<<

25 9

Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R435–R452 Cancer back

REVIEW The impact of transcription on metabolism in prostate and breast cancers

Ninu Poulose1, Ian G Mills1,2,* and Rebecca E Steele1,*

1Centre for Cancer Research and Cell Biology, Queen’s University of Belfast, Belfast, UK 2Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK

Correspondence should be addressed to I G Mills: [email protected]

*(I G Mills and R E Steele contributed equally to this work)

Abstract

Metabolic dysregulation is regarded as an important driver in cancer development and Key Words progression. The impact of transcriptional changes on metabolism has been intensively ff androgen studied in hormone-dependent cancers, and in particular, in prostate and breast cancer. ff androgen receptor These cancers have strong similarities in the function of important transcriptional ff breast drivers, such as the oestrogen and androgen receptors, at the level of dietary risk and ff oestrogen epidemiology, genetics and therapeutically. In this review, we will focus on the function ff endocrine therapy of these nuclear hormone receptors and their downstream impact on metabolism, with a resistance particular focus on metabolism. We go on to discuss how remains dysregulated as the cancers progress. We conclude by discussing the opportunities that this presents for drug repurposing, imaging and the development and testing of new Endocrine-Related Cancer therapeutics and treatment combinations. (2018) 25, R435–R452

Introduction: prostate and breast cancer

Sex hormones act through nuclear hormone receptors Early-stage PCa is dependent on androgens for survival and induce distinct transcriptional programmes essential and can be treated by androgen deprivation therapy; to male and female physiology. They exert their effects however, with the advancement of cancer, it becomes on target tissues such as the mammary gland, ovary and refractory to hormone treatment. This later stage of PCa the uterus in females and the testis and the prostate is known as castrate-resistant PCa (CRPC) or androgen- gland in males. Studies indicate that these hormones indifferent PCa, meaning that the cancer thrives despite a also play pivotal roles in the development of endocrine- reduction in serum androgen levels (Feldman & Feldman related cancers. The highest incidence cancers affecting 2001, Beltran et al. 2011). The prostate is also a target for hormone-dependent organs are prostate cancer (PCa) oestrogens among which oestradiol-17β is considered and breast cancer (BCa). PCa is the second most common the most potent inducer of prostatic proliferation and male malignancy in many western industrialised promotes epithelial-to-mesenchymal transition in countries and fifth leading cause of cancer death in men benign prostatic epithelial cells (Shi et al. 2017). Studies worldwide (GLOBOCAN 2012). BCa is the most frequently also suggest that oestrogen–androgen balance may be a diagnosed female malignancy and the fifth most common key determinant in the development of aggressive PCa cause of death from cancer overall (GLOBOCAN 2012). (Black et al. 2014). Likewise androgens play important

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access

-18-0048 Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R436 Cancer back roles in the development and progression of BCa subtypes tissues bound to serum sex hormone-binding globulin such as molecular apocrine tumours (Robinson et al. and albumin, where it is converted to the more active 2011). Endogenous oestrogen levels have been linked metabolite, DHT by the 5α-reductase (Heinlein & to an increased risk of breast and endometrial cancers Chang 2004, Eacker et al. 2008). (Brown & Hankinson 2015). AR also known as NR3C4 (nuclear receptor subfamily The predominant BCa subtype is luminal tumours, 3, group C, member 4) is a ligand-dependent TF that accounting for approximately two-thirds of all diagnosed regulates the expression of genes involved in male BCa cases (Ignatiadis & Sotiriou 2013). Originating from sexual phenotype. AR gene encodes a protein of 110 KDa breast luminal epithelial cells, the majority of luminal with three major domains, the N-terminal domain, tumours are oestrogen receptor (ER) positive, and clinical the DNA-binding domain and the C-terminal ligand- studies have highlighted a strong ovarian-derived binding domain (LBD). Binding of androgens to LBD hormone-driven biology, in mammary gland tumour causes dissociation of heat shock proteins, its nuclear development, particularly associated with oestradiol- translocation through binding of importin-α, subsequent 17β (Pike et al. 1993, Perou et al. 2000). ER-positive BCa dimerisation and binding to androgen response elements patients have the most favourable BCa subtype prognosis, in the promoter regions of target genes, such as prostate- generally responding well to endocrine-targeted therapies; specific antigen (PSA) and transmembrane protease serine however, endocrine therapy resistance does occur (García- 2 (Tan et al. 2015). AR is expressed mostly in the secretory Becerra et al. 2013). In recent years, an important role for epithelial cells and to some extent in the stroma, whereas the androgen receptor (AR) in breast tumourigenesis has basal cells are AR negative (Prins et al. 1991). In a fully emerged, with between 70 and 90% of tumours shown to formed prostate, the androgens continue to function to harbour AR positivity (Moinfar et al. 2003, Niemeier et al. promote the survival of secretory epithelia. Physiological 2010, Collins et al. 2011). In addition, two gene expression testosterone and DHT levels prevented in these studies have identified an AR-positive BCa subtype; secretory epithelial cells, whereas castration resulted in a molecular apocrine BCa tumours, also characterised by loss of prostate secretory epithelial cells due to apoptosis ER negativity, they constitute 8–12% of all BCa cases in rat models, which was preceded by degeneration of (Farmer et al. 2005, Doane et al. 2006). Despite their prostatic capillaries (Heinlein & Chang 2004). Hence, ER-negative status, molecular apocrine tumours express a AR signalling remains essential to the structural and number of genes that are usually expressed in ER-positive functional integrity of prostate gland. tumours such as XBP-1, SCUBE2, SPDEF and FOXA1 ER signalling is important for a number of normal (Doane et al. 2006, Robinson et al. 2011). It is proposed cellular proliferative processes and the maintenance of that in such tumours, in the absence of ER, AR can bind to lipid and carbohydrate metabolic , primarily ER cis-regulatory genomic elements and transcriptionally in reproduction-related tissues (Nilsson & Gustafsson activate canonical ER target genes (Robinson et al. 2011). 2011, Vrtačnik et al. 2014). In addition, the role of Consequently, patients with molecular apocrine tumours the ER in both BCa and PCa development has been display poor clinical response to ER antagonists, and extensively studied. Like testosterone, oestrogen is a it is believed that the use of anti-androgens may be of steroid hormone, derived from ; it mediates increasing therapeutic benefit Farmer( et al. 2005). its function through binding to one of two cognate ERs – alpha (ERα) and beta (ERβ). Two ERα isoforms ERα36 (Wang et al. 2005) and ER 46 (Flouriot et al. 2000) Nuclear hormone receptors – AR and ER α have additionally been identified. Although there are Nuclear hormone receptors such as the AR and ER are two subtypes, ERα is the main driver in ER-positive ligand-activated transcription factors (TFs), which induce BCa and the main subtype of focus in this review. distinct transcriptional programmes conducive to the Upon ligand activation, ERα dimerises and interacts development and differentiation of prostate and breast with transcriptional coregulators to bind to oestrogen- tissues, respectively. The growth and maintenance of responsive elements upstream of ERα target genes (May the prostate is dependent on androgens, testosterone & Westley 1988, Hayashi et al. 2003). ERα is primarily and 5α-dihydrotestosterone (DHT), acting through expressed in the mammary gland and is a ligand- their cognate receptor, AR (Heinlein & Chang 2004). inducible TF and signal transducer that has functional Testosterone is synthesised from cholesterol, primarily by roles in proliferation, differentiation and migration and the Leydig cells in the testes and transported to the target is known to transcriptionally regulate genes involved

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R437 Cancer back in mitochondrial biogenesis, the TCA cycle and lipid Metabolic reprogramming in prostate and metabolism (Huss et al. 2004, Manavathi et al. 2013). breast cancer Consequently, the overexpression of ER-α, as observed in many Luminal A BCa tumours, leads to hormone- Understanding the unique metabolic landscape of normal dependent breast tumourigenesis (Barnes et al. 2004). prostate glandular epithelium is imperative to better AR and ER transcriptional activity is critical to the understanding the metabolic changes in PCa (Fig. 1). progression of PCa and BCa and are the main targets of Unlike other mammalian cells, normal prostate glandular hormonal therapies. Apart from cell-cycle regulators and epithelial cells are characterised by synthesis and signalling molecules, central metabolism and metabolic secretion of enormously high levels of citrate, the major gene signatures form a core set of AR and ER target genes component of the prostatic fluid. Citrate is produced (Abba et al. 2005, Massie et al. 2011). Altered cellular from acetyl CoA and oxaloacetate (OAA) by the enzyme metabolism is a hallmark of virtually all cancers. Both in citrate synthase. In a typical mammalian cell, citrate is vitro and in vivo studies support a major role for metabolic oxidised via Krebs cycle to regenerate OAA, whereas in transformation in the progression of PCa and BCa. The prostate cells, citrate is an end product of metabolism, importance of metabolism is further reinforced by the which is not further oxidised but exported to the cytosol. fact that both PCa and BCa are influenced by metabolic This is achieved through cellular accumulation of zinc diseases like type 2 diabetes, metabolic syndrome and that inhibits the mitochondrial aconitase activity and obesity (Hsing et al. 2007, Vona-Davis et al. 2007). truncates Krebs cycle at the first step of citrate oxidation

Figure 1 Schematic diagram showing the metabolic landscape in (A) the normal prostate gland and (B) prostate cancer. ACLY, ATP citrate lyase; ACON, aconitase; CS, citrate synthase; EAAC1, excitatory amino acid carrier 1; MAAT, aspartate aminotransferase; PDH, pyruvate dehydrogenase. Pathways are shown in black arrows and inhibitory steps in red lines.

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R438 Cancer back

(Costello & Franklin 2006). The active transport of zinc accumulation (Costello & Franklin 2006, Makhov et al. in prostate cells is mainly mediated through SLC39A1 2011). Interestingly, the expression of ZIP1 and ZIP2 or ZIP1. The continued supply of acetyl CoA and OAA are comparatively lower in normal prostate tissue from for citrate production are met by a high rate of aerobic African-Americans patients who are at a higher risk of glycolysis and aspartate accumulation, respectively. PCa incidence when compared to age-matched Caucasian Glycolysis converts glucose to pyruvate, which enters men (Rishi et al. 2003). Epigenetic changes especially mitochondria and gets decarboxylated to acetyl CoA by methylation is one of the most recurrent events in locally pyruvate dehydrogenase (PDH). OAA is produced from the advanced and metastatic PCa with AR promoter CpG transamination of aspartate, which is actively transported islands reported to be hotspots of aberrant methylation into the prostate cells through the specific receptor and consequent loss of AR expression (Kinoshita et al. 2000, excitatory amino acid carrier 1 (EAAC1) (Franklin et al. Massie et al. 2017). DNA methylation is intimately linked 2006). Testosterone stimulates the expression/activity of to central metabolism as well as one carbon metabolism. EAAC1, aspartate aminotransferase and mitochondrial Androgens regulate the expression of key biosynthetic PDH thus facilitating citrate production by regulating involved in the polyamine synthesis in the both acetyl CoA and OAA levels (Franklin et al. 1986, 2006, prostate, which requires one carbon metabolism and Costello & Franklin 1993). In addition, testosterone affects share the DNA methylation donor, S-adenosylmethionine citrate oxidation by differentially regulating m-aconitase (SAM). Therefore, a decline in polyamine biosynthesis gene expression in rat lateral and ventral prostate cells during cancer progression may enhance the availability with an inhibitory effect on former and stimulatory of SAM pool for epigenetic modification Massie( et al. effect on latter (Costello et al. 1995, 2000). Although 2017). The absence of the Warburg effect in early stage bio-energetically less efficient, the citrate production is PCa is clinically relevant as these tumours do not appear essential to maintain the pH of the semen, chelation of on fluorodeoxyglucose positron emission tomography calcium ions by citrate (Ford & Harrison 1984) and may (FDG-PET) scans (Liu et al. 2001). However, in the later have more complex roles. Furthermore, a relatively low stages, PCa cells start exhibiting Warburg effect, and abundance or absence of ATP citrate lyase (ACLY) by FDG-PET may be useful in monitoring treatment response negative regulation through miR-22 (Xin et al. 2016) may in advanced metastatic PCa (Eidelman et al. 2017). Early- be preventing the degradation of citrate to acetyl CoA and stage PCa appears to rely on for energy production, OAA in the cytosol thereby promoting its secretion into and this property is exploited in visualising tumours the prostatic fluid. Interestingly, ACLY, a key enzyme in by 11C-acetate or 11C-choline accumulation, which the de novo lipogenesis has been shown to be upregulated seems to be a more sensitive imaging technique than in clinical samples from PCa, BCa osteosarcoma, cervical FDG-PET in the detection of primary and metastatic PCas cancer and lung cancer with a concomitant reduction in (Jadvar 2012). 11C-sarcosine is also gaining interest as a miRNA-22 (Xin et al. 2016). To summarise, normal prostate radiotracer for PCa imaging as sarcosine, a metabolic cells are characterised by a high net citrate production product of choline is associated with PCa aggressiveness associated with a low level of oxidative phosphorlyation and progression (Piert et al. 2017). In BCa, most studies (OXPHOS) and high rate of glycolysis. have focused on18F-labelled thymidine analogues to A metabolic transformation from a zinc-accumulating assess cell proliferation, although promising results from citrate-secreting cell to citrate-oxidising cell is a prominent using 11C-choline have been reported in BCa patients feature of prostate malignancy. Patient-derived tumour (Kenny 2016). metabolomics data demonstrate strikingly low citrate The metabolic characteristics of mammary epithelial levels in CRPC tumours when compared to normal cells change with tumour onset and metastatic disease prostate and in androgen-dependent PCa (Shafi et al. progression to sustain augmented biosynthetic demand. 2015). ZIP1 gene expression is markedly downregulated One metabolomic study in a syngeneic mouse mammary in adenocarcinomatous glands coupled with a loss of tumour model observed metabolite alterations in a number zinc accumulation (Franklin et al. 2005). Two other of pathways involved in glycolysis, the pentose phosphate zinc transporter isoforms ZIP2 and ZIP3 are also pathway and a decrease in GSH-dependent antioxidative downregulated in PCa (Desouki et al. 2007). Cancer cells pathway, and intermediates in lipid biosynthesis like tend to evade the inefficient process of citrate production synthase (FASN) (Lu et al. 2010). Moreover, through epigenetic silencing of zinc transporters by metabolomic analysis of 270 BCa samples and 97 normal promoter hypermethylation thereby inhibiting zinc breast samples has highlighted a switch from a positive to

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R439 Cancer back

Figure 2 (A) Key metabolic differences between ER-positive and ER-negative breast tumours. (B) Schematic diagram showing role of glutamine and cholesterol metabolite, 27-OHC in supporting tumour growth. AI, aromatase inhibition; CYP27A1, cytochrome P450 family 27 subfamily A member 1; CYP7B1, cytochrome P450 family 7 subfamily B member 1; GLS, glutaminase; GLUD, glutamate dehydrogenase; SLC1A5, solute carrier family 1 member 5; SLC7A5, SLC family 7 member 5; 27-OHC, 27-hydroxyl cholesterol. negative correlation between glutamate and glutamine in and metabolic studies are corroborative of the role of AR tumour vs normal, indicating that BCa cells, particularly as a core regulator of an anabolic transcriptional network ER-negative tumours have a metabolic dependency on in PCa. Studies by Massie et al. have sought to derive a glutamine metabolism (Fig. 2) (Budczies et al. 2015). detailed map of AR-regulated genes in PCa cell lines Notably, c-MYC amplification is found in 15.7% of all representing distinct molecular subtype of the disease, by BCa s (Deming et al. 2000). In ER-positive breast tumours, employing ChIP seq and transcript profiling Massie( et al. it is known to confer response to endocrine treatment 2011). Stimulation of AR by androgen upregulated genes (Ellis et al. 2012) and implicated in the development of involved in cell cycle, glucose uptake and glycolysis, lipid treatment resistance (Venditti et al. 2002, McNeil et al. turnover, nucleotide and amino acid metabolism. This 2006, Miller et al. 2011). Similarly, in the absence of study identified calcium/calmodulin-dependent protein ER, one characteristic of triple-negative BCa (TNBC) is kinase 2 (CAMKK2) as a hormone-dependent modulator increased transcriptional activity of the c-MYC oncogene of anabolic metabolism in PCa in an AMP-activated (Alles et al. 2009). It is established that high expression protein kinase (AMPK)-dependent manner (Massie et al. of c-MYC can augment glutamine uptake and maintain 2011). CAMKK2 is also consistently overexpressed in the elevated bioenergetic demands of a tumourigenic cell clinical samples from both hormone sensitive and CRPC. (Anso et al. 2013). Interestingly, a recent study by Mishra AR-mediated activation of cellular fuel sensor, AMPK and et al. reported elevated levels of the oncometabolite D-2- downstream AMPK-PGC1α signalling axis confers distinct hydroxyglutarate in ER-negative breast tumours driven growth advantage to the tumour cells (Tennakoon et al. by the c-MYC-induced mitochondrial enzyme, alcohol 2014). Consequently, genetic changes in AR form one of dehydrogenase iron-containing protein 1, promoting the core contributors to altered cellular metabolism in metabolic reprogramming, reductive glutamine PCa. C-MYC is another TF overexpressed in PCa, which metabolism and disease progression (Mishra et al. 2018). partially overlaps with AR-binding sites and antagonises Akin to lipid metabolism in PCa, glutamine metabolism AR-mediated transcriptional output (Barfeld et al. 2017). requires mitochondrial function and oxidative However, in molecular apocrine BCa, MYC cooperates with phosphorylation. AR in androgen-responsive gene transcription contrary The role of sex hormones and nuclear hormone to the antagonistic relationship seen in PCa (Ni et al. receptors in the metabolic transformation of prostate or 2013). In vitro studies provide important insights into the breast is quite complex and inadequately understood, with intricate transcriptional and gene regulatory networks several caveats in existing information linking genetic and and target genes relevant to the progression of PCa and molecular changes to metabolic transformation. Genomic BCa. However, in order to get a comprehensive picture of

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R440 Cancer back the mechanisms underpinning the disease, it is important in monocarboxylate transporter (MCT4) expression and to simultaneously derive and interpret data from in vivo lactate dehydrogenase activity (LDH) (Vaz et al. 2012). models, tissue explants and clinical specimens. GLUTs and MCTs may be functioning synchronously to maintain glycolysis at a steady rate in solid tumours. However, in PCa, a reciprocal metabolic relationship seems Metabolic pathways in prostate and breast to exist between cancer-associated fibroblast (CAFs) and cancers cancer cells. When co-cultured, CAFs undergo metabolic reprogramming to a Warburg phenotype with increased Metabolite transporters GLUT1, lactate production and efflux through MCT4, An increase in glucose uptake and switching to aerobic whereas PCa cells are shifted to aerobic metabolism with glycolysis or Warburg effect is a prominent metabolic an increase in lactate upload via MCT1. The allocation alteration in most cancers. However, the prostate has of glycolytic metabolism to CAFs and ‘lactate shuttling’ a unique metabolic landscape that favours switching appears to be one of the strategies employed by PCa cells to an increased OXPHOS during cancer initiation and for growth and progression (Fiaschi et al. 2012). MCT2 is progression. Glucose uptake and increased glycolysis yet another MCT isoform overexpressed in PCa through appears to be a metabolic adaptation in the late stage epigenetic regulation (Pertega-Gomes et al. 2015b). These of the disease correlated with poor prognosis (Pertega- studies indicate that MCTs could be promising targets in Gomes et al. 2015a). Studies from our group and others the treatment of PCa. have shown androgen-stimulated glucose uptake and lactate production in PCa cell lines (Massie et al. 2011, Amino acid metabolism Vaz et al. 2012). The expression of ubiquitous glucose transporter, GLUT1, was elevated by androgen treatment Many cancers show increased dependency on glutamine in these cells. A recent study has further validated the uptake and metabolism, which not only serves the critical role played by GLUT1 in PCa, the knockdown of protein synthesis requirements but also fuels TCA cycle which inhibited glycolysis, cell proliferation and induced (anaplerosis) and acts a source of fatty acid production cycle arrest at G2/M phase (Xiao et al. 2018). Other GLUT through reductive carboxylation (Mullen et al. 2011, isoforms like GLUT3 (Vaz et al. 2012), insulin-sensitive Eidelman et al. 2017). Glutaminolysis involves conversion isoforms GLUT4, GLUT12 (Chandler et al. 2003) and of glutamine to glutamate by glutaminase, which is then fructose transporter GLUT5 (Reinicke et al. 2012) are also transformed into alpha-ketoglutarate that is fed into the reported in PCa cells. Similarly, GLUT1, GLUT2, GLUT3, Krebs cycle. Glutaminase 1 expression is augmented in GLUT4, GLUT5 and GLUT12 have been reported to be PCa patients and is highly correlated with the stage of expressed in BCa cells (Rogers et al. 2003, Godoy et al. the disease (Pan et al. 2015). The expression of major 2006, Garrido et al. 2013). GLUT1 expression has been glutamine transporter in cancer cells, alanine–serine– shown to positively correlate with ERα positivity and cysteine transporter‐2 (ASCT2), is also elevated in PCa confer a more aggressive disease and poorer prognosis patient samples (Wang et al. 2015). Indeed, inhibition in BCa patient tumour samples (Kang et al. 2002). of ASCT2 limited PCa growth and metastasis in vivo and Downregulation of GLUT1 with the multi-kinase hence a putative therapeutic target. inhibitor sorafenib has been shown to induce apoptosis Among the other metabolic pathways, MYC-regulated through mitochondrial membrane depolarisation and purine biosynthesis has been shown to be significant AMPK-dependent inhibition of the mTORC1 pathway in in PCa with two reported biomarkers in the pathway BCa (Fumarola et al. 2013). Further studies are needed to namely, phosphoribosylaminoimidazole carboxylase and delineate the specific roles of these GLUT isoforms in PCa phosphoribosylaminoimidazole-succinocarboxamide and BCa. synthase and inosine monophosphate dehydrogenase Monocarboxylate transporters (MCTs) are proton 2 (IMPDH2); inhibition of IMPDH2 impaired cell symporters involved in lactate efflux to maintain the high proliferation in vitro by inducing nucleolar stress glycolytic rate seen in solid tumours and to prevent the (Barfeld et al. 2015). Recent studies have shown that cytotoxicity from acidosis (Pinheiro et al. 2012). Androgen- enzymes in the hexosamine biosynthetic pathway responsive (LNCaP) and androgen-nonresponsive (PC3) are androgen regulated and upregulated in PCa cell lines exhibited distinct glycolytic profiles with a higher transcriptionally and metabolically. The enzyme involved lactate production in PC3 and a concomitant increase in O-linked conjugation, O-linked β-N-acetylglucosamine

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R441 Cancer back transferase (OGT) is overexpressed in PCa and correlate in order to meet the increased demand for membrane with disease progression. Interestingly c-MYC is a target biogenesis. In addition, de novo lipogenesis also serves to of OGT whose OGlcNAcylation site overlaps with provide building blocks for lipid signalling molecules such phosphorylation site preventing its polyubiquitination as phosphatidylinositol-3,4,5-trisphosphate, ceramide- and proteosomal degradation (Itkonen et al. 2013). Some 1-phosphate, phosphatidic acid, diacylglycerol, and studies support a role for one carbon metabolism in PCa, lysophosphatidic acid all of which fuel cancer cell growth specifically choline and vitamin B12 show a positive (Ray & Roy 2017). Another advantage of lipogenesis correlation with PCa risk (Johansson et al. 2009). Apart is availability of substrates for posttranslational from these, dysregulated lipid metabolism is one of the modification of proteins like prenylation, palmitoylation main aspects of metabolic reprogramming in PCa as well and GPI modification and lipids also serve as an energy as BCa. reserve during times of nutrient limitation (Zhang & Du 2012). De novo lipogenesis ensures a constant supply of fatty acids in poorly vascularised tumour tissues, thereby Lipid metabolism maintaining a high proliferative rate. Aberrant lipogenesis is a prominent feature of cancer cells Several key enzymes involved in the lipid biosynthetic and increasingly recognised as an important factor in pathway are frequently upregulated in PCa. Kadhi et al. tumour growth and metastases (Fig. 3). Most mammalian made an interesting observation that the peripheral zone cells meet their lipid requirements through diet, and lipid of prostate are naturally endowed with a higher capacity for biosynthesis is restricted to tissues like adipose, liver and de novo lipogenesis and fatty acid oxidation, which makes lactating breast tissue. Studies by Medes et al. in the early it susceptible to oncogenesis and explains the frequent 1950s demonstrated that de novo lipogenesis is induced in occurrence of cancer in this zone (Al Kadhi et al. 2017). neoplastic tissues (Medes et al. 1953). It is not surprising Androgens by coordinating the expression of enzymes that lipid synthesis is induced in proliferating cancer cells involved in lipogenesis play a central role in mediating

Figure 3 Schematic diagram showing SREBP-mediated lipogenic programme and pathways involved in lipid and cholesterol biosynthesis. The pathways are shown in blue arrows, key metabolic enzymes upregulated in PCa and BCa in blue boxes, inhibitors of the enzymes are in red.

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R442 Cancer back the lipogenic switch in PCa cells. One of the mechanisms programme in a MAPK-dependent manner and can be by which androgens exert this lipogenic switch is through blocked by SREBP inhibitor, fatostatin. Intriguingly, a activation of sterol regulatory element-binding proteins high-fat diet was sufficient to drive metastasis in PTEN-null (SREBPs), master regulators of lipid and cholesterol model highlighting the importance of dietary intervention biosynthesis. SREBP family of TFs encompasses SREBP-1a, in regulating PCa progression (Chen et al. 2018a). SREBP SREBP-1c and SREBP-2, which share similar structure and pathways are also activated by mutant p53, often seen in mechanism of activation. In brief, SREBP precursors are metastatic PCa. In fact, fatostatin alone or in combination anchored to endoplasmic reticulum membrane where it with docetaxel displayed significant antitumor effects forms a complex with SREBP cleavage-activating protein in PCa xenograft models (Li et al. 2015). Studies in an (SCAP), which in turn interacts with a retention protein LNCaP xenograft model of human PCa indicate increased complex. The loss of interaction between SCAP and lipogenesis as an adaptive mechanism during progression retention complex leads to the migration of SREBP/SCAP to androgen independence (Ettinger et al. 2004). These complex to the golgi where it is subjected to proteolytic studies significantly enhance our understanding of the cleavage by resident proteases and subsequent release of mechanistic basis for dysregulated lipid metabolism in N-terminal SREBP segment. NSREBP further translocates PCa and could open doors to new approaches for patient to the nucleus and binds to SREs in the promoter region stratification and molecular subtyping. of key lipogenic genes involved in both the synthesis of FASN, an AR target gene and a key enzyme in the fatty acids and cholesterol (Heemers et al. 2006). SREBP- lipid biosynthetic pathway, is frequently upregulated in dependent de novo lipogenesis promotes PCa growth and human PCa and BCa and has been widely studied. The metastases. Huang et al. has shown a positive correlation recent development of Fasnall, a FASN inhibitor with between SREBP1 protein expression and clinical anti-tumour activity (Alwarawrah et al. 2016), provides an Gleason score in PCa (Huang et al. 2012). Androgen- interesting therapeutic opportunity. FASN expression is induced SREBP activation is proposed to occur through an early event in the development of PCa with a gradual increased AR-induced expression of SCAP, which favours increase with the stage of cancer and a higher expression SREBP/SCAP translocation to the golgi and activation correlates with poor prognosis and patient survival, (Heemers et al. 2006). SREBP1 in turn regulates the suggesting the use of FASN as a general PCa marker expression of AR by binding to its promoter region (Huang (Swinnen et al. 2002, Rossi et al. 2003). It is noteworthy et al. 2012) thereby maintaining the lipogenic phenotype. that many of the oncogenic signalling pathways are Two recent studies have identified novel genomic involved in the regulation of lipid metabolism. Growth drivers of lipid metabolism in PCa with implications factor-associated signalling pathways like PI3K/Akt and for subtyping and treatment of the disease. Both studies MAPK regulates FASN expression through SREBP1 (Zhang employed PTEN-null transgenic mouse model of PCa, & Du 2012). Other TFs such as Sp1, members of p53 which gives high-grade intraepithelial prostate tumours family and the lipogenesis-related nuclear protein S14 at an early age and invasive PCa at a late stage. The first are also known to modulate FASN expression (Zhang & study focused on PDH complex (PDC), a complex with Du 2012). FASN is also regulated post-translationally in a gatekeeper function in converting pyruvate into acetyl PCa by androgen-regulated ubiquitin-specific protease-2a CoA for entry into the TCA cycle in mitochondria, (USP2a) that removes ubiquitin residues from FASN and demonstrates how cellular metabolism is intimately linked stabilises the protein. USP2a is overexpressed in PCa and to the regulation of gene expression. By inactivating represents another therapeutic target (Graner et al. 2004). pyruvate dehydrogenase A1 (PDHA1), prostate tumour Other enzymes involved in fatty acid synthesis, which growth was restrained at an early stage mainly through are frequently upregulated in PCa are ACLY (Xin et al. suppressing lipid biosynthesis. Mechanistically, this is 2016), acetyl CoA carboxylase (ACC) that catalyses the due to reduced histone acetylation at regulatory regions conversion of acetyl CoA to malonyl CoA and stearoyl- bound by SREBP owing to a reduction in the activity CoA desaturases (SCDs), rate-limiting enzyme in the nuclear compartmentalised PDHA1 (Chen et al. 2018b). biosynthesis of monounsaturated fatty acids (Kim et al. Furthermore, PDHA1 is frequently amplified and 2011). It is interesting to note that SCD peptides derived overexpressed in these tumours signifying its potential as from proteolytic cleavage of SCD can transactivate AR and a therapeutic target. The second study utilises the same facilitate cell proliferation (Kim et al. 2011). genetic background and found that co-deletion of Pml SREBP-1 has additionally been shown to confer poor leads to hyperactivation of an SREBP dependent lipogenic clinical prognosis in BCa and promote invasive and

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R443 Cancer back metastatic potential, correlating with a greater tumour- The biosynthesis of cholesterol, an important node metastasis and lymph node metastasis stage in component of the lipid bilayer, occurs through the BCa patients (Bao et al. 2016). SREBP-1 transcriptionally mevalonate pathway using the same precursor, acetyl activates a number of enzymes involved in lipid CoA, used in fatty acid synthesis. Enhanced cholesterol metabolism and lipogenesis, including FASN, ACC and biosynthesis regulated by SREBP-2 is a major player in SCD-1 (Song et al. 2012, Lee et al. 2013, Bao et al. 2016). In the initiation and progression of PCa with an increase in normal breast cells, circulating lipids are used to synthesise PCa stem cell population with SREBP-2 overexpression lipids, whilst in breast tumour cells, FASN is utilised in (Li et al. 2016). Aberrant cholesteryl ester accumulation lipid generation and FASN upregulation is frequently in lipid droplets aggravates the cancer invasiveness and observed in BCa (Mashima et al. 2009, Zhang & Du 2012). is found in high-grade PCa with PTEN loss and PI3K/Akt In addition to an increase in lipogenesis, PCa cells activation (Yue et al. 2014). Squalene monooxygenase heavily rely on lipid oxidation for their growth and (SQLE), the second rate-limiting enzyme of cholesterol survival. A recent study by Itkonen et al. has demonstrated synthesis, is highly expressed in lethal PCa and may be enoyl-CoA delta isomerase 2 (ECI2), an enzyme involved in a therapeutic target in high-risk patients (Stopsack et al. lipid degradation, as a direct AR target and overexpressed 2017). SQLE is also considered a bonafide oncogene in in clinical PCa. Consequently, PCa cells are vulnerable BCa (Brown et al. 2016). Cholesterol forms the structural to inhibitors of lipid degradation like perhexiline or by backbone for steroid hormone biosynthesis (Fig. 4). ECI2 knockdown and respond by activating incomplete Intratumoral steroid biosynthesis has been recognised followed by cell death response. Moreover, as crucial factor in the progression of both PCa and BCa. the clinically approved drug perhexiline exhibited potent Although serum androgen levels are diminished following anti-tumour activity in combination with antiandrogen, hormone ablation therapy, the intraprostatic androgen enzalutamide and abiraterone acetate (Itkonen et al. 2017). levels remain moderately high as the tumour cells start Fatty acid oxidation is also an important bioenergetic synthesising their own androgens, and this could be the pathway in MYC-overexpressing triple-negative BCa driving force in CRPC progression (Locke et al. 2008). (Camarda et al. 2016). Hence, targeting lipid metabolism This finding has paved way for the development of novel would be a promising approach in PCa and BCa treatment. therapeutic approaches to treat PCa such as abiraterone

Figure 4 Steroid hormone biosynthesis pathway. A schematic representation highlighting the structural similarities of cholesterol, testosterone, oestradiol and DHEA in the biosynthesis of cholesterol-derived steroid hormones and the key enzymes involved. CYP11A1, cytochrome P450 family 11 subfamily A; CYP17A1, cytochrome P450 family 17α-hydroxylase/17,20-lyase; 3β-HSD, 3β-hydroxysteroid dehydrogenase.

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R444 Cancer back acetate, an inhibitor of 17α-hydroxylase and C17,20 Treatment resistance in AR-positive and lyase (CYP17A1) for the treatment of men with advanced ER-positive cancers CRPC (Ryan et al. 2013). In fact, genetic polymorphisms of CYP17A1 are associated with higher aggressiveness AR plays a central role in the pathogenesis of PCa and and risk of progression to CRPC in patients receiving is the main therapeutic target. Several studies indicate androgen deprivation therapy (Robles-Fernandez et al. that polymorphisms/mutations in the AR predispose men 2017). The importance of cholesterol in PCa progression to PCa (Gottlieb et al. 2012). For instance, polymorphic is further established by a clinical study by Platz et al. variation in the length of CAG repeats in the amino where the use of statins (cholesterol-lowering drugs) terminus of AR is found to be inversely correlated to AR in patients is found to be associated with reduced risk transcriptional activity, with short repeats associated of metastatic PCa (Platz et al. 2006). The cholesterol with an increased risk of PCa (Heinlein & Chang 2004). metabolite, 27-hydroxyl-cholesterol (27-OHC) is a known Although disputed, this association may partly explain the selective ER modulator, which promotes tumourigenesis ethnic differences seen in PCa incidence, where African- in ER-positive BCa (Fig. 2) (Warner & Gustafsson 2014). American men have short CAG repeat length and higher Higher levels of 27-OHC have been reported in ERα- incidence, and the vice versa with Asian men (Heinlein positive breast tumours in comparison to normal breast & Chang 2004). The initiation of PCa may involve gene tissue, along with an observed reduction in the 27-OHC- rearrangements that activate common growth-promoting metabolising enzyme CYP7B1 (Wu et al. 2013). In vivo pathways (Tomlins et al. 2005, Hermans et al. 2006). xenograft experiments have additionally shown that Dysregulation of PI3K/Akt and RAS/RAF pathways are 27-OHC alone is sufficient to support oestrogenic activity also implicated in PCa initiation and progression. Early- in ER-dependent BCa models, providing rationale for stage PCa can be treated by castration through surgical 27-OHC targeting (Wu et al. 2013). Similar to the Chen (orchiectomy) or chemical means. Chemical castration et al. study in PCa, a high-cholesterol diet alone increased involves use of gonadotropin-releasing hormone (GnRH) the growth and metastasis of ER-positive tumours in a analogues such as leuprolide and goserelin or GnRH mouse BCa model (Nelson et al. 2013). antagonist such as degarelix. Patients are also subjected Obesity is major challenge faced by the modern to androgen deprivation therapy (ADT) by treatment society and associated with pathogenesis of cardiovascular with anti-androgens, which are AR ligands that compete diseases and type 2 diabetes. Obesity is also a driving force for AR-binding sites. The steroidal anti-androgens are in incidence and increased recurrence of PCa and BCa. now replaced with non-steroidal ones such as flutamide, Besides de novo adipogenesis induced in these cancers, bicalutamide and nilutamide (Tan et al. 2015). The adipocyte-derived fatty acids also accelerate cancer second-generation AR antagonist enzalutamide is a progression. Although the tumour microenvironment of more potent antagonist with higher affinity than the PCa and BCa differ, emerging evidence suggests a similar first-generation counterparts. Abiraterone acetate that relationship between local adipocytes and PCa and BCa blocks androgen biosynthesis is clinically approved and cells. The lipid content of peri-prostatic and mammary used in combination with prednisone as a treatment for adipocytes is proportionately increased during obesity. metastatic CRPC (Auchus et al. 2014). An in vitro co-culture of obese adipocytes with breast Although ADT or chemical castration remains first or PCa cell lines, showed a higher rate of transfer of line of treatment of PCa, disease eventually progresses to FA from adipocytes to the cancer cells correlated with castration-resistant mode, which is ultimately fatal. AR is increased proliferation and survival (Balaban et al. 2017). expressed throughout cancer progression and remains a Interestingly, it is the peripheral zone of the prostate, major driving force for CRPC. Clinical and experimental which is more susceptible to becoming cancerous and evidence suggests a positive correlation between AR is enriched in genes regulating lipid metabolism like expression and lower recurrence-free survival and disease FASN, ACACA, ACSL1 and ACSL3. Together, these studies progression. Different mechanisms have been proposed underscore the importance of targeting intermediates to explain the progression to CRPC like (1) AR gene in the lipid/cholesterol biosynthetic pathway in the amplifications enabling increased sensitivity of the AR treatment of PCa. De novo lipid biosynthesis requires the to its agonists under conditions of androgen deprivation; substrates acetyl CoA and NADPH. Therefore, lipogenesis (2) overexpression of AR coregulators such as SRC1, SRC-3, is coupled to other metabolic pathways like glucose and CDC25B, TIP60 and NMT55, allowing AR sensitisation at glutamine to derive these substrates (Zhang & Du 2012). low levels of androgens; (3) AR mutations that render the

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R445 Cancer back receptor responsive to alternate, non-androgen ligands like endocrine therapy resistance. Consequently, a number oestrogen, progesterone, cortisol and anti-androgens; (4) of clinical trials combining such therapies with those ligand-independent AR activation such as that mediated that target HER2 such as gefitinib have been undertaken, by IL6, the circulating levels of which is often higher in yet unpublished to date (Baselga et al. 2005; https:// metastatic PCa (Hobisch et al. 1998); (5) interaction with clinicaltrials.gov/ct2/show/NCT00080743; https:// tumour suppressors like PTEN, the loss of which in PCa clinicaltrials.gov/ct2/show/NCT00632723). The growth may increase AR turnover and hence transcriptional factors and signalling pathways driven by these tumours, activity and (6) AR-independent mechanisms (Heinlein despite their growth factor dependency switch, regulate & Chang 2004, Tan et al. 2015). The acquisition of some BCa tumour metabolism. AR mutations that renders AR transcriptional activity by Alterations in both tumour epigenomic and allowing anti-androgens like bicalutamide (Hara et al. transcriptomic profiles post endocrine treatment have 2003) and adrenal androgens DHEA and androstenediol been observed in vitro and in vivo by the Magnani to function as transcriptional agonists is a frequent group following the generation of endocrine therapy cause of failure of ADT. Consequently, antiandrogen resistance BCa cell lines and NOD-SCID mouse metastatic withdrawal syndrome is observed in a proportion of model generation (Nguyen et al. 2015). The epigenetic patients with failure of ADT, with tumour regression reprogramming of ERα-positive BCas, in particular after discontinuation of antiandrogen treatment. For a H3K27ac modifications in promoter-proximal regions detailed description of various mechanisms leading to has been shown to occur and potentiate endocrine failure of ADT, audience is directed to reviews by Heinlein therapy resistance mechanisms following treatment & Chang (Heinlein & Chang 2004) and Karantanos et al. and particularly augment metastatic development. A (Karantanos et al. 2013). number of H3K27ac alterations in close proximity to the In BCa, endocrine therapies such as tamoxifen, promoters of several genes, which are already known to be aromatase inhibitors (AIs), including letrozole and involved in endocrine therapy resistance were observed anastrozole and the ER antagonists Faslodex (fulvestrant) (Magnani et al. 2013). In addition, the upregulation of target circulating oestrogen and consequently, deprive the cholesterol biosynthesis (CB) pathway in AI-resistant ER-dependent tumour cells of ligand activation. However, BCa cells was also reported, including the upregulation both intrinsic and acquired de novo resistance to endocrine of a number of genes, which are known to regulate the therapies occurs in a subset of patients. Breast tumour synthesis of 27-OHC (Nguyen et al. 2015). It is known that resistance to ER-targeted therapies has clinically been 27-OHC facilitates the oestrogen-independent binding of observed to occur via a plethora of mechanisms, with ERα to a plethora of genomic regulatory loci and hence such tumours, reverting to low expression of ERα and can drive ER-dependent transcription when endogenous thus losing their ER-driven phenotype (Osborne & Schiff levels of oestrogen are low following endocrine therapy 2011). ER-positive BCas have clinically been shown to (Nelson et al. 2013). Furthermore, endocrine therapy revert to a HER2-driven dependency following endocrine can directly regulate cellular cholesterol metabolism by treatment, predominately, via HER2 gene amplification or targeting the cholesterol epoxide hydrolase enzymatic overexpression (Meng et al. 2004, Gutierrez et al. 2005, complex and the generation of the oncometabolite Lipton et al. 2005). Several pre-clinical studies have oxysterol 6-oxo-cholestan-3β,5α-diol, otherwise referred highlighted that breast tumours can alternate between an to as OCDO (Leignadier et al. 2017, Voisin et al. 2017). ER and HER2 dependency following prolonged targeted OCDO drives BCa progression by facilitating the nuclear therapy exposure (Meng et al. 2004, Gutierrez et al. 2005, translocation of the glucocorticoid receptor (GR) and Lipton et al. 2005, Massarweh et al. 2008). Growth factor consequently, leads to the transcriptional activation receptor overexpression is one method by which tumours of a number of GR target genes involved in endocrine can circumvent endocrine therapy responsiveness, in therapy resistance (Voisin et al. 2017). The Magnani addition to HER2, IGF, FGF, VEGF and Src have also been group has developed a CB signature that could be used identified as ER-independent drivers of ER-positive BCa in ERα-positive BCa patients to stratify before adjuvant in response to hormone-targeted therapy (Arpino et al. treatment, to determine whether they are likely to 2008, Morgan et al. 2009, Chakraborty et al. 2010). Similar develop endocrine therapy tumour resistance via the to HER2, the pathways regulated by such receptors and upregulation of the CB pathway and potentially benefit their corresponding ligands are frequently overexpressed from adjuvant metabolic inhibitor treatment (Nguyen or genetically amplified in a number of clinical cases of et al. 2015).

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R446 Cancer back

Table 1 Metabolic inhibitors in pre-clinical and clinical studies.

Clinical trial Drug Pathway target Mechanism of action status Source Metformin Energy production AMPK activator inhibits Ongoing Chen et al. (2018a), Chae pathways mitochondrial complex I, lipid and clinical trials et al. (2016), NCT01980823 protein synthesis and glycolysis (www.clinicaltrials.gov) Sorafenib Lipid metabolism Multi-kinase inhibitor Phase III Zhu et al. (2015) clinical trials Fatostatin Lipogenesis SREBP1 and SREBP2 inhibitor Pre-clinical Guo et al. (2014), Li et al. (2014) Fasnall, TVB-2640, Fatty acid synthesis FASN inhibitors Ongoing NCT03179904 (www. Carulenin, C75, clinical trials clinicaltrials.gov) Orlistat BMS 303141, SB Glycolysis and lipid ATP citrate lyase (ACLY) inhibitors Pre-clinical Hatzivassiliou et al. (2005), 204990 synthesis Zhao et al. (2016) Phloretin Glucose transport GLUT1 and GLUT4 Pre-clinical Ma et al. (2016), Min et al. (2015), Wu et al. (2009) 3-bromopyruvate Glycolysis Hexokinase inhibition and inhibition Pre-clinical Ho et al. (2016) of other glycolytic enzymes Statins; atorvastatin Cholesterol/lipid HMG-CoA synthase (HMGCS) Clinical trials Chen et al. (2018a) metabolism inhibitors SCD1 inhibitor Lipid metabolism Stearoyl-CoA desaturase 1 (SCD1) Pre-clinical Chen et al. (2016), Mason inhibitor et al. (2012) ACC inhibitor; Fatty acid synthesis Acetyl CoA carboxylase (ACC) Phase III Chen et al. (2018b), Griffith ND-630, GS-0976 inhibitor clinical trials et al. (2014), NCT02781584 (www.clinicaltrials.gov) CYP17A1 inhibitors; Steroid hormone Cytochrome P450 Phase III NCT00268476, NCT01842321 abiraterone metabolism 17α-hydroxylase/17,20-lyase clinical trials (www.clinicaltrials.gov) (CYP17A1) inhibitors

Conclusion and future directions: biochemical measures such as insulin, IGF, leptin and stratification of prostate and breast cancer adipokine concentrations, assessment of obesity and BMI of the patient along with etiologic heterogeneity. The use of Studies have highlighted the importance of studying metabolic inhibitors would warrant a careful examination metabolic characteristics of PCa and BCa in order to of patient metabolic history as these cancers are influenced subtype the diseases and accelerate the discovery of novel by metabolic disorders. Therefore, it is important to metabolic biomarkers in the diagnosis of these cancers. consider the medical history of patients, existing drugs used PSA is a widely used biomarker in PCa but is also elevated like metformin and statins and the presented side effects. in men with benign prostatic hyperplasia and prostatitis, Thus, the successful clinical application of metabolic drugs which limits its usefulness (Nicholson & Ricke 2011). A lies in clinical trial patient stratification and combination combinatorial approach involving transcriptomic, ChIP, therapy with existing drugs. proteomic and metabolomic data are currently employed to study novel biomarkers for early detection of these cancers. Novel approaches to quantify lipids like vibrational Raman Declaration of interest microspectroscopy are being developed, which could be The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review. a sensitive technique for tumour diagnosis and staging (O’Malley et al. 2017). Further stratification incorporating metabolite and lipogenic gene markers might prove Funding beneficial in the accurate diagnosis and treatment of the The article does not present primary research; however, the authors disease. The overexpression of SREBP and FASN in PCa would like to acknowledge the support of Prostate Cancer UK/Movember (R E S and I G M), the Norwegian Research Council (N P) and the John Black and BCa makes them attractive therapeutic targets. Other Foundation (I G M). potential therapeutic targets include ACLY, PDH, ACC and SCD. A more detailed understanding of dysregulated lipid metabolism will help in designing better drugs or Author contributions statement N P and R E S wrote the text and made the figures and table. I G M repurposing those that are already developed (Table 1). conceived the structure of the review and contributed to the writing of It would also be beneficial to incorporate a spectrum of the text.

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R447 Cancer back

Baselga J, Albanell J, Ruiz A, Lluch A, Gascón P, Guillém V, González S, Acknowledgements Sauleda S, Marimón I, Tabernero JM, et al. 2005 Phase II and tumor The authors would like to acknowledge the support of all within the pharmacodynamic study of gefitinib in patients with advanced Centre for Cancer Research and Cell Biology and particularly Dr Paul breast cancer. Journal of Clinical Oncology 23 5323–5333. (https://doi. Mullan and Prof. David Waugh. org/10.1200/JCO.2005.08.326) Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, Wang Y, Sheikh KL, Terry S, Tagawa ST, et al. 2011 Molecular References characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discovery 1 487–495. (https://doi. Abba MC, Hu Y, Sun H, Drake JA, Gaddis S, Baggerly K, Sahin A & org/10.1158/2159-8290.CD-11-0130) Aldaz CM 2005 Gene expression signature of estrogen receptor alpha Black A, Pinsky PF, Grubb RL 3rd, Falk RT, Hsing AW, Chu L, Meyer T, status in breast cancer. BMC Genomics 6 37. (https://doi. Veenstra TD, Xu X, Yu K, et al. 2014 Sex steroid hormone org/10.1186/1471-2164-6-37) metabolism in relation to risk of aggressive prostate cancer. Cancer Al Kadhi O, Traka MH, Melchini A, Troncoso-Rey P, Jurkowski W, Epidemiology, Biomarkers and Prevention 23 2374–2382. (https://doi. Defernez M, Pachori P, Mills RD, Ball RY & Mithen RF 2017 org/10.1158/1055-9965.EPI-14-0700) Increased transcriptional and metabolic capacity for lipid Brown SB & Hankinson SE 2015 Endogenous estrogens and the risk of metabolism in the peripheral zone of the prostate may underpin its breast, endometrial, and ovarian cancers. 99 8–10. (https:// increased susceptibility to cancer. Oncotarget 8 84902–84916. (https:// doi.org/10.1016/j.steroids.2014.12.013) doi.org/10.18632/oncotarget.17926) Brown DN, Caffa I, Cirmena G, Piras D, Garuti A, Gallo M, Alberti S, Alles MC, Gardiner-Garden M, Nott DJ, Wang Y, Foekens JA, Nencioni A, Ballestrero A & Zoppoli G 2016 Squalene epoxidase is a Sutherland RL, Musgrove EA & Ormandy CJ 2009 Meta-analysis and bona fide oncogene by amplification with clinical relevance in breast gene set enrichment relative to er status reveal elevated activity of cancer. Scientific Reports 6 19435. (https://doi.org/10.1038/srep19435) MYC and E2F in the “basal” breast cancer subgroup. PLoS ONE 4 Budczies J, Pfitzner BM, Gyorffy B, Winzer KJ, Radke C, Dietel M, e4710. (https://doi.org/10.1371/journal.pone.0004710) Fiehn O & Denkert C 2015 Glutamate enrichment as new diagnostic Alwarawrah Y, Hughes P, Loiselle D, Carlson DA, Darr DB, Jordan JL, opportunity in breast cancer. International Journal of Cancer 136 Xiong J, Hunter LM, Dubois LG, Thompson JW, et al. 2016 Fasnall, a 1619–1628. (https://doi.org/10.1002/ijc.29152) selective FASN inhibitor, shows potent anti-tumor activity in the Camarda R, Zhou AY, Kohnz RA, Balakrishnan S, Mahieu C, Anderton B, MMTV-Neu model of HER2(+) breast cancer. Cell Chemical Biology 23 Eyob H, Kajimura S, Tward A, Krings G, et al. 2016 Inhibition of 678–688. (https://doi.org/10.1016/j.chembiol.2016.04.011) fatty acid oxidation as a therapy for MYC-overexpressing triple- Anso E, Mullen AR, Felsher DW, Mates JM, Deberardinis RJ & negative breast cancer. Nature Medicine 22 427–432. (https://doi. Chandel NS 2013 Metabolic changes in cancer cells upon org/10.1038/nm.4055) suppression of MYC. Cancer and Metabolism 1 7. (https://doi. Chae YK, Arya A, Malecek MK, Shin DS, Carneiro B, Chandra S, org/10.1186/2049-3002-1-7) Kaplan J, Kalyan A, Altman JK, Platanias L, et al. 2016 Repurposing Arpino G, Wiechmann L, Osborne CK & Schiff R 2008 Crosstalk metformin for cancer treatment: current clinical studies. Oncotarget 7 between the estrogen receptor and the HER tyrosine kinase receptor 40767–40780. (https://doi.org/10.18632/oncotarget.8194) family: molecular mechanism and clinical implications for endocrine Chakraborty AK, Welsh A & Digiovanna MP 2010 Co-targeting the therapy resistance. Endocrine Reviews 29 217–233. (https://doi. insulin-like growth factor I receptor enhances growth-inhibitory and org/10.1210/er.2006-0045) pro-apoptotic effects of anti-estrogens in human breast cancer cell Auchus RJ, Yu MK, Nguyen S & Mundle SD 2014 Use of prednisone lines. Breast Cancer Research and Treatment 120 327–335. (https://doi. with abiraterone acetate in metastatic castration-resistant prostate org/10.1007/s10549-009-0382-5) cancer. Oncologist 19 1231–1240. (https://doi.org/10.1634/ Chandler JD, Williams ED, Slavin JL, Best JD & Rogers S 2003 Expression theoncologist.2014-0167) and localization of GLUT1 and GLUT12 in prostate carcinoma. Balaban S, Shearer RF, Lee LS, van Geldermalsen M, Schreuder M, Cancer 97 2035–2042. (https://doi.org/10.1002/cncr.11293) Shtein HC, Cairns R, Thomas KC, Fazakerley DJ, Grewal T, et al. 2017 Chen L, Ren J, Yang L, Li Y, Fu J, Tian Y, Qiu F, Liu Z & Qiu Y 2016 Adipocyte lipolysis links obesity to breast cancer growth: adipocyte- Stearoyl-CoA desaturase-1 mediated cell apoptosis in colorectal derived fatty acids drive breast cancer cell proliferation and migration. cancer by promoting ceramide synthesis. Scientific Reports 6 article id Cancer and Metabolism 5 1. (https://doi.org/10.1186/s40170-016-0163-7) 19665. (https://doi.org/10.1038/srep19665) Bao J, Zhu L, Zhu Q, Su J, Liu M & Huang W 2016 SREBP-1 is an Chen J, Guccini I, Mitri DD, Brina D, Revandkar A, Sarti M, Pasquini E, independent prognostic marker and promotes invasion and Alajati A, Pinton S, Losa M, et al. 2018a Compartmentalized migration in breast cancer. Oncology Letters 12 2409–2416. (https:// activities of the pyruvate dehydrogenase complex sustain lipogenesis doi.org/10.3892/ol.2016.4988) in prostate cancer. Nature Genetics 50 219–228. (https://doi. Barfeld SJ, Fazli L, Persson M, Marjavaara L, Urbanucci A, org/10.1038/s41588-017-0026-3) Kaukoniemi KM, Rennie PS, Ceder Y, Chabes A, Visakorpi T, et al. Chen M, Zhang J, Sampieri K, Clohessy JG, Mendez L, Gonzalez- 2015 Myc-dependent purine biosynthesis affects nucleolar stress and Billalabeitia E, Liu XS, Lee YR, Fung J, Katon JM, et al. 2018b An therapy response in prostate cancer. Oncotarget 6 12587–12602. aberrant SREBP-dependent lipogenic program promotes metastatic (https://doi.org/10.18632/oncotarget.3494) prostate cancer. Nature Genetics 50 206–218. (https://doi.org/10.1038/ Barfeld SJ, Urbanucci A, Itkonen HM, Fazli L, Hicks JL, Thiede B, s41588-017-0027-2) Rennie PS, Yegnasubramanian S, DeMarzo AM & Mills IG 2017 Collins LC, Cole KS, Marotti JD, Hu R, Schnitt SJ & Tamimi RM 2011 c-Myc antagonises the transcriptional activity of the androgen Androgen receptor expression in breast cancer in relation to receptor in prostate cancer affecting key gene networks. eBioMedicine molecular phenotype: results from the Nurses’ Health Study. Modern 18 83–93. (https://doi.org/10.1016/j.ebiom.2017.04.006) Pathology 24 924–931. (https://doi.org/10.1038/modpathol.2011.54) Barnes CJ, Vadlamudi RK & Kumar R 2004 Novel estrogen receptor Costello LC & Franklin RB 1993 Testosterone regulates pyruvate coregulators and signaling molecules in human diseases. Cellular and dehydrogenase activity of prostate mitochondria. Hormone and Molecular Life Sciences 61 281–291. (https://doi.org/10.1007/s00018- Metabolic Research 25 268–270. (https://doi. 003-3222-5) org/10.1055/s-2007-1002094)

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R448 Cancer back

Costello LC & Franklin RB 2006 The clinical relevance of the proposed mechanism for regulation of net citrate production in metabolism of prostate cancer; zinc and tumor suppression: prostate. Hormone and Metabolic Research 18 177–181. (https://doi. connecting the dots. Molecular Cancer 5 17. (https://doi. org/10.1055/s-2007-1012264) org/10.1186/1476-4598-5-17) Franklin RB, Feng P, Milon B, Desouki MM, Singh KK, Kajdacsy-Balla A, Costello LC, Liu Y & Franklin RB 1995 Testosterone stimulates the Bagasra O & Costello LC 2005 hZIP1 zinc uptake transporter down biosynthesis of m-aconitase and citrate oxidation in prostate regulation and zinc depletion in prostate cancer. Molecular Cancer 4 epithelial cells. Molecular and Cellular Endocrinology 112 45–51. 32. (https://doi.org/10.1186/1476-4598-4-32) (https://doi.org/10.1016/0303-7207(95)03582-R) Franklin RB, Zou J, Yu Z & Costello LC 2006 EAAC1 is expressed in rat Costello LC, Liu Y, Zou J & Franklin RB 2000 Mitochondrial aconitase and human prostate epithelial cells; functions as a high-affinity gene expression is regulated by testosterone and prolactin in prostate L-aspartate transporter; and is regulated by prolactin and epithelial cells. Prostate 42 196–202. (https://doi.org/10.1002/ testosterone. BMC Biochemistry 7 10. (https://doi.org/10.1186/1471- (SICI)1097-0045(20000215)42:3<196::AID-PROS5>3.0.CO;2-8) 2091-7-10) Deming SL, Nass SJ, Dickson RB & Trock BJ 2000 C-myc amplification in Fumarola C, Caffarra C, La Monica S, Galetti M, Alfieri RR, Cavazzoni A, breast cancer: a meta-analysis of its occurrence and prognostic Galvani E, Generali D, Petronini PG & Bonelli MA 2013 Effects of relevance. British Journal of Cancer 83 1688–1695. (https://doi. sorafenib on energy metabolism in breast cancer cells: role of AMPK- org/10.1054/bjoc.2000.1522) mTORC1 signaling. Breast Cancer Research and Treatment 141 67–78. Desouki MM, Geradts J, Milon B, Franklin RB & Costello LC 2007 hZip2 (https://doi.org/10.1007/s10549-013-2668-x) and hZip3 zinc transporters are down regulated in human prostate García-Becerra R, Santos N, Díaz L & Camacho J 2013 Mechanisms of adenocarcinomatous glands. Molecular Cancer 6 37. (https://doi. resistance to endocrine therapy in breast cancer: focus on signaling org/10.1186/1476-4598-6-37) pathways, miRNAs and genetically based resistance. International Doane AS, Danso M, Lal P, Donaton M, Zhang L, Hudis C & Gerald WL Journal of Molecular Sciences 14 108–145. (https://doi.org/10.3390/ 2006 An estrogen receptor-negative breast cancer subset ijms14010108) characterized by a hormonally regulated transcriptional program and Garrido P, Moran J, Alonso A, Gonzalez S & Gonzalez C 2013. 17beta- response to androgen. Oncogene 25 3994–4008. (https://doi. estradiol activates glucose uptake via GLUT4 translocation and PI3K/ org/10.1038/sj.onc.1209415) Akt signaling pathway in MCF-7 cells. Endocrinology 154 1979–1989. Eacker SM, Agrawal N, Qian K, Dichek HL, Gong EY, Lee K & Braun RE (https://doi.org/10.1210/en.2012-1558) 2008 Hormonal regulation of testicular steroid and cholesterol Godoy A, Ulloa V, Rodriguez F, Reinicke K, Yanez AJ, Garcia Mde L, homeostasis. Molecular Endocrinology 22 623–635. (https://doi. Medina RA, Carrasco M, Barberis S, Castro T, et al. 2006 Differential org/10.1210/me.2006-0534) subcellular distribution of glucose transporters GLUT1-6 and GLUT9 Eidelman E, Twum-Ampofo J, Ansari J & Siddiqui MM 2017 The in human cancer: ultrastructural localization of GLUT1 and GLUT5 metabolic phenotype of prostate cancer. Frontiers in Oncology 7 131. in breast tumor tissues. Journal of Cellular Physiology 207 614–627. (https://doi.org/10.3389/fonc.2017.00131) (https://doi.org/10.1002/jcp.20606) Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, Wallis JW, Van Tine BA, Gottlieb B, Beitel LK, Nadarajah A, Paliouras M & Trifiro M 2012 The Hoog J, Goiffon RJ, Goldstein TC, et al. 2012 Whole-genome analysis androgen receptor gene mutations database: 2012 update. Human informs breast cancer response to aromatase inhibition. Nature 486 Mutation 33 887–894. (https://doi.org/10.1002/humu.22046) 353–360. (https://doi.org/10.1038/nature11143) Graner E, Tang D, Rossi S, Baron A, Migita T, Weinstein LJ, Ettinger SL, Sobel R, Whitmore TG, Akbari M, Bradley DR, Gleave ME & Lechpammer M, Huesken D, Zimmermann J, Signoretti S, et al. 2004 Nelson CC 2004 Dysregulation of sterol response element-binding The isopeptidase USP2a regulates the stability of fatty acid synthase proteins and downstream effectors in prostate cancer during in prostate cancer. Cancer Cell 5 253–261. (https://doi.org/10.1016/ progression to androgen independence. Cancer Research 64 S1535-6108(04)00055-8) 2212–2221. (https://doi.org/10.1158/0008-5472.CAN-2148-2) Griffith DA, Kung DW, Esler WP, Amor PA, Bagley SW, Beysen C, Farmer P, Bonnefoi H, Becette V, Tubiana-Hulin M, Fumoleau P, Carvajal-Gonzalez S, Doran SD, Limberakis C, Mathiowetz AM, et al. Larsimont D, Macgrogan G, Bergh J, Cameron D, Goldstein D, et al. 2014 Decreasing the rate of metabolic ketone reduction in the 2005 Identification of molecular apocrine breast tumours by discovery of a clinical acetyl-CoA carboxylase inhibitor for the microarray analysis. Oncogene 24 4660–4671. (https://doi. treatment of diabetes. Journal of Medicinal Chemistry 57 org/10.1038/sj.onc.1208561) 10512–10526. (https://doi.org/10.1021/jm5016022) Feldman BJ & Feldman D 2001 The development of androgen- Guo D, Bell EH, Mischel P & Chakravarti A 2014 Targeting SREBP-1- independent prostate cancer. Nature Reviews Cancer 1 34–45. (https:// driven lipid metabolism to treat cancer. Current Pharmaceutical Design doi.org/10.1038/35094009) 20 2619–2626. (https://doi.org/10.2174/13816128113199990486) Fiaschi T, Marini A, Giannoni E, Taddei ML, Gandellini P, De Donatis A, Gutierrez MC, Detre S, Johnston S, Mohsin SK, Shou J, Allred DC, Lanciotti M, Serni S, Cirri P & Chiarugi P 2012 Reciprocal metabolic Schiff R, Osborne CK & Dowsett M 2005 Molecular changes in reprogramming through lactate shuttle coordinately influences tamoxifen-resistant breast cancer: relationship between estrogen tumor-stroma interplay. Cancer Research 72 5130–5140. (https://doi. receptor, HER-2, and p38 mitogen-activated protein kinase. Journal of org/10.1158/0008-5472.CAN-12-1949) Clinical Oncology 23 2469–2476. (https://doi.org/10.1200/ Flouriot G, Brand H, Denger S, Metivier R, Kos M, Reid G, Sonntag- JCO.2005.01.172) Buck V & Gannon F 2000 Identification of a new isoform of the Hara T, Miyazaki J, Araki H, Yamaoka M, Kanzaki N, Kusaka M & human estrogen receptor-alpha (hER-alpha) that is encoded by Miyamoto M 2003 Novel mutations of androgen receptor: a possible distinct transcripts and that is able to repress hER-alpha activation mechanism of bicalutamide withdrawal syndrome. Cancer Research function 1. EMBO Journal 19 4688–4700. (https://doi.org/10.1093/ 63 149–153. emboj/19.17.4688) Hatzivassiliou G, Zhao F, Bauer DE, Andreadis C, Shaw AN, Dhanak D, Ford WC & Harrison A 1984 The role of citrate in determining the Hingorani SR, Tuveson DA & Thompson CB 2005 ATP citrate lyase activity of calcium ions in human semen. International Journal of inhibition can suppress tumor cell growth. Cancer Cell 8 311–321. Andrology 7 198–202. (https://doi.org/10.1111/j.1365-2605.1984. (https://doi.org/10.1016/j.ccr.2005.09.008) tb00777.x) Hayashi SI, Eguchi H, Tanimoto K, Yoshida T, Omoto Y, Inoue A, Franklin RB, Kahng MW, Akuffo V & Costello LC 1986 The effect of Yoshida N & Yamaguchi Y 2003 The expression and function of testosterone on citrate synthesis and citrate oxidation and a estrogen receptor alpha and beta in human breast cancer and its

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R449 Cancer back

clinical application. Endocrine-Related Cancer 10 193–202. (https:// Kenny L 2016 The use of novel PET tracers to image breast cancer doi.org/10.1677/erc.0.0100193) biologic processes such as proliferation, DNA damage and repair, and Heemers HV, Verhoeven G & Swinnen JV 2006 Androgen activation of angiogenesis. Journal of Nuclear Medicine 57 (Supplement 1) 89S–95S. the sterol regulatory element-binding protein pathway: current (https://doi.org/10.2967/jnumed.115.157958) insights. Molecular Endocrinology 20 2265–2277. (https://doi. Kim SJ, Choi H, Park SS, Chang C & Kim E 2011 Stearoyl CoA org/10.1210/me.2005-0479) desaturase (SCD) facilitates proliferation of prostate cancer cells Heinlein CA & Chang C 2004 Androgen receptor in prostate cancer. through enhancement of androgen receptor transactivation. Endocrine Reviews 25 276–308. (https://doi.org/10.1210/er.2002-0032) Molecules and Cells 31 371–377. (https://doi.org/10.1007/s10059-011- Hermans KG, van Marion R, van Dekken H, Jenster G, van 0043-5) Weerden WM & Trapman J 2006 TMPRSS2: ERG fusion by Kinoshita H, Shi Y, Sandefur C, Meisner LF, Chang C, Choon A, translocation or interstitial deletion is highly relevant in androgen- Reznikoff CR, Bova GS, Friedl A & Jarrard DF 2000 Methylation of dependent prostate cancer, but is bypassed in late-stage androgen the androgen receptor minimal promoter silences transcription in receptor-negative prostate cancer. Cancer Research 66 10658–10663. human prostate cancer. Cancer Research 60 3623–3630. (https://doi.org/10.1158/0008-5472.CAN-06-1871) Lee JS, Sul JY, Park JB, Lee MS, Cha EY, Song IS, Kim JR & Chang ES Ho N, Morrison J, Silva A & Coomber BL 2016 The effect of 2013 Fatty acid synthase inhibition by amentoflavone suppresses 3-bromopyruvate on human colorectal cancer cells is dependent on HER2/neu (erbB2) oncogene in SKBR3 human breast cancer cells. glucose concentration but not hexokinase II expression. Bioscience Phytotherapy Research 27 713–720. (https://doi.org/10.1002/ Reports 36 e00299. (https://doi.org/10.1042/BSR20150267) ptr.4778) Hobisch A, Eder IE, Putz T, Horninger W, Bartsch G, Klocker H & Leignadier J, Dalenc F, Poirot M & Silvente-Poirot S 2017 Improving the Culig Z 1998 Interleukin-6 regulates prostate-specific protein efficacy of hormone therapy in breast cancer: the role of cholesterol expression in prostate carcinoma cells by activation of the androgen metabolism in SERM-mediated autophagy, cell differentiation and receptor. Cancer Research 58 4640–4645. death. Biochemical Pharmacology 144 18–28. (https://doi. Hsing AW, Sakoda LC & Chua S Jr 2007 Obesity, metabolic syndrome, org/10.1016/j.bcp.2017.06.120) and prostate cancer. American Journal of Clinical Nutrition 86 Li X, Chen YT, Hu P & Huang WC 2014 Fatostatin displays high anti- s843–s857. (https://doi.org/10.1093/ajcn/86.3.843S) tumor activity in prostate cancer by blocking SREBP-regulated Huang WC, Li X, Liu J, Lin J & Chung LW 2012 Activation of androgen metabolic pathways and androgen receptor signaling. Molecular receptor, lipogenesis, and oxidative stress converged by SREBP-1 is Cancer Therapeutics 13 855–866. (https://doi.org/10.1158/1535-7163. responsible for regulating growth and progression of prostate cancer MCT-13-0797) cells. Molecular Cancer Research 10 133–142. (https://doi. Li X, Wu JB, Chung LW & Huang WC 2015 Anti-cancer efficacy of org/10.1158/1541-7786.MCR-11-0206) SREBP inhibitor, alone or in combination with docetaxel, in prostate Huss JM, Torra IP, Staels B, Giguere V & Kelly DP 2004 Estrogen- cancer harboring p53 mutations. Oncotarget 6 41018–41032. (https:// related receptor alpha directs proliferator-activated doi.org/10.18632/oncotarget.5879) receptor alpha signaling in the transcriptional control of energy Li X, Wu JB, Li Q, Shigemura K, Chung LW & Huang WC 2016 SREBP-2 metabolism in cardiac and skeletal muscle. Molecular and Cellular promotes stem cell-like properties and metastasis by transcriptional Biology 24 9079–9091. (https://doi.org/10.1128/MCB.24.20.9079- activation of c-Myc in prostate cancer. Oncotarget 7 12869–12884. 9091.2004) (https://doi.org/10.18632/oncotarget.7331) Ignatiadis M & Sotiriou C 2013 Luminal breast cancer: from biology to Lipton A, Leitzel K, Ali SM, Demers L, Harvey HA, Chaudri-Ross HA, treatment. Nature Reviews Clinical Oncology 10 494–506. (https://doi. Evans D, Lang R, Hackl W, Hamer P, et al. 2005 Serum HER-2/neu org/10.1038/nrclinonc.2013.124) conversion to positive at the time of disease progression in patients Itkonen HM, Minner S, Guldvik IJ, Sandmann MJ, Tsourlakis MC, with breast carcinoma on hormone therapy. Cancer 104 257–263. Berge V, Svindland A, Schlomm T & Mills IG 2013 O-GlcNAc (https://doi.org/10.1002/cncr.21202) transferase integrates metabolic pathways to regulate the stability of Liu IJ, Zafar MB, Lai YH, Segall GM & Terris MK 2001 c-MYC in human prostate cancer cells. Cancer Research 73 Fluorodeoxyglucose positron emission tomography studies in 5277–5287. (https://doi.org/10.1158/0008-5472.CAN-13-0549) diagnosis and staging of clinically organ-confined prostate cancer. Itkonen HM, Brown M, Urbanucci A, Tredwell G, Ho Lau C, Barfeld S, Urology 57 108–111. (https://doi.org/10.1016/S0090-4295(00)00896-7) Hart C, Guldvik IJ, Takhar M, Heemers HV, et al. 2017 Lipid Locke JA, Guns ES, Lubik AA, Adomat HH, Hendy SC, Wood CA, degradation promotes prostate cancer cell survival. Oncotarget 8 Ettinger SL, Gleave ME & Nelson CC 2008 Androgen levels increase 38264–38275. (https://doi.org/10.18632/oncotarget.16123) by intratumoral de novo steroidogenesis during progression of Jadvar H 2012 Molecular imaging of prostate cancer: PET radiotracers. castration-resistant prostate cancer. Cancer Research 68 6407–6415. American Journal of Roentgenology 199 278–291. (https://doi. (https://doi.org/10.1158/0008-5472.CAN-07-5997) org/10.2214/AJR.12.8816) Lu X, Bennet B, Mu E, Rabinowitz J & Kang Y 2010 Metabolomic Johansson M, Van Guelpen B, Vollset SE, Hultdin J, Bergh A, Key T, changes accompanying transformation and acquisition of metastatic Midttun O, Hallmans G, Ueland PM & Stattin P 2009 One-carbon potential in a syngeneic mouse mammary tumor model. Journal of metabolism and prostate cancer risk: prospective investigation of Biological Chemistry 285 9317–9321. (https://doi.org/10.1074/jbc. seven circulating B vitamins and metabolites. Cancer Epidemiology, C110.104448) Biomarkers and Prevention 18 1538–1543. (https://doi. Ma L, Wang R, Nan Y, Li W, Wang Q & Jin F 2016 Phloretin exhibits an org/10.1158/1055-9965.EPI-08-1193) anticancer effect and enhances the anticancer ability of cisplatin on Kang SS, Chun YK, Hur MH, Lee HK, Kim YJ, Hong SR, Lee JH, Lee SG non-small cell lung cancer cell lines by regulating expression of & Park YK 2002 Clinical significance of glucose transporter 1 apoptotic pathways and matrix metalloproteinases. International (GLUT1) expression in human breast carcinoma. Japanese Journal of Journal of Oncology 48 843–853. (https://doi.org/10.3892/ Cancer Research 93 1123–1128. (https://doi. ijo.2015.3304) org/10.1111/j.1349-7006.2002.tb01214.x) Magnani L, Stoeck A, Zhang X, Lanczky A, Mirabella AC, Wang TL, Karantanos T, Corn PG & Thompson TC 2013 Prostate cancer Gyorffy B & Lupien M 2013 Genome-wide reprogramming of the progression after androgen deprivation therapy: mechanisms of chromatin landscape underlies endocrine therapy resistance in breast castrate-resistance and novel therapeutic approaches. Oncogene 32 cancer. PNAS 110 E1490–E1499. (https://doi.org/10.1073/ 5501–5511. (https://doi.org/10.1038/onc.2013.206) pnas.1219992110)

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R450 Cancer back

Makhov PB, Golovine KV, Kutikov A, Canter DJ, Rybko VA, Morgan L, Gee J, Pumford S, Farrow L, Finlay P, Robertson J, Ellis I, Roshchin DA, Matveev VB, Uzzo RG & Kolenko VM 2011 Reversal of Kawakatsu H, Nicholson R & Hiscox S 2009 Elevated Src kinase epigenetic silencing of AP-2alpha results in increased zinc uptake in activity attenuates Tamoxifen response in vitro and is associated DU-145 and LNCaP prostate cancer cells. Carcinogenesis 32 with poor prognosis clinically. Cancer Biology and Therapy 8 1773–1781. (https://doi.org/10.1093/carcin/bgr212) 1550–1558. (https://doi.org/10.4161/cbt.8.16.8954) Manavathi B, Dey O, Gajulapalli VN, Bhatia RS, Bugide S & Kumar R Mullen AR, Wheaton WW, Jin ES, Chen PH, Sullivan LB, Cheng T, 2013 Derailed estrogen signaling and breast cancer: an authentic Yang Y, Linehan WM, Chandel NS & DeBerardinis RJ 2011 Reductive couple. Endocrine Reviews 34 1–32. (https://doi.org/10.1210/er.2011- carboxylation supports growth in tumour cells with defective 1057) mitochondria. Nature 481 385–388. (https://doi.org/10.1038/ Mashima T, Seimiya H & Tsuruo T 2009 De novo fatty-acid synthesis nature10642) and related pathways as molecular targets for cancer therapy. British Nelson ER, Wardell SE, Jasper JS, Park S, Suchindran S, Howe MK, Journal of Cancer 100 1369–1372. (https://doi.org/10.1038/sj. Carver NJ, Pillai RV, Sullivan PM, Sondhi V, et al. 2013. bjc.6605007) 27-hydroxycholesterol links hypercholesterolemia and breast cancer Mason P, Liang B, Li L, Fremgen T, Murphy E, Quinn A, Madden SL, pathophysiology. Science 342 1094–1098. (https://doi.org/10.1126/ Biemann HP, Wang B, Cohen A, et al. 2012 SCD1 inhibition causes science.1241908) cancer cell death by depleting mono-unsaturated fatty acids. PLoS Nguyen VT, Barozzi I, Faronato M, Lombardo Y, Steel JH, Patel N, ONE 7 e33823. (https://doi.org/10.1371/journal.pone.0033823) Darbre P, Castellano L, Gyorffy B, Woodley L, et al. 2015 Differential Massarweh S, Osborne CK, Creighton CJ, Qin L, Tsimelzon A, Huang S, epigenetic reprogramming in response to specific endocrine Weiss H, Rimawi M & Schiff R 2008 Tamoxifen resistance in breast therapies promotes cholesterol biosynthesis and cellular invasion. tumors is driven by growth factor receptor signaling with Nature Communications 6 10044. (https://doi.org/10.1038/ repression of classic estrogen receptor genomic function. Cancer ncomms10044) Research 68 826–833. (https://doi.org/10.1158/0008-5472.CAN-07- Ni M, Chen Y, Fei T, Li D, Lim E, Liu XS & Brown M 2013 Amplitude 2707) modulation of androgen signaling by c-MYC. Genes and Development Massie CE, Lynch A, Ramos-Montoya A, Boren J, Stark R, Fazli L, 27 734–748. (https://doi.org/10.1101/gad.209569.112) Warren A, Scott H, Madhu B, Sharma N, et al. 2011 The androgen Nicholson TM & Ricke WA 2011 Androgens and estrogens in benign receptor fuels prostate cancer by regulating central metabolism and prostatic hyperplasia: past, present and future. Differentiation 82 biosynthesis. EMBO Journal 30 2719–2733. (https://doi.org/10.1038/ 184–199. (https://doi.org/10.1016/j.diff.2011.04.006) emboj.2011.158) Niemeier LA, Dabbs DJ, Beriwal S, Striebel JM & Bhargava R 2010 Massie CE, Mills IG & Lynch AG 2017 The importance of DNA Androgen receptor in breast cancer: expression in estrogen receptor- methylation in prostate cancer development. Journal of Steroid positive tumors and in estrogen receptor-negative tumors with Biochemistry and Molecular Biology 166 1–15. (https://doi. apocrine differentiation. Modern Pathology 23 205–212. (https://doi. org/10.1016/j.jsbmb.2016.04.009) org/10.1038/modpathol.2009.159) May FE & Westley BR 1988 Identification and characterization of Nilsson S & Gustafsson JA 2011 Estrogen receptors: therapies targeted to estrogen-regulated RNAs in human breast cancer cells. Journal of receptor subtypes. Clinical Pharmacology and Therapeutics 89 44–55. Biological Chemistry 263 12901–12908. (https://doi.org/10.1038/clpt.2010.226) McNeil CM, Sergio CM, Anderson LR, Inman CK, Eggleton SA, O’Malley J, Kumar R, Kuzmin AN, Pliss A, Yadav N, Balachandar S, Murphy NC, Millar EK, Crea P, Kench JG, Alles MC, et al. 2006 Wang J, Attwood K, Prasad PN & Chandra D 2017 Lipid c-Myc overexpression and endocrine resistance in breast cancer. quantification by Raman microspectroscopy as a potential biomarker Journal of Steroid Biochemistry and Molecular Biology 102 147–155. in prostate cancer. Cancer Letters 397 52–60. (https://doi.org/10.1016/j.jsbmb.2006.09.028) Osborne CK & Schiff R 2011 Mechanisms of endocrine resistance in Medes G, Thomas A & Weinhouse S 1953 Metabolism of neoplastic breast cancer. Annual Review of Medicine 62 233–247. (https://doi. tissue. IV. A study of lipid synthesis in neoplastic tissue slices in org/10.1146/annurev-med-070909-182917) vitro. Cancer Research 13 27–29. Pan T, Gao L, Wu G, Shen G, Xie S, Wen H, Yang J, Zhou Y, Tu Z & Meng S, Tripathy D, Shete S, Ashfaq R, Haley B, Perkins S, Beitsch P, Qian W 2015 Elevated expression of glutaminase confers glucose Khan A, Euhus D, Osborne C, et al. 2004 HER-2 gene amplification utilization via glutaminolysis in prostate cancer. Biochemical and can be acquired as breast cancer progresses. PNAS 101 9393–9398. Biophysical Research Communications 456 452–458. (https://doi. (https://doi.org/10.1073/pnas.0402993101) org/10.1016/j.bbrc.2014.11.105) Miller TW, Balko JM, Ghazoui Z, Dunbier A, Anderson H, Dowsett M, Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Gonzalez-Angulo AM, Mills GB, Miller WR, Wu H, et al. 2011 A gene Pollack JR, Ross DT, Johnsen H, Akslen LA, et al. 2000 Molecular expression signature from human breast cancer cells with acquired portraits of human breast tumours. Nature 406 747–752. (https://doi. hormone independence identifies MYC as a mediator of antiestrogen org/10.1038/35021093) resistance. Clinical Cancer Research 17 2024–2034. (https://doi. Pertega-Gomes N, Felisbino S, Massie CE, Vizcaino JR, Coelho R, org/10.1158/1078-0432.CCR-10-2567) Sandi C, Simoes-Sousa S, Jurmeister S, Ramos-Montoya A, Asim M, et Min J, Li X, Huang K, Tang H, Ding X, Qi C, Qin X & Xu Z 2015 al. 2015a A glycolytic phenotype is associated with prostate cancer Phloretin induces apoptosis of non-small cell lung carcinoma A549 progression and aggressiveness: a role for monocarboxylate cells via JNK1/2 and p38 MAPK pathways. Oncology Reports 34 transporters as metabolic targets for therapy. Journal of Pathology 236 2871–2879. (https://doi.org/10.3892/or.2015.4325) 517–530. (https://doi.org/10.1002/path.4547) Mishra P, Tang W, Putluri V, Dorsey TH, Jin F, Wang F, Zhu D, Amable L, Pertega-Gomes N, Vizcaino JR, Felisbino S, Warren AY, Shaw G, Kay J, Deng T, Zhang S, et al. 2018 ADHFE1 is a breast cancer oncogene Whitaker H, Lynch AG, Fryer L, Neal DE, et al. 2015b Epigenetic and and induces metabolic reprogramming. Journal of Clinical oncogenic regulation of SLC16A7 (MCT2) results in protein over- Investigation 128 323–340. (https://doi.org/10.1172/JCI93815) expression, impacting on signalling and cellular phenotypes in Moinfar F, Okcu M, Tsybrovskyy O, Regitnig P, Lax SF, Weybora W, prostate cancer. Oncotarget 6 21675–21684. (https://doi.org/10.18632/ Ratschek M, Tavassoli FA & Denk H 2003 Androgen receptors oncotarget.4328) frequently are expressed in breast carcinomas: potential relevance to Piert M, Shao X, Raffel D, Davenport MS, Montgomery J, Kunju LP, new therapeutic strategies. Cancer 98 703–711. (https://doi. Hockley BG, Siddiqui J, Scott PJH, Chinnaiyan AM, et al. 2017 org/10.1002/cncr.11532) Preclinical evaluation of (11)C-sarcosine as a substrate of proton-

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R451 Cancer back

coupled amino acid transporters and first human application in Stopsack KH, Gerke TA, Andren O, Andersson SO, Giovannucci EL, prostate cancer. Journal of Nuclear Medicine 58 1216–1223. (https:// Mucci LA & Rider JR 2017 Cholesterol uptake and regulation in doi.org/10.2967/jnumed.116.173179) high-grade and lethal prostate cancers. Carcinogenesis 38 806–811. Pike MC, Spicer DV, Dahmoush L & Press MF 1993 Estrogens, (https://doi.org/10.1093/carcin/bgx058) progestogens, normal breast cell proliferation, and breast cancer risk. Swinnen JV, Roskams T, Joniau S, Van Poppel H, Oyen R, Baert L, Epidemiologic Reviews 15 17–35. (https://doi.org/10.1093/ Heyns W & Verhoeven G 2002 Overexpression of fatty acid synthase oxfordjournals.epirev.a036102) is an early and common event in the development of prostate Pinheiro C, Longatto-Filho A, Azevedo-Silva J, Casal M, Schmitt FC & cancer. International Journal of Cancer 98 19–22. (https://doi. Baltazar F 2012 Role of monocarboxylate transporters in human org/10.1002/ijc.10127) cancers: state of the art. Journal of Bioenergetics and Biomembranes 44 Tan MH, Li J, Xu HE, Melcher K & Yong EL 2015 Androgen receptor: 127–139. (https://doi.org/10.1007/s10863-012-9428-1) structure, role in prostate cancer and drug discovery. Acta Platz EA, Leitzmann MF, Visvanathan K, Rimm EB, Stampfer MJ, Pharmacologica Sinica 36 3–23. (https://doi.org/10.1038/ Willett WC & Giovannucci E 2006 Statin drugs and risk of advanced aps.2014.18) prostate cancer. Journal of the National Cancer Institute 98 1819–1825. Tennakoon JB, Shi Y, Han JJ, Tsouko E, White MA, Burns AR, Zhang A, (https://doi.org/10.1093/jnci/djj499) Xia X, Ilkayeva OR, Xin L, et al. 2014 Androgens regulate prostate Prins GS, Birch L & Greene GL 1991 Androgen receptor localization in cancer cell growth via an AMPK-PGC-1alpha-mediated metabolic different cell types of the adult rat prostate. Endocrinology 129 switch. Oncogene 33 5251–5261. (https://doi.org/10.1038/ 3187–3199. (https://doi.org/10.1210/endo-129-6-3187) onc.2013.463) Ray U & Roy SS 2017 Aberrant lipid metabolism in cancer cells – the Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, role of oncolipid-activated signaling. FEBS Journal 285 432–443. Varambally S, Cao X, Tchinda J, Kuefer R, et al. 2005 Recurrent (https://doi.org/10.1111/febs.14281) fusion of TMPRSS2 and ETS transcription factor genes in prostate Reinicke K, Sotomayor P, Cisterna P, Delgado C, Nualart F & Godoy A cancer. Science 310 644–648. (https://doi.org/10.1126/ 2012 Cellular distribution of Glut-1 and Glut-5 in benign and science.1117679) malignant human prostate tissue. Journal of Cellular Biochemistry 113 Vaz CV, Alves MG, Marques R, Moreira PI, Oliveira PF, Maia CJ & 553–562. (https://doi.org/10.1002/jcb.23379) Socorro S 2012 Androgen-responsive and nonresponsive prostate Rishi I, Baidouri H, Abbasi JA, Bullard-Dillard R, Kajdacsy-Balla A, cancer cells present a distinct glycolytic metabolism profile. Pestaner JP, Skacel M, Tubbs R & Bagasra O 2003 Prostate cancer in International Journal of Biochemistry and Cell Biology 44 2077–2084. African American men is associated with downregulation of zinc (https://doi.org/10.1016/j.biocel.2012.08.013) transporters. Applied Immunohistochemistry and Molecular Morphology Venditti M, Iwasiow B, Orr FW & Shiu RP 2002 C-myc gene expression 11 253–260. (https://doi.org/10.1097/00129039-200309000-00009) alone is sufficient to confer resistance to antiestrogen in human Robinson JL, Macarthur S, Ross-Innes CS, Tilley WD, Neal DE, Mills IG breast cancer cells. International Journal of Cancer 99 35–42. (https:// & Carroll JS 2011 Androgen receptor driven transcription in doi.org/10.1002/ijc.10269) molecular apocrine breast cancer is mediated by FoxA1. EMBO Voisin M, de Medina P, Mallinger A, Dalenc F, Huc-Claustre E, Journal 30 3019–3027. (https://doi.org/10.1038/emboj.2011.216) Leignadier J, Serhan N, Soules R, Segala G, Mougel A, et al. 2017 Robles-Fernandez I, Martinez-Gonzalez LJ, Pascual-Geler M, Cozar JM, Identification of a tumor-promoter cholesterol metabolite in human Puche-Sanz I, Serrano MJ, Lorente JA & Alvarez-Cubero MJ 2017 breast cancers acting through the glucocorticoid receptor. PNAS 114 Association between polymorphisms in sex hormones synthesis and E9346–E9355. (https://doi.org/10.1073/pnas.1707965114) metabolism and prostate cancer aggressiveness. PLoS ONE 12 Vona-Davis L, Howard-McNatt M & Rose DP 2007 Adiposity, type 2 e0185447. (https://doi.org/10.1371/journal.pone.0185447) diabetes and the metabolic syndrome in breast cancer. Obesity Rogers S, Docherty SE, Slavin JL, Henderson MA & Best JD 2003 Reviews 8 395–408. (https://doi. Differential expression of GLUT12 in breast cancer and normal org/10.1111/j.1467-789X.2007.00396.x) breast tissue. Cancer Letters 193 225–233. (https://doi.org/10.1016/ Vrtačnik P, Ostanek B, Mencej-Bedrač S & Marc J 2014 The many faces S0304-3835(03)00010-7) of estrogen signaling. Biochemical Medicine 24 329–342. Rossi S, Graner E, Febbo P, Weinstein L, Bhattacharya N, Onody T, Wang Z, Zhang X, Shen P, Loggie BW, Chang Y & Deuel TF 2005 Bubley G, Balk S & Loda M 2003 Fatty acid synthase expression Identification, cloning, and expression of human estrogen receptor- defines distinct molecular signatures in prostate cancer. Molecular alpha36, a novel variant of human estrogen receptor-alpha66. Cancer Research 1 707–715. Biochemical and Biophysical Research Communications 336 1023–1027. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, (https://doi.org/10.1016/j.bbrc.2005.08.226) Fizazi K, Mainwaring P, Piulats JM, Ng S, et al. 2013 Abiraterone in Wang Q, Hardie RA, Hoy AJ, van Geldermalsen M, Gao D, Fazli L, metastatic prostate cancer without previous chemotherapy. New Sadowski MC, Balaban S, Schreuder M, Nagarajah R, et al. 2015 England Journal of Medicine 368 138–148. (https://doi.org/10.1056/ Targeting ASCT2-mediated glutamine uptake blocks prostate cancer NEJMoa1209096) growth and tumour development. Journal of Pathology 236 278–289. Shafi AA, Putluri V, Arnold JM, Tsouko E, Maity S, Roberts JM, Coarfa C, (https://doi.org/10.1002/path.4518) Frigo DE, Putluri N, Sreekumar A, et al. 2015 Differential regulation Warner M & Gustafsson JA 2014 On estrogen, cholesterol metabolism, of metabolic pathways by androgen receptor (AR) and its and breast cancer. New England Journal of Medicine 370 572–573. constitutively active splice variant, AR-V7, in prostate cancer cells. (https://doi.org/10.1056/NEJMcibr1315176) Oncotarget 6 31997–32012. (https://doi.org/10.18632/oncotarget.5585) Wu CH, Ho YS, Tsai CY, Wang YJ, Tseng H, Wei PL, Lee CH, Liu RS & Shi X, Peng Y, Du X, Liu H, Klocker H, Lin Q, Shi J & Zhang J 2017 Lin SY 2009 In vitro and in vivo study of phloretin-induced Estradiol promotes epithelial-to-mesenchymal transition in human apoptosis in human liver cancer cells involving inhibition of type II benign prostatic epithelial cells. Prostate 77 1424–1437. (https://doi. glucose transporter. International Journal of Cancer 124 2210–2219. org/10.1002/pros.23404) (https://doi.org/10.1002/ijc.24189) Song HJ, Sneddon AA, Heys SD & Wahle KW 2012 Regulation of fatty Wu Q, Ishikawa T, Sirianni R, Tang H, McDonald JG, Yuhanna IS, acid synthase (FAS) and apoptosis in estrogen-receptor positive and Thompson B, Girard L, Mineo C, Brekken RA, et al. 2013 negative breast cancer cells by conjugated linoleic acids. 27-Hydroxycholesterol promotes cell-autonomous, ER-positive breast Prostaglandins, Leukotrienes and Essential Fatty Acids 87 197–203. cancer growth. Cell Reports 5 637–645. (https://doi.org/10.1016/j. (https://doi.org/10.1016/j.plefa.2012.09.002) celrep.2013.10.006)

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access Endocrine-Related N Poulose et al. From hormones to fats and 25:9 R452 Cancer back

Xiao H, Wang J, Yan W, Cui Y, Chen Z, Gao X, Wen X & Chen J 2018 Zhang F & Du G 2012 Dysregulated lipid metabolism in cancer. World GLUT1 regulates cell glycolysis and proliferation in prostate cancer. Journal of Biological Chemistry 3 167–174. (https://doi.org/10.4331/ Prostate 78 86–94. (https://doi.org/10.1002/pros.23448) wjbc.v3.i8.167) Xin M, Qiao Z, Li J, Liu J, Song S, Zhao X, Miao P, Tang T, Wang L, Zhao S, Torres A, Henry RA, Trefely S, Wallace M, Lee JV, Carrer A, Liu W, et al. 2016 miR-22 inhibits tumor growth and metastasis by Sengupta A, Campbell SL, Kuo YM, et al. 2016 ATP-citrate lyase targeting ATP citrate lyase: evidence in osteosarcoma, prostate controls a glucose-to-acetate metabolic switch. Cell Reports 17 cancer, cervical cancer and lung cancer. Oncotarget 7 44252–44265. 1037–1052. (https://doi.org/10.1016/j.celrep.2016.09.069) (https://doi.org/10.18632/oncotarget.10020) Zhu AX, Rosmorduc O, Evans TR, Ross PJ, Santoro A, Carrilho FJ, Yue S, Li J, Lee SY, Lee HJ, Shao T, Song B, Cheng L, Masterson TA, Bruix J, Qin S, Thuluvath PJ, Llovet JM, et al. 2015 SEARCH: a phase Liu X, Ratliff TL, et al. 2014 Cholesteryl ester accumulation induced III, randomized, double-blind, placebo-controlled trial of sorafenib by PTEN loss and PI3K/AKT activation underlies human prostate plus erlotinib in patients with advanced hepatocellular carcinoma. cancer aggressiveness. Cell Metabolism 19 393–406. (https://doi. Journal of Clinical Oncology 33 559–566. (https://doi.org/10.1200/ org/10.1016/j.cmet.2014.01.019) JCO.2013.53.7746)

Received in final form 10 May 2018 Accepted 14 May 2018 Accepted Preprint published online 14 May 2018

http://erc.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/ERC-18-0048 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 06:03:19AM via free access