+ All Categories
Home > Documents > Regulation of lipid metabolism in breast cancer provides diagnostic and therapeutic opportunities

Regulation of lipid metabolism in breast cancer provides diagnostic and therapeutic opportunities

Date post: 11-Oct-2016
Category:
Upload: matej
View: 213 times
Download: 1 times
Share this document with a friend
12
177 ISSN 1758-4299 10.2217/CLP.12.10 © 2012 Future Medicine Ltd Clin. Lipidol. (2012) 7(2), 177–188 REVIEW Regulation of lipid metabolism in breast cancer provides diagnostic and therapeutic opportunities Cancer cells are characterized by extensive metabolic changes, since the rapid growth of tumors leads to lack of oxygen and hypoxia in the cells. One crucial feature is also the Warburg effect, a process in which glycolysis occurs despite the presence of oxygen in the tumor [1] . Alterations of many other meta- bolic pathways, such as increased nucleotide synthesis, are also well-known hallmarks of rapidly proliferating cancer cells [2] . Another profound phenotypic shift in malignant cells occurs in fatty acid and lipid metabo- lism [3] . The differences in lipid metabolism between normal and malignant tissues make these pathways attractive targets for cancer therapeutics. The goal of this article is to provide an over- all perspective on the changes in lipid metabo- lism in breast cancer, and more focused reviews on specific topics can be found in the refer- ences. The article focuses on fatty acid and phospholipid metabolism and also presents other cancer-related lipids including lysophos- phatidic acid (LPA), eicosanoids and (glyco) sphingolipids. The changes of these lipids in breast cancer, as well as emerging diagnostic and therapeutic opportunities, are discussed. Steroids are also important cancer-related lipid molecules, but since these compounds have already been utilized for a long time in breast cancer treatment, this field is excluded from the present review. Fatty acid metabolism in normal & malignant tissues In healthy adults, cells obtain the needed fatty acids mainly from nutrition. De novo synthesis of fatty acids occurs only in a very limited num- ber of tissues (i.e., in the liver, adipose tissue and lactating mammary gland). In liver and adipose tissue, high carbohydrate diet together with increased insulin levels stimulate de novo fatty acid synthesis to convert excess sugars into fatty acids and triglycerides for energy storage [4] . In the lactating mammary gland, fatty acid synthase (FASN) produces medium- chain fatty acids, which are easily digested by the offspring [5] . This key enzyme in lipogen- esis, illustrated in Figure 1A, catalyzes the synthesis of palmitate from acetyl-CoA and malonyl-CoA. Another important enzyme in fatty acid synthesis is acetyl-CoA-carboxylase (ACC1, gene name ACACA), whose function is to feed FASN with malonyl-CoA by cata- lyzing the ATP-dependent carboxylation of acetyl-CoA to malonyl-CoA, and thus acting as the rate-limiting enzyme in the fatty acid synthesis pathway. One of the key proteins regulating the expression of these enzymes and of membrane phospholipid synthesis is SREBP-1, which is stimulated by insulin, and which has a well-established role in hepato- cytes [6] . In adipocytes, however, PPAR- g is an important regulator of lipogenesis [7] , and the role of SREBP-1 still remains unclear, since on During malignant transformation, the lipid metabolism of cells changes radically and therefore alterations in lipid metabolism are a prominent feature of solid tumors. Lipid metabolism has been investigated at the gene and protein expression levels for several decades, but recent advances in lipidomics technology have also enabled the investigation of pathways at the level of molecular lipids. This review provides an overview of the changes of global lipid metabolism (i.e., fatty acid, phospholipid, eicosanoid and sphingolipid metabolism) in breast cancer, and discusses the diagnostic and therapeutic potential of these pathways. KEYWORDS: breast cancer n diagnostics n eicosanoids n glycolipids n lipid metabolism n lysophosphatidic acid n phospholipids n sphingolipids n therapeutics Mika Hilvo* & Matej Orešič Biotechnology for Health & Well-being, VTT Technical Research Centre of Finland, PO Box 1000, FI-02044 VTT, Espoo, Finland *Author for correspondence: Tel.: +358 20 7226131 Fax: +358 20 7227071 mika.hilvo@vtt.fi part of For reprint orders, please contact: [email protected]
Transcript

177ISSN 1758-429910.2217/CLP.12.10 © 2012 Future Medicine Ltd Clin. Lipidol. (2012) 7(2), 177–188

Review

Regulation of lipid metabolism in breast cancer provides diagnostic and therapeutic opportunities

Cancer cells are characterized by extensive metabolic changes, since the rapid growth of tumors leads to lack of oxygen and hypoxia in the cells. One crucial feature is also the Warburg effect, a process in which glycolysis occurs despite the presence of oxygen in the tumor [1]. Alterations of many other meta-bolic pathways, such as increased nucleotide synthesis, are also well-known hallmarks of rapidly proliferating cancer cells [2]. Another profound phenotypic shift in malignant cells occurs in fatty acid and lipid metabo-lism [3]. The differences in lipid metabolism between normal and malignant tissues make these pathways attractive targets for cancer therapeutics.

The goal of this article is to provide an over-all perspective on the changes in lipid metabo-lism in breast cancer, and more focused reviews on specific topics can be found in the refer-ences. The article focuses on fatty acid and phospholipid metabolism and also presents other cancer-related lipids including lysophos-phatidic acid (LPA), eicosanoids and (glyco)sphingolipids. The changes of these lipids in breast cancer, as well as emerging diagnostic and therapeutic opportunities, are discussed. Steroids are also important cancer-related lipid molecules, but since these compounds have already been utilized for a long time in breast cancer treatment, this field is excluded from the present review.

Fatty acid metabolism in normal & malignant tissuesIn healthy adults, cells obtain the needed fatty acids mainly from nutrition. De novo synthesis of fatty acids occurs only in a very limited num-ber of tissues (i.e., in the liver, adipose tissue and lactating mammary gland). In liver and adipose tissue, high carbohydrate diet together with increased insulin levels stimulate de novo fatty acid synthesis to convert excess sugars into fatty acids and triglycerides for energy storage [4]. In the lactating mammary gland, fatty acid synthase (FASN) produces medium-chain fatty acids, which are easily digested by the offspring [5]. This key enzyme in lipogen-esis, illustrated in Figure 1A, catalyzes the synthesis of palmitate from acetyl-CoA and malonyl-CoA. Another important enzyme in fatty acid synthesis is acetyl-CoA-carboxylase (ACC1, gene name ACACA), whose function is to feed FASN with malonyl-CoA by cata-lyzing the ATP-dependent carboxylation of acetyl-CoA to malonyl-CoA, and thus acting as the rate-limiting enzyme in the fatty acid synthesis pathway. One of the key proteins regulating the expression of these enzymes and of membrane phospholipid synthesis is SREBP-1, which is stimulated by insulin, and which has a well-established role in hepato-cytes [6]. In adipocytes, however, PPAR-g is an important regulator of lipogenesis [7], and the role of SREBP-1 still remains unclear, since on

During malignant transformation, the lipid metabolism of cells changes radically and therefore alterations in lipid metabolism are a prominent feature of solid tumors. Lipid metabolism has been investigated at the gene and protein expression levels for several decades, but recent advances in lipidomics technology have also enabled the investigation of pathways at the level of molecular lipids. This review provides an overview of the changes of global lipid metabolism (i.e., fatty acid, phospholipid, eicosanoid and sphingolipid metabolism) in breast cancer, and discusses the diagnostic and therapeutic potential of these pathways.

KEYWORDS: breast cancer n diagnostics n eicosanoids n glycolipids n lipid metabolism n lysophosphatidic acid n phospholipids n sphingolipids n therapeutics

Mika Hilvo* & Matej OrešičBiotechnology for Health & Well-being, VTT Technical Research Centre of Finland, PO Box 1000, FI-02044 VTT, Espoo, Finland *Author for correspondence: Tel.: +358 20 7226131 Fax: +358 20 7227071 [email protected]

part of

For reprint orders, please contact: [email protected]

Clin. Lipidol. (2012) 7(2)178 future science groupClin. Lipidol. (2012) 7(2)

the one hand, for example, mice overexpressing SREBP-1 do show a phenotype in adipose tissue [8], but on the other hand, it has been reported that SREBP-1 does not affect the expression of lipogenic genes in adipocytes [9].

In normal tissues the activity of the de novo synthetic pathway is regulated by nutrition, whereas in cancers the pathway is dysregulated and out of nutritional control. The coordinated upregulation of FASN and ACC1 is an early event in tumor progression, as was shown by expression studies in the 1990s [10–12]. In nor-mal breast tissue the expression of these enzymes occurs in scattered cells in the lobules and ter-minal ducts, whereas in carcinomas in situ the expression becomes intense and highest expres-sion is found in high-grade ductal carcinomas in situ [10]. FASN has also been shown to pre-dict poorer disease-free and overall survival in breast cancer patients, and thus be associated with a higher risk of recurrence [11,12]. There are very few studies considering SREBP-1, the key regulator of FASN and ACACA, in breast cancer, and the existing studies are mainly per-formed in vitro. The correlation found between SREBP-1 and FASN expression is weak [13], although it has to be kept in mind that the activ-ity of SREBP-1 is regulated at the post-transla-tional level. Yang et al. have performed in vitro studies indicating that SREBP-1 is regulated by the MAPK and PI3K pathways and that the transcription of FASN is regulated by SREBP-1 [13,14]. However, more recently, Yoon et al. have reported that in breast cancer cells FASN and ACACA are not regulated by SREBP-1, instead,

the key regulator is HER2, and these proteins are regulated at the translational level by the mTOR signaling pathway [15]. Therefore, the exact role of SREBP-1 in regulating FASN and ACACA is still unclear in breast cancer.

In addition, new players have been identified that regulate the activity of ACC1 and hence the whole lipogenic pathway. One of the enzymes regulated by PI3K/AKT together with SREBP-1 is stearoyl-CoA desaturase 1 (SCD1), whose function is to produce monounsaturated fatty acids from saturated ones. The expression of SCD1 is increased in several cancers and it has been reported in lung cancer cells that SCD1 can regulate the activity of ACC1 by regulating the cellular content of palmitic acid, which is a negative regulator of ACC1, and also by con-trolling the phosphorylation status of AMPK, which in turn can regulate ACC1 activity by phosphorylation [16]. Inhibition of expression or activity of SCD1 has been reported to inhibit breast cancer cell growth [17]. SPOT14 (S14, THRSP) is amplified in 15–20% of breast cancers, and its expression correlates with the expression of ACC1 as well as with tumor grade and decreased disease-free survival [18,19]. The expression of SPOT14 is regulated by hormones and SREBP-1 [20] and the link between the lipogenic phenotype (i.e., ACC1 and SPOT14 expression) became more evident when the crys-tal structure of SPOT14 was recently revealed [21]. It was shown that SPOT14 can form het-erodimers with MIG12 protein, which can then induce the polymerization of ACC1 and thereby regulate its activity [21]. The link between SCD1

De novofatty acid synthesis

ACC1

Acetyl-CoA Malonyl-CoA

FASN

Palmitate

Regulation ofredox balance

Membranesynthesis

Proteinpalmitoylation

Formation oflipid rafts

Increased resistance to chemotherapy

PC branch of theKennedy pathway

Choline

ChoK

CT

CDP-choline

CHPT1

PCho

PC

oo–

Figure 1. Production of fatty acids and membrane phospholipids in cancer cells. (A) De novo synthesis of fatty acids may contribute to several different cellular processes in cancer. (B) Illustration of the PC branch of the Kennedy pathway that produces PC. ACC1: Acetyl-CoA carboxylase; ChoK: Choline kinase; CHPT: CDP-choline diacylglycerolphosphocholinetransferase; CT: CTP-choline cytidylyltransferase; FASN: Fatty acid synthase; PC: Phosphatidylcholine; PCho: Phosphocholine.

Review | Hilvo & Orešič

www.futuremedicine.com 179future science group

and SPOT14 and ACC1 activity may raise the possibility of exploiting these enzymes to reduce lipogenesis in cancer cells.

Upregulation of membrane phospholipid metabolism in breast cancerThe glycerophospholipids, phosphatidyl choline (PC) and phosphatidylethanolamine, account for more than half of the total phospholipid spe-cies in eukaryotic cell membranes, and de novo synthesis of these phospholipids occurs via the so-called Kennedy pathway [22]. The pathway has analogous branches for PC and phospha-tidylethanolamine production, but as the PC branch is investigated more heavily in cancer, this article focuses on the PC branch only (Figure 1B). Choline kinase (ChoK) catalyzes the phosphorylation of free choline using ATP as a phosphate donor, and thus produces phos-phocholine. At least three isoforms of ChoK exist and among these the a-isoform is tumor related [23]. CTP-choline cytidylyltransferase then catalyzes the synthesis of CDP-choline from phosphocholine and CTP. CDP-choline is used by CDP-choline diacyl glycerol phos-phocholinetransferase as a substrate to pro-duce PC. The activity and expression of ChoK is increased in approximately 40% of breast tumors when compared with levels in healthy tissues, and the overexpression is highest in tumors of high grade and estrogen recep-tor (ER)-negative status [24]. It has also been demonstrated in cell line studies that ChoK is both necessary and sufficient for growth factor-induced proliferation and that ChoK inhibi-tors show strong antitumor activity in human breast cancer xenografts [25]. In addition to its recognized therapeutic value, choline metabo-lism also offers diagnostic opportunities. Not only the PC content but also the total choline-containing compounds, consisting of choline, phosphocholine and glycerophosphocholine, are increased in tumors, and these compounds can be detected noninvasively by magnetic reso-nance spectroscopy. Using magnetic resonance spectroscopy combined with MRI, breast can-cer can be separated from benign cases with rea-sonable specificity and sensitivity [26]. Another method for imaging is PET, in which, tradi-tionally, the tumors have been visualized using fluorodeoxyglucose (18F), although labeled [11C]choline has recently become an alternative to this compound. A study in ER-positive breast cancer patients showed that breast tumors can

be visualized using [11C]choline [27], which may also be used to observe patient responses to treatment with trastuzumab [28].

The regulation of lipid metabolism in breast cancer has been investigated in many stud-ies at the level of genes and proteins. Studies of molecular lipids, the end product of lipid metabolism, have become possible over the past decade with developments in the emerg-ing field of lipid omics. High-throughput lipid-omic methods allow for profiling of hundreds of molecular lipids in a single experiment [29]. For example, the authors’ investigations on the lipidomic changes in breast cancer tis-sue samples revealed that the phospholipids were increased, especially in the grade 3 and ER-negative tumors [30]. Specifically, the PCs containing major or minor products of FASN (i.e., saturated fatty acids C14:0, C16:0 and C18:0) were found at higher levels in these aggressive tumors, and also predicted poorer overall survival for the patients. The finding that these phospho lipids were particularly increased in ER-negative tumors was surpris-ing, as it has been shown that estrogen should upregulate de novo synthesis of fatty acids [31]. However, these results were consistent with the above-mentioned findings that ChoK activity is associated with ER negativity. In addition, in the lipidomic study HER2 was not shown to affect the lipid profiles in the clinical breast cancer samples, although the interconnection between HER2 signaling and FASN activity has been shown, as mentioned earlier [15]. Thus, it is evident that more studies, especially in vivo and clinical studies, of the lipid metabolism at gene, protein and metabolite levels, are needed to solve the discrepancies between the different studies considering the regulation of fatty acid and lipid metabolism in breast cancer.

The role & therapeutic potential of de novo fatty acid synthesis in breast cancerThe most obvious reason for cells to overproduce fatty acids by the de novo pathway is that these fatty acids are used to build membrane phospho-lipids, which are needed in rapidly proliferat-ing cancer cells. In addition, the phospholipids are needed in building lipid rafts, which have an important role in cancer cell signaling, and thereby de novo fatty acid synthesis may also indirectly affect cell signaling processes [32]. Protein palmitoylation affects, for example, the Wnt1/b-catenin pathway in prostate cancer, and

Regulation of lipid metabolism in breast cancer provides diagnostic & therapeutic opportunities | Review

Clin. Lipidol. (2012) 7(2)180 future science group

also in this way the major product of the lipo-genic pathway, palmitate, may contribute to cell signaling [33]. However, the production of fatty acids may not be the only role of the de novo pathway, as this pathway has also been suggested to have relevance in cellular redox balance regu-lation. Cancer cells are usually under glycolytic and hypoxic conditions, in which the cells can-not fully oxidize the reducing equivalents that are formed. However, in lipogenesis NADPH is oxidized and therefore it may consume the exces-sive amounts of reducing compounds present in the cancer cell [3].

Lipogenesis may also have a profound role in the sensitivity of cancers to treatment, as shown by a recent study considering prostate cancer [34]. The study revealed that increased levels of satu-rated fatty acids in cell membranes make cancer cells less susceptible to oxidative stress. This is important because oxidized phospholipids have been shown to be crucial mediators for apopto-sis and unsaturated fatty acids are more easily oxidized [35]. Moreover, increased membrane saturation decreases both the lateral (within the plane of one membrane leaflet) and trans-versal (movement from one membrane leaflet to another) mobility of lipids, which then leads to diminished uptake of chemotherapeutic reagents into cancer cells [34]. The different roles of this pathway are summarized in Figure 1A.

The upregulation of the de novo fatty acid synthesis pathway in cancer, as compared with normal tissue, naturally suggests that target-ing this pathway may be a therapeutic option. Indeed, inhibitors for the FASN enzyme have been generated, including cerulenin, C75 and orlistat. Cerulenin is an antibiotic derived from the fungus Cephalosporum caerulens, which has shown promising antitumoral effects in xeno-graft models, although its chemical instability unfortunately limits its clinical use. C75, a syn-thetic derivative of cerulenin, is more stable and also effective in xenografts [5,36]. The use of C75, however, is limited by the fact that this inhibi-tor has been reported to cause dramatic weight loss, which is explained by the fact that, besides FASN inhibition, C75 also stimulates CPT1 protein, which catalyzes fatty acid oxidation in mitochondria [37,38]. Orlistat is a US FDA-approved drug used for treating obesity, which also inhibits FASN [39]. In vitro breast cancer studies have shown interesting results for this inhibitor but development of its chemical struc-ture would be needed because orlistat possesses poor solubility [40]. Other potential FASN

inhibitors are the antibiotic triclosan and certain polyphenolic compounds [40].

There are several mechanisms by which FASN inhibitors can reduce cancer cell growth. One hypothesis is that the inhibition of FASN reduces de novo fatty acid synthesis, and that the cancer cells are therefore lacking an adequate pool of fatty acids for building membranes for new cells. This hypothesis is supported by stud-ies showing that the addition of palmitate or oleate to the cell culture medium restores cell viability despite fatty acid inhibition, whereas this effect is not seen with non-FASN-related fatty acids [41]. Another hypothesis is that accu-mulation of malonyl-CoA, the product of ACC1 and the reactant of FASN, is cytotoxic to cancer cells. This hypothesis is supported by a study in which ACC1 inhibition was not cytotoxic to breast cancer cells, and inhibition of both ACC1 and FASN showed reduced cytotoxicity as com-pared with the inhibition of FASN alone [36]. However, another study using RNAi showed that knock-down of both ACACA and FASN reduces proliferation and induces apoptosis in cancer cells, supporting the idea that malonyl-CoA is not a prerequisite for cytotoxicity but that fatty acid synthesis is the crucial event [42]. There is also evidence for other cytotoxic mechanisms. It has been shown that inhibition of FASN can increase chemotherapy-induced cytotoxicity by activating proapoptotic and inactivating antiapoptotic signaling cascades in breast cancer cells [43]. p53 protein plays an important role in cellular responses to nongeno-toxic metabolic stress, and it also has connection to FASN, as in breast cancer cells FASN inhibi-tion together with p53 silencing leads to apopto-sis, whereas FASN inhibition with functioning p53 leads only to growth arrest [44]. FASN gene silencing causes wide changes in the expression of genes related to metabolism, cell survival and proliferation as well as DNA replication processes [45]. Finally, inhibition of FASN may also decrease HER2 activity, because silencing of FASN leads to downregulation of HER2 mRNA [46], although other data show that actually HER2 regulates FASN [15]. Data from colon cancer studies indicate that inhibition of FASN causes stress in the endoplasmic reticu-lum, which may also explain the antitumoral properties of FASN inhibitors [47]. It is likely that several nonexclusive mechanisms exist, and therefore more studies are still needed to elabo-rate their relative importance in the antitumoral properties of FASN inhibition.

Review | Hilvo & Orešič

www.futuremedicine.com 181future science group

LPA regulates several cellular processesThe synthesized phospholipids contain fatty acyl chains coming from both de novo synthesis and nutrition. The phospholipids are not only static molecules residing in the cell membrane, as, for example, cleavage of their fatty acyl chains has important consequences in cell signaling. This section discusses LPA and the next section dis-cusses eicosanoids, both of which can be formed by cleavage of membrane phospholipids and have important roles in cancer cell signaling.

The role of LPA in the initiation and progres-sion of several cancers has been investigated, and thorough reviews of this lipid and cancer have been published [48,49]. LPA consists of a glycerol backbone, a fatty acid bonded at the sn1 or sn2 position, and a phosphate group bonded at the sn3 position. LPA can be produced in two ways; either by hydrolysis of one fatty acyl chain from phosphatidic acid or by hydrolysis of the choline group from lysophosphatidylcholine [48]. The latter reaction is catalyzed by the lysophospholi-pase D activity of the extracellular enzyme auto-taxin (ATX). Even before the lysophospholipase activity of ATX was known, ATX was reported to be associated with cancer progression. LPA exerts its effect by binding to G protein-coupled receptors, and at the moment six of these have been reported (LPA

1–6) [50]. The LPA receptors

affect many cancer signaling pathways, such as PI3K, MAPK and Wnt/b-catenin, and thereby affect processes related to, for example, cell pro-liferation and survival, angiogenesis and inflam-mation [49], as shown in Figure 2. Both ATX and the LPA receptors are overexpressed in breast cancer as compared with normal mammary epi-thelial cells, and the overexpression of ATX and LPA

1–3 receptors has been reported to induce

mammary carcinomas in mice [51]. LPA1 is also

associated with cytokine production and bone metastasis in breast cancer [52]. In vitro studies have revealed that ATX protects breast cancer cells against Taxol-induced apoptosis [53].

Because of their crucial role in cancer, LPA, ATX and LPA receptors provide attractive tar-gets for cancer therapeutics. Zhang et al. have generated LPA analogs that perform a dual role; they inhibit the lysophospholipase D activity of ATX as well as abrogate signaling through the LPA receptor. These inhibitors were found to reduce cell migration and invasion by breast tumors in vivo [54]. Lpath Inc. (CA, USA) has generated monoclonal antibodies recognizing LPA. These antibodies are in preclinical trials;

the concept is that the monoclonal antibody neutralizes the action of LPA, thereby prevent-ing its function in cancer [201]. LPA is also a potential diagnostic biomarker, as most patients with ovarian or other gynecologic cancer show increased concentrations of LPA circulating in the plasma [55]. However, in breast cancer patients no elevated levels have been observed [55], and therefore, based on the available evi-dence, LPA is more likely to be beneficial as a therapeutic target rather than a diagnostic tool in this cancer type.

Role of eicosanoids in breast cancerThe term eicosanoid is used to refer to a group of oxygenated, 20-carbon fatty acids, and these molecules have an important role in cellular signaling. Eicosanoids differ from the lipids in previous sections in that they are essential (i.e., the precursors come solely from the diet). Even today, reliable measurements of eicosanoids from in vivo and clinical samples are very chal-lenging – despite the availability of sensitive instruments and analytical techniques – due to the sensitivity of many eicosanoids to sample collection and pretreatment. Most of the infor-mation regarding these molecules in cancer, therefore, still comes from studies investigat-ing the enzymes and receptors related to these signaling molecules, rather than the molecules themselves.

Eicosanoids have been investigated extensively in cancer, and a thorough review considering these molecules has been published by Wang and DuBois [56]. The precursors of eicosanoids are stored as fatty acyl chains in membrane phospholipids, from which they are cleaved for eicosanoid synthesis. Arachidonic acid, an w-6 fatty acid with four double bonds, is the major precursor for various eicosanoids and it can be converted by the COX pathway to prostanoids (prostaglandins, thromboxanes and prostacy-clins), by the lipoxygenase (LOX) pathway to leukotrienes and hydroxyeicosatetraenoic acids and by the P450 pathway to epoxyeicosatrienoic acids and hydroxyperoxyeicosatetraenoic acids.

Under normal conditions in healthy cells COX-2 is mainly unexpressed but during inflammation its levels are rapidly elevated lead-ing to increased production of prostaglandins. The significance of COX-2 in breast cancer is still highly controversial, although expression studies were carried out in the 1990s [57,58]. Some studies have reported high expression of COX-2 in most breast cancer samples with no expression

Regulation of lipid metabolism in breast cancer provides diagnostic & therapeutic opportunities | Review

Clin. Lipidol. (2012) 7(2)182 future science group

in normal breast tissue [57], whereas other studies have shown that COX-2 is upregulated only in some breast cancer samples [58] or downregulated in breast cancer as compared with healthy tis-sue [59]. The use of NSAIDs inhibiting COX in reducing breast cancer risk is also under debate, although there is evidence that NSAIDs may be beneficial in reducing cancer risk [60].

Regardless of the specific role of COX-2, it is nevertheless clear that one of the prostaglan-dins, prostaglandin E2 (PGE2), has many roles in breast cancer. It is known that PGE2 can increase the breast cancer cell proliferation rate by inducing aromatase enzyme expression, and thus local synthesis of estrogens [61], as well as promote invasion of breast cancer cells to lymph nodes by upregulating the CC chemokine recep-tor CCR7 [62]. Myeloid-derived suppressor cells, which are found in cancers and shown to inhibit T-cell activity, thus leading to immunosuppres-sion, are induced by PGE2 in a breast cancer mouse model [63]. Finally, PGE2 can promote angiogenesis in breast cancer by stimulating angiogenic regulatory genes [64] and affecting endothelial cell motility [65]. Based on this evi-dence, which is far from comprehensive, it can be concluded that PGE2 affects the processes of cell proliferation, cell invasion, immunology and angiogenesis in breast cancer (Figure 2). However, the role of PGE2 may be even more complex. Recently the authors have shown that

the key enzyme degrading PGE2, 15-hydroxy-prostaglandin dehydrogenase, has a dual role in breast cancer. On the one hand previous stud-ies have indicated HPGD as a tumor-suppressor gene and downregulated in cancer as compared with normal tissues [66], but on the other hand it is highly upregulated in those breast cancer patients with poor outcome [67].

The LOX enzymes metabolize arachi-donic acid to hydroxyperoxyeicosatetraenoic acids that can be further reduced to hydroxy-eicosatetraenoic acids. In the case of 5-LOX, hydroxyperoxyeicosatetraenoic acid is further metabolized to leukotriene A4, which can be further transformed to other leukotrienes [68]. 15-LOX-1 and 15-LOX-2 are suggested to have tumor-suppressor roles in breast cancer, as their expression is reduced in malignant breast cells and tissues compared with healthy ones [69,70], whereas the expression of 5-LOXAP is increased in breast cancer and together with 12-LOX also associated with poor survival of the patient [71]. Indeed, inhibition of both 5-LOX and 12-LOX reduces cell proliferation and induces apoptosis in breast cancer cells in vitro [72]. By contrast, the activity of 15-LOX-2 has been implicated in the increased adhesion of breast cancer cells to the extracellular matrix [73]. Thus, in summary, 5-LOX and 12-LOX possess protumorigenic activity while 15-LOX-1 and 15-LOX-2 pos-sess antitumorigenic activity in breast cancer (Figure 3) [68].w-6 fatty acids form the majority of the poly-

unsaturated fatty acids in the western diet. In addition to the pathways derived from arachi-donic acid, there can also be parallel competing pathways derived from eicosapentaenoic acid, which is an w-3 fatty acid. In general, arachi-donic acid-derived eicosanoids promote inflam-mation, whereas eicosapentaenoic acid-derived eicosanoids are less inflammatory or even anti-inflammatory, and eicosapentaenoic acid com-petes with arachidonic acid for prostaglandin and leukotriene synthesis [74]. w-3 fatty acids have also shown promising results in breast cancer. It was shown two decades ago, in mice, that diets rich in w-3 fatty acids can reduce breast cancer cell growth and metastasis [75]. It has been shown that dietary w-3 fatty acids can be found in breast tis-sue [76], and that w-3 fatty acids have a protective effect in breast adipose tissue [77]. This suggests that lipid metabolism can be seen not only as a promising source of targets for drug development but also as a source of novel disease prevention strategies via nutritional interventions.

Signaling

Proliferation

Arachidonic acid PGE2

COX-2

AngiogenesisInvasion

Inflammation andimmunologicalprocesses

LPA receptor

LPALPCATX/LPC

Figure 2. Effects of lysophosphatidic acid and prostaglandin E2 in breast cancer. LPA: Lysophosphatidic acid; LPC: Lysophosphatidylcholine.

Review | Hilvo & Orešič

www.futuremedicine.com 183future science group

Sphingolipids & glycosphingolipids are either pro- or antitumorigenicSphingolipids form a class of lipids of which the common structural feature is the sphingosine backbone. It has been suggested that the balance between different sphingolipids affects the char-acteristics of a cancer cell (Figure 3). For exam-ple, ceramides and sphingosines are associated with cell growth arrest and cell death, whereas glycosylated sphingolipids together with sphin-gosine-1-phosphate (S1P) are involved in cellular proliferation, survival and drug resistance [78]. Actually, the so-called ceramide–sphingosine–S1P rheostat was proposed some time ago [79]. In this model, ceramide and sphingosine are apoptotic molecules while S1P promotes cell survival [79].

In cells, ceramides can be generated by de novo synthesis, either by condensation of serine and palmitoyl-CoA, by catabolism of sphingomyelin or glycosphingolipids or by desphosphorylation of phosphorylated ceramides [78]. In many can-cers chemotherapeutic agents increase ceramide levels, thereby leading to cell death. Ceramide levels can also be increased exogenously, and exogenous ceramide analogs induce apopto-sis in several cancers, including breast cancer, and also show selectivity towards cancer cells versus normal breast epithelial cells [80,81]. The hydrophobicity and physicochemical properties of short-chain ceramides limit their use in vivo, but delivery of these ceramides using liposomal carriers was reported to be effective in a breast cancer mouse model [82]. The downregulation of ceramides, however, is not straightforward, as it has also been reported that ceramide synthase, and thereby some long-chain ceramides, are upregulated in breast cancer tissue as compared with benign breast tissue [83].

Ceramide can be converted to sphingosine by the enzyme ceramidase and then sphingo-sine kinase (SK) converts the sphingosine to S1P. SK exists as two isozymes, SK1 and SK2, and of these two isozymes the role of SK1 in cancer is clearer. In breast cancer, SK1 is highly expressed, especially in ER-negative tumors, but also among ER-positive patients high expression of SK1 is associated with poor outcome [84]. High expression of SK2 in breast cancer has not been reported in the literature, and there are also controversial results on its effect on cancer cells. On the one hand SK2 can induce apoptosis in cells [85] and may also have an opposing role in the regulation of ceramide biosynthesis com-pared with SK1 [86], but on the other hand the

activity of SK2 has been implicated in enhanc-ing breast cancer tumorigenesis, for instance by affecting tumor-associated macrophages [87] or by participating in the EGF-catalyzed migration of cells [88].

S1P exerts its effects by binding to five G pro-tein-coupled receptors, named S1PR

1–5. S1P has

numerous roles in the cell; it is associated with cancer cell survival, growth, motility, trans-formation, neoangiogenesis and inflammation [89]. There seems to be an interesting interac-tion between S1P and hypoxia. The expression of SK1 is activated by hypoxia-inducible factors [90], but SK1 also regulates the levels of HIF1-a, as shown in breast cancer cells [91]. Studies in glioma cells have revealed that HIF2-a has an important role in the regulation of SK1 expres-sion and, interestingly, S1P released to the extra-cellular environment induces neoangiogenesis [92]. In fact, S1P has been shown to display a synergistic effect with VEGF in neoangiogenesis [93] and, indeed, monoclonal antibodies against S1P have been shown to inhibit angiogenesis in tumor xenograft models [94]. In breast cancer S1P is associated with hormonal regulation, since it is involved in a pathway in which estrogen stimu-lates the activity of the EGF receptor [95]. In addition, recent reports indicate a role of S1P in the generation of tamoxifen resistance in breast cancer. Studies with cell lines showed that over-expression of SK1 results in enhanced cell prolif-eration and resistance to tamoxifen-induced cell growth arrest and apoptosis, and moreover that tamoxifen-resistant cells exhibit higher expres-sion of SK1 [96]. Tamoxifen responsiveness can be rescued by silencing SK1 expression with RNAi [96]. These results are supported by clini-cal findings showing that high expression of SK1 and the S1P

1 and S1P

3 receptors predict a poorer

Prot

umor

igen

ic Proapoptotic

Multidrug resistanceGlycosylated ceramides

S1P

5-LOX

12-LOX

Ceramides

Sphingosine

15-LOX-1

15-LOX-2

Figure 3. The balance of sphingolipids and the activity of lipoxygenase enzymes affect the malignancy state of the cell. Glycosylated ceramides are associated with multidrug resistance of breast cancer cells.

Regulation of lipid metabolism in breast cancer provides diagnostic & therapeutic opportunities | Review

Clin. Lipidol. (2012) 7(2)184 future science group

prognosis in ER-positive breast cancer patients [97]. Interestingly, S1P has also been implicated in epigenetic regulation, as SK2 associates with histone H3 and histone deacetylases to produce S1P, which regulates histone acetylation and can thereby affect cell cycle progression [98]. The role of S1P in several cellular processes has made it an interesting target for cancer therapeutics. There are three major strategies for drug development: S1P-specific antibodies that neutralize the extra-cellular action of S1P; SK inhibitors that inhibit the production of S1P; and inhibitors that block the activity of S1P receptors. These approaches have yielded promising results in vitro and in vivo, but so far it seems that, in the context of cancer, the only one that has proceeded to a Phase I clinical trial is the antibody approach, which has also shown promising results in mouse breast cancer models [201].

Glycosphingolipids include cerebrosides, gan-gliosides and globosides. Cerebrosides consist of a ceramide with a hexose at the 1-hydroxyl moiety. The sugar residue can be either glu-cose or galactose, and therefore cerebrosides can be classified as either glucocerebrosides or galacto cerebrosides [99]. Glucosylceramide syn-thase (GCS) is the enzyme producing glucosyl-ceramide (glucocerebroside) by catalyzing the transfer of glucose to ceramide [78]. The activity of GCS, and thereby the increased concentration of glycosylated ceramides, is thought to poten-tiate the development of multidrug resistance in cancer cells [100,101]. Interestingly, however, in clinical breast cancer samples the expression of GCS is associated with lower histological grading and ER-positive status [102]. This is also supported by the authors’ own findings show-ing dramatically decreased concentrations of cer-tain glycosylated ceramides in tumors of higher grade and ER-negative status [30]. Nevertheless, inhibition of the activity of GCS downregulates P-glycoprotein and resensitizes breast cancer cells to, for example, vinblastine [103].

Globosides are glycosphingolipids with two or more sugars, usually glucose, galactose or N-acetylgalactosamine, but they have not been investigated extensively in the context of breast cancer. Acidic glycosphingolipids containing one or more sialic acid (e.g., N-acetylneuraminic acid) residue(s) in their carbohydrate moiety are called gangliosides [104]. Gangliosides are the most complex sphingolipids, and those with one sialic acid residue are referred to as the GM series, those with two as the GD series, those with three, the GT series, and those with four,

the GQ series. Gangliosides have diverse roles in cancer, as they have been implicated in immuno-logical processes, angiogenesis, cell adhesion and motility as well as cell signaling [105]. However, gangliosides have not been investigated heavily in breast cancer. It has been reported that the overall ganglioside levels are higher as compared with normal breast tissue, and GM3-, GD3- and GT3-containing gangliosides, not present in nor-mal breast tissue, are found in malignant tissues [106]. Indeed, ganglioside-based vaccines have been developed for cancer treatment, and two of these are racotumomab and NGcGM3/VSSP vaccine [107]. Racotumomab is an anti-idiotype monoclonal antibody, which means that it is a mirror image of the original antibody, and can act as an antigen, inducing a response against the original antigen. The NGcGM3/VSSP vaccine is a conjugation of the ganglioside with bacte-rial proteoliposomes. These therapies, developed by the RECOMBIO company (Madrid, Spain), have not caused severe side effects in patients and have progressed into Phase III clinical trials [107].

ConclusionIt is apparent that the regulation of lipid metabo-lism in malignant tissues is very different from that occurring in normal tissues. The observed changes in lipids, such as membrane lipids, may also provide an explanation for the comorbidity of obesity and cancer. In fact, during the last few years, interesting studies have emerged sug-gesting a link between the metabolic syndrome and cancer. Hirsch et al. investigated, in cell lines, the changes in gene expression that occur during the malignant transformation [108]. This study, which also included transformed mam-mary epithelial cells, revealed that many of the genes differentially regulated during transforma-tion are also related to lipid metabolism, inflam-mation, obesity and the metabolic syndrome. Moreover, several drugs designed for the treat-ment of metabolic diseases showed either com-plete suppression or delay in tumor growth in mouse models [108].

Therefore, lipid metabolism provides thera-peutic opportunities not only for drug develop-ment but also for lifestyle changes, including nutrition, which may be considered in cancer prevention.

Future perspectiveThe importance of lipid metabolism in the cancer field has been largely neglected until recently, possibly because the other metabolic

Review | Hilvo & Orešič

www.futuremedicine.com 185future science group

changes in cancer are highly prominent and due to technological limitations. However, recent developments in the lipidomics field have made lipid metabolism investigations more tempt-ing, and therefore the authors believe that in the future lipid metabolism will become a more important field in cancer drug develop-ment and diagnostics. The authors believe that in the future:

nThe development of analytical methods will lead to increased knowledge of lipid metabo-lism in cancer and also provide diagnostic opportunities;

nThe integration of data from gene, protein and lipid studies will enable a more comprehensive understanding of the metabolism in malig-nant cells and also reveal new signaling pathways contributing to carcinogenesis;

nThe importance of nutrition on lipid metabo-lism and cancer pathogenesis will become more clear, which may lead to new nutritional recommendations;

nThe first clinical trials of drugs targeting lipid metabolism will be finished and prove that lipid metabolism is an attractive target for future drug development.

Financial & competing interests disclosureM Hilvo was financially supported by the Academy of Finland postdoctoral researcher’s fellowship 138201. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Executive summary

Fatty acid metabolism in normal & malignant tissues � In normal tissues, de novo synthesis of fatty acids occurs in a limited number of tissues and is under nutritional control.

� In tumors, upregulation of the de novo fatty acid synthesis pathway is a universal feature and out of nutritional control.Upregulation of membrane phospholipid metabolism in breast cancer � The activity and expression of choline kinase and the Kennedy pathway is increased in breast tumors and leads to increased membrane phospholipid synthesis.

� Phospholipids containing saturated de novo synthesized fatty acids are especially increased in grade 3 and estrogen receptor-negative breast cancers.

The role & therapeutic potential of de novo fatty acid synthesis in breast cancer � Inhibitors of fatty acid synthase may exert their antitumoral activities by affecting membrane phospholipid synthesis, malonyl-CoA accumulation, apoptotic pathways, HER2 activity and stress in the endoplasmic reticulum.

Lysophosphatidic acid regulates several cellular processes � Lysophosphatidic acid exerts its function on cells via G protein-coupled receptors, and affects cell proliferation and survival, angiogenesis and inflammation.

Role of eicosanoids in breast cancer � The role of COX-2 in breast cancer is controversial.

� Prostaglandin E2 promotes carcinogenesis by multiple mechanisms.

� Lipoxygenase isozymes are either pro- or antitumorigenic.Sphingolipids & glycosphingolipids are either pro- or antitumorigenic � The balance between different sphingolipids determines the malignancy of cells.

� According to the so-called ceramide–sphingosine–sphingosine-1-phosphate rheostat model, ceramide and sphingosine are apoptotic molecules while sphingosine-1-phosphate promotes cell survival.

� Glycosphingolipids are associated with the multidrug resistant phenotype of cancer cells.

ReferencesPapers of special note have been highlighted as:n of interestnn of considerable interest

1 Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 4(11), 891–899 (2004).

2 Griffin JL, Shockcor JP. Metabolic profiles of cancer cells. Nat. Rev. Cancer 4(7), 551–561 (2004).

3 Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat. Rev. Cancer 7(10), 763–777 (2007).

4 Sul HS, Wang D. Nutritional and hormonal regulation of enzymes in fat synthesis: studies of fatty acid synthase and mitochondrial glycerol-3-phosphate acyltransferase gene transcription. Annu. Rev. Nutr. 18, 331–351 (1998).

5 Kuhajda FP, Pizer ES, Li JN, Mani NS, Frehywot GL, Townsend CA. Synthesis and antitumor activity of an inhibitor of fatty acid synthase. Proc. Natl Acad. Sci. USA 97(7), 3450–3454 (2000).

6 Foretz M, Guichard C, Ferre P, Foufelle F. Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes. Proc. Natl Acad. Sci. USA 96(22), 12737–12742 (1999).

Regulation of lipid metabolism in breast cancer provides diagnostic & therapeutic opportunities | Review

Clin. Lipidol. (2012) 7(2)186 future science group

7 Kersten S. Mechanisms of nutritional and hormonal regulation of lipogenesis. EMBO Rep. 2(4), 282–286 (2001).

8 Shimomura I, Hammer RE, Richardson JA et al. Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adipose tissue: model for congenital generalized lipodystrophy. Genes Dev. 12(20), 3182–3194 (1998).

9 Sekiya M, Yahagi N, Matsuzaka T et al.SREBP-1-independent regulation of lipogenic gene expression in adipocytes. J. Lipid Res. 48(7), 1581–1591 (2007).

10 Milgraum LZ, Witters LA, Pasternack GR, Kuhajda FP. Enzymes of the fatty acid synthesis pathway are highly expressed in in situ breast carcinoma. Clin. Cancer Res. 3(11), 2115–2120 (1997).

11 Alo PL, Visca P, Marci A, Mangoni A, Botti C, Di Tondo U. Expression of fatty acid synthase (FAS) as a predictor of recurrence in stage I breast carcinoma patients. Cancer 77(3), 474–482 (1996).

12 Alo PL, Visca P, Trombetta G et al. Fatty acid synthase (FAS) predictive strength in poorly differentiated early breast carcinomas. Tumori 85(1), 35–40 (1999).

13 Yang YA, Morin PJ, Han WF et al. Regulation of fatty acid synthase expression in breast cancer by sterol regulatory element binding protein-1c. Exp. Cell Res. 282(2), 132–137 (2003).

14 Yang YA, Han WF, Morin PJ, Chrest FJ, Pizer ES. Activation of fatty acid synthesis during neoplastic transformation: role of mitogen-activated protein kinase and phosphatidylinositol 3-kinase. Exp. Cell Res. 279(1), 80–90 (2002).

15 Yoon S, Lee MY, Park SW et al. Up-regulation of acetyl-CoA carboxylase alpha and fatty acid synthase by human epidermal growth factor receptor 2 at the translational level in breast cancer cells. J. Biol. Chem. 282(36), 26122–26131 (2007).

16 Scaglia N, Chisholm JW, Igal RA. Inhibition of stearoylCoA desaturase-1 inactivates acetyl-CoA carboxylase and impairs proliferation in cancer cells: role of AMPK. PLoS ONE 4(8), e6812 (2009).

17 Luyimbazi D, Akcakanat A, McAuliffe PF et al. Rapamycin regulates stearoyl CoA desaturase 1 expression in breast cancer. Mol. Cancer Ther. 9(10), 2770–2784 (2010).

18 Moncur JT, Park JP, Memoli VA, Mohandas TK, Kinlaw WB. The ‘Spot 14’ gene resides on the telomeric end of the 11q13 amplicon and is expressed in lipogenic breast cancers: implications for control of tumor metabolism. Proc. Natl Acad. Sci. USA 95(12), 6989–6994 (1998).

19 Wells WA, Schwartz GN, Morganelli PM, Cole BF, Gibson JJ, Kinlaw WB. Expression of ‘Spot 14’ (THRSP) predicts disease free survival in invasive breast cancer: immunohistochemical ana lysis of a new molecular marker. Breast Cancer Res. Treat. 98(2), 231–240 (2006).

20 Martel PM, Bingham CM, McGraw CJ et al. S14 protein in breast cancer cells: direct evidence of regulation by SREBP-1c, superinduction with progestin, and effects on cell growth. Exp. Cell Res. 312(3), 278–288 (2006).

21 Colbert CL, Kim CW, Moon YA et al. Crystal structure of Spot 14, a modulator of fatty acid synthesis. Proc. Natl Acad. Sci. USA 107(44), 18820–18825 (2010).

22 Gibellini F, Smith TK. The Kennedy pathway – de novo synthesis of phosphatidylethanolamine and phosphatidylcholine. IUBMB Life 62(6), 414–428 (2010).

23 Glunde K, Bhujwalla ZM, Ronen SM. Choline metabolism in malignant transformation. Nat. Rev. Cancer 11(12), 835–848 (2011).

24 Ramirez de Molina A, Gutierrez R, Ramos MA et al. Increased choline kinase activity in human breast carcinomas: clinical evidence for a potential novel antitumor strategy. Oncogene 21(27), 4317–4322 (2002).

25 Ramirez de Molina A, Banez-Coronel M, Gutierrez R et al. Choline kinase activation is a critical requirement for the proliferation of primary human mammary epithelial cells and breast tumor progression. Cancer Res. 64(18), 6732–6739 (2004).

26 Katz-Brull R, Lavin PT, Lenkinski RE. Clinical utility of proton magnetic resonance spectroscopy in characterizing breast lesions. J. Natl Cancer Inst. 94(16), 1197–1203 (2002).

27 Contractor KB, Kenny LM, Stebbing J et al. [11C]choline positron emission tomography in estrogen receptor-positive breast cancer. Clin. Cancer Res. 15(17), 5503–5510 (2009).

28 Kenny LM, Contractor KB, Hinz R et al. Reproducibility of [11C]choline-positron emission tomography and effect of trastuzumab. Clin. Cancer Res. 16(16), 4236–4245 (2010).

29 Orešič M, Hänninen VA, Vidal-Puig A. Lipidomics: a new window to biomedical frontiers. Trends Biotechnol. 26(12), 647–652 (2008).

30 Hilvo M, Denkert C, Lehtinen L et al. Novel theranostic opportunities offered by characterization of altered membrane lipid

metabolism in breast cancer progression. Cancer Res. 71(9), 3236–3245 (2011).

n Describes the global lipidomic changes in clinical breast cancer samples.

31 Rezaul K, Thumar JK, Lundgren DH et al. Differential protein expression profiles in estrogen receptor-positive and -negative breast cancer tissues using label-free quantitative proteomics. Genes Cancer 1(3), 251–271 (2010).

32 Swinnen JV, Van Veldhoven PP, Timmermans L et al. Fatty acid synthase drives the synthesis of phospholipids partitioning into detergent-resistant membrane microdomains. Biochem. Biophys. Res. Commun. 302(4), 898–903 (2003).

33 Fiorentino M, Zadra G, Palescandolo E et al. Overexpression of fatty acid synthase is associated with palmitoylation of Wnt1 and cytoplasmic stabilization of beta-catenin in prostate cancer. Lab. Invest. 88(12), 1340–1348 (2008).

34 Rysman E, Brusselmans K, Scheys K et al. De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res. 70(20), 8117–8126 (2010).

n Provides important evidence on the mechanism by which de novo fatty acid synthesis may affect the sensitivity of cells to chemotherapy.

35 Fruhwirth GO, Hermetter A. Mediation of apoptosis by oxidized phospholipids. Subcell. Biochem. 49, 351–367 (2008).

36 Pizer ES, Thupari J, Han WF et al. Malonyl-coenzyme-A is a potential mediator of cytotoxicity induced by fatty-acid synthase inhibition in human breast cancer cells and xenografts. Cancer Res. 60(2), 213–218 (2000).

37 Loftus TM, Jaworsky DE, Frehywot GL et al. Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science 288(5475), 2379–2381 (2000).

38 Kuhajda FP, Landree LE, Ronnett GV. The connections between C75 and obesity drug-target pathways. Trends Pharmacol. Sci. 26(11), 541–544 (2005).

39 Kridel SJ, Axelrod F, Rozenkrantz N, Smith JW. Orlistat is a novel inhibitor of fatty acid synthase with antitumor activity. Cancer Res. 64(6), 2070–2075 (2004).

40 Lupu R, Menendez JA. Pharmacological inhibitors of fatty acid synthase (FASN) – catalyzed endogenous fatty acid biogenesis: a new family of anti-cancer agents? Curr. Pharm. Biotechnol. 7(6), 483–493 (2006).

Review | Hilvo & Orešič

www.futuremedicine.com 187future science group

41 Menendez JA, Mehmi I, Atlas E, Colomer R, Lupu R. Novel signaling molecules implicated in tumor-associated fatty acid synthase-dependent breast cancer cell proliferation and survival: role of exogenous dietary fatty acids, p53-p21WAF1/CIP1, ERK1/2 MAPK, p27KIP1, BRCA1, and NF-kappaB. Int. J. Oncol. 24(3), 591–608 (2004).

42 Chajès V, Cambot M, Moreau K, Lenoir GM, Joulin V. Acetyl-CoA carboxylase alpha is essential to breast cancer cell survival. Cancer Res. 66(10), 5287–5294 (2006).

43 Menendez JA, Vellon L, Colomer R, Lupu R. Pharmacological and small interference RNA-mediated inhibition of breast cancer-associated fatty acid synthase (oncogenic antigen-519) synergistically enhances Taxol (paclitaxel)-induced cytotoxicity. Int. J. Cancer 115(1), 19–35 (2005).

44 Menendez JA, Lupu R. RNA interference-mediated silencing of the p53 tumor-suppressor protein drastically increases apoptosis after inhibition of endogenous fatty acid metabolism in breast cancer cells. Int. J. Mol. Med. 15(1), 33–40 (2005).

45 Knowles LM, Smith JW. Genome-wide changes accompanying knockdown of fatty acid synthase in breast cancer. BMC Genomics 8, 168 (2007).

46 Menendez JA, Vellon L, Mehmi I et al. Inhibition of fatty acid synthase (FAS) suppresses HER2/neu (erbB-2) oncogene overexpression in cancer cells. Proc. Natl Acad. Sci. USA 101(29), 10715–10720 (2004).

47 Little JL, Wheeler FB, Fels DR, Koumenis C, Kridel SJ. Inhibition of fatty acid synthase induces endoplasmic reticulum stress in tumor cells. Cancer Res. 67(3), 1262–1269 (2007).

48 Mills GB, Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nat. Rev. Cancer 3(8), 582–591 (2003).

49 Panupinthu N, Lee HY, Mills GB. Lysophosphatidic acid production and action: critical new players in breast cancer initiation and progression. Br. J. Cancer 102(6), 941–946 (2010).

50 Yanagida K, Masago K, Nakanishi H et al. Identification and characterization of a novel lysophosphatidic acid receptor, p2y5/LPA

6.

J. Biol. Chem. 284(26), 17731–17741 (2009).

51 Liu S, Umezu-Goto M, Murph M et al. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell 15(6), 539–550 (2009).

nn Shows how overexpression of autotaxin or lysophosphatidic acid receptors induces breast cancer in mice.

52 Boucharaba A, Serre CM, Gres S et al. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J. Clin. Invest. 114(12), 1714–1725 (2004).

53 Samadi N, Gaetano C, Goping IS, Brindley DN. Autotaxin protects MCF-7 breast cancer and MDA-MB-435 melanoma cells against Taxol-induced apoptosis. Oncogene 28(7), 1028–1039 (2009).

54 Zhang H, Xu X, Gajewiak J et al. Dual activity lysophosphatidic acid receptor pan-antagonist/autotaxin inhibitor reduces breast cancer cell migration in vitro and causes tumor regression in vivo. Cancer Res. 69(13), 5441–5449 (2009).

n Interesting study describing how inhibition of lysophosphatidic acid–autotaxin signaling causes tumor regression in vivo.

55 Xu Y, Shen Z, Wiper DW et al. Lysophosphatidic acid as a potential biomarker for ovarian and other gynecologic cancers. JAMA 280(8), 719–723 (1998).

56 Wang D, Dubois RN. Eicosanoids and cancer. Nat. Rev. Cancer 10(3), 181–193 (2010).

57 Parrett M, Harris R, Joarder F, Ross M, Clausen K, Robertson F. Cyclooxygenase-2 gene expression in human breast cancer. Int. J. Oncol. 10(3), 503–507 (1997).

58 Hwang D, Scollard D, Byrne J, Levine E. Expression of cyclooxygenase-1 and cyclooxygenase-2 in human breast cancer. J. Natl Cancer Inst. 90(6), 455–460 (1998).

59 Zhao X, Goswami M, Pokhriyal N et al. Cyclooxygenase-2 expression during immortalization and breast cancer progression. Cancer Res. 68(2), 467–475 (2008).

60 Agrawal A, Fentiman IS. NSAIDs and breast cancer: a possible prevention and treatment strategy. Int. J. Clin. Pract. 62(3), 444–449 (2008).

61 Zhou J, Suzuki T, Kovacic A et al. Interactions between prostaglandin E(2), liver receptor homologue-1, and aromatase in breast cancer. Cancer Res. 65(2), 657–663 (2005).

62 Pan MR, Hou MF, Chang HC, Hung WC. Cyclooxygenase-2 up-regulates CCR7 via EP2/EP4 receptor signaling pathways to enhance lymphatic invasion of breast cancer cells. J. Biol. Chem. 283(17), 11155–11163 (2008).

63 Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 67(9), 4507–4513 (2007).

64 Chang SH, Liu CH, Conway R et al. Role of prostaglandin E2-dependent angiogenic switch in cyclooxygenase 2-induced breast cancer progression. Proc. Natl Acad. Sci. USA 101(2), 591–596 (2004).

65 Kamiyama M, Pozzi A, Yang L, DeBusk LM, Breyer RM, Lin PC. EP2, a receptor for PGE2, regulates tumor angiogenesis through direct effects on endothelial cell motility and survival. Oncogene 25(53), 7019–7028 (2006).

66 Wolf I, O’Kelly J, Rubinek T et al. 15-hydroxyprostaglandin dehydrogenase is a tumor suppressor of human breast cancer. Cancer Res. 66, 7818–7823 (2006).

67 Lehtinen L, Vainio P, Wikman H et al. 15-hydroxyprostaglandin dehydrogenase associates with poor prognosis in breast cancer, induces epithelial–mesenchymal transition, and promotes cell migration in cultured breast cancer cells. J. Pathol. 226(4), 674–686 (2012).

68 Pidgeon GP, Lysaght J, Krishnamoorthy S et al. Lipoxygenase metabolism: roles in tumor progression and survival. Cancer Metastasis Rev. 26(3–4), 503–524 (2007).

69 Subbarayan V, Xu XC, Kim J et al. Inverse relationship between 15-lipoxygenase-2 and PPAR-gamma gene expression in normal epithelia compared with tumor epithelia. Neoplasia 7(3), 280–293 (2005).

70 Jiang WG, Watkins G, Douglas-Jones A, Mansel RE. Reduction of isoforms of 15-lipoxygenase (15-LOX)-1 and 15-LOX-2 in human breast cancer. Prostaglandins Leukot. Essent. Fatty Acids 74(4), 235–245 (2006).

71 Jiang WG, Douglas-Jones AG, Mansel RE. Aberrant expression of 5-lipoxygenase-activating protein (5-LOXAP) has prognostic and survival significance in patients with breast cancer. Prostaglandins Leukot. Essent. Fatty Acids 74(2), 125–134 (2006).

72 Tong WG, Ding XZ, Adrian TE. The mechanisms of lipoxygenase inhibitor-induced apoptosis in human breast cancer cells. Biochem. Biophys. Res. Commun. 296(4), 942–948 (2002).

73 Nony PA, Kennett SB, Glasgow WC, Olden K, Roberts JD. 15(S)-lipoxygenase-2 mediates arachidonic acid-stimulated adhesion of human breast carcinoma cells through the activation of TAK1, MKK6, and p38 MAPK. J. Biol. Chem. 280(36), 31413–31419 (2005).

74 Simopoulos AP. Omega-3 fatty acids in inflammation and autoimmune diseases. J. Am. Coll. Nutr. 21(6), 495–505 (2002).

75 Rose DP, Connolly JM, Rayburn J, Coleman M. Influence of diets containing eicosapentaenoic or docosahexaenoic acid on growth and metastasis of breast cancer cells in nude mice. J. Natl Cancer Inst. 87(8), 587–592 (1995).

Regulation of lipid metabolism in breast cancer provides diagnostic & therapeutic opportunities | Review

Clin. Lipidol. (2012) 7(2)188 future science group

76 Bagga D, Capone S, Wang HJ et al. Dietary modulation of omega-3/omega-6 polyunsaturated fatty acid ratios in patients with breast cancer. J. Natl Cancer Inst. 89(15), 1123–1131 (1997).

77 Maillard V, Bougnoux P, Ferrari P et al. N-3 and N-6 fatty acids in breast adipose tissue and relative risk of breast cancer in a case–control study in Tours, France. Int. J. Cancer 98(1), 78–83 (2002).

78 Ryland LK, Fox TE, Liu X, Loughran TP, Kester M. Dysregulation of sphingolipid metabolism in cancer. Cancer Biol. Ther. 11(2), 138–149 (2011).

79 Cuvillier O, Pirianov G, Kleuser B et al. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature 381(6585), 800–803 (1996).

80 Bieberich E, Hu B, Silva J et al. Synthesis and characterization of novel ceramide analogs for induction of apoptosis in human cancer cells. Cancer Lett. 181(1), 55–64 (2002).

81 Crawford KW, Bittman R, Chun J, Byun HS, Bowen WD. Novel ceramide analogues display selective cytotoxicity in drug-resistant breast tumor cell lines compared with normal breast epithelial cells. Cell. Mol. Biol. (Noisy-le-grand) 49(7), 1017–1023 (2003).

82 Stover TC, Sharma A, Robertson GP, Kester M. Systemic delivery of liposomal short-chain ceramide limits solid tumor growth in murine models of breast adenocarcinoma. Clin. Cancer Res. 11(9), 3465–3474 (2005).

83 Schiffmann S, Sandner J, Birod K et al. Ceramide synthases and ceramide levels are increased in breast cancer tissue. Carcinogenesis 30(5), 745–752 (2009).

84 Ruckhaberle E, Rody A, Engels K et al. Microarray ana lysis of altered sphingolipid metabolism reveals prognostic significance of sphingosine kinase 1 in breast cancer. Breast Cancer Res. Treat. 112(1), 41–52 (2008).

85 Liu H, Toman RE, Goparaju SK et al. Sphingosine kinase type 2 is a putative BH3-only protein that induces apoptosis. J. Biol. Chem. 278(41), 40330–40336 (2003).

86 Maceyka M, Sankala H, Hait NC et al. SphK1 and SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism. J. Biol. Chem. 280(44), 37118–37129 (2005).

87 Weigert A, Schiffmann S, Sekar D et al. Sphingosine kinase 2 deficient tumor xenografts show impaired growth and fail to polarize macrophages towards an anti-

inflammatory phenotype. Int. J. Cancer 125(9), 2114–2121 (2009).

88 Hait NC, Bellamy A, Milstien S, Kordula T, Spiegel S. Sphingosine kinase type 2 activation by ERK-mediated phosphorylation. J. Biol. Chem. 282(16), 12058–12065 (2007).

89 Pyne NJ, Pyne S. Sphingosine 1-phosphate and cancer. Nat. Rev. Cancer 10(7), 489–503 (2010).

90 Schwalm S, Doll F, Romer I, Bubnova S, Pfeilschifter J, Huwiler A. Sphingosine kinase-1 is a hypoxia-regulated gene that stimulates migration of human endothelial cells. Biochem. Biophys. Res. Commun. 368(4), 1020–1025 (2008).

91 Ader I, Brizuela L, Bouquerel P, Malavaud B, Cuvillier O. Sphingosine kinase 1: a new modulator of hypoxia inducible factor 1alpha during hypoxia in human cancer cells. Cancer Res. 68(20), 8635–8642 (2008).

92 Anelli V, Gault CR, Cheng AB, Obeid LM. Sphingosine kinase 1 is up-regulated during hypoxia in U87MG glioma cells. Role of hypoxia-inducible factors 1 and 2. J. Biol. Chem. 283(6), 3365–3375 (2008).

93 Licht T, Tsirulnikov L, Reuveni H, Yarnitzky T, Ben-Sasson SA. Induction of pro-angiogenic signaling by a synthetic peptide derived from the second intracellular loop of S1P3 (EDG3). Blood 102(6), 2099–2107 (2003).

94 Visentin B, Vekich JA, Sibbald BJ et al. Validation of an anti-sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell 9(3), 225–238 (2006).

95 Sukocheva O, Wadham C, Holmes A et al. Estrogen transactivates EGFR via the sphingosine 1-phosphate receptor Edg-3: the role of sphingosine kinase-1. J. Cell Biol. 173(2), 301–310 (2006).

96 Sukocheva O, Wang L, Verrier E, Vadas MA, Xia P. Restoring endocrine response in breast cancer cells by inhibition of the sphingosine kinase-1 signaling pathway. Endocrinology 150(10), 4484–4492 (2009).

97 Watson C, Long JS, Orange C et al. High expression of sphingosine 1-phosphate receptors, S1P1 and S1P3, sphingosine kinase 1, and extracellular signal-regulated kinase-1/2 is associated with development of tamoxifen resistance in estrogen receptor-positive breast cancer patients. Am J. Pathol. 177(5), 2205–2215 (2010).

98 Hait NC, Allegood J, Maceyka M et al. Regulation of histone acetylation in the

nucleus by sphingosine-1-phosphate. Science 325(5945), 1254–1257 (2009).

nn Provides evidence of how lipids, in this case sphingosine-1-phosphate, can regulate gene expression epigenetically.

99 Tan RX, Chen JH. The cerebrosides. Nat. Prod. Rep. 20(5), 509–534 (2003).

100 Liu YY, Han TY, Giuliano AE, Cabot MC. Ceramide glycosylation potentiates cellular multidrug resistance. FASEB J. 15(3), 719–730 (2001).

101 Morjani H, Aouali N, Belhoussine R, Veldman RJ, Levade T, Manfait M. Elevation of glucosylceramide in multidrug-resistant cancer cells and accumulation in cytoplasmic droplets. Int. J. Cancer 94(2), 157–165 (2001).

102 Ruckhaberle E, Karn T, Hanker L et al. Prognostic relevance of glucosylceramide synthase (GCS) expression in breast cancer. J. Cancer Res. Clin. Oncol. 135(1), 81–90 (2009).

103 Gouaze V, Liu YY, Prickett CS, Yu JY, Giuliano AE, Cabot MC. Glucosylceramide synthase blockade down-regulates P-glycoprotein and resensitizes multidrug-resistant breast cancer cells to anticancer drugs. Cancer Res. 65(9), 3861–3867 (2005).

104 Yu RK, Tsai YT, Ariga T, Yanagisawa M. Structures, biosynthesis, and functions of gangliosides – an overview. J. Oleo. Sci. 60(10), 537–544 (2011).

105 Birkle S, Zeng G, Gao L, Yu RK, Aubry J. Role of tumor-associated gangliosides in cancer progression. Biochimie 85(3–4), 455–463 (2003).

106 Marquina G, Waki H, Fernandez LE et al. Gangliosides expressed in human breast cancer. Cancer Res. 56(22), 5165–5171 (1996).

107 Fernandez LE, Gabri MR, Guthmann MD et al. NGcGM3 ganglioside: a privileged target for cancer vaccines. Clin. Dev. Immunol. 2010, 814397 (2010).

108 Hirsch HA, Iliopoulos D, Joshi A et al. A transcriptional signature and common gene networks link cancer with lipid metabolism and diverse human diseases. Cancer Cell 17(4), 348–361 (2010).

nn Elucidates pathways that connect the metabolic syndrome and cancer with each other.

n Website201 Lpath Incorporated.

www.lpath.com

Review | Hilvo & Orešič


Recommended