm leads to ERa dimerization. This dimerized ERa passes through the

tumor promoting genes and leads to their expression. The high level of leptin in the

s cell surface receptor and leads to activation of several oncogenic pathways such as

/STAT3 pathway, the major pathway activated by leptin). The phosphorylated JAK2

ultimately leads to phosphorylation of STAT3. The dimerization of STAT3 takes place

.g. c-myc, EGFR, and src), cell cycle promoting genes (e.g. Cyclin D1) or anti-apoptotic

leptin receptor but it also has crosstalk signaling with estrogen receptor-a. Leptin

e absence of estrogen, leptin also stimulates signaling through ERa through MAPK-

ignaling through activation of MAPK pathway and inhibition of AMPK pathway. The

l and/or decreased apoptosis and ultimately the induction of breast cancer.

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513 507

targeted therapies such as estrogen antagonist (e.g. tamoxifen andfulvestrant) in estrogen receptor (ER) positive cases and ofmonoclonal antibody trastazumab (Herceptin) for the inhibitionof signaling through HER-2 receptor have contributed significantlyto these advances [103]. Mammalian target of rapamycin (mTOR)inhibitors (e.g. everolimus) which target mTOR pathway is a newlyadded targeted therapy in breast cancer treatment [104].

Combination therapy has proven to be more effective approachthan the mono-agent therapy for breast cancer treatment.Targeting HER-2 with trastazumab and VEGF with bevacizumabin combination with chemotherapy is more effective than singleagent in the treatment of breast cancer [105]. The combination ofaromatase inhibitor (letrozole) and HER-2 inhibitor (trastazumab)is an effective therapy in hormonal-refractory breast cancer due tothe fact that the signaling through HER-2 increases in aromataseinhibitor resistant breast tumors [106,107]. Lapatinib, a dual-EGFR/HER-2 selective reversible inhibitor in combination withtrastazumab significantly improved the progression-free survival(PFS) in HER-2 over expressing breast cancer cells [105]. Thus, thiscombination of lapatinib and trastazumab can be effective in thetreatment of trastazumab-refractory metastatic breast cancer.

Reducing estrogen synthesis by targeting aromatase orinhibiting signaling through estrogen receptor could be atherapeutic option in obesity-associated breast cancer. Thearomatase inhibitors inhibit the estrogen synthesis and antiestro-gens inhibit and/or degrade estrogen receptor and consequentlyabrogate oncogenic signaling. Therefore, these two classes ofcompounds may be effective for treating obesity-related breastcancer. Tamoxifen shown to reduce serum IGF-I levels in obesewomen. Aromatase inhibitors such as letrozole reduce plasmaestrogen levels in obese postmenopausal women [108]. Thus, theseantiestrogens could be used to treat obesity-related breast canceras the levels of both IGF-I and estrogen were found to be increasedin obesity-associated breast cancer. Roscovitine is a selectivecyclin-dependent kinase (CDK) inhibitor which has found toreduce estrogen-induced phosphorylation of ERa at Ser118 inMCF-7 cells and thus inhibited signaling through ERa [109]. Theinhibition of signaling through JAK-2/STAT3 pathway by inhibitingSTAT3 or its upstream kinases may also be beneficial. Agents thatcould inhibit signaling through the insulin receptor might bepotential therapeutics in reducing insulin-mediated tumorgrowth.

FDA approved drugs such as phentermine, diethylpropion,phendimetrazine, orlistat and lorcaserin which are used in thetreatment of hyperlipidemia/hypercholesterolemia and obesitycan also be effective candidates for the treatment of obesity-related breast malignancies. Pitavastatin, a drug used for thetreatment of hyperlipidemia significantly reduced the develop-ment of diethylnitrosamine (DEN)-induced liver carcinogenesis in

Table 1Therapeutic agents in targeting hormones and growth factors for breast cancer treatm

Drug class Drug name Mode of action

Aromatase inhibitors Letrozole, anastrozole,

exemestane

Act through the inhib

aromatase, an enzym

the synthesis of estro

precursor hormones

Antiestrogens and estrogen

antagonists

Tamoxifen, fulvestrant Tamoxifen mainly ac

inhibition of ER. Fulve

through ER degradati

CDK-inhibitors Roscovitine Reduces phosphoryla

thereby inhibits dow

Anti-obesity and

anti-hyperlipidemia drugs

Orlistat, Pitavastatin Orlistat acts through

cycle progression, pro

apoptosis, and repres

db/db-obese mice. Orlistat has been found to inhibit growth ofbreast cancer cells through blockade of cell cycle progression,promotion of apoptosis, and repression of HER-2/neu [110,111].The therapeutic agents for the treatment of obesity-associatedbreast cancer targeting hormones and growth factors are listed inTable 1.

Dietary and physical interventions such as reduction in high-fatdiet and increased physical activity can reduce the risk of breastcancer in postmenopausal women. A balanced energy budget ofbody controlled by limitation of calorie intake and/or by elevationof energy expenditure could be key ways to reduce obesity [115].The activation of AMPK by bioactive food components may be animportant strategy for the prevention of several cancers causeddue to increased BMI. AMPK plays an important role in cellularenergy homeostasis through the stimulation of fatty acid oxida-tion, inhibition of lipogenesis and modulation of insulin secretionby pancreatic beta-cells [116]. Thus, the phytochemicals thatactivate AMPK may also be useful in reducing the risk of obesity-mediated breast cancer in postmenopausal women.

3.2. Adipokines- and cytokines-targeted

Inflammatory cytokines and adipokines are potent targets forthe treatment of obesity-associated breast malignancy. Animportant strategy for the treatment of this class of cancer isthe development of pharmacological compounds that targetobesity-related pathways. There are various compounds that arebeing tested in clinical trials for obesity [117]. These compoundscan also be useful to treat obesity-associated breast tumorigenesispossibly in combination with anti-breast cancer agents. Fig. 2illustrates the various therapeutic agents and their targets inobesity and obesity-associated carcinogenesis.

The antagonists of leptin that can inhibit signaling through theleptin receptor and, adiponectin agonists that can mimic the actionof adiponectin could also be used to inhibit the growth of epithelialbreast cells. Pegylated leptin peptide receptor antagonist 2 (PEG-LPrA2) reduced the growth of breast cancer cells particularly ER-positive cells in xenograft mice model by reducing the expressionof pro-angiogenic (e.g. VEGF/VEGFR-2) and pro-proliferative (e.g.proliferating cell nuclear antigen and cyclin D1) molecules [118].ADP-355 is a peptide-based adiponectin receptor agonist whichshows growth inhibitory effects on breast cancer cell lines as wellas on orthotopic xenograft breast cancer model [119].

Metformin, an anti-diabetic drug was found to reduce IL-6mRNA expression level and to enhance mRNA expression level ofIL-1R, the receptor for a naturally occurring anti-inflammatorycytokine. Action of metformin may be attributable to bemediated through the activation of AMPK pathway as silencingof AMPK-a1 results in lipopolysaccharide (LPS)-induced IL-6 and

ent.

Status References

ition of

e which catalyze

gen from its

Clinical phase III (with or without

chemotherapy) for invasive breast

cancer in postmenopausal women

[112]

ts through

strant mainly acts

on.

Tamoxifen is in clinical phase I/II for

triple negative breast cancer (in

combination with decitabine and

LBH589) Fulvestrant is in Clinical phase

III (for postmenopausal breast cancer)

[113,114]

tion of ERa,

nstream signaling

Preclinical [108]

blockade of cell

motion of

sion of HER2/neu

Preclinical [111,112]

Fig. 2. Schematic diagram illustrating therapeutic agents targeting different signaling molecules involved in obesity and obesity-associated breast cancer. In obesity, the

activity of aromatase enzyme increases which leads to increased production of estrogen (E2) by the adipose tissue. Estrogen signaling activates several downstream genes

such as cyclin D1 and c-myc. Aromatase inhibitors and antiestrogens can be used to target this pathway in obesity-associated breast cancer. Obesity also causes chronic

inflammation which leads to an increased expression and secretion of several inflammatory molecules such as TNF-a and IL-6. TNF-a promotes downstream NFkB pathway

in which NFkB binds to its response elements in the nucleus and increases the expression of its target genes. TNF-a further enhances IL-6 expression, IL-6 activates JAK2/

STAT3 signaling which leads to the expression of STAT3 target genes. In obesity, Wnt/b-catenin signaling leads to the activation of b-catenin target genes involved in breast

carcinogenesis. The activation of these oncogenic pathways through obesity-inflammation axis finally leads to breast carcinogenesis. Increased adiponectin activity activates

AMPK which causes fatty acid oxidation and leads to adipose tissue reduction. The therapeutic agents targeting these molecules are depicted in the figure. Abbreviations: E2,

estrogen; ER, estrogen receptor; ERE, estrogen response element; RE, response element; FASN, fatty acid synthase; PM, plasma membrane; Adn, adiponectin.

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513508

IL-8 expression [32]. Preclinical studies have demonstrated thatmetformin can inhibit the growth of breast cancer cells.Metformin mainly acts through the inhibition of signaling bymTOR pathway and leads to reduced expression of HER-2 proteinon breast cancer cells [31]. Thus, metformin may also showpromising results in the treatment of obesity-related breastcancer.

Table 2Synthetic molecules in targeting adipokines and inflammatory cytokines involved in o

Name of the compound Category Mode of action

Pegylated leptin peptide

receptor antagonist 2 (PEG-LPrA2)

Synthetic peptide Reduces the expres

angiogenic and pro-

ADP-355 Synthetic peptide Mimics the action

modulating key sig

an adiponectin-like

Aspirin Synthetic Anti-inflammatory

upregulation of pro

and downregulatio

proteins,

Celecoxib Synthetic Anti-inflammatory

Exemestane in combination

with celecoxib

Synthetic Aromatase inhibito

inhibitor

Infliximab Monoclonal antibody Anti-inflammatory

Metformin Synthetic Acts through either

of pro-inflammator

activation of AMPK

of mTOR pathway,

levels

Anti-inflammatory drugs which act through the inhibition ofkey inflammatory molecules such as IL-6, TNF-a and COX-2 canalso be used for treating obesity-associated breast cancer. Non-steroidal anti-inflammatory drugs (NSAIDs) such as aspirin couldprevent the occurrence of cancers of epithelial origin includingbreast cancer as evidenced by in vitro, in vivo and epidemiologicalstudies. Aspirin has been found to promote apoptotic pathways in

besity-associated breast cancer.

Target/pathway Status Refs.

sion of pro-

proliferative factors

Leptin Preclinical [118]

of adiponectin by

naling pathways in

manner

Adiponectin Preclinical [119]

acts through

-apoptotic proteins

n of anti-apoptotic

COX-1 and COX-2 Preclinical [31]

COX2 Phase II completed [120]

r with COX-2 Aromatase And COX-2 Phase II completed [122]

TNF-a Phase II completed [123]

reduced expression

y cytokines and/or

pathway, inhibition

lowering IGF-1

AMPK, IGF-1, mTOR Preclinical [31,32]

Table 3Phytochemicals in targeting adipokines and inflammatory cytokines involved in obesity-associated breast cancer.

Name of the

compound

Category Mode of action Target/pathway Status Refs.

Curcumin Polyphenol Anti-inflammatory, fatty acid oxidation NFkB, and COX-2, PPARg Preclinical [129]

(-)-Catechin Flavanol Increases expression of adiponectin Adiponectin Preclinical [146,147]

EGCG Flavanol Promotes fat oxidation and inhibit adipocyte

differentiation, decreases fat absorption

AMPK, pancreatic lipases Preclinical [143,144]

Genistein Isoflavonoid Inhibitor of adipocyte differentiation AMPK, Fatty acid synthase Preclinical [139–141]

Isoquercitrin Flavones Prevents preadipocyte differentiation into adipocyte Wnt/b-catenin Preclinical [148,149]

Myricetin Flavones Inhibits lipid droplet accumulation in adipocytes Peroxisome proliferator-activated

receptor-a (PPARa)

Preclinical [150]

Quercetin Flavone Anti-inflammatory, inhibits adipocyte cell growth COX-2, NFkB and TNF-a Phase I [136]

Resveratrol Polyphenol Anti-inflammatory, inhibits adipocyte differentiation IL-6, TNF-a and NFkB, AMPK, PPARg Preclinical [131]

Ursolic acid Triterpenoid Anti-inflammatory NFkB, and COX-2 Preclinical [127,137,138]

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513 509

cancer cell lines by up regulation of pro-apoptotic and downregulation of anti-apoptotic proteins [31]. There are several otherdrugs in clinical trials for the treatment of obesity and/or obesityassociated breast cancer targeting cytokines and inflammatorymediators, which are further listed in Table 2. Celecoxib, a COX-2inhibitor successfully completed phase II clinical trial for thetreatment of postmenopausal breast cancer [120]. Exemestane, anaromatase inhibitor in combination with avandamet (combinationof rosiglitazone and metformin) was found to be effective in phase Iclinical trial to treat breast cancer in obese postmenopausalwomen [121]. The combination of Exemestane and Celecoxib wastested in phase II clinical trial for the treatment of breast cancer inobese postmenopausal women and found to be effective [122].Infliximab, a monoclonal antibody targeting TNF-a completedphase II clinical trial for the treatment of cancer related fatigue inpatients who had undergone breast cancer treatment [123]. In apilot study, docosahexaenoic acid (DHA), an omega-3 fatty acid,reduced inflammation and aromatase expression in obesepostmenopausal women [124]. Further clinical studies on theefficacy of DHA against breast cancer are currently ongoing. Greentea extract is being tested in phase II clinical trial in healthypostmenopausal women [125]. (-)-Catechin, one of the compo-nents of green tea is in phase I clinical trial for treating women withhormone receptor-negative stages I–III breast cancer [126]. Theefficacy of these drugs could also be tested in obesity-associatedbreast cancer possibly in combination with anti-breast cancerdrugs.

Phytochemicals found in fruits, vegetables have also showntheir efficacy against obesity and obesity-associated cancers.Curcumin, phytochemical found in turmeric, is an anti-inflamma-tory agent and shown to modulates NFkB, Stat3, COX-2, Akt, andmTOR pathways [127,128]. By targeting these obesity-relatedpathways, curcumin reduces the symptoms of obesity such ashyperglycemia and hyperlipidemia and thus inhibits obesity-associated tumor growth [129,130]. Resveratrol, a dietary poly-phenol generally found in grapes, shown to inhibit inflammatorycytokines and chemokines [131–133]. Quercetin generally foundin citrus fruits, onions, tea and red wine possesses anti-inflammatory and anti-oxidant activities. It alters Akt/mTORpathway, COX-2 expression, NFkB signaling, TNF-a expression,and VEGF expression [134,135]. A phase I clinical study showedthat quercetin inhibits tumor growth and inflammation [136].Ursolic acid is a triterpenoid found in rose-mary, shown to be anti-inflammatory and insulin sensitizing in nature [127,137]. Ursolicacid also inhibits VEGF and other inflammatory growth factors inmice, thus it could also be an anti-angiogenic agent [138]. Hence,these anti-inflammatory and anti-angiogenic phytochemicalscould be designed to test their efficacy either alone or incombination with available therapeutic agents against obesity-associated breast cancer.

Dietary flavonoids such as genistein and (�)-catechin exertsanti-obesity effects either by promoting fatty acid oxidation or byinhibiting adipocyte differentiation [139–147]. Genistein is foundin soy (Glycine max) and soy food products. Genistein inhibitsadipocyte differentiation probably by promoting AMPK-activation[139,140]. Genistein was also found to inhibit fatty acid synthaseexpression and JAK2 phosphorylation which indirectly leads tosuppression of adipocyte differentiation [141]. EGCG and (�)-catechin are components of green tea and known as green teapolyphenols. EGCG was reported to have pancreatic lipaseinhibitory activity and was also found to inhibit adipocytedifferentiation by AMPK-activation [142–145]. Similar to EGCG,(-)-catechin also activates AMPK and inhibits adipocyte differenti-ation. In addition to this, (-)-catechin also increases the expressionof adiponectin [146,147]. Isoquercitrin and myricetin are flavonesthat also exert anti-obesity effects. Isoquercitrin activates wnt/b-catenin signaling, thus inhibits preadipocyte differentiation intoadipocytes [148,149]. Myricetin activates PPARa and inhibits lipiddeposition in adipocytes [150]. Phytochemicals with differentphases of their preclinical and clinical development against obesityand obesity-associated breast cancer are further listed in Table 3.

4. Future directions and perspectives

The saying ‘‘Prevention is better than cure’’ needs to be followedto reduce the number of obesity-associated breast cancer cases.Reduction of high calories in diet by reducing the consumption offat-rich diet and increased physical activity are the two mostimportant prevention strategies for obesity and ultimately forobesity-associated breast cancer. Development of more effectivedrugs that can selectively inhibit the estrogen synthesis withminimal side effects might be useful candidates against obesity-associated breast cancer. More research needs to be focused on thedevelopment of effective leptin antagonists and adiponectinagonists. Therapeutic agents that can reduce energy metabolismand induce adipocyte degeneration might also prove to be effectivecandidate drugs. Combination of chemotherapy and hormonaltherapy or different hormonal therapies is to be tested in theclinical trials. Future research on the efficacy of anti-obesity drugssuch as pitavastatin and orlistat in the treatment of obesity-relatedmammary carcinogenesis is also needed. The efficacy of anti-diabetic drug metformin against breast cancer needs to be tested inclinical trials as it has shown its efficacy in preclinical trials. Moreresearch is also needed on the efficacy of anti-inflammatory drugsagainst obesity-associated breast cancer.

5. Conclusion

Obesity is a strong risk factor for the increased breast cancerincidence and mortality in postmenopausal women. The detailed

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513510

molecular mechanisms involved in obesity-associated breastcancer need to be studied. However, the known signaling pathwaysinvolved in obesity-associated breast cancer are mediated throughestrogen, insulin, leptin, adiponectin and inflammatory cytokines.In fat cells, increased aromatase activity leads to an increase inestrogen synthesis which further decreases the level of SHBG, bothcausally linked with obesity-associated breast cancer. The levels ofInsulin and insulin like growth factors are increased in obesitywhich increases the growth rate of cancer cells via activation of keytumorigenic pathways such as PI3K/Akt/mTOR and Ras/Raf/MAPK.The increased leptin and/or decreased adiponectin levels are thestrong risk factors for the development of postmenopausal breastcancer. Obesity-inflammation axis plays an important role in thedevelopment of breast cancer. In obese condition, increasedproduction of inflammatory cytokines has been found which arepotentially linked to breast cancer development via severalmechanisms including cellular proliferation and genetic damage.The therapeutic regimens for obesity-associated breast cancerinclude aromatase inhibitors, estrogen antagonists, leptin antago-nists, adiponectin agonists, and anti-inflammatory drugs. Howevertheir trial and use is warranted with clinical significance.

Acknowledgements

This work was supported by the EpiHeD-Network Scheme(BSC0118) and Research fellowship grants (SK, SS) from theCouncil of Scientific and Industrial Research (CSIR), Government ofIndia, India. Part of this work was also supported by grants toS.M.M. from the Science & Engineering Research Board (SERB), NewDelhi, India. We thank Ms. Isha Soni for her assistance in proof-reading the manuscript. CSIR-CDRI communication Number-8554.No conflict of interest.

References

[1] Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancerstatistics. CA Cancer J Clin 2011;61:69–90.

[2] Hulka BS, Moorman PG. Breast cancer: hormones and other risk factors.Maturitas 2001;38:103–13. discussion 13–6.

[3] Key J, Hodgson S, Omar RZ, Jensen TK, Thompson SG, Boobis AR, et al. Meta-analysis of studies of alcohol and breast cancer with consideration of themethodological issues. Cancer Causes Control 2006;17:759–70.

[4] Baan R, Straif K, Grosse Y, Secretan B, El Ghissassi F, Bouvard V, et al.Carcinogenicity of alcoholic beverages. Lancet Oncol 2007;8:292–3.

[5] Donohoe CL, Doyle SL, Reynolds JV. Visceral adiposity, insulin resistance andcancer risk. Diabetol Metab Syndr 2011;3:12.

[6] Kanasaki K, Koya D. Biology of obesity: lessons from animal models of obesity.J Biomed Biotechnol 2011;2011:197636.

[7] Vainio H, Kaaks R, Bianchini F. Weight control and physical activity in cancerprevention: international evaluation of the evidence. Eur J Cancer Prev2002;11(Suppl. 2):S94–100.

[8] Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index andincidence of cancer: a systematic review and meta-analysis of prospectiveobservational studies. Lancet 2008;371:569–78.

[9] Rack B, Andergassen U, Neugebauer J, Salmen J, Hepp P, Sommer H, et al. TheGerman SUCCESS C Study – The First European Lifestyle Study on breastcancer. Breast Care (Basel) 2010;5:395–400.

[10] Danaei G, Vander Hoorn S, Lopez AD, Murray CJ, Ezzati M. (Cancers) CRAcg.Causes of cancer in the world: comparative risk assessment of nine beha-vioural and environmental risk factors. Lancet 2005;366:1784–93.

[11] Carmichael AR. Obesity as a risk factor for development and poor prognosis ofbreast cancer. BJOG 2006;113:1160–6.

[12] Abrahamson PE, Gammon MD, Lund MJ, Flagg EW, Porter PL, Stevens J, et al.General and abdominal obesity and survival among young women withbreast cancer. Cancer Epidemiol Biomarkers Prev 2006;15:1871–7.

[13] Porter GA, Inglis KM, Wood LA, Veugelers PJ. Effect of obesity on presentationof breast cancer. Ann Surg Oncol 2006;13:327–32.

[14] Calle EE, Thun MJ. Obesity and cancer. Oncogene 2004;23:6365–78.[15] Maccio A, Madeddu C, Mantovani G. Adipose tissue as target organ in the

treatment of hormone-dependent breast cancer: new therapeutic perspec-tives. Obes Rev 2009;10:660–70.

[16] Roberts DL, Dive C, Renehan AG. Biological mechanisms linking obesity andcancer risk: new perspectives. Annu Rev Med 2010;61:301–16.

[17] Jones JI, Clemmons DR. Insulin-like growth factors and their binding proteins:biological actions. Endocr Rev 1995;16:3–34.

[18] Renier G, Clement I, Desfaits AC, Lambert A. Direct stimulatory effect ofinsulin-like growth factor-I on monocyte and macrophage tumor necrosisfactor-alpha production. Endocrinology 1996;137:4611–8.

[19] Heemskerk VH, Daemen MA, Buurman WA. Insulin-like growth factor-1 (IGF-1) and growth hormone (GH) in immunity and inflammation. CytokineGrowth Factor Rev 1999;10:5–14.

[20] Law JH, Habibi G, Hu K, Masoudi H, Wang MY, Stratford AL, et al. Phosphory-lated insulin-like growth factor-i/insulin receptor is present in all breastcancer subtypes and is related to poor survival. Cancer Res 2008;68:10238–46.

[21] Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors andneoplasia. Nat Rev Cancer 2004;4:505–18.

[22] Chan BT, Lee AV. Insulin receptor substrates (IRSs) and breast tumorigenesis. JMammary Gland Biol Neoplasia 2008;13:415–22.

[23] LeRoith D, Roberts CT. The insulin-like growth factor system and cancer.Cancer Lett 2003;195:127–37.

[24] Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, et al. Adipocytedeath defines macrophage localization and function in adipose tissue ofobese mice and humans. J Lipid Res 2005;46:2347–55.

[25] Murano I, Barbatelli G, Parisani V, Latini C, Muzzonigro G, Castellucci M, et al.Dead adipocytes, detected as crown-like structures, are prevalent in visceralfat depots of genetically obese mice. J Lipid Res 2008;49:1562–8.

[26] Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW.Obesity is associated with macrophage accumulation in adipose tissue. J ClinInvest 2003;112:1796–808.

[27] Aggarwal BB, Gehlot P. Inflammation and cancer: how friendly is the rela-tionship for cancer patients? Curr Opin Pharmacol 2009;9:351–69.

[28] Subbaramaiah K, Howe LR, Bhardwaj P, Du B, Gravaghi C, Yantiss RK, et al.Obesity is associated with inflammation and elevated aromatase expressionin the mouse mammary gland. Cancer Prev Res (Phila) 2011;4:329–46.

[29] Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK, et al. Inflam-mation and increased aromatase expression occur in the breast tissue ofobese women with breast cancer. Cancer Prev Res (Phila) 2011;4:1021–9.

[30] Nicolini A, Carpi A, Rossi G. Cytokines in breast cancer. Cytokine GrowthFactor Rev 2006;17:325–37.

[31] Maccio A, Madeddu C. Obesity, inflammation, and postmenopausal breastcancer: therapeutic implications. ScientificWorldJournal 2011;11:2020–36.

[32] Grisouard J, Dembinski K, Mayer D, Keller U, Muller B, Christ-Crain M.Targeting AMP-activated protein kinase in adipocytes to modulate obesi-ty-related adipokine production associated with insulin resistance and breastcancer cell proliferation. Diabetol Metab Syndr 2011;3:16.

[33] Zappala G, Rechler MM. IGFBP-3, hypoxia and TNF-alpha inhibit adiponectintranscription. Biochem Biophys Res Commun 2009;382:785–9.

[34] Kim KY, Kim JK, Jeon JH, Yoon SR, Choi I, Yang Y. c-Jun N-terminal kinase isinvolved in the suppression of adiponectin expression by TNF-alpha in 3T3-L1 adipocytes. Biochem Biophys Res Commun 2005;327:460–7.

[35] Vona-Davis L, Rose DP. Adipokines as endocrine, paracrine, and autocrinefactors in breast cancer risk and progression. Endocr Relat Cancer 2007;14:189–206.

[36] Collins S, Kuhn CM, Petro AE, Swick AG, Chrunyk BA, Surwit RS. Role of leptinin fat regulation. Nature 1996;380:677.

[37] Lindheim SR, Sauer MV, Carmina E, Chang PL, Zimmerman R, Lobo RA.Circulating leptin levels during ovulation induction: relation to adiposityand ovarian morphology. Fertil Steril 2000;73:493–8.

[38] Green ED, Maffei M, Braden VV, Proenca R, DeSilva U, Zhang Y, et al. Thehuman obese (OB) gene: RNA expression pattern and mapping on thephysical, cytogenetic, and genetic maps of chromosome 7. Genome Res1995;5:5–12.

[39] Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positionalcloning of the mouse obese gene and its human homologue. Nature 1994;372:425–32.

[40] Ahima RS, Osei SY. Leptin signaling. Physiol Behav 2004;81:223–41.[41] Hynes GR, Jones PJ. Leptin and its role in lipid metabolism. Curr Opin Lipidol

2001;12:321–7.[42] Huang L, Wang Z, Li C. Modulation of circulating leptin levels by its soluble

receptor. J Biol Chem 2001;276:6343–9.[43] Hamilton BS, Paglia D, Kwan AY, Deitel M. Increased obese mRNA expression

in omental fat cells from massively obese humans. Nat Med 1995;1:953–6.[44] Harris HR, Tworoger SS, Hankinson SE, Rosner BA, Michels KB. Plasma leptin

levels and risk of breast cancer in premenopausal women. Cancer Prev Res(Phila) 2011;4:1449–56.

[45] Garofalo C, Koda M, Cascio S, Sulkowska M, Kanczuga-Koda L, Golaszewska J,et al. Increased expression of leptin and the leptin receptor as a marker ofbreast cancer progression: possible role of obesity-related stimuli. ClinCancer Res 2006;12:1447–53.

[46] Ishikawa M, Kitayama J, Nagawa H. Enhanced expression of leptin and leptinreceptor (OB-R) in human breast cancer. Clin Cancer Res 2004;10:4325–31.

[47] Cleary MP, Phillips FC, Getzin SC, Jacobson TL, Jacobson MK, Christensen TA,et al. Genetically obese MMTV-TGF-alpha/Lep(ob)Lep(ob) female mice do notdevelop mammary tumors. Breast Cancer Res Treat 2003;77:205–15.

[48] Cleary MP, Juneja SC, Phillips FC, Hu X, Grande JP, Maihle NJ. Leptin receptor-deficient MMTV-TGF-alpha/Lepr(db)Lepr(db) female mice do not developoncogene-induced mammary tumors. Exp Biol Med (Maywood) 2004;229:182–93.

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513 511

[49] Bjørbaek C, Uotani S, da Silva B, Flier JS. Divergent signaling capacities of thelong and short isoforms of the leptin receptor. J Biol Chem 1997;272:32686–95.

[50] Yin N, Wang D, Zhang H, Yi X, Sun X, Shi B, et al. Molecular mechanismsinvolved in the growth stimulation of breast cancer cells by leptin. Cancer Res2004;64:5870–5.

[51] Saxena NK, Vertino PM, Anania FA, Sharma D. leptin-induced growth stimu-lation of breast cancer cells involves recruitment of histone acetyltrans-ferases and mediator complex to CYCLIN D1 promoter via activation of Stat3.J Biol Chem 2007;282:13316–25.

[52] Bjørbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS. Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol Cell 1998;1:619–25.

[53] Bjørbaek C, El-Haschimi K, Frantz JD, Flier JS. The role of SOCS-3 in leptinsignaling and leptin resistance. J Biol Chem 1999;274:30059–65.

[54] Palianopoulou M, Papanikolaou V, Stefanou N, Tsezou A. The activation ofleptin-mediated survivin is limited by the inducible suppressor SOCS-3 inMCF-7 cells. Exp Biol Med (Maywood) 2011;236:70–6.

[55] Park H, Kim M, Kwon GT, Lim DY, Yu R, Sung MK, et al. A high-fat dietincreases angiogenesis, solid tumor growth, and lung metastasis of CT26colon cancer cells in obesity-resistant BALB/c mice. Mol Carcinog 2011.

[56] Garonna E, Botham KM, Birdsey GM, Randi AM, Gonzalez-Perez RR, Wheeler-Jones CP. Vascular endothelial growth factor receptor-2 couples cyclo-oxy-genase-2 with pro-angiogenic actions of leptin on human endothelial cells.PLoS One 2011;6:e18823.

[57] Gonzalez RR, Cherfils S, Escobar M, Yoo JH, Carino C, Styer AK, et al. Leptinsignaling promotes the growth of mammary tumors and increases theexpression of vascular endothelial growth factor (VEGF) and its receptortype two (VEGF-R2). J Biol Chem 2006;281:26320–28.

[58] Kitawaki J, Kusuki I, Koshiba H, Tsukamoto K, Honjo H. Leptin directlystimulates aromatase activity in human luteinized granulosa cells. MolHum Reprod 1999;5:708–13.

[59] Falk RT, Brinton LA, Madigan MP, Potischman N, Sturgeon SR, Malone KE, et al.Interrelationships between serum leptin, IGF-1, IGFBP3, C-peptide and pro-lactin and breast cancer risk in young women. Breast Cancer Res Treat2006;98:157–65.

[60] Fusco R, Galgani M, Procaccini C, Franco R, Pirozzi G, Fucci L, et al. Cellular andmolecular crosstalk between leptin receptor and estrogen receptor-{alpha} inbreast cancer: molecular basis for a novel therapeutic setting. Endocr RelatCancer 2010;17:373–82.

[61] Jarde T, Caldefie-Chezet F, Damez M, Mishellany F, Penault-Llorca F, Guillot J,et al. Leptin and leptin receptor involvement in cancer development: a studyon human primary breast carcinoma. Oncol Rep 2008;19:905–11.

[62] Ozet A, Arpaci F, Yilmaz MI, Ayta H, Ozturk B, Komurcu S, et al. Effects oftamoxifen on the serum leptin level in patients with breast cancer. Jpn J ClinOncol 2001;31:424–7.

[63] Catalano S, Mauro L, Marsico S, Giordano C, Rizza P, Rago V, et al. Leptininduces, via ERK1/ERK2 signal, functional activation of estrogen receptoralpha in MCF-7 cells. J Biol Chem 2004;279:19908–15.

[64] Garofalo C, Sisci D, Surmacz E. Leptin interferes with the effects of theantiestrogen ICI 182,780 in MCF-7 breast cancer cells. Clin Cancer Res2004;10:6466–75.

[65] Nunez NP, Jelovac D, Macedo L, Berrigan D, Perkins SN, Hursting SD, et al.Effects of the antiestrogen tamoxifen and the aromatase inhibitor letrozoleon serum hormones and bone characteristics in a preclinical tumor model forbreast cancer. Clin Cancer Res 2004;10:5375–80.

[66] Geisler J, Lønning PE, Krag LE, Løkkevik E, Risberg T, Hagen AI, et al. Changes inbone and lipid metabolism in postmenopausal women with early breastcancer after terminating 2-year treatment with exemestane: a randomised,placebo-controlled study. Eur J Cancer 2006;42:2968–75.

[67] Fiorio E, Mercanti A, Terrasi M, Micciolo R, Remo A, Auriemma A, et al. Leptin/HER2 crosstalk in breast cancer: in vitro study and preliminary in vivoanalysis. BMC Cancer 2008;8:305.

[68] Rahmati-Yamchi M, Zarghami N, Rahbani M, Montazeri A. Plasma leptin,hTERT gene expression, and anthropometric measures in obese andnon-obese women with breast cancer. Breast Cancer (Auckl) 2011;5:27–35.

[69] Ren H, Zhao T, Wang X, Gao C, Wang J, Yu M, et al. Leptin upregulatestelomerase activity and transcription of human telomerase reverse transcrip-tase in MCF-7 breast cancer cells. Biochem Biophys Res Commun 2010;394:59–63.

[70] Chagnon YC, Wilmore JH, Borecki IB, Gagnon J, Perusse L, Chagnon M, et al.Associations between the leptin receptor gene and adiposity in middle-agedCaucasian males from the HERITAGE family study. J Clin Endocrinol Metab2000;85:29–34.

[71] Mammes O, Betoulle D, Aubert R, Herbeth B, Siest G, Fumeron F. Associationof the G-2548A polymorphism in the 50 region of the LEP gene with over-weight. Ann Hum Genet 2000;64:391–4.

[72] Yiannakouris N, Yannakoulia M, Melistas L, Chan JL, Klimis-Zacas D, Man-tzoros CS. The Q223R polymorphism of the leptin receptor gene is signifi-cantly associated with obesity and predicts a small percentage of bodyweight and body composition variability. J Clin Endocrinol Metab 2001;86:4434–9.

[73] Terrasi M, Fiorio E, Mercanti A, Koda M, Moncada CA, Sulkowski S, et al.Functional analysis of the -2548G/A leptin gene polymorphism in breastcancer cells. Int J Cancer 2009;125:1038–44.

[74] Cleveland RJ, Gammon MD, Long CM, Gaudet MM, Eng SM, Teitelbaum SL,et al. Common genetic variations in the LEP and LEPR genes, obesity andbreast cancer incidence and survival. Breast Cancer Res Treat 2010;120:745–52.

[75] Kadowaki T, Yamauchi T. Adiponectin and adiponectin receptors. Endocr Rev2005;26:439–51.

[76] Saito K, Tobe T, Yoda M, Nakano Y, Choi-Miura NH, Tomita M. Regulation ofgelatin-binding protein 28 (GBP28) gene expression by C/EBP. Biol PharmBull 1999;22:1158–62.

[77] Bełtowski J, Jamroz-Wisniewska A, Widomska S. Adiponectin and its role incardiovascular diseases. Cardiovasc Hematol Disord Drug Targets 2008;8:7–46.

[78] Yamauchi T, Hara K, Kubota N, Terauchi Y, Tobe K, Froguel P, et al. Dual rolesof adiponectin/Acrp30 in vivo as an anti-diabetic and anti-atherogenicadipokine. Curr Drug Targets Immune Endocr Metabol Disord 2003;3:243–54.

[79] Grossmann ME, Nkhata KJ, Mizuno NK, Ray A, Cleary MP. Effects of adipo-nectin on breast cancer cell growth and signaling. Br J Cancer 2008;98:370–9.

[80] Jarde T, Caldefie-Chezet F, Goncalves-Mendes N, Mishellany F, Buechler C,Penault-Llorca F, et al. Involvement of adiponectin and leptin in breastcancer: clinical and in vitro studies. Endocr Relat Cancer 2009;16:1197–210.

[81] Nakayama S, Miyoshi Y, Ishihara H, Noguchi S. Growth-inhibitory effect ofadiponectin via adiponectin receptor 1 on human breast cancer cells throughinhibition of S-phase entry without inducing apoptosis. Breast Cancer ResTreat 2008;112:405–10.

[82] Miyoshi Y, Funahashi T, Kihara S, Taguchi T, Tamaki Y, Matsuzawa Y, et al.Association of serum adiponectin levels with breast cancer risk. Clin CancerRes 2003;9:5699–704.

[83] Mantzoros C, Petridou E, Dessypris N, Chavelas C, Dalamaga M, Alexe DM,et al. Adiponectin and breast cancer risk. J Clin Endocrinol Metab 2004;89:1102–7.

[84] Chen DC, Chung YF, Yeh YT, Chaung HC, Kuo FC, Fu OY, et al. Serumadiponectin and leptin levels in Taiwanese breast cancer patients. CancerLett 2006;237:109–14.

[85] Hou WK, Xu YX, Yu T, Zhang L, Zhang WW, Fu CL, et al. Adipocytokines andbreast cancer risk. Chin Med J (Engl) 2007;120:1592–6.

[86] Tworoger SS, Eliassen AH, Kelesidis T, Colditz GA, Willett WC, Mantzoros CS,et al. Plasma adiponectin concentrations and risk of incident breast cancer. JClin Endocrinol Metab 2007;92:1510–6.

[87] Kang JH, Yu BY, Youn DS. Relationship of serum adiponectin and resistinlevels with breast cancer risk. J Korean Med Sci 2007;22:117–21.

[88] Miyatani Y, Yasui T, Uemura H, Yamada M, Matsuzaki T, Kuwahara A, et al.Associations of circulating adiponectin with estradiol and monocyte chemo-tactic protein-1 in postmenopausal women. Menopause 2008;15:536–41.

[89] Combs TP, Berg AH, Rajala MW, Klebanov S, Iyengar P, Jimenez-Chillaron JC,et al. Sexual differentiation, pregnancy, calorie restriction, and aging affectthe adipocyte-specific secretory protein adiponectin. Diabetes 2003;52:268–76.

[90] Dieudonne MN, Bussiere M, Dos Santos E, Leneveu MC, Giudicelli Y, PecqueryR. Adiponectin mediates antiproliferative and apoptotic responses in humanMCF7 breast cancer cells. Biochem Biophys Res Commun 2006;345:271–9.

[91] Chen X, Wang Y. Adiponectin and breast cancer. Med Oncol 2011;28:1288–95.

[92] Kim KY, Baek A, Hwang JE, Choi YA, Jeong J, Lee MS, et al. Adiponectin-activated AMPK stimulates dephosphorylation of AKT through protein phos-phatase 2A activation. Cancer Res 2009;69:4018–26.

[93] Motoshima H, Wu X, Mahadev K, Goldstein BJ. Adiponectin suppressesproliferation and superoxide generation and enhances eNOS activity inendothelial cells treated with oxidized LDL. Biochem Biophys Res Commun2004;315:264–71.

[94] Arita Y, Kihara S, Ouchi N, Maeda K, Kuriyama H, Okamoto Y, et al. Adipocyte-derived plasma protein adiponectin acts as a platelet-derived growth factor-BB-binding protein and regulates growth factor-induced common postre-ceptor signal in vascular smooth muscle cell. Circulation 2002;105:2893–8.

[95] Wang Y, Lam KS, Xu JY, Lu G, Xu LY, Cooper GJ, et al. Adiponectin inhibits cellproliferation by interacting with several growth factors in an oligomeriza-tion-dependent manner. J Biol Chem 2005;280:18341–47.

[96] Schaffler A, Scholmerich J, Buechler C. Mechanisms of Disease: adipokinesand breast cancer—endocrine and paracrine mechanisms that connect adi-posity and breast cancer. Nat Rev Endocrinol 2007;3:345–54.

[97] Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for chronicinflammation and adiponectin reduction in adipose tissue of ob/ob anddietary obese mice. Am J Physiol Endocrinol Metab 2007;293:E1118–28.

[98] Vaupel P. The role of hypoxia-induced factors in tumor progression. Oncolo-gist 2004;9(Suppl. 5):10–7.

[99] Feldser D, Agani F, Iyer NV, Pak B, Ferreira G, Semenza GL. Reciprocal positiveregulation of hypoxia-inducible factor 1alpha and insulin-like growth factor2. Cancer Res 1999;59:3915–8.

[100] Fukuda R, Hirota K, Fan F, Jung YD, Ellis LM, Semenza GL. Insulin-like growthfactor 1 induces hypoxia-inducible factor 1-mediated vascular endothelialgrowth factor expression, which is dependent on MAP kinase and phospha-tidylinositol 3-kinase signaling in colon cancer cells. J Biol Chem 2002;277:38205–11.

[101] Seifeddine R, Dreiem A, Blanc E, Fulchignoni-Lataud MC, Le Frere Belda MA,Lecuru F, et al. Hypoxia down-regulates CCAAT/enhancer binding protein-alpha expression in breast cancer cells. Cancer Res 2008;68:2158–65.

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513512

[102] Pal P, Lochab S, Kanaujiya J, Sanyal S, Trivedi AK. Ectopic expression of hC/EBPs in breast tumor cells induces apoptosis. Mol Cell Biochem 2010;337:111–8.

[103] Johnston SR. Fulvestrant (’Faslodex’): extending the reach of endocrinetherapy? Breast Cancer Res Treat 2005;93(Suppl. 1):S1–2.

[104] Margariti N, Fox SB, Bottini A, Generali D. ‘‘Overcoming breast cancer drugresistance with mTOR inhibitors’’. Could it be a myth or a real possibility inthe short-term future? Breast Cancer Res Treat 2011;128:599–606.

[105] Alvarez RH, Booser DJ, Cristofanilli M, Sahin AA, Strom EA, Guerra L, et al.Phase 2 trial of primary systemic therapy with doxorubicin and docetaxelfollowed by surgery, radiotherapy, and adjuvant chemotherapy with cyclo-phosphamide, methotrexate, and 5-fluorouracil based on clinical and patho-logic response in patients with stage IIB to III breast cancer: long-term resultsfrom the University of Texas M. D. Anderson Cancer Center Study ID97-099.Cancer 2010;116:1210–7.

[106] Sabnis G, Goloubeva O, Jelovac D, Schayowitz A, Brodie A. Inhibition of thephosphatidylinositol 3-kinase/Akt pathway improves response of long-termestrogen-deprived breast cancer xenografts to antiestrogens. Clin Cancer Res2007;13:2751–7.

[107] Jelovac D, Sabnis G, Long BJ, Macedo L, Goloubeva OG, Brodie AM. Activationof mitogen-activated protein kinase in xenografts and cells during prolongedtreatment with aromatase inhibitor letrozole. Cancer Res 2005;65:5380–9.

[108] Lorincz AM, Sukumar S. Molecular links between obesity and breast cancer.Endocr Relat Cancer 2006;13:279–92.

[109] Wesierska-Gadek J, Gritsch D, Zulehner N, Komina O, Maurer M. Roscovitine,a selective CDK inhibitor, reduces the basal and estrogen-induced phosphor-ylation of ER-a in human ER-positive breast cancer cells. J Cell Biochem2011;112:761–72.

[110] Shimizu M, Yasuda Y, Sakai H, Kubota M, Terakura D, Baba A, et al. Pitavas-tatin suppresses diethylnitrosamine-induced liver preneoplasms in maleC57BL/KsJ-db/db obese mice. BMC Cancer 2011;11:281.

[111] Menendez JA, Vellon L, Lupu R. Antitumoral actions of the anti-obesity drugorlistat (XenicalTM) in breast cancer cells: blockade of cell cycle progression,promotion of apoptotic cell death and PEA3-mediated transcriptional re-pression of Her2/neu (erbB-2) oncogene. Ann Oncol 2005;16:1253–67.

[112] http://clinicaltrials.gov/ct2/show/NCT00944918?term=aromatase+inhibi-tors+phase+3&rank=3.

[113] Leo AD, Biganzoli L, Bohm S, Lupi G, Oriana S, Riboldi G, et al. An intensivetreatment with mitoxantrone and ifosfamide in second-line therapy ofepithelial ovarian cancer. Tumori 1994;80:443–7.

[114] http://clinicaltrials.gov/ct2/show/NCT00241449?term=fulvestrant+in+post-menopausal+women+phase+3&rank=14.

[115] Chen JQ, Brown TR, Russo J. Regulation of energy metabolism pathways byestrogens and estrogenic chemicals and potential implications in obesityassociated with increased exposure to endocrine disruptors. Biochim Bio-phys Acta 2009;1793:1128–43.

[116] Winder WW, Hardie DG. AMP-activated protein kinase, a metabolic masterswitch: possible roles in type 2 diabetes. Am J Physiol 1999;277:E1–0.

[117] Rodgers RJ, Tschop MH, Wilding JP. Anti-obesity drugs: past, present andfuture. Dis Model Mech 2012;5:621–6.

[118] Rene Gonzalez R, Watters A, Xu Y, Singh UP, Mann DR, Rueda BR, et al. Leptin-signaling inhibition results in efficient anti-tumor activity in estrogen recep-tor positive or negative breast cancer. Breast Cancer Res 2009;11:R36.

[119] Otvos L, Haspinger E, La Russa F, Maspero F, Graziano P, Kovalszky I, et al.Design and development of a peptide-based adiponectin receptor agonist forcancer treatment. BMC Biotechnol 2011;11:90.

[120] http://clinicaltrials.gov/ct2/show/NCT00201773?term=celecoxib+pha-se+2+clinical+trial+in+postmenopausal+women&rank=2.

[121] http://clinicaltrials.gov/ct2/show/NCT00933309?term=exemestane%2C+a-vandamet&rank=1.

[122] http://clinicaltrials.gov/ct2/show/NCT00073073?term=celecoxib+postme-nopausal+breast+cancer+phase+2&rank=3.

[123] http://clinicaltrials.gov/ct2/show/NCT00112749?term=infliximab+breast+-cancer+phase+2&rank=1.

[124] http://clinicaltrials.gov/ct2/show/NCT01127867?term=dha+breast+can-cer&rank=2.

[125] http://clinicaltrials.gov/ct2/show/NCT00917735?term=green+tea+extract+-breast+cancer&rank=1.

[126] http://clinicaltrials.gov/ct2/show/NCT00516243?term=green+tea+extract+-breast+cancer&rank=2.

[127] Aggarwal BB, Shishodia S. Molecular targets of dietary agents for preventionand therapy of cancer. Biochem Pharmacol 2006;71:1397–421.

[128] Zhou H, Luo Y, Huang S. Updates of mTOR inhibitors. Anticancer Agents MedChem 2010;10:571–81.

[129] Shehzad A, Khan S, Sup Lee Y. Curcumin molecular targets in obesity andobesity-related cancers. Future Oncol 2012;8:179–90.

[130] Aggarwal BB. Targeting inflammation-induced obesity and metabolic dis-eases by curcumin and other nutraceuticals. Annu Rev Nutr 2010;30:173–99.

[131] Ghanim H, Sia CL, Abuaysheh S, Korzeniewski K, Patnaik P, Marumganti A,et al. An antiinflammatory and reactive oxygen species suppressive effects ofan extract of Polygonum cuspidatum containing resveratrol. J Clin EndocrinolMetab 2010;95:E1–8.

[132] Morselli E, Maiuri MC, Markaki M, Megalou E, Pasparaki A, Palikaras K, et al.Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death Dis 2010;1:e10.

[133] Lanzilli G, Fuggetta MP, Tricarico M, Cottarelli A, Serafino A, Falchetti R, et al.Resveratrol down-regulates the growth and telomerase activity of breastcancer cells in vitro. Int J Oncol 2006;28:641–8.

[134] Zhang X, Chen LX, Ouyang L, Cheng Y, Liu B. Plant natural compounds:targeting pathways of autophagy as anti-cancer therapeutic agents. CellProlif 2012;45:466–76.

[135] Pratheeshkumar P, Sreekala C, Zhang Z, Budhraja A, Ding S, Son YO, et al.Cancer prevention with promising natural products: mechanisms of actionand molecular targets. Anticancer Agents Med Chem 2012;12:1159–84.

[136] Ferry DR, Smith A, Malkhandi J, Fyfe DW, deTakats PG, Anderson D, et al.Phase I clinical trial of the flavonoid quercetin: pharmacokinetics and evi-dence for in vivo tyrosine kinase inhibition. Clin Cancer Res 1996;2:659–68.

[137] De Angel RE, Smith SM, Glickman RD, Perkins SN, Hursting SD. Antitumoreffects of ursolic acid in a mouse model of postmenopausal breast cancer.Nutr Cancer 2010;62:1074–86.

[138] Kanjoormana M, Kuttan G. Antiangiogenic activity of ursolic acid. IntegrCancer Ther 2010;9:224–35.

[139] Hwang JT, Park IJ, Shin JI, Lee YK, Lee SK, Baik HW, et al. Genistein, EGCG, andcapsaicin inhibit adipocyte differentiation process via activating AMP-acti-vated protein kinase. Biochem Biophys Res Commun 2005;338:694–9.

[140] Naaz A, Yellayi S, Zakroczymski MA, Bunick D, Doerge DR, Lubahn DB, et al.The soy isoflavone genistein decreases adipose deposition in mice. Endocri-nology 2003;144:3315–20.

[141] Zhang M, Ikeda K, Xu JW, Yamori Y, Gao XM, Zhang BL. Genistein suppressesadipogenesis of 3T3-L1 cells via multiple signal pathways. Phytother Res2009;23:713–8.

[142] Grove KA, Sae-tan S, Kennett MJ, Lambert JD. (�)-Epigallocatechin-3-gallateinhibits pancreatic lipase and reduces body weight gain in high fat-fed obesemice. Obesity (Silver Spring) 2012;20:2311–3.

[143] Kim H, Hiraishi A, Tsuchiya K, Sakamoto K. (�)-Epigallocatechin gallatesuppresses the differentiation of 3T3-L1 preadipocytes through transcriptionfactors FoxO1 and SREBP1c. Cytotechnology 2010;62:245–55.

[144] Klaus S, Pultz S, Thone-Reineke C, Wolfram S. Epigallocatechin gallateattenuates diet-induced obesity in mice by decreasing energy absorptionand increasing fat oxidation. Int J Obes (Lond) 2005;29:615–23.

[145] Ross SA. Evidence for the relationship between diet and cancer. Exp Oncol2010;32:137–42.

[146] Cho SY, Park PJ, Shin HJ, Kim YK, Shin DW, Shin ES, et al. (�)-Catechinsuppresses expression of Kruppel-like factor 7 and increases expression andsecretion of adiponectin protein in 3T3-L1 cells. Am J Physiol EndocrinolMetab 2007;292:E1166–72.

[147] Si H, Fu Z, Babu PV, Zhen W, Leroith T, Meaney MP, et al. Dietary epicatechinpromotes survival of obese diabetic mice and Drosophila melanogaster. JNutr 2011;141:1095–100.

[148] Ross SE, Hemati N, Longo KA, Bennett CN, Lucas PC, Erickson RL, et al.Inhibition of adipogenesis by Wnt signaling. Science 2000;289:950–3.

[149] Lee SH, Kim B, Oh MJ, Yoon J, Kim HY, Lee KJ, et al. Persicaria hydropiper (L.)spach and its flavonoid components, isoquercitrin and isorhamnetin, activatethe Wnt/b-catenin pathway and inhibit adipocyte differentiation of 3T3-L1cells. Phytother Res 2011;25:1629–35.

[150] Chang CJ, Tzeng TF, Liou SS, Chang YS, Liu IM. Myricetin increases hepaticperoxisome proliferator-activated receptor a protein expression anddecreases plasma lipids and adiposity in rats. Evid Based ComplementAlternat Med 2012;2012:787152.

Syed Musthapa Meeran Ph.D is currently a senior sci-entist at the Division of Endocrinology of CSIR-CentralDrug Research Institute. In his doctoral work, he studiedthe role of asbestos and other biomass pollutants-me-diated genetic and cytogentic changes in the pulmonarysystem. To further in his postdoctoral study, he evalu-ated the role of inflammatory cytokines for the skincancer risk at the Department of Dermatology, Univer-sity of Alabama at Birmingham. Later he became afaculty at the Department of Biology, University ofAlabama at Birmingham, where he studied the epige-netic and chemoprevention of breast cancer. He is cur-rently working on the novel genetic and epigenetic

targets for breast and lung cancer prevention as well as therapy with bioactivedietary supplements. His research interest are also focused on the role of variouscytokine and chemokine mediated intra cellular signaling in the cancer progression,especially breast cancer.

Sajid Khan obtained his Masters degree in Biotechnol-ogy from Aligarh Muslim University, Aligarh, India in2011. Currently, he is working as a doctoral researchfellow under the guidance of Dr. Syed Musthapa Meeranin the Endocrinology Division of CSIR-Central Drug Re-search Institute, Lucknow, India. His research work isprimarily focused on the role of leptin and adiponectinin obesity-associated breast cancer. He also obtainedSenior Research Fellowship (CSIR-SRF) from the Councilof Scientific and Industrial Research, New Delhi, Indiafor his doctoral study.

S. Khan et al. / Cytokine & Growth Factor Reviews 24 (2013) 503–513 513

Samriddhi Shukla has received her Masters degree inBiotechnology from Dr. R.M.L. Avadh University, Faiza-bad, India with university ranking and gold medal. She iscurrently pursuing her doctoral research under thementorship of Dr. Syed Musthapa Meeran, CSIR-CentralDrug Research Institute, Lucknow, India. Her researchwork mainly focuses on the genetics and epigenetics oflung cancer. She also obtained Senior Research Fellow-ship (CSIR-SRF) from the Council of Scientific and In-dustrial Research, New Delhi, India for her doctoralresearch work.

Sonam Sinha earned her Masters in Biotechnology fromAMITY University, Lucknow, India in 2011. She is cur-rently engaged as a Project Assistant under the super-vision of Dr. Syed Musthapa Meeran at CSIR-CentralDrug Research Institute, Lucknow, India. Her researchinterest is mainly focused on studying novel genetic andepigenetic targets for breast cancer prevention andtherapy with bioactive dietary supplements.