M C Bruce et al. Kinome associated with 21:5 R357–R370 Review ER-positive status

The kinome associated with estrogen receptor-positive status in human breast cancer

Correspondence M Christine Bruce, Danielle McAllister and Leigh C Murphy should be addressed to L C Murphy Department of Biochemistry and Medical Genetics, Manitoba Institute of Cell Biology, University of Manitoba Email and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9 Leigh.Murphy@ med.umanitoba.ca

Abstract

Estrogen receptor alpha (ERa) regulates and is regulated by involved in several Key Words functions associated with the hallmarks of cancer. The following literature review strongly " estrogen receptors suggests that distinct kinomes exist for ERa-positive and -negative human breast cancers. " breast cancer Importantly, consistent with the known heterogeneity of ERa-positive cancers, different " phosphorylation subgroups exist, which can be defined by different kinome signatures, which in turn are " kinases correlated with clinical outcome. Strong evidence supports the interplay of networks, " endocrine therapy sensitivity suggesting that targeting a single node may not be sufficient to inhibit the network. " biomarker Therefore, identifying the important hubs/nodes associated with each clinically relevant kinome in ERC tumors could offer the ability to implement the best therapy options at diagnosis, either endocrine therapy alone or together with other targeted therapies, Endocrine-Related Cancer for improved overall outcome. Endocrine-Related Cancer (2014) 21, R357–R370

Introduction

The idea of personalized approaches to therapy in breast therapy was used for breast cancer at least in the form of cancer based on the molecular nature of the tumor can be ablation surgery such as ovariectomy dates back well over traced back to the late 1960s and early 1970s, when it was a century, the identification of ERs in breast cancers discovered that some but not all human breast tumors provided a molecular mechanism and rationale for the use express estrogen receptors (ERs; Jensen et al. 1971). This of hormonal therapies (Jensen & Jordan 2003). This led heralded the beginning of an era of targeted therapies for directly to the development of the successful modern breast cancer, identified the first biomarker used clinically endocrine therapies such as tamoxifen and other selective to predict the biological behavior of breast cancer, and estrogen receptor modulators (SERMs), which bind to the established the beginnings of understanding molecular ERs and induce conformational changes that modify and mechanisms by which the ovarian hormone, estrogen, in some cases inactivate the ERs (Jensen & Jordan 2003). drives the growth and survival of the majority of human The newer endocrine therapies, the aromatase inhibitors breast cancers (Jensen & Jordan 2003), at least initially. (AIs), which inhibit the aromatase , eliminate the Inhibiting the activity of ERs with the antiestrogen production of estrogen and therefore inhibit estrogen’s tamoxifen was the first targeted therapy in breast cancer. proliferative action (Goss et al. 2011). Although the knowledge that female hormones were It was evident from the start that not all ERC breast involved in breast cancer and the hormonal/endocrine tumors were created equal. Although a little more than

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R358 ER-positive status

70% of all breast tumors express ER, only about half of (Yamnik et al. 2009, Yamnik & Holz 2010, Murphy et al. patients with ERC tumors respond to tamoxifen. There- 2011), as well as the demonstration that a clinically fore, ERC breast cancers exhibit heterogeneity associated relevant phosphorylation profile of ERs can be identified with prognosis and treatment outcomes. The first step to in human breast cancer (Skliris et al. 2010a), suggests that resolve this heterogeneity came from the idea that the kinases and/or phosphatases associated with ERC breast measurement of a downstream target of estrogen- cancer could provide a wealth of potential drug targets to dependent ER signaling such as the progesterone receptor complement existing endocrine therapies or generate new (PR) would increase confidence that the pathway was endocrine therapies. The following is a review of kinases intact (Horwitz et al. 1975). This increased the accuracy of that have been identified as associated with ER status in treatment prediction, but was obviously still imprecise as breast tumors or those that have been implicated in the some 20–30% of ERC/PRC tumors are de novo resistant to regulation of estrogen signaling and/or modifying sensi- the endocrine therapies. Furthermore, initial response to tivity to estrogen and its antagonists in breast cancer. endocrine therapies is often followed by acquired resist- ance despite the continued expression of ER (Encarnacion Kinases identified as mutated or structurally et al. 1993, Bachleitner-Hofmann et al. 2002). altered in breast cancer in large breast cancer The next significant insight into the heterogeneity of cohorts ERC breast cancer came with the identification of HER2 (ERBB2) amplification. Approximately 20% of all breast Genome-wide analyses of large cohorts of breast cancer cancers have amplified, overexpressed HER2, and 40–50% cases are providing detailed, comprehensive analyses of of these will also be ERC. Interestingly, ERC/HER2C genomic aberrations in breast cancer at a population level. tumors are more likely to be resistant to endocrine Some studies have also provided data concerning their therapy, in particular tamoxifen, thus providing impact on clinical characteristics. These studies have important insight into the relevance of crosstalk of shown that many of the frequently altered (amplified, signaling pathways initiated at the level of the plasma fused, deleted, or mutated) encode kinases. Some of membrane with many aspects of the ER signaling these frequently altered genes, such as HER2,were pathway. This may cause ligand-independent activation previously known but others, such as MAP3K1 and its of ER signaling and hormone therapy resistance. substrate MAP2K4, have not previously been identified to The molecular detailing that has become possible have functional roles in breast cancer. However, given that Endocrine-Related Cancer through the Project and new high- , kinases in particular, have proven to be clinically throughput/high-content technologies in the last decade efficacious therapeutic targets, a wealth of data has now initially established five intrinsic molecular subgroups been generated not only to understand the complex (Sorlie et al. 2003) of which three were significantly biology of the disease but also to identify new treatments populated with ERC tumors. Most recently, at least ten for specific cohorts. molecular subgroups of human breast cancer have been The recently published METABRIC cohort of breast described (Curtis et al.2012) and eight of these appeared to cancers, in which 2000 individual breast cancers were be significantly populated with ERC tumors (Curtis et al. interrogated, identified ten integrative clusters, each with 2012). Associated with these studies has been the frequent distinct molecular characteristics associated with clinical identification of kinases, either mutated and/or structurally outcome (Curtis et al.2012). Often, altered genes encoding altered in large cohorts of breast tumors (Banerji et al.2012, kinases and phosphatases dominate individual clusters Curtis et al.2012, Shah et al.2012, Stephens et al.2012). (Table 1). Furthermore, the original intrinsic subtypes, in Some of these have also been found frequently altered and particular the ERC luminal A and luminal B (Perou et al. associated with AI sensitivity (Ellis et al.2012). 2000, Ignatiadis & Sotiriou 2013) subtypes, have been ER and its many coactivators are regulated by further subdivided due to the METABRIC study (Curtis et al. phosphorylation as well as other post-translational 2012, Dawson et al.2013). Therefore, a brief description of modifications (PTMs; Rowan et al. 2000, York et al. 2010, this follows as it relates to clusters that have significant Le Romancer et al. 2011, Zhang et al. 2013). The discovery ERC components. that the ER and its coactivators are substrates of several METABRIC integrative cluster 1 (IntClust1), represent- kinases (enzymes causing phosphorylation of specific ing 7% of breast cancer (Curtis et al.2012, Dawson et al. substrates), which are regulated by signaling pathways 2013), contains predominantly ERC tumors with luminal B frequently mutated or structurally altered in breast cancer features and is characterized by amplification of the

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R359 ER-positive status

Table 1 Kinases and phosphatases altered in ERC-dominated its amplification in this region. A high frequency (w50%) breast cancer clusters from METABRIC (Curtis et al.2012, of PI3K catalytic subunit p110a (PIK3CA) mutations is also Dawson et al. 2013) observed in this group. ERC tumors with luminal A characteristics predo- METABRIC Kinases or phosphatases minate integrative cluster 3 (IntClust3), which represents integrative clusters identified Alteration 15% of all breast tumors (Dawson et al. 2013). Tumors in IntClust1 Ribosomal S6 Amplification/ this cluster tend to have few structural alterations, low kinase 1 (RPS6KB1) overexpression Protein phosphatase Amplification/ genomic instability, and usually an excellent prognosis. 1D (PPM1D) overexpression Interestingly, they also have the highest frequency of IntClust2 PAK1 Amplification/ PIK3CA mutations (w58%). As well, similar to other overexpression C PIK3CA Mutation clusters dominated by ER tumors, a high level of IntClust3 PIK3CA Mutation MAP3K1 mutations (w15%) is observed in IntClust3. MAP3K1 Mutation IntClust5 represents 10% of all breast tumors and is IntClust5 HER2 Amplification/ w overexpression characterized by HER2 amplification with 42% of them PIK3CA Mutation also expressing ERa (Dawson et al. 2013). Approximately IntClust6 FGFR1 Amplification 30% of tumors in this cohort also have PIK3CA mutations. IntClust7 PI3K Mutation C MAP3K1 Mutation ER tumors with overexpressed HER2 are more likely to IntClust8 MAP2K4 Mutation be resistant to endocrine therapies. One reason for this is PI3K Mutation thought to be the constitutive activation of several kinases IntClust9 Regulatory subunit B Deletion of protein downstream of HER2, such as ERK1/2, PI3K, and AKT, all phosphatase 2 of which can phosphorylate ERa and several ER coacti- alpha (PPP2R2A) vators to enhance the ligand-independent activity of ERa as well as to enhance the agonist activity of tamoxifen (Wu et al. 2005, Le Romancer et al. 2011). 17q23 region. Two genes in this amplicon associated with IntClust6, representing 4% of breast tumors, is another increased expression are ribosomal protein S6 kinase beta 1 predominantly ERC group with both luminal A and (RPS6KB1 and p70S6K1) and protein phosphatase 1D luminal B characteristics (Dawson et al. 2013). Tumors of this PPM1D ( ). Interestingly, expression of both of these genes group show amplification of the 8p12 locus and Endocrine-Related Cancer C has been shown to be regulated by estrogen in ER breast are genomically unstable with an intermediate prognosis. cancer cells at the transcriptional level (Han et al.2009, Less than 20% of tumors in this group have PIK3CA Yamnik et al.2009, Yamnik & Holz 2010). In addition, mutations, which is low compared to other ERC groups. the protein product of RPS6KB1 (p70S6K1) has The for FGFR1 is located in the chromosome 8p12 been shown to phosphorylate ERa (Yamnik & Holz 2010) region. FGFR1 amplification and overexpression have been and under IGF1 stimulation p70S6K1 can be directly linked to endocrine therapy resistance (Turner et al. C co-immunoprecipitated with ERa in ER breast cancer 2010, Balko et al. 2012). FGFR1 also regulates several kinases cells (Becker et al.2011). PPM1D/Wip1 is known for its p53 such as PI3K and AKT, which are known to phosphorylate C inhibitory effects and, in ER MCF7 cells, has been shown ERa and its coactivators (Le Romancer et al. 2011). to be regulated by estrogen and to increase the transcrip- IntClust7 (10% of breast tumors) and IntClust8 (15% tional activity of ERa (ESR1) as well as other steroid hormone of breast tumors) are mainly ERC groups and while similar receptors (Proia et al. 2006), possibly by modulating in many ways tumors in Intclust8 display higher genomic phosphorylation of coactivators such as SRC1 and/or instability, with an associated inferior prognosis than p300/CBP (Proia et al.2006). tumors in IntClust7 (Dawson et al. 2013). Interestingly, Integrative cluster 2 (IntClust2), representing 4% of both again have a high frequency of PI3K (PIK3CA) breast tumors, is also dominated by ERC tumors with mutations. IntClust7 has a high frequency of MAP3K1 characteristics of both luminal A and luminal B subtypes mutations and IntClust8 has the highest frequency of (Dawson et al. 2013). Amplification of chromosome region MAP2K4 mutations, a downstream target of MAP3K1. 11q13/14 characterizes this cluster and high expression of The structural alterations usually found in MAP3K1 another kinase, PAK1, known to affect ERa phosphory- and MAP2K4 are deletions and mutations associated lation and activity (Wang et al. 2002, Mazumdar & Kumar with loss of function (Teng et al. 1997, Pham et al. 2013), 2003, Holm et al. 2009, Kok et al. 2010) is associated with suggesting that the pathways they regulate have

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R360 ER-positive status

tumor-suppressor function. Steroid hormone receptor also have a high frequency of PIK3CA mutations while by signaling, including estrogen and androgen receptors, contrast IntClust6 has less than a 20% frequency of PI3K shows crosstalk with MAP3K1 in some systems, and it is pathway mutations (Dawson et al.2013). the ERC tumors, in particular luminal A type, that have Experimental and clinical data suggest that hyper- the highest level of mutation and/or deletion of the activation of the PI3K/AKT/mTOR pathway is associated MAP3K1/MAP2K4 pathway (Pham et al. 2013). This with endocrine resistance. However, gain-of-function pathway has been implicated in cell death pathways, PIK3CA mutations in ERC tumors are often associated which under normal conditions is important for mam- with good prognosis (Miller et al. 2011a). Such apparently mary gland involution, hence its loss of function in many contradictory results may be due to complexities associ- ERC breast cancers may in part be responsible for the ated with feedforward and feedback mechanisms involved dissociation of estrogen-induced proliferative and survival in the PI3K/AKT/mTOR pathway and other alterations pathways from growth-inhibiting differentiation co-occurring in regulators of the pathway. This latter idea pathways (Pham et al. 2013). This pathway has been is supported by the finding that most often the associ- described as having a molecular switch type function ations of PIK3CA mutations with good outcome in ERC between survival and cell death, possibly dependent on tumors are restricted to those tumors that are also HER2 cell-type background, subcellular localization, and caspase negative (Fu et al. 2013). 3 activity (Pham et al. 2013). Increased ERa expression As discussed above, the PIK3CA gene, encoding the combined with a decreased activity of the MAP3K1/ PI3K catalytic subunit p110a, is one of the most frequently MAP2K4 pathway may, in part, underlie the change in mutated genes in ERC breast cancers (Dawson et al. 2013). the balance of estrogen signaling regulation of survival/ Many of the mutations occur in ‘hot spots’ with the most proliferation vs cell cycle inhibition that is often frequent mutations being E542K, E545K (both in the associated with differentiation. helical domain), and H1047R (in the kinase domain) IntClust9 (7% of breast tumors) is characterized by (Barbareschi et al. 2007). Generally, the mutations result in a high frequency of PPP2R2A deletions (Dawson et al. constitutive activation of the kinase (Di Cosimo & Baselga 2013). PPP2R2A is the regulatory subunit B of protein 2009). As mentioned above, the published data largely phosphatase 2 alpha (PP2A). While IntClust9 contains a suggest that mutations in PIK3CA occur more frequently mixture of intrinsic subtypes, its ERC members are mainly in ERC breast cancer with good prognosis and may also be of the luminal B type, and loss of PPP2R2A expression is associated with better clinical outcome in patients treated Endocrine-Related Cancer associated with a high mitotic index. Interestingly, PP2A with endocrine therapy (Di Cosimo & Baselga 2009, Miller also has been implicated in the regulation of ERa et al. 2011a). However, this is not a universal finding phosphorylation and activity (Lu et al. 2003a). (Cuorvo et al. 2014). Recent reviews have discussed this The above data underscore that distinctly altered aspect in detail (Fu et al. 2013). kinase and phosphatase mutation patterns occur frequently AKT1 mutations, often associated with PI3K-inde- in ERC breast cancer subgroups with altered clinical pendent constitutive activity, occur in w4% of breast outcome. This would support the idea that combining cancers and are often associated with ERC tumors. endocrine therapies with other targeted therapies informed However, the relationship of AKT expression and/or by each subgroup’s distinct kinase pattern could provide activation to outcome in breast cancer is inconsistent better clinical outcome for breast cancer patients. (Badve et al. 2010, Aleskandarany et al. 2011). Consi- dering that the different AKT isoforms have distinct Kinases associated with the PI3K/AKT/mTOR functions (Dillon & Muller 2010) and that a recent study and ER-positive breast cancer using proximity ligation assay (PLA) to distinguish between pAKT1 and pAKT2 (Spears et al. 2012) METABRIC and similar studies (Curtis et al.2012, Ellis et al. determined the former to be associated with poor 2012, Stephens et al.2012) as well as previous smaller scale prognosis and the latter with better prognosis, it seems studies (Miller et al.2010, 2011a,b) have identified the possible that the relative levels of each isoform may frequent structural and mutational alterations in the determine the final read out. Furthermore, the inability PI3K/AKT/mTOR pathway that occur in ERC breast cancer. to distinguish the different pAKT isoforms may, at least in IntClust1, IntClust2, IntClust3, and IntClust5 (Dawson part, underlie the previous lack of consistency (Badve et al.2013)havew25, 50, 58, and 30% frequencies of et al. 2010, Aleskandarany et al. 2011)aroundtheir PIK3CA mutations respectively. IntClust7 and IntClust8 association with breast cancer outcome.

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R361 ER-positive status

Owing to the frequency with which components of the SRPK1, TTK, and VRK1. Many of these kinases have been PI3K/AKT/mTOR pathway are altered in ERC tumors, and previously reported to be involved in the G2 and M phases the fact that the pathway is a central hub receiving growth of the cell cycle (Malumbres & Barbacid 2009, Knight et al. and survival signals from many factors, the detailed 2010). Notably, these kinases include the 12 kinases molecular nature of the pathway in different ERC tumors making up the mitosis metagene cluster identified by may be important to guide appropriate therapy options. Bianchini et al. (2010). Similarly, Finetti et al. (2008) found that overexpression of kinases from this 16-kinase signature in luminal A (ERC) tumors was associated with Kinases associated with ER status by a poor clinical outcome. The association of mitosis and expression analyses proliferation signatures with poorer prognosis in the Several studies have now been published in which different luminal breast cancer subgroups is a consistent theme kinase expression patterns were found in ERC vs other (Ribelles et al. 2013). clinical subtypes of breast cancer. Using publically available Unique kinases associated with ER status in breast RNA expression databases, Bianchini et al.(2010)identified cancer cell lines have also been reported (Michalides et al. 16 kinases that were overexpressed in ERC/HER2K breast 2002, Finetti et al. 2008, Midland et al. 2012), some of cancer biopsy samples. These were IGF1R, STK32B, ERBB4, which show overlap with studies in tumors (Finetti et al. FGFR3, BMPR1B, MAST4, HSPB8, IKBKB, DCLK1, ERBB3, 2008, Midland et al. 2012), therefore potentially providing STK39, MAP3K1, PTK6, PLK2, TEX14,andMST1. Kinases models of ERC breast cancer kinomes in vitro. These data uniquely overexpressed in ERK and HER2C breast cancer support the existence of distinct kinomes, not only subtypes were also identified. Two robust kinase clusters associated with ER status per se, but also further define were recognized: a mitosis metagene cluster (12 distinct heterogeneity within ERC breast tumors. kinases) and an immune kinase cluster (15 distinct kinases), which were present in all clinical subgroups. Overexpression Kinase overexpression associated with of kinases composing the mitosis metagene cluster was sensitivity to endocrine therapies mostly found in ERK tumors and was not prognostic in this subgroup. However, ERC/HER2K tumors that presented a There are many different kinases that when either over- high mitosis kinase score were associated with a worse expressed or underexpressed in ERC breast cancer cells prognosis but showed a higher frequency of pathological lead to altered cell growth and survival responses to both Endocrine-Related Cancer complete response (pCR) to neoadjuvant chemotherapy. On SERMs such as estrogen and tamoxifen, and AIs. Many of the other hand, overexpression of kinases from the immune these are listed in Table 2. kinase cluster was associated with better survival in Such data underscore the importance of kinase net- ERC/HER2K and HER2C subgroups. Interestingly, the works, as many of the kinases listed in Table 2 converge on authors also observed that in ERC tumors, many of the common hubs associated with cell growth, survival and overexpressed kinases were transmembrane growth factor cell death. It is not surprising that the interaction of receptors, while ERK tumors mostly overexpressed intra- multiple kinases with ER signaling is observed in breast cellular kinases associated with cell proliferation (Bianchini cancer, as many of these kinases are key components of et al.2010). proliferation, survival and cell death pathways and would Finetti et al. (2008) undertook a genome-wide be expected to be regulated by estrogen, through the ER, expression analysis of a cohort of primary human breast which is the primary mitogenic/survival signal in the tumors focusing mainly on the basal and luminal A majority of breast cancers, at least initially (Musgrove & intrinsic breast cancer subtypes. From this analysis, they Sutherland 2009, Osborne & Schiff 2011). extracted kinase genes whose differential expression was In ERC breast cancer, gene signatures that are based on associated with a clinical outcome. Not unexpectedly, proliferation often strongly correlate with Ki67 expression there were substantial differences in the pattern of kinase (Gao et al.2014), a well-known marker of proliferation. gene expression between the basal and the luminal Furthermore, in ERC breast tumors in particular, it is A intrinsic subtypes. Of more interest, however, was the Ki67 and not pCR that is the early correlative endpoint discovery that 16 kinases were overexpressed in most of for predicting efficacy of both hormonal therapy or the basal group and in a few luminal A tumors. These chemotherapy in ERC breast cancer (Yerushalmi et al. kinases were AURKA, AURKB, BUB1, BUB1B, CDC2 (CDK1), 2010, Dowsett et al. 2011, Ellis et al.2011, CDC7, CHEK1, MASTL, MELK, NEK2, PBK, , PLK4, von Minckwitz et al. 2012, Gao et al. 2014). Recently, using

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Endocrine-Related Cancer O:1.50EC1-22Pitdi ra Britain Great in Printed 10.1530/ERC-14-0232 DOI: http://erc.endocrinology-journals.org

Table 2 Kinases experimentally manipulated and shown to influence tamoxifen (Tam) or aromatase inhibitor (AI) sensitivity Review

Kinases Gain of function Loss of function References Effects Clinical correlation

EGFR Ectopic overexpression Agthoven et al. (1992) Tam-resistant growth Yes (Giltnane et al. 2007 HER2 Ectopic overexpression Benz et al. (1992) Tam-resistant growth Yes (Osborne et al. 2003) AKT Ectopic overexpression Campbell et al. (2001), Tam-resistant growth Yes (Perez-Tenorio et al. 2002) deGraffenried et al. (2004) and Silva et al. (2007) Akt3 Ectopic overexpression Faridi et al. (2003) E2-independent, Yes (Nakatani et al. 1999) Tam-stimulated growth as xenograft ERK1/2 MAPK Yes (Gee et al. 2001) PKCa Ectopic overexpression Chisamore et al. (2001) and Tam-resistant growth Yes (Kalstad et al. 2010)

Frankel et al. (2007) with associated Kinome al. et Bruce C M q PKCd Ectopic overexpression Nabha et al. (2005) Tam-resistant growth 04SceyfrEndocrinology for Society 2014 PKA Overexpression by reducing Michalides et al. (2004) Tam-resistant growth Yes (Michalides et al. 2004) expression of negative regulatory subunit PKA-RIalpha MKP3 Ectopic overexpression Cui et al. (2006) Tam-resistant growth Yes (Cui et al. 2006) CDK10 siRNA Iorns et al. (2008) Tam-resistant growth Yes (Iorns et al. 2008) inhibition CRK7 siRNA Iorns et al. (2009) Tam-resistant growth No inhibition IKK3 Ectopic overexpression Guo et al. (2010) Protection against No

Tam-induced cell status ER-positive ulse yBocetfiaLtd. Bioscientifica by Published death SphK1 Ectopic overexpression Sukocheva et al. (2009) Tam-resistant growth Yes (Ruckhaberle et al. 2008) inhibition c-ABL siRNA Zhao et al. (2010) Sensitizes cells to Yes (Zhao et al. 2010) Tam inhibition Ron receptor tyrosine Ectopic overexpression McClaine et al. (2010) Tamoxifen resistance No kinase (MST1R) EphA2 receptor tyrosine Ectopic overexpression Lu et al. (2003b) Tamoxifen resistance Yes (Brantley-Sieders et al. 2011) kinase (EPHA2) cRET siRNA Plaza-Menacho et al. (2010) Increased sensitivity Yes (Morandi et al. 2013) Downloaded fromBioscientifica.com at09/26/202101:37:02PM to Tam HSPB8 Ectopic overexpression Gonzalez-Malerva et al. (2011) Tam resistance Yes (Gonzalez-Malerva et al. 2011) LMTK3 siRNA Giamas et al. (2011) Increased sensitivity Yes (Giamas et al. 2011) to Tam IGF-R1 Ectopic overexpression Zhang et al. (2011) Tam and fulvestrant Yes (Fox et al. 2011, Winder resistance et al. 2014) 21 FGFR3 Ectopic overexpression Tomlinson et al. (2012) Tam and fulvestrant Yes (Tomlinson et al. 2012) :5 resistance TBK1 Ectopic overexpression Wei et al. (2014) Tam resistance Yes (Wei et al. 2014) R362 via freeaccess Review M C Bruce et al. Kinome associated with 21:5 R363 ER-positive status

neoadjuvant antiestrogen therapy (particularly AIs), it kinases with known functions in breast cancer develop- was observed that decreased detection of survival pathway ment and progression. Dysregulation including mutation activation markers (e.g. phosphorylated AKT and phos- and structural alterations of kinases themselves or the phorylated mTOR) in tumors after treatment, compared signaling pathways in which they function is thought, in with pretreatment levels, was predictive of a better clinical part, to contribute to the dysregulation of ER signaling outcome (Generali et al. 2008, 2011), suggesting that that is associated with the development of breast cancer as estrogen via the ER was regulating these pathways, directly well as its progression to endocrine resistance in vivo. or indirectly in vivo. Many preclinical models have shown Multiple aspects of ER activity can be affected by that estrogen, via ER signaling, at least in part, regulates the phosphorylation; however, the focus of the majority of activities of PI3K/AKT/mTOR, p38MAPK, and MAPK/ studies addressing this issue has been on the transcrip- ERK1/2 (Zhang et al. 2002, Lee et al. 2005, Yu & Henske tional activity (Le Romancer et al.2011)and,not 2006, Kazi et al.2009, Maruani et al.2012). This suggests that surprisingly, it has been observed that differential phos- the use of hormonal therapies in combination with the phorylation of ERa (e.g. PKA activation) can alter the most appropriately targeted kinase inhibitors may be more chromatin-binding pattern (cistrome) of the receptor, in beneficial at the onset of therapy than application in a turn altering the ERa transcriptome (de Leeuw et al. 2013). sequential fashion. Indeed, there are ongoing clinical trials The function of phosphorylation on ER activity and that evaluating the use of combination therapies with endo- of other steroid hormone receptors have been recently crine therapies as first-line approaches (Ciruelos 2014). extensively reviewed (Le Romancer et al. 2011, Trevino & A major challenge of using kinase inhibitors is that Weigel 2013) and will not be reviewed herein. kinase networks especially those associated with growth and Multiple different kinases can phosphorylate the same survival are often interconnected and inhibition of one may ERa residue (e.g. serine amino acid residue 167 (S167) can have effects beyond the immediate targets. Adaptive be phosphorylated by AKT, p70S6K, Aurora A, and reprogramming of cancer kinomes due to single kinase p90RSK) (Le Romancer et al. 2011). Similarly, one kinase inhibitors has been demonstrated (Duncan et al.2012)and can phosphorylate several different residues on ERa an argument for targeted combination therapy approaches (e.g. MAPK can phosphorylate S104/106, S118, and S167) initially has been put forward (Stuhlmiller et al.2014). The (Le Romancer et al.2011). Many kinases known to development of resistance to hormonal therapies most phosphorylate ERa can also phosphorylate various often occurs despite the continued expression of ERa Endocrine-Related Cancer (Encarnacion et al. 1993, Bachleitner-Hofmann et al. S554 2002). In addition, many laboratory studies suggest that S106 Y219 S294 S104 S154 S212 S282 S305 adaptive responses of breast cancer cells, rather than S102 S118 S167 S236 T311 Y537 S559 Y52 selection of existing ER-negative cells in the original S46/47 heterogeneous population, frequently occur (Santen et al. 1 180 263 302 595 2003, Browne et al.2013). In such cases, increased expression/activity of kinases also frequently occurs (Coutts A/B CDE F &Murphy1998, Santen et al.2003, 2004), supportive of the concept that adaptive reprogramming of the breast cancer kinome, in part, underlies some mechanisms of estrogen Figure 1 independence and anti-estrogen resistance (Stuhlmiller Known phosphorylation sites on estrogen receptor a. Sites identified by stars are those that have been determined in ERaC breast cancer cases in et al.2014). The challenge will be to understand this and the Manitoba Breast Tumor Bank (MBTB) and are the basis of the P7-ERa its potential plasticity within the underlying heterogeneity score. The black stars are those residues which when phosphorylated are of ERC breast cancers and at the individual patient level. associated with a good clinical outcome in patients treated with tamoxifen. The open/white stars are those residues which when phosphorylated are associated with a poor clinical outcome in patients treated with tamoxifen (Skliris et al. 2010a). It should be noted that others have determined that Kinases known to phosphorylate ERa S305 when phosphorylated is associated with a poor clinical outcome in tamoxifen-treated patients (Le Romancer et al. 2011) and we have found In addition to ER signaling regulating kinase pathway that tyrosine (Y) 537 when phosphorylated is also associated with a poor expression and/or activity, ER activity can also be clinical outcome in tamoxifen-treated patients (Skliris et al. 2010b). These regulated by kinases. latter two results (textured stars) are consistent with our observation that phosphorylation in N-terminal residues is associated with a good clinical ERa can be phosphorylated on many residues (Fig. 1) outcome, but phosphorylation in C-terminal residues is associated with a and ERa has been identified as a substrate for several poor clinical outcome.

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R364 ER-positive status

coactivators and coregulators (Wu et al. 2005), impacting and fluorescence resonance energy transfer (FRET) (Zwart ER (ESR1) transcriptional activity. This provides a et al.2007a,b). Direct interaction of ERa with kinases can potential mechanism allowing the integration of multiple occur in the cytoplasm (Poulard et al.2012), at the plasma pathways to regulate the biology and/or patho-biology of membrane (Poulard et al.2012), as well as in the nucleus on ERa in the mammary gland. Support for this idea, at least chromatin (Madak-Erdogan et al.2011, 2014, Wierer et al. in breast cancer, is provided by the existence of a 2013), with some methodologies, such as PLA, allowing the phosphorylation code for ERa (Skliris et al. 2010a) and at determination of the subcellular localization of any least one of its coactivators, SRC3/AIB1 (Wu et al. 2004, interactions that occur (Poulard et al.2012). York et al. 2010). As ERa can undergo several other types of There is no doubt that phosphorylation and other PTMs, e.g. acetylation, methylation, ubiquitylation, etc. PTMs (Wu et al. 2007, Le Romancer et al. 2011) are (Le Romancer et al. 2011), one can envisage a broader ERa important for the regulation of ER highlighted by the PTM or epigenetic-like code. existence of a phosphorylation code for ERa in human The identification of kinases phosphorylating ERa has breast tumors (Skliris et al. 2010a). However, it is still been achieved using multiple methods, alone or in unclear as to which kinases and phosphatases are involved combination: in vitro studies using recombinant , in regulating ERa in vivo. Correlations of expression and/or mass spectroscopy, the use of selective small-molecular- activity of some kinases with ER or its phosphorylated weight kinase activators or inhibitors, and overexpression forms in human breast tumors have been reported, or knockdown of expression using cells in culture or as supporting a possible role in vivo (Murphy et al. 2004, xenografts. Furthermore, it is reasonable to assume, while Gee et al. 2005, Jiang et al. 2007, Shrivastav et al. 2014). not proving, that if ER can be directly phosphorylated by a Many of the kinases that can directly phosphorylate kinase, the two proteins would directly bind under ERa and its coregulators can also be regulated by estrogen appropriate circumstances. Table 3 lists a number of kinases signaling under some circumstances. Many such kinases are that have been found to undergo protein:protein components of key signaling pathways involved in interactions with ERa using different methodologies, development/differentiation, proliferation, cell cycle, e.g. co-immunoprecipitation (Wierer et al. 2013) and motility/invasion, and cell death. Whether or not they are other pull-down-type assays (Kanaujiya et al.2013), PLA regulated by estrogen or in turn regulate ERa may depend on (Poulard et al.2012), yeast-2-hybrid assay (Paul et al.2014), levels of expression of both substrate and kinase, and on Endocrine-Related Cancer Table 3 Kinases shown to interact directly with estrogen receptor a

Kinases Assays References

ERK1/2 CoIP; ChIP/reChIP Madak-Erdogan et al. (2011) p70S6K1 Co-IP Becker et al. (2011) GSK3 Co-IP Medunjanin et al. (2005) CDK2 GST pull-down, in vitro kinase assay Rogatsky et al. (1999) IKKa Co-IP ChIP-reChIP Park et al. (2005) CDK7 In vitro Co-IP Chen et al. (2000) AKT In vitro kinase assay Campbell et al. (2001) and Sun et al. (2001) PI3K Proximity ligation assay Co-IP Sun et al. (2001) and Poulard et al. (2012) pp90RSK1 In vitro kinase assays Co-IP Joel et al. (1998) PKA FRET in vitro kinase assay Chen et al. (1999), Zwart et al. (2007a,b) ILK GST-down experiments Co-IP Acconcia et al. (2006) EGFR Co-IP in vitro kinase assay Marquez et al. (2001) IGFR Co-IP Mendez et al. (2003) DNA-PK Co-IP Medunjanin et al. (2010) cAbl Co-IP in vitro kinase assay He et al. (2010) CK2 In vitro kinase assay Williams et al. (2009) IKK3 Co-IP in vitro kinase assay Guo et al. (2010) PAK1 In vitro kinase assay GST-pull-down Wang et al. (2002) experiments Co-IP c-SRC Proximity ligation assay Co-IP in vitro Arnold et al. (1995b, Poulard et al. (2012) and kinase assay Sun et al. (2012)) p38 MAPK Immunocomplex kinase assay in vitro Lee & Bai (2002) kinase assay shows no phosphorylation PLK1 Co-IP ChIP/reChIP Wierer et al. (2013)

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R365 ER-positive status

mutations or structural alterations that may eliminate or the earliest alterations occurring in ‘precursors’ of breast introduce new regulatory factors. The ability of the cancer (Shoker et al.1999, Lee et al.2006). ERa is highly estrogen/ERa complex to activate kinases that in turn expressed in most atypical ductal hyperplasia (ADH), ductal can phosphorylate ER and modify function provides a carcinoma in situ (DCIS), and invasive breast cancers powerful feed-forward amplification system that may (Shoker et al.1999, Allred et al.2001). However, the drive many human breast cancers. Understanding how mechanisms leading to increased ERa expression during this occurs and how it can be perturbed to drive tumorigenesis are poorly understood. Interestingly, beside progression to therapy resistance would provide funda- alterations in ER gene transcription (Muscat et al.2013), mental information to inform early combination altered kinase expression affecting ERa protein stability has approaches, for preventing and/or treating resistant tumors. been suggested (Henrich et al.2003, Antoon et al.2013) Interestingly, although in primary breast tumors, few and, therefore, highlights the potential link between ERa mutations have been found, one known mutation phosphorylation and estrogen signaling. K303R changes the amino acid lysine that can be The challenge now is to identify those kinases and acetylated (another type of PTM) to an arginine residue their associated networks that phosphorylate and regulate that cannot be acetylated. This in turn alters regulation of ERa function in ERC tumors and potentially normal S305 phosphorylation on ERa, a site of ERa phosphory- breast in vivo, as such information will identify the most lation with established relevance in vivo and in AI efficacious targeted treatment approaches upfront and resistance (Herynk & Fuqua 2004, Barone et al. 2010). may inform better prevention approaches. There are also other examples of the co-regulation of phosphorylation with other PTMs to regulate ERa Conclusions expression and/or activity. For example, c-Src can phos- ERa regulates and is regulated by kinases involved in several a phorylate ER at Y537 that in turn facilitates its functions associated with the hallmarks of cancer (Hanahan interaction with E3 ubiquitin such as E6AP, & Weinberg 2011).Thepresenceofaphosphorylationcode a resulting in ubiquitin-dependent degradation of ER for ERa in breast cancers associated with clinical outcome (Zhou & Slingerland 2014). Most recently, however, (Skliris et al.2010a) and the knowledge that there are also a frequent somatic mutations in ER have been identified other PTMs associated with ERa (Le Romancer et al.2011) C in metastases from ER breast cancer. This is more in vivo suggest that there is an epigenetic-like code for ERa, frequently associated with metastases that occur during Endocrine-Related Cancer which can regulate and is in part regulated by distinct or following endocrine therapy (Zhang et al. 1997, kinomes. The literature reviewed above strongly suggests that Robinson et al. 2013, Jeselsohn et al. 2014, Segal & Dowsett distinct kinomes exist for ER-positive and -negative breast 2014). The most frequent mutation identified was at Y537 cancers. Even within ERC cancers, different subgroups exist, or the residue beside it, D538; therefore, affecting directly defined by different kinome signatures, which can be a or indirectly a well-known phosphorylation site on ER correlated with clinical outcome. Strong evidence supports (Arnold et al. 1995a, Nettles et al. 2008, Skliris et al. 2010b) the interplay of kinase networks, suggesting that targeting a with known clinical relevance (Skliris et al. 2010b). Such single node may not be sufficient to inhibit the network. a data underscore the importance of ER phosphorylation Therefore, identifying the important hubs/nodes associated and other PTMs to breast cancer progression. with each clinically relevant kinome in ERC tumors could a Our understanding of estrogen-regulated ER signaling offer the ability to implement the best therapy options at in human breast cells comes from model systems that are all diagnosis, either endocrine therapy alone or together with C K cancer cells. Moreover, all ER and ER cell lines were other targeted therapies for an improved overall outcome. originally obtained from breast cancer metastases usually pleural and ascitic effusions (Soule et al.1973, Lippman et al a .1976). We have little if any understanding of ER Declaration of interest signaling in normal human breast epithelial cells, mainly The authors declare that there is no conflict of interest that could be due to the lack of appropriate ERC models. This remains a perceived as prejudicing the impartiality of the review. large gap in our understanding, potentially limiting the development of more efficacious prevention strategies. This is especially important as increased ERa expression in Funding This work was made possible through grant support to L C Murphy from the normal human breast is associated with an increased cancer Canadian Institutes of Health Research (CIHR), the Canadian Breast Cancer risk (Khan et al.2005) and elevated ERa expression is one of Foundation (CBCF), and the Canadian Cancer Society Research Institute

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R366 ER-positive status

(CCSRI). M C Bruce is funded by a CBCF-Prairies/NWT region Postdoctoral Benz CC, Scott GK, Sarup JC, Johnson RM, Tripathy D, Coronado E, Fellowship. Morerover, some of published works referred to in this review Shepard HM & Osborne CK 1992 Estrogen-dependent, tamoxifen- used breast tumor samples from the Manitoba Breast Tumor Bank, a member resistant tumorigenic growth of MCF-7 cells transfected with HER2/neu. of the Canadian Tumor Repository Network, which are funded in part by the Breast Cancer Research and Treatment 24 85–95. (doi:10.1007/BF01961241) CancerCare Manitoba Foundation (CCMF) and CIHR, respectively. Bianchini G, Iwamoto T, Qi Y, Coutant C, Shiang CY, Wang B, Santarpia L, Valero V, Hortobagyi GN, Symmans WF et al. 2010 Prognostic and therapeutic implications of distinct kinase expression patterns in different subtypes of breast cancer. Cancer Research 70 8852–8862. References (doi:10.1158/0008-5472.CAN-10-1039) Brantley-Sieders DM, Jiang A, Sarma K, Badu-Nkansah A, Walter DL, Shyr Y Acconcia F, Manavathi B, Mascarenhas J, Talukder AH, Mills G & Kumar R & Chen J 2011 Eph/ephrin profiling in human breast cancer reveals 2006 An inherent role of integrin-linked kinase-estrogen receptor a significant associations between expression level and clinical outcome. interaction in cell migration. Cancer Research 66 11030–11038. PLoS ONE 6 e24426. (doi:10.1371/journal.pone.0024426) (doi:10.1158/0008-5472.CAN-06-2676) Browne BC, Hochgrafe F, Wu J, Millar EK, Barraclough J, Stone A, McCloy RA, van Agthoven T, van Agthoven TL, Portengen H, Foekens JA & Dorssers LC Lee CS, Roberts C, Ali NA et al. 2013 Global characterization of 1992 Ectopic expression of epidermal growth factor receptors induces signalling networks associated with tamoxifen resistance in breast hormone independence in ZR-75-1 human breast cancer cells. Cancer cancer. FEBS Journal 280 5237–5257. (doi:10.1111/febs.12441) Research 52 5082–5088. Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S & Aleskandarany MA, Rakha EA, Ahmed MA, Powe DG, Ellis IO & Green AR Nakshatri H 2001 Phosphatidylinositol 3-kinase/AKT-mediated activation 2011 Clinicopathologic and molecular significance of phospho-Akt of estrogen receptor a: a new model for anti-estrogen resistance. Journal of expression in early invasive breast cancer. Breast Cancer Research and Biological Chemistry 276 9817–9824. (doi:10.1074/jbc.M010840200) Treatment 127 407–416. (doi:10.1007/s10549-010-1012-y) Chen D, Pace P, Coombes R & Ali S 1999 Phosphorylation of human Allred D, Mohsin S & Fuqua S 2001 Histological and biological evolution of estrogen receptor a by regulates dimerization. human premalignant breast disease. Endocrine-Related Cancer 8 47–61. Molecular and Cellular Biology 19 1002–1015. (doi:10.1677/erc.0.0080047) Chen D, Washbrook E, Sarwar N, Pace P, Coombes R, Egly J & Ali S 2000 Antoon JW, Martin EC, Lai R, Salvo VA,TangY, NitzchkeAM, Elliott S,Nam SY, Activation of estrogen receptor a by S118 phosphorylation involves a Xiong W, Rhodes LV et al. 2013 MEK5/ERK5 signaling suppresses estrogen ligand-dependent interaction with TFIIH and participation of CDK7. receptor expression and promotes hormone-independent tumorigenesis. Molecular and Cellular Biology 6 127–137. PLoS ONE 8 e69291. (doi:10.1371/journal.pone.0069291) Chisamore MJ, Ahmed Y, Bentrem DJ, Jordan VC & Tonetti DA 2001 Novel Arnold S, Vorojeikina D & Notides A 1995a Phosphorylation of tyrosine antitumor effect of estradiol in athymic mice injected with a T47D 537 on the human estrogen receptor is required for binding to an breast cancer cell line overexpressing protein kinase Ca. Clinical Cancer estrogen response element. Journal of Biological Chemistry 270 Research 7 3156–3165. 30205–30212. (doi:10.1074/jbc.270.4.1850) Ciruelos G 2014 Targeting the PI3K/AKT/mTOR pathway in estrogen Arnold S, Obourn J, Jaffe H & Notides A 1995b Phosphorylation of the receptor-positive breast cancer. Cancer Treatment Reviews 40 862–871. human estrogen receptor on tyrosine 537 in vivo and by Src family (doi:10.1016/j.ctrv.2014.03.004) tyrosines in vitro. Molecular Endocrinology 9 24–33. Coutts A & Murphy L 1998 Elevated mitogen activated protein kinase Bachleitner-Hofmann T, Pichler-Gebhard B, Rudas M, Gnant M, Taucher S, activity in estrogen non-responsive human breast cancer cells. Cancer Endocrine-Related Cancer KandiolerD,JanschekE,DubskyP,RokaS,SpornEet al. 2002 Pattern of Research 58 4071–4074. hormone receptor status of secondary contralateral breast cancers in patients Cui Y, Parra I, Zhang M, Hilsenbeck SG, Tsimelzon A, Furukawa T, Horii A, receiving adjuvant tamoxifen. Clinical Cancer Research 8 3427–3432. Zhang ZY, Nicholson RI & Fuqua SA 2006 Elevated expression of Badve S, Collins NR, Bhat-Nakshatri P, Turbin D, Leung S, Thorat M, Dunn SE, mitogen-activated protein kinase phosphatase 3 in breast tumors: a Geistlinger TR, Carroll JS, Brown M et al. 2010 Subcellular localization of mechanism of tamoxifen resistance. Cancer Research 66 5950–5959. activated AKT in estrogen receptor- and progesterone receptor-expressing (doi:10.1158/0008-5472.CAN-05-3243) breast cancers: potential clinical implications. American Journal of Path- Cuorvo LV,VerderioP,Ciniselli CM, Girlando S, DecarliN,Leonardi E, FerroA, ology 176 2139–2149. (doi:10.2353/ajpath.2010.090477) Caldara A, Triolo R, Eccher C et al. 2014 PI3KCA mutation status is of Balko JM, Mayer IA, Sanders ME, Miller TW, Kuba MG, Meszoely IM, limited prognostic relevance in ER-positive breast cancer patients Wagle N, Garraway LA & Arteaga CL 2012 Discordant cellular response treated with hormone therapy. Virchows Archiv: An International to presurgical letrozole in bilateral synchronous ERC breast cancers Journal of Pathology 464 85–93. (doi:10.1007/s00428-013-1500-7) with a KRAS mutation or FGFR1 gene amplification. Molecular Cancer Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Therapeutics 11 2301–2305. (doi:10.1158/1535-7163.MCT-12-0511) Lynch AG, Samarajiwa S, Yuan Y et al. 2012 The genomic and Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, transcriptomic architecture of 2,000 breast tumours reveals novel Lawrence MS, Sivachenko AY, Sougnez C, Zou L et al. 2012 Sequence subgroups. Nature 486 346–352. (doi:10.1038/nature10983) analysis of mutations and translocations across breast cancer subtypes. Dawson SJ, Rueda OM, Aparicio S & Caldas C 2013 A new genome-driven Nature 486 405–409. (doi:10.1038/nature11154) integrated classification of breast cancer and its implications. Barbareschi M, Buttitta F, Felicioni L, Cotrupi S, Barassi F, Del Grammastro M, EMBO Journal 32 617–628. (doi:10.1038/emboj.2013.19) Ferro A, Dalla Palma P, Galligioni E & Marchetti A 2007 Different Di Cosimo S & Baselga J 2009 Phosphoinositide 3-kinase mutations in prognostic roles of mutations in the helical and kinase domains of the breast cancer: a “good” activating mutation? Clinical Cancer Research 15 PIK3CA gene in breast carcinomas. Clinical Cancer Research 13 5017–5019. (doi:10.1158/1078-0432.CCR-09-1173) 6064–6069. (doi:10.1158/1078-0432.CCR-07-0266) Dillon RL & Muller WJ 2010 Distinct biological roles for the akt family in Barone I, Iacopetta D, Covington KR, Cui Y, Tsimelzon A, Beyer A, Ando S & mammary tumor progression. Cancer Research 70 4260–4264. Fuqua SA 2010 Phosphorylation of the mutant K303R estrogen (doi:10.1158/0008-5472.CAN-10-0266) receptor a at serine 305 affects aromatase inhibitor sensitivity. Oncogene Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J, Ellis M, 29 2404–2414. (doi:10.1038/onc.2009.520) Henry NL, Hugh JC, Lively T et al. 2011 Assessment of Ki67 in breast Becker MA, Ibrahim YH, Cui X, Lee AV & Yee D 2011 The IGF pathway cancer: recommendations from the International Ki67 in Breast Cancer regulates ERa through a S6K1-dependent mechanism in breast cancer working group. Journal of the National Cancer Institute 103 1656–1664. cells. Molecular Endocrinology 25 516–528. (doi:10.1210/me.2010-0373) (doi:10.1093/jnci/djr393)

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R367 ER-positive status

Duncan JS, Whittle MC, Nakamura K, Abell AN, Midland AA, Zawistowski JS, GiamasG,Filipovic A, Jacob J, Messier W, ZhangH, Yang D, Zhang W, Shifa BA, Johnson NL, Granger DA, Jordan NV, Darr DB et al. 2012 Dynamic Photiou A, Tralau-Stewart C et al. 2011 Kinome screening for regulators of reprogramming of the kinome in response to targeted MEK inhibition in theestrogenreceptor identifies LMTK3 asa new therapeutic target in breast triple-negative breast cancer. Cell 149 307–321. (doi:10.1016/j.cell. cancer. Nature Medicine 17 715–719. (doi:10.1038/nm.2351) 2012.02.053) Giltnane JM, Ryden L, Cregger M, Bendahl PO, Jirstrom K & Rimm DL 2007 Ellis MJ, Suman VJ, Hoog J, Lin L, Snider J, Prat A, Parker JS, Luo J, Quantitative measurement of epidermal growth factor receptor is a DeSchryver K, Allred DC et al. 2011 Randomized phase II neoadjuvant negative predictive factor for tamoxifen response in hormone receptor comparison between letrozole, anastrozole, and exemestane for positive premenopausal breast cancer. Journal of Clinical Oncology 25 postmenopausal women with estrogen receptor-rich stage 2 to 3 breast 3007–3014. (doi:10.1200/JCO.2006.08.9938) cancer: clinical and biomarker outcomes and predictive value of the Gonzalez-Malerva L, Park J, Zou L, Hu Y, Moradpour Z, Pearlberg J, Sawyer J, baseline PAM50-based intrinsic subtype – ACOSOG Z1031. Journal of Stevens H, Harlow E & LaBaer J 2011 High-throughput ectopic Clinical Oncology 29 2342–2349. (doi:10.1200/JCO.2010.31.6950) expression screen for tamoxifen resistance identifies an atypical kinase Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, Wallis JW, Van Tine BA, Hoog J, that blocks autophagy. PNAS 108 2058–2063. (doi:10.1073/pnas. Goiffon RJ, Goldstein TC et al. 2012 Whole-genome analysis informs 1018157108) breast cancer response to aromatase inhibition. Nature 486 353–360. Goss PE, Ingle JN, Ales-Martinez JE, Cheung AM, Chlebowski RT, (doi:10.1038/nature11143) Wactawski-Wende J, McTiernan A, Robbins J, Johnson KC, Martin LW Encarnacion CA, Ciocca DR, McGuire WL, Clark GM, Fuqua SA & et al. 2011 Exemestane for breast-cancer prevention in postmenopausal Osborne CK 1993 Measurement of steroid hormone receptors in breast women. New England Journal of Medicine 364 2381–2391. (doi:10.1056/ cancer patients on tamoxifen. Breast Cancer Research and Treatment 26 NEJMoa1103507) 237–246. (doi:10.1007/BF00665801) deGraffenried LA, Friedrichs WE, Russell DH, Donzis EJ, Middleton AK, Faridi J, Wang L, Endemann G & Roth RA 2003 Expression of constitutively Silva JM, Roth RA & Hidalgo M 2004 Inhibition of mTOR activity active Akt-3 in MCF-7 breast cancer cells reverses the estrogen and restores tamoxifen response in breast cancer cells with aberrant Akt tamoxifen responsivity of these cells in vivo. Clinical Cancer Research 9 activity. Clinical Cancer Research 10 8059–8067. (doi:10.1158/1078- 2933–2939. 0432.CCR-04-0035) Finetti P, Cervera N, Charafe-Jauffret E, Chabannon C, Charpin C, Guo JP, Shu SK, Esposito NN, Coppola D, Koomen JM & Cheng JQ 2010 Chaffanet M, Jacquemier J, Viens P, Birnbaum D & Bertucci F 2008 IKKepsilon, phosphorylation of estrogen receptor a Ser-167 and Sixteen-kinase gene expression identifies luminal breast cancers with contribution to tamoxifen resistance in breast cancer. Journal of poor prognosis. Cancer Research 68 767–776. (doi:10.1158/0008-5472. Biological Chemistry 285 3676–3684. (doi:10.1074/jbc.M109.078212) CAN-07-5516) Han HS, Yu E, Song JY, Park JY, Jang SJ & Choi J 2009 The estrogen receptor a Fox EM, Miller TW, Balko JM, Kuba MG, Sanchez V, Smith RA, Liu S, pathway induces oncogenic Wip1 phosphatase gene expression. Molecular Gonzalez-Angulo AM, Mills GB, Ye F et al. 2011 A kinome-wide screen Cancer Research 7 713–723. (doi:10.1158/1541-7786.MCR-08-0247) identifies the insulin/IGF-I receptor pathway as a mechanism of escape Hanahan D & Weinberg RA 2011 Hallmarks of cancer: the next generation. from hormone dependence in breast cancer. Cancer Research 71 Cell 144 646–674. (doi:10.1016/j.cell.2011.02.013) 6773–6784. (doi:10.1158/0008-5472.CAN-11-1295) He X, Zheng Z, Song T, Wei C, Ma H, Ma Q, Zhang Y, Xu Y, Shi W, Ye Q et al. Frankel LB, Lykkesfeldt AE, Hansen JB & Stenvang J 2007 Protein kinase Ca 2010 c-Abl regulates estrogen receptor a transcription activity is a marker for antiestrogen resistance and is involved in the growth of through its stabilization by phosphorylation. Oncogene 29 2238–2251. tamoxifen resistant human breast cancer cells. Breast Cancer Research (doi:10.1038/onc.2009.513) Endocrine-Related Cancer and Treatment 104 165–179. (doi:10.1007/s10549-006-9399-1) Henrich L, Smith J, Kitt D, Errington T, Nguyen B, Traish A & Lannigan D Fu X, Osborne CK & Schiff R 2013 Biology and therapeutic potential of PI3K 2003 Extracellular signal-regulated kinase 7, a regulator of hormone- signaling in ERC/HER2-negative breast cancer. Breast 22 (Suppl 2) dependent estrogen receptor destruction. Molecular and Cellular Biology S12–S18. (doi:10.1016/j.breast.2013.08.001) 23 5979–5988. (doi:10.1128/MCB.23.17.5979-5988.2003) Gao Q, Patani N, Dunbier AK, Ghazoui Z, Zvelebil M, Martin LA & Dowsett M Herynk M & Fuqua S 2004 Estrogen receptor mutations in human disease. 2014 Effect of aromatase inhibition on functional gene modules in Endocrine Reviews 25 869–898. (doi:10.1210/er.2003-0010) oestrogen receptor positive breast cancer and their relationship with Holm C, Kok M, Michalides R, Fles R, Koornstra RH, Wesseling J, antiproliferative response. Clinical Cancer Research 20 2485–2494. Hauptmann M, Neefjes J, Peterse JL, Stal O et al. 2009 Phosphorylation (doi:10.1158/1078-0432.CCR-13-2602) of the oestrogen receptor a at serine 305 and prediction of tamoxifen Gee J, Robertson J, Ellis I & Nicholson R 2001 Phosphorylation of ERK1/2 resistance in breast cancer. Journal of Pathology 217 372–379. mitogen-activated protein kinase is associated with poor response to (doi:10.1002/path.2455) anti-hormonal therapy and decreased patient survival in clinical breast Horwitz KB, McGuire WL, Pearson OH & Segaloff A 1975 Predicting cancer. International Journal of Cancer 95 247–254. (doi:10.1002/ response to endocrine therapy in human breast cancer: a hypothesis. 1097-0215(20010720)95:4!247::AID-IJC1042O3.0.CO;2-S) Science 189 726–727. (doi:10.1126/science.168640) Gee J, Robertson J, Gutteridge E, Ellis I, Pinder S, Rubini M & Nicholson R Ignatiadis M & Sotiriou C 2013 Luminal breast cancer: from biology to 2005 Epidermal growth factor receptor/HER2/insulin-like growth treatment. Nature Reviews. Clinical Oncology 10 494–506. (doi:10.1038/ factor receptor signalling and oestrogen receptor activity in clinical nrclinonc.2013.124) breast cancer. Endocrine-Related Cancer 12 (Suppl 1) S99–S111. Iorns E, Turner NC, Elliott R, Syed N, Garrone O, Gasco M, Tutt AN, Crook T, (doi:10.1677/erc.1.01005) Lord CJ & Ashworth A 2008 Identification of CDK10 as an important Generali D, Fox SB, Brizzi MP, Allevi G, Bonardi S, Aguggini S, Milani M, determinant of resistance to endocrine therapy for breast cancer. Cancer Bersiga A, Campo L, Dionisio R et al. 2008 Down-regulation of Cell 13 91–104. (doi:10.1016/j.ccr.2008.01.001) phosphatidylinositol 30-kinase/AKT/molecular target of rapamycin Iorns E, Martens-de Kemp SR, Lord CJ & Ashworth A 2009 CRK7 modifies metabolic pathway by primary letrozole-based therapy in human breast the MAPK pathway and influences the response to endocrine therapy. cancer. Clinical Cancer Research 14 2673–2680. (doi:10.1158/ Carcinogenesis 30 1696–1701. (doi:10.1093/carcin/bgp187) 1078-0432.CCR-07-1046) Jensen E & Jordan V 2003 The estrogen receptor: a model for molecular Generali D, Symmans WF, Berruti A & Fox SB 2011 Predictive immuno- medicine. Clinical Cancer Research 9 1980–1989. histochemical biomarkers in the context of neoadjuvant therapy for Jensen E, Block G, Smith S, Kyser K & DeSombre E 1971 Estrogen receptors breast cancer. Journal of the National Cancer Institute. Monographs 2011 and breast cancer response to adrenalectomy. Journal of the National 99–102. (doi:10.1093/jncimonographs/lgr030) Cancer Institute. Monographs 34 55–70.

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R368 ER-positive status

Jeselsohn R, Yelensky R, Buchwalter G, Frampton G, Meric-Bernstam F, Madak-Erdogan Z, Lupien M, Stossi F, Brown M & Katzenellenbogen BS Gonzalez-Angulo AM, Ferrer-Lozano J, Perez-Fidalgo JA, Cristofanilli M, 2011 Genomic collaboration of estrogen receptor a and extracellular Gomez H et al. 2014 Emergence of constitutively active estrogen signal-regulated kinase 2 in regulating gene and proliferation programs. receptor-a mutations in pretreated advanced estrogen receptor-positive Molecular and Cellular Biology 31 226–236. (doi:10.1128/MCB.00821-10) breast cancer. Clinical Cancer Research 20 1757–1767. (doi:10.1158/ Madak-Erdogan Z, VentrellaR, Petry L & Katzenellenbogen BS2014 Novel roles 1078-0432.CCR-13-2332) for ERK5 and cofilin as critical mediators linking ERa-driven transcription, Jiang J, Sarwar N, Peston D, Kulinskaya E, Shousha S, Coombes R & Ali S 2007 actin reorganization and invasiveness in breast cancer. Molecular Cancer Phosphorylation of estrogen receptor-a at Ser167 is indicative of longer Research 12 714–727. (doi:10.1158/1541-7786.MCR-13-0588) disease-free and overall survival in breast cancer patients. Clinical Cancer Malumbres M & Barbacid M 2009 Cell cycle, CDKs and cancer: a changing Research 13 5769–5776. (doi:10.1158/1078-0432.CCR-07-0822) paradigm. Nature Reviews. Cancer 9 153–166. (doi:10.1038/nrc2602) Joel PB, Smith J, Sturgill TW, Fisher TL, Blenis J & Lannigan DA 1998 Marquez DC, Lee J, Lin T & Pietras RJ 2001 Epidermal growth factor pp90rsk1 regulates estrogen receptor-mediated transcription through receptor and tyrosine phosphorylation of estrogen receptor. Endocrine phosphorylation of Ser-167. Molecular and Cellular Biology 18 1978–1984. 16 73–81. (doi:10.1385/ENDO:16:2:073) Kalstad G, Cornmark L, Zahirovic I, Landberg G, Jirstrom K & Larsson C Maruani DM, Spiegel TN, Harris EN, Shachter AS, Unger HA, Herrero- 2010 PKCa expression is a marker for breast cancer aggressiveness. Gonzalez S & Holz MK 2012 Estrogenic regulation of S6K1 expression Molecular Cancer 9 76–90. (doi:10.1186/1476-4598-9-76) creates a positive regulatory loop in control of breast cancer cell Kanaujiya JK, Lochab S, Kapoor I, Pal P, Datta D, Bhatt ML, Sanyal S, Behre G proliferation. Oncogene 31 5073–5080. (doi:10.1038/onc.2011.657) & Trivedi AK 2013 Proteomic identification of Profilin1 as a corepressor Mazumdar A & Kumar R 2003 Estrogen regulation of Pak1 and FKHR of estrogen receptor a in MCF7 breast cancer cells. Proteomics 13 pathways in breast cancer cells. FEBS Letters 535 6–10. (doi:10.1016/ 2100–2112. (doi:10.1002/pmic.201200534) S0014-5793(02)03846-2) Kazi AA, Molitoris KH & Koos RD 2009 Estrogen rapidly activates the McClaine RJ, Marshall AM, Wagh PK & Waltz SE 2010 Ron receptor PI3K/AKT pathway and hypoxia-inducible factor 1 and induces tyrosine kinase activation confers resistance to tamoxifen in breast vascular endothelial growth factor A expression in luminal epithelial cancer cell lines. Neoplasia 12 650–658. cells of the rat uterus. Biology of Reproduction 81 378–387. (doi:10.1095/ Medunjanin S, Hermani A, Servi BD, Grisouard J, Rincke G & Mayer D 2005 Glycogen synthase kinase-3 interacts with and phosphorylates estrogen biolreprod.109.076117) receptor a and is involved in the regulation of receptor activity. Journal of Khan SA, Bhandare D & Chatterton RT Jr 2005 The local hormonal Biological Chemistry 280 33006–33014. (doi:10.1074/jbc.M506758200) environment and related biomarkers in the normal breast. Endocrine- Medunjanin S, Weinert S, Schmeisser A, Mayer D & Braun-Dullaeus RC Related Cancer 12 497–510. (doi:10.1677/erc.1.00732) 2010 Interaction of the double-strand break repair kinase DNA-PK Knight Z, Lin H & Shokat K 2010 Targeting the cancer kinome through and estrogen receptor-a. Molecular Biology of the Cell 21 1620–1628. polypharmacology. Nature Reviews. Cancer 10 130–137. (doi:10.1038/ (doi:10.1091/mbc.E09-08-0724) nrc2787) Mendez P, Azcoitia I & Garcia-Segura LM 2003 Estrogen receptor a forms Kok M, Zwart W, Holm C, Fles R, Hauptmann M, Van’t Veer LJ, Wessels LF, estrogen-dependent multimolecular complexes with insulin-like Neefjes J, Stal O, Linn SC et al. 2010 PKA-induced phosphorylation of growth factor receptor and phosphatidylinositol 3-kinase in the adult ERa at serine 305 and high PAK1 levels is associated with sensitivity to rat brain. Brain Research. Molecular Brain Research 112 170–176. tamoxifen in ER-positive breast cancer. Breast Cancer Research and Michalides R, van Tinteren H, Balkenende A, Vermorken JB, Benraadt J, Treatment 125 1–12. (doi:10.1007/s10549-010-0798-y) Huldij J & van Diest P 2002 Cyclin A is a prognostic indicator in early Endocrine-Related Cancer Lee H & Bai W 2002 Regulation of estrogen receptor nuclear export by ligand- stage breast cancer with and without tamoxifen treatment. British induced and p38-mediated receptor phosphorylation. Molecular and Journal of Cancer 86 402–408. (doi:10.1038/sj.bjc.6600072) Cellular Biology 22 5835–5845. (doi:10.1128/MCB.22.16.5835-5845.2002) Michalides R, Griekspoor A, Balkenende A, Verwoerd D, Janssen L, Jalink K, Lee YR, Park J, Yu HN, Kim JS, Youn HJ & Jung SH 2005 Up-regulation of Floore A, Velds A, van’t Veer L & Neefjes J 2004 Tamoxifen resistance by PI3K/Akt signaling by 17b-estradiol through activation of estrogen a conformational arrest of the estrogen receptor a after PKA activation receptor-a, but not estrogen receptor-b, and stimulates cell growth in in breast cancer. Cancer Cell 5 597–605. (doi:10.1016/j.ccr.2004.05.016) breast cancer cells. Biochemical and Biophysical Research Communications Midland AA, Whittle MC, Duncan JS, Abell AN, Nakamura K, Zawistowski JS, 336 1221–1226. (doi:10.1016/j.bbrc.2005.08.256) Carey LA, Earp HS III, Graves LM, Gomez SM et al. 2012 Defining Lee S, Mohsin SK, Mao S, Hilsenbeck SG, Medina D & Allred DC 2006 the expressed breast cancer kinome. Cell Research 22 620–623. Hormones, receptors, and growth in hyperplastic enlarged lobular (doi:10.1038/cr.2012.25) units: early potential precursors of breast cancer. Breast Cancer Research Miller TW, Hennessy BT, Gonzalez-Angulo AM, Fox EM, Mills GB, Chen H, 8 R6. (doi:10.1186/bcr1367) Higham C, Garcia-Echeverria C, Shyr Y & Arteaga CL 2010 Hyperacti- de Leeuw R, Flach K, Bentin Toaldo C, Alexi X, Canisius S, Neefjes J, vation of phosphatidylinositol-3 kinase promotes escape from hormone Michalides R & Zwart W 2013 PKA phosphorylation redirects ERa to dependence in estrogen receptor-positive human breast cancer. promoters of a unique gene set to induce tamoxifen resistance. Journal of Clinical Investigation 120 2406–2413. (doi:10.1172/JCI41680) Oncogene 32 3543–3551. (doi:10.1038/onc.2012.361) Miller TW, Balko JM & Arteaga CL 2011a Phosphatidylinositol 3-kinase and Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM & Corbo L 2011 antiestrogen resistance in breast cancer. Journal of Clinical Oncology 29 Cracking the estrogen receptor’s posttranslational code in breast 4452–4461. (doi:10.1200/JCO.2010.34.4879) tumors. Endocrine Reviews 32 597–622. (doi:10.1210/er.2010-0016) Miller TW, Rexer BN, Garrett JT & Arteaga CL 2011b Mutations in the Lippman M, Bolan G & Huff K 1976 Effects of estrogens and antiestrogens phosphatidylinositol 3-kinase pathway: role in tumor progression and on hormone-responisve human breast cancer in long-term tissue therapeutic implications in breast cancer. Breast Cancer Research 13 224. culture. Cancer Research 36 4595–4601. (doi:10.1186/bcr3039) Lu Q, Surks H, Ebling H, Baur W, Brown D, Pallas D & Karas R 2003a von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Regulation of estrogen receptor a-mediated transcription by a direct Gerber B, Eiermann W, Hilfrich J, Huober J et al. 2012 Definition and interaction with protein phosphatase 2A. Journal of Biological Chemistry impact of pathologic complete response on prognosis after neoadjuvant 278 4639–4645. (doi:10.1074/jbc.M210949200) chemotherapy in various intrinsic breast cancer subtypes. Journal of Lu M, Miller KD, Gokmen-Polar Y, Jeng MH & Kinch MS 2003b EphA2 Clinical Oncology 30 1796–1804. (doi:10.1200/JCO.2011.38.8595) overexpression decreases estrogen dependence and tamoxifen Morandi A, Martin LA, Gao Q, Pancholi S, Mackay A, Robertson D, Zvelebil M, sensitivity. Cancer Research 63 3425–3429. Dowsett M, Plaza-Menacho I & Isacke CM 2013 GDNF-RET signaling in

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R369 ER-positive status

ER-positive breast cancers is a key determinant of response and Proia DA, Nannenga BW, Donehower LA & Weigel NL 2006 Dual roles for the resistance to aromatase inhibitors. Cancer Research 73 3783–3795. phosphatase PPM1D in regulating progesterone receptor function. (doi:10.1158/0008-5472.CAN-12-4265) Journal of Biological Chemistry 281 7089–7101. (doi:10.1074/jbc. Murphy LC, Cherlet T, Adeyinka A, Niu Y, Snell L & Watson P 2004 M511839200) Phospho-serine-118 estrogen receptor-a detection in human breast Ribelles N, Perez-Villa L, Jerez JM, Pajares B, Vicioso L, Jimenez B, de Luque V, tumors in vivo. Clinical Cancer Research 10 1354–1359. (doi:10.1158/ Franco L, Gallego E, Marquez A et al. 2013 Pattern of recurrence of early 1078-0432.CCR-03-0112) breast cancer is different according to intrinsic subtype and proliferation Murphy LC, Seekallu SV & Watson PH 2011 Clinical significance of index. Breast Cancer Research 15 R98. (doi:10.1186/bcr3559) estrogen receptor phosphorylation. Endocrine-Related Cancer 18 Robinson DR, Wu YM, Vats P, Su F, Lonigro RJ, Cao X, Kalyana-Sundaram S, R1–R14. (doi:10.1677/ERC-10-0070) Wang R, Ning Y, Hodges L et al. 2013 Activating ESR1 mutations in Muscat GE, Eriksson NA, Byth K, Loi S, Graham D, Jindal S, Davis MJ, Clyne C, hormone-resistant metastatic breast cancer. Nature Genetics 45 Funder JW, Simpson ER et al. 2013 Research resource: nuclear receptors as 1446–1451. (doi:10.1038/ng.2823) transcriptome: discriminant and prognostic value in breast cancer. Rogatsky I, Trowbridge J & Garabedian M 1999 Potentiation of human Molecular Endocrinology 27 350–365. (doi:10.1210/me.2012-1265) estrogen receptor a transcriptional activation through phosphorylation Musgrove EA & Sutherland RL 2009 Biological determinants of endocrine of serines 104 and 106 by the cyclin A–CDK2 complex. Journal of resistance in breast cancer. Nature Reviews. Cancer 9 631–643. Biological Chemistry 274 22296–22302. (doi:10.1074/jbc.274.32.22296) (doi:10.1038/nrc2713) Rowan B, Weigel N & O’Malley B 2000 Phosphorylation of steroid Nabha SM, Glaros S, Hong M, Lykkesfeldt AE, Schiff R, Osborne K & Reddy KB receptor coactivator-1. Journal of Biological Chemistry 275 4475–4483. 2005 Upregulation of PKC-delta contributes to antiestrogen resistance (doi:10.1074/jbc.275.6.4475) in mammary tumor cells. Oncogene 24 3166–3176. (doi:10.1038/sj.onc. Ruckhaberle E, Rody A, Engels K, Gaetje R, von Minckwitz G, Schiffmann S, 1208502) Grosch S, Geisslinger G, Holtrich U, Karn T et al. 2008 Microarray Nakatani K, Thompson DA, Barthel A, Sakaue H, Liu W, Weigel RJ & Roth RA analysis of altered sphingolipid metabolism reveals prognostic signi- 1999 Up-regulation of Akt3 in estrogen receptor-deficient breast ficance of sphingosine kinase 1 in breast cancer. Breast Cancer Research cancers and androgen-independent prostate cancer lines. Journal of and Treatment 112 41–52. (doi:10.1007/s10549-007-9836-9) Biological Chemistry 274 21528–21532. (doi:10.1074/jbc.274.31.21528) Santen RJ, Song RX, Zhang Z, Kumar R, Jeng MH, Masamura S, Yue W & Nettles KW, Bruning JB, Gil G, Nowak J, Sharma SK, Hahm JB, Kulp K, Berstein L 2003 Adaptive hypersensitivity to estrogen: mechanism for Hochberg RB, Zhou H, Katzenellenbogen JA et al. 2008 NFkBselectivityof superiority of aromatase inhibitors over selective estrogen receptor estrogen receptor ligands revealed by comparative crystallographic modulators for breast cancer treatment and prevention. Endocrine- analyses. Nature Chemical Biology 4 241–247. (doi:10.1038/nchembio.76) Related Cancer 10 111–130. (doi:10.1677/erc.0.0100111) Osborne CK & Schiff R 2011 Mechanisms of endocrine resistance in breast Santen R, Song R, Zhang Z, Yue W & Kumar R 2004 Adaptive cancer. Annual Review of Medicine 62 233–247. (doi:10.1146/annurev- hypersensitivity to estrogen: mechanism for sequential responses to med-070909-182917) hormonal therapy in breast cancer. Clinical Cancer Research 10 Osborne C, Bardou V, Hopp T, Chamness G, Hilsenbeck S, Fuqua S, Wong J, 337S–345S. (doi:10.1158/1078-0432.CCR-031207) Allred D, Clark G & Schiff R 2003 Role of the estrogen receptor Segal CV & Dowsett M 2014 Estrogen receptor mutations in breast cancer- coactivator AIB1 (SRC-3) and HER-2/neu in tamoxifen resistance in new focus on an old target. Clinical Cancer Research 20 1724–1726. breast cancer. Journal of the National Cancer Institute 95 353–361. (doi:10.1158/1078-0432.CCR-14-0067)

Endocrine-Related Cancer (doi:10.1093/jnci/95.5.353) Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, Turashvili G, Ding J, Tse K, Park K, Krishnan V, O’Malley B, Yamamoto Y & Gaynor R 2005 Formation Haffari G et al. 2012 The clonal and mutational evolution spectrum of of an IKKa-dependent transcription complex is required for estrogen primary triple-negative breast cancers. Nature 486 395–399. receptor-mediated gene activation. Molecular Cell 18 71–82. (doi:10.1038/nature10933) (doi:10.1016/j.molcel.2005.03.006) Shoker BS, Jarvis C, Sibson DR, Walker C & Sloane JP 1999 Oestrogen Paul A, Garcia YA, Zierer B, Patwardhan C, Gutierrez O, Hildenbrand Z, receptor expression in the normal and pre-cancerous breast. Journal of Harris DC, Balsiger HA, Sivils JC, Johnson JL et al. 2014 The Pathology 188 237–244. (doi:10.1002/(SICI)1096-9896(199907)188:3 cochaperone SGTA (small glutamine-rich tetratricopeptide repeat- !237::AID-PATH343O3.0.CO;2-8) containing protein a) demonstrates regulatory specificity for the Shrivastav A, Bruce M, Jaksic D, Bader T, Seekallu S, Penner C, Nugent Z, androgen, glucocorticoid and progesterone receptors. Journal of Watson P & Murphy L 2014 The mechanistic target for rapamycin is Biological Chemistry 289 15297–15308. (doi:10.1074/jbc.M113.535229) related to the phosphorylation score for estrogen receptor a in human Perez-Tenorio G, Stal O & Group MotSSBC 2002 Activation of AKT/PKB in breast tumors, in vivo. Breast Cancer Research 16 R49. (doi:10.1186/ breast cancer predicts a worse outcome among endocrine treated patients. bcr3660) British Journal of Cancer 86 540–545. (doi:10.1038/sj.bjc.6600126) Silva J, Cavazos DA, Donzis E, Friedrichs WE, Marciniak R & Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, deGraffenried LA 2007 Akt-induced tamoxifen resistance is associated Ross DT, Johnsen H, Akslen LA et al. 2000 Molecular portraits of human with altered FKHR regulation. Cancer Investigation 25 569–573. breast tumors. Nature 406 747–752. (doi:10.1038/35021093) (doi:10.1080/07357900701513538) Pham TT, Angus SP & Johnson GL 2013 MAP3K1: genomic alterations in Skliris GP, Nugent ZJ, Rowan BG, Penner CR, Watson PH & Murphy LC cancer and function in promoting cell survival or apoptosis. Genes & 2010a A phosphorylation code for oestrogen receptor-a predicts Cancer 4 419–426. (doi:10.1177/1947601913513950) clinical outcome to endocrine therapy in breast cancer. Endocrine- Plaza-Menacho I, Morandi A, Robertson D, Pancholi S, Drury S, Dowsett M, Related Cancer 17 589–597. (doi:10.1677/ERC-10-0030) Martin LA & Isacke CM 2010 Targeting the RET Skliris GP, Nugent ZJ, Watson PH & Murphy LC 2010b Estrogen receptor a sensitizes breast cancer cells to tamoxifen treatment and reveals a role phosphorylated at tyrosine 537 is associated with poor clinical for RET in endocrine resistance. Oncogene 29 4648–4657. (doi:10.1038/ outcome in breast cancer patients treated with tamoxifen. Hormones & onc.2010.209) Cancer 1 215–221. (doi:10.1007/s12672-010-0049-z) Poulard C, Treilleux I, Lavergne E, Bouchekioua-Bouzaghou K, Goddard- Sorlie T, Tibshirani R, Parker J, Hastie T, Marron J, Nobel A, Deng S, Johnsen H, Leon S, Chabaud S, Tredan O, Corbo L & Le Romancer M 2012 Activation Pesich R, Geisler S et al. 2003 Repeated observation of breast tumor of rapid oestrogen signalling in aggressive human breast cancers. EMBO subtypes in independent gene expression data sets. PNAS 100 Molecular Medicine 4 1200–1213. (doi:10.1002/emmm.201201615) 8418–8423. (doi:10.1073/pnas.0932692100)

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access Review M C Bruce et al. Kinome associated with 21:5 R370 ER-positive status

Soule HD, Vazguez J, Long A, Albert S & Brennan M 1973 A human cell line polymorphism defines clinical outcome in estrogen receptor-positive from a pleural effusion derived from a breast carcinoma. Journal of the breast cancer patients treated with tamoxifen. Pharmacogenomics National Cancer Institute 51 1409–1416. Journal 14 28–34. (doi:10.1038/tpj.2013.8) Spears M,Cunningham CA, TaylorKJ,Mallon EA,Thomas JS,Kerr GR, Jack WJ, Wu RC, Qin J, Yi P, Wong J, Tsai SY, Tsai MJ & O’Malley BW 2004 Selective Kunkler IH, Cameron DA, Chetty U et al. 2012 Proximity ligation assays phosphorylations of the SRC-3/AIB1 coactivator integrate genomic for isoform-specific Akt activation in breast cancer identify activated reponses to multiple cellular signaling pathways. Molecular Cell 15 Akt1 as a driver of progression. Journal of Pathology 227 481–489. 937–949. (doi:10.1016/j.molcel.2004.08.019) (doi:10.1002/path.4022) Wu RC, Smith CL & O’Malley BW 2005 Transcriptional regulation by Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, steroid receptor coactivator phosphorylation. Endocrine Reviews 26 Nik-Zainal S, Martin S, Varela I, Bignell GR et al. 2012 The landscape of 393–399. (doi:10.1210/er.2004-0018) cancer genes and mutational processes in breast cancer. Nature 486 Wu RC, Feng Q, Lonard DM & O’Malley BW 2007 SRC-3 coactivator 400–404. (doi:10.1038/nature11017) functional lifetime is regulated by a phospho-dependent ubiquitin time Stuhlmiller TJ, Earp HS & Johnson GL 2014 Adaptive reprogramming of the clock. Cell 129 1125–1140. (doi:10.1016/j.cell.2007.04.039) breast cancer kinome. Clinical Pharmacology and Therapeutics 95 Yamnik RL & Holz MK 2010 mTOR/S6K1 and MAPK/RSK signaling pathways 413–415. (doi:10.1038/clpt.2014.8) coordinately regulate estrogen receptor a serine 167 phosphorylation. Sukocheva O, Wang L, Verrier E, Vadas MA & Xia P 2009 Restoring FEBS Letters 584 124–128. (doi:10.1016/j.febslet.2009.11.041) endocrine response in breast cancer cells by inhibition of the Yamnik RL, Digilova A, Davis DC, Brodt ZN, Murphy CJ & Holz MK 2009 S6 sphingosine kinase-1 signaling pathway. Endocrinology 150 4484–4492. kinase 1 regulates estrogen receptor a in control of breast cancer cell (doi:10.1210/en.2009-0391) proliferation. Journal of Biological Chemistry 284 6361–6369. Sun M, Paciga J, Feldman R, Yuan Z, Coppola D, Lu Y, Shelley S, Nicosia S & (doi:10.1074/jbc.M807532200) Cheng J 2001 Phosphatidylinositol-3-OH kinase (PI3K)/AKT2, activated Yerushalmi R, Woods R, Ravdin PM, Hayes MM & Gelmon KA 2010 Ki67 in in breast cancer, regulates and is induced by estrogen receptor a (ERa) breast cancer: prognostic and predictive potential. Lancet Oncology 11 via interaction between ERa and PI3K. Cancer Research 61 5985–5991. 174–183. (doi:10.1016/S1470-2045(09)70262-1) Sun J, Zhou W, Kaliappan K, Nawaz Z & Slingerland JM 2012 ERa York B, Yu C, Sagen JV, Liu Z, Nikolai BC, Wu RC, Finegold M, Xu J & phosphorylation at Y537 by src triggers E6-AP-ERa binding, ERa O’Malley BW 2010 Reprogramming the posttranslational code of SRC-3 ubiquitylation, promoter occupancy, and target gene expression. confers a switch in mammalian systems biology. PNAS 107 Molecular Endocrinology 26 1567–1577. (doi:10.1210/me.2012-1140) 11122–11127. (doi:10.1073/pnas.1005262107) Teng DH, Perry WL III, Hogan JK, Baumgard M, Bell R, Berry S, Davis T, Frank D, Yu J & Henske EP 2006 Estrogen-induced activation of mammalian target Frye C, Hattier T etal.1997 Human mitogen-activated protein kinase kinase of rapamycin is mediated via tuberin and the small GTPase Ras 4 as a candidate tumor suppressor. Cancer Research 57 4177–4182. homologue enriched in brain. Cancer Research 66 9461–9466. Tomlinson DC, Knowles MA & Speirs V 2012 Mechanisms of FGFR3 actions (doi:10.1158/0008-5472.CAN-06-1895) in endocrine resistant breast cancer. International Journal of Cancer 130 Zhang Q-X, Borg A, Wolf D, Oesterreich S & Fuqua S 1997 An estrogen 2857–2866. (doi:10.1002/ijc.26304) receptor mutant with strong hormone independent activity from a Trevino LS & Weigel NL 2013 Phosphorylation: a fundamental regulator of metastatic breast cancer. Cancer Research 57 1244–1249. steroid receptor action. Trends in Endocrinology and Metabolism 24 Zhang Z, Maier B, Santen RJ & Song RX 2002 Membrane association of 515–524. (doi:10.1016/j.tem.2013.05.008) estrogen receptor a mediates estrogen effect on MAPK activation. Turner N, Pearson A, Sharpe R, Lambros M, Geyer F, Lopez-Garcia MA, Biochemical and Biophysical Research Communications 294 926–933. Endocrine-Related Cancer Natrajan R, Marchio C, Iorns E, Mackay A et al. 2010 FGFR1 (doi:10.1016/S0006-291X(02)00348-0) amplification drives endocrine therapy resistance and is a therapeutic Zhang Y, Moerkens M, Ramaiahgari S, de Bont H, Price L, Meerman J & target in breast cancer. Cancer Research 70 2085–2094. (doi:10.1158/ van de Water B 2011 Elevated insulin-like growth factor 1 receptor signaling 0008-5472.CAN-09-3746) induces antiestrogen resistance through the MAPK/ERK and PI3K/Akt Wang RA, Mazumdar A, Vadlamudi RK & Kumar R 2002 P21-activated signaling routes. Breast Cancer Research 13 R52. (doi:10.1186/bcr2883) kinase-1 phosphorylates and transactivates estrogen receptor-a and Zhang F, Wang Y, Wang L, Luo X, Huang K, Wang C, Du M, Liu F, Luo T & promotes hyperplasia in mammary epithelium. EMBO Journal 21 Huang D 2013 Poly(ADP-ribose) polymerase 1 is a key regulator of 5437–5447. (doi:10.1093/emboj/cdf543) estrogen receptor a-dependent gene transcription. Journal of Biological Wei C, Cao Y, Yang X, Zheng Z, Guan K, Wang Q, Tai Y, Zhang Y, Ma S, Ge X Chemistry 288 11348–11357. (doi:10.1074/jbc.M112.429134) et al. 2014 Elevated expression of TANK-binding kinase 1 enhances Zhao H, Ou-Yang F, Chen IF, Hou MF, Yuan SS, Chang HL, Lee YC, Plattner R, tamoxifen resistance in breast cancer. PNAS 111 E601–E610. (doi:10. Waltz SE, Ho SM et al. 2010 Enhanced resistance to tamoxifen by the 1073/pnas.1316255111) c-ABL proto-oncogene in breast cancer. Neoplasia 12 214–223. Wierer M, Verde G, Pisano P, Molina H, Font-Mateu J, Di Croce L & Beato M Zhou W & Slingerland JM 2014 Links between oestrogen receptor 2013 PLK1 signaling in breast cancer cells cooperates with estrogen activation and proteolysis: relevance to hormone-regulated cancer receptor-dependent gene transcription. Cell Reports 3 2021–2032. therapy. Nature Reviews. Cancer 14 26–38. (doi:10.1038/nrc3622) (doi:10.1016/j.celrep.2013.05.024) Zwart W, Griekspoor A, Berno V, Lakeman K, Jalink K, Mancini M, Neefjes J Williams CC, Basu A, El-Gharbawy A, Carrier LM, Smith CL & Rowan BG & Michalides R 2007a PKA-induced resistance to tamoxifen is 2009 Identification of four novel phosphorylation sites in estrogen associated with an altered orientation of ERa towards co-activator receptor a: impact on receptor-dependent gene expression and SRC-1. EMBO Journal 26 3534–3544. (doi:10.1038/sj.emboj.7601791) phosphorylation by protein kinase CK2. BMC Biochemistry 10 36. Zwart W, Griekspoor A, Rondaij M, Verwoerd D, Neefjes J & Michalides R (doi:10.1186/1471-2091-10-36) 2007b Classification of anti-estrogens according to intramolecular FRET Winder T, Giamas G, Wilson PM, Zhang W, Yang D, Bohanes P, Ning Y, effects on phospho-mutants of estrogen receptor a. Molecular Cancer Gerger A, Stebbing J & Lenz HJ 2014 Insulin-like growth factor receptor Therapeutics 6 1526–1533. (doi:10.1158/1535-7163.MCT-06-0750)

Received in final form 9 July 2014 Accepted 23 July 2014 Made available online as an Accepted Preprint 23 July 2014

http://erc.endocrinology-journals.org q 2014 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/ERC-14-0232 Printed in Great Britain Downloaded from Bioscientifica.com at 09/26/2021 01:37:02PM via free access