12
Luminal B Breast Cancer: Molecular Characterization, Clinical Management, and Future Perspectives Felipe Ades, Dimitrios Zardavas, Ivana Bozovic-Spasojevic, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Giuseppe Viale, Christos Sotiriou, and Martine Piccart Felipe Ades, Lina Pugliano, Debora Fumagalli, Evandro de Azambuja, Chris- tos Sotiriou, and Martine Piccart, Insti- tut Jules Bordet, Universite ´ Libre de Bruxelles; Dimitrios Zardavas, Breast International Group, Brussels, Belgium; Ivana Bozovic-Spasojevic, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia; and Giuseppe Viale, European Institute of Oncology, Univer- sity of Milan, Milan, Italy. Published online ahead of print at www.jco.org on July 21, 2014. Authors’ disclosures of potential con- flicts of interest and author contribu- tions are found at the end of this article. Corresponding author: Martine Piccart, MD, PhD, Institut Jules Bordet, 121 Boulevard de Waterloo, Bruxelles, Belgium 1000; e-mail: [email protected]. © 2014 by American Society of Clinical Oncology 0732-183X/14/3299-1/$20.00 DOI: 10.1200/JCO.2013.54.1870 A B S T R A C T Gene expression profiling has reshaped our understanding of breast cancer by defining and characterizing four main intrinsic molecular subtypes: human epidermal growth factor receptor 2– enriched, basal-like, luminal A, and luminal B subtypes. Luminal B breast cancer has been reported to have lower expression of hormone receptors, higher expression of proliferation markers, and higher histologic grade than luminal A. It also exhibits worse prognosis and has a distinct profile of response to hormone therapy and chemotherapy. Although luminal cancers share similarities, the studies conducted in recent years using next-generation sequencing technology show that luminal A and B breast cancers should be perceived as distinct entities, with specific oncogenic drivers, rather than more proliferative varieties of luminal tumors. This review discusses the definition and molecular characterization of luminal B breast cancer and presents the available clinical evidence for chemotherapy and endocrine therapy patterns of response. It also provides an overview of ongoing research on molecularly targeted agents for this disease. J Clin Oncol 32. © 2014 by American Society of Clinical Oncology INTRODUCTION In the early 2000s, high-throughput technology en- abled the interrogation of the expression of thou- sands of genes in a single experiment. This led to the determination that breast cancer comprises at least four molecularly distinct diseases with different fea- tures, clinical behaviors, and treatment response profiles. 1-4 These intrinsic molecular subtypes were defined as: basal-like, human epidermal growth fac- tor receptor 2 (HER2) – enriched, and luminal A and B subtypes. 1 Additional studies separated luminal breast cancers into at least two subgroups: luminal A and luminal B. 2 Luminal A tumors were characterized as having the highest expression of estrogen-related and low expression of proliferation-related genes, 2 whereas luminal B cancers showed lower expression of estrogen receptor (ER) 2,3,5 as well as low ex- pression of progesterone receptor (PgR) genes 6 and higher expression of proliferation cluster genes (MKI67) and cell cycle–associated genes (CCNB1 and MYBL2). 7,8 Luminal B tumors showed increased expression of growth factor re- ceptor genes, with approximately 20% being HER2 positive by mRNA levels and immunohis- tochemistry (IHC). 9 Table 1 lists proposed surro- gate IHC definitions for the classification of luminal B breast cancer. 6,10-13 MOLECULAR CHARACTERIZATION OF LUMINAL B Luminal B breast cancer is also unique with regard to the profile of gene copy number alterations (CNAs), DNA methylation, and somatic point mutations. 14 High-level DNA amplifications and chromosomal aberrations were more frequently detected in lumi- nal B cancer than in the other subtypes (Fig 1). 15,16 Identifying recurrent genome CNAs in luminal B breast cancer led to the discovery of oncogenes that function as the molecular drivers of this subtype (eg, ZNF703). 17 This oncogene has been shown to in- duce cellular proliferation independently of estro- gen stimulation in cell lines 17 and is associated with worse clinical outcome. 18 It stimulates genes related to WNT and NOTCH signaling pathways, involved in regulating the self-renewal of breast tumor– initiating cells (TICs). 17 Similarly, when the SIX1 oncogene is expressed, it promotes breast TICs and is related to negative prognosis. 18,19 The METABRIC (Molecular Taxonomy of Breast Cancer International Consortium) study, in- tegrating genomic and transcriptomic profiling of 2,000 breast tumors, led to a refined breast cancer molecular taxonomy. 20 Ten molecular subtypes (defined as integrative clusters and labeled IntClusts 1 to 10) with different clinical behaviors were iden- tified. Luminal B breast cancers clustered in several JOURNAL OF CLINICAL ONCOLOGY R E V I E W A R T I C L E © 2014 by American Society of Clinical Oncology 1 http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2013.54.1870 The latest version is at Published Ahead of Print on July 21, 2014 as 10.1200/JCO.2013.54.1870 Copyright 2014 by American Society of Clinical Oncology 129.94.53.232 Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

Luminal B Breast Cancer: Molecular Characterization,Clinical Management, and Future PerspectivesFelipe Ades, Dimitrios Zardavas, Ivana Bozovic-Spasojevic, Lina Pugliano, Debora Fumagalli,Evandro de Azambuja, Giuseppe Viale, Christos Sotiriou, and Martine Piccart

Felipe Ades, Lina Pugliano, DeboraFumagalli, Evandro de Azambuja, Chris-tos Sotiriou, and Martine Piccart, Insti-tut Jules Bordet, Universite Libre deBruxelles; Dimitrios Zardavas, BreastInternational Group, Brussels, Belgium;Ivana Bozovic-Spasojevic, Institute forOncology and Radiology of Serbia,Belgrade, Serbia; and Giuseppe Viale,European Institute of Oncology, Univer-sity of Milan, Milan, Italy.

Published online ahead of print atwww.jco.org on July 21, 2014.

Authors’ disclosures of potential con-flicts of interest and author contribu-tions are found at the end of thisarticle.

Corresponding author: Martine Piccart, MD,PhD, Institut Jules Bordet, 121 Boulevardde Waterloo, Bruxelles, Belgium 1000;e-mail: [email protected].

© 2014 by American Society of ClinicalOncology

0732-183X/14/3299-1/$20.00

DOI: 10.1200/JCO.2013.54.1870

A B S T R A C T

Gene expression profiling has reshaped our understanding of breast cancer by defining andcharacterizing four main intrinsic molecular subtypes: human epidermal growth factor receptor2–enriched, basal-like, luminal A, and luminal B subtypes. Luminal B breast cancer has beenreported to have lower expression of hormone receptors, higher expression of proliferationmarkers, and higher histologic grade than luminal A. It also exhibits worse prognosis and has adistinct profile of response to hormone therapy and chemotherapy. Although luminal cancersshare similarities, the studies conducted in recent years using next-generation sequencingtechnology show that luminal A and B breast cancers should be perceived as distinct entities,with specific oncogenic drivers, rather than more proliferative varieties of luminal tumors. Thisreview discusses the definition and molecular characterization of luminal B breast cancer andpresents the available clinical evidence for chemotherapy and endocrine therapy patterns ofresponse. It also provides an overview of ongoing research on molecularly targeted agents forthis disease.

J Clin Oncol 32. © 2014 by American Society of Clinical Oncology

INTRODUCTION

In the early 2000s, high-throughput technology en-abled the interrogation of the expression of thou-sands of genes in a single experiment. This led to thedetermination that breast cancer comprises at leastfour molecularly distinct diseases with different fea-tures, clinical behaviors, and treatment responseprofiles.1-4 These intrinsic molecular subtypes weredefined as: basal-like, human epidermal growth fac-tor receptor 2 (HER2) –enriched, and luminal Aand B subtypes.1

Additional studies separated luminal breastcancers into at least two subgroups: luminal A andluminal B.2 Luminal A tumors were characterized ashaving the highest expression of estrogen-relatedand low expression of proliferation-related genes,2

whereas luminal B cancers showed lower expressionof estrogen receptor (ER)2,3,5 as well as low ex-pression of progesterone receptor (PgR) genes6

and higher expression of proliferation clustergenes (MKI67) and cell cycle–associated genes(CCNB1 and MYBL2).7,8 Luminal B tumorsshowed increased expression of growth factor re-ceptor genes, with approximately 20% beingHER2 positive by mRNA levels and immunohis-tochemistry (IHC).9 Table 1 lists proposed surro-gate IHC definitions for the classification ofluminal B breast cancer.6,10-13

MOLECULAR CHARACTERIZATION OFLUMINAL B

Luminal B breast cancer is also unique with regard tothe profile of gene copy number alterations (CNAs),DNA methylation, and somatic point mutations.14

High-level DNA amplifications and chromosomalaberrations were more frequently detected in lumi-nal B cancer than in the other subtypes (Fig 1).15,16

Identifying recurrent genome CNAs in luminal Bbreast cancer led to the discovery of oncogenes thatfunction as the molecular drivers of this subtype (eg,ZNF703).17 This oncogene has been shown to in-duce cellular proliferation independently of estro-gen stimulation in cell lines17 and is associated withworse clinical outcome.18 It stimulates genes relatedto WNT and NOTCH signaling pathways, involvedin regulating the self-renewal of breast tumor–initiating cells (TICs).17 Similarly, when the SIX1oncogene is expressed, it promotes breast TICs andis related to negative prognosis.18,19

The METABRIC (Molecular Taxonomy ofBreast Cancer International Consortium) study, in-tegrating genomic and transcriptomic profiling of2,000 breast tumors, led to a refined breast cancermolecular taxonomy.20 Ten molecular subtypes(defined as integrative clusters and labeled IntClusts1 to 10) with different clinical behaviors were iden-tified. Luminal B breast cancers clustered in several

JOURNAL OF CLINICAL ONCOLOGY R E V I E W A R T I C L E

© 2014 by American Society of Clinical Oncology 1

http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2013.54.1870The latest version is at Published Ahead of Print on July 21, 2014 as 10.1200/JCO.2013.54.1870

Copyright 2014 by American Society of Clinical Oncology129.94.53.232

Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 fromCopyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 2: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

IntClusts. IntClusts 1, 6, and 9 were enriched for luminal B cancerscharacterized by 17q23/20q cis-acting aberrations, 8p12 cis-acting ab-errations, and 8q cis-acting aberrations/20q amplifications, respec-tively. IntClust 2, which exhibited a high mortality rate, was enrichedin both luminal A and B cancers bearing cis-acting aberrations in the11q13/14 region, where several driver genes reside. Recurrent lossof transcript expression in PPP2R2A, a B-regulatory subunit of thePP2A mitotic exit holoenzyme complex, was observed in luminal Bcancers, a significant pathophysiologic molecular alteration forthis subtype.20

Next-generation sequencing (NGS) studies have helped toelucidate the molecular uniqueness of luminal B breast cancer. TheCancer Genome Atlas (TCGA) Network initiative characterized ahigh number of primary breast cancers using a wide variety of

platforms. This analysis provided an abundance of molecular char-acteristics distinguishing luminal B cancer from the other sub-types. Luminal A and luminal B breast cancers are distinct entities,albeit with overlap. An unsupervised clustering analysis of methyl-ation data showed that the group with a hypermethylated profilewas enriched for luminal B cancers. This group had also a lownumber of MAP3K1 and MAP2K4 mutations. It was shown thatluminal B cancers had lower frequency of PIK3CA mutations (29%v 45%) and higher frequency of TP53 mutations (29% v 12%) thanluminal A cancers (Fig 1).14 Another gene found to be mutated to asignificant degree in both luminal A and B cancers was GATA3,with a frequency of 14% and 15%, respectively. However, differenttypes of mutations were also identified for the two subtypes, withhotspot CA intron 4 deletions associated with luminal A cancerand exon 5 frame shifts associated with luminal B cancer.21 Of note,GATA3 mutations were shown to confer endocrine sensitivity inthe neoadjuvant setting and are positively correlated with Ki67decline on aromatase inhibition (P � .01).22

RUNX1 was identified through genome-wide analysis as a tran-scription factor involved in mediating ER genomic recruitment andregulating tethering genes.21-24 Loss-of-function mutations affectingthis gene, as well as its dimerization partner CBFB, could lead to aphenotype of endocrine resistance. RUNX1 deletion was associatedwith poorly differentiated breast cancer25; interestingly, pathway rep-resentation and analysis by direct reference on graphical models(PARADIGM) –inferred pathway analysis associated RUNX1 loss-of-function mutations with the luminal B subtype, as well as features ofendocrine resistance.22

A

B

Freq

uenc

y

90

80

70

60

50

40

20

30

10

0

Luminal ALuminal BHER2 enrichedBasal like

TP53 PIK3CA GATA3 MLL3 MAP3K1 CDH1 PTEN TBX3

Copy NumberAberrations

Luminal A Luminal B HER2 Enriched Basal Like

Increased copy number:

1q and 16p 1q, 8q, 17q, and 20q

1q, 7p, 8q, 16p, and 20q

3q, 8q, and 10p

Decreased copynumber:

16q 1p, 8p, 13q, 16q, 17p, and 22q

1p, 8p, 13q, and 18q

3p, 4p, 4q, 5q, 12q, 13q, 14q, and 15q

High-levelamplifications:

8p11-12, 11q13-14, 12q13-14, 17q11-12, 17q21-24, and 20q13

8p11-12, 8q, and 11q13-14

17q Rare Fig 1. Percentage of somatic mutationsand molecular alterations across differentintrinsic subtypes of breast cancer. (A)Reported copy number alteration acrossdifferent breast cancer intrinsic subtypes.(B) Frequencies of the most commonlymutated cancer-related genes across dif-ferent breast cancer intrinsic subtypes.HER2, human epidermal growth factorreceptor 2.

Table 1. Proposed Surrogate IHC Markers for Classification of Luminal BBreast Cancer

StudyER

StatusPgR

Status (%)HER2Status

Ki67Level (%)

Blows et al10� Positive Positive PositiveCheang et al11� Positive Positive Negative � 13.25Prat et al6 Positive � 20 Negative � 14Goldhirsch et al12� Positive Positive Negative � 14Harbeck et al13� Positive Positive Negative � 20 to 25

Abbreviations: ER, estrogen receptor; HER2, human epidermal growthfactor receptor 2; IHC, immunohistochemistry; PgR, progesteronereceptor.

�ER positive and/or PgR positive.

Ades et al

2 © 2014 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 3: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

LUMINAL B DEFINITION IN CLINICAL PRACTICE

Subtype Definition Using Gene Expression Classifiers

Several molecular signatures have been developed in an effort topredict the molecular subtype in clinical practice, but their perfor-mance is controversial, and their use in daily practice is limited be-cause of the relatively high cost and technical complexity. One of thesemolecular signatures, prediction of microarray (PAM) 50, is gener-ated from an expanded intrinsic set of 1,906 genes derived from fourmicroarray studies.2,3,8,26 After applying several minimization meth-ods, 50 genes involved in proliferation, or related to ER, HER2,myoepithelial, and basal characteristics, were selected. The PAMmethod was used to generate the algorithm. The original signature hasbeen implemented in a clinical assay (Prosigna) based on NanoString(Seattle, WA) technology. Because this technology allows the use ofmaterial extracted from formalin-fixed paraffin-embedded samples,the test can be widely applied in the clinical setting. This assay is able toidentify the same intrinsic subtypes as the original work,27 and itsprognostic value has been validated in two studies.28,29

Surrogate Histologic and IHC Markers

In clinical practice, luminal B cancers are differentiated fromluminal A cancers based on proliferation markers.9,11,30 ER and HER2expression show a bimodal distribution with clear cutoff points forassessment.31 However, proliferation is determined by several geneswith a continuous distribution; thus, its status cannot be identifieddichotomously. Therefore, establishing a cutoff to determine high andlow proliferative tumors in the clinical setting remains a challenge.

Proliferation status has been assessed in clinical practice by dif-ferent techniques, with tumor grade being the most widely used.30

Nonetheless, technical issues limit its use as a surrogate marker. In onestudy of 675 breast cancer samples, the concordance rate for gradeclassification among three different pathologists was as low as 43%.32

Ki67 is a nuclear marker expressed in all cell-cycle phases exceptG0.33 It is evaluated by IHC, with results expressed in a quantitativeway, meaning the percentage of stained cells. Important intratumorvariation occurs, depending on the tumor section analyzed and thepathologist evaluation.34 In a meta-analysis including 46 studies and12,155 patients, high Ki67 labeling was correlated with an increasedrisk of relapse and worse survival. The threshold for Ki67 in thesestudies varied between 3.5% and 35%, and the criteria to define thethreshold were diverse.35 In a study aimed at defining a Ki67 cutoff,tumors of 357 patients were profiled by PAM50 and compared withKi67 results; 101 breast cancers were classified as luminal A and 69 asluminal B. The optimal Ki67 level discriminating luminal A from Bwas 13.25%. This cutoff point was rounded up to 14%, and recom-mended as a surrogate marker for luminal cancer classification. Thehigh rate of misclassification, occurring in 25% of the cases,11 is worthnoting. The 14% level was endorsed by the 2011 St Gallen Early BreastCancer Conference consensus panel.12 In 2013, major disagreementsemerged among the panelists; the majority voted for a cutoff of� 20%, whereas others recommended a lower, local laboratory–specific cutoff or the use of multigene expression assay.13

PgR status has also been used to define luminal B cancer. PgRnegativity is a marker of worse prognosis in luminal cancers36,37; inbreast tumors assessed as ER positive and having Ki67 � 14%, loss ofPgR was identified as an adverse prognostic factor (relapse: hazard

ratio [HR], 1.96; 95% CI, 1.44 to 2.68).38 In another study, luminal Bcancers classified as ER positive, HER2 negative, Ki67 � 14%, andPgR � 20% had worse prognosis than tumors with PgR � 20%.6

Despite the multiple attempts to establish a clinically useful,IHC-based luminal B surrogate marker, efforts so far have failed toaccurately reproduce the PAM50 classification39 or to define a goldstandard, which in large part stems from the controversies about themost reliable way to quantify proliferation.

Prognostic Signatures Within Intrinsic Subtypes

Gene expression signatures are used to provide prognostic infor-mation beyond that provided by the clinicopathologic parameters andare particularly useful in the discrimination of luminal breast cancers.Most luminal A tumors are classified at low genomic risk, whereasluminal B cancers are often classified at high genomic risk40-42 (Table2). Several gene expression profiling (GEP) prognostic signatures havebeen developed with little overlap between their constituent genes.Despite this, a significant overlap in their performance is observed.40

In a meta-analysis of publicly available breast cancer gene expressionand clinical data, gene coexpression modules related to proliferation,ER, and HER2 were used to evaluate the role of constituent genes. Thestudy identified 524 genes associated with survival; of these, 71% werestrongly coexpressed with proliferation, 26% with ER, and 2.2% withHER2,9 revealing the importance of proliferation genes as a commondriving force behind signature performance.

Actually, these multigene prognosticators do not really identifydifferent risk groups within intrinsic subtypes; they rather dividebreast tumors according to their proliferation pattern. Because HER2-enriched and basal-like cancers are almost all high proliferative can-cers, GEP prognostic signatures add little information in thesesubtypes. However, in luminal breast cancers, the signatures are ableto identify low and high proliferative groups, roughly correlated withthe luminal A and luminal B subtypes.9,40,43

Table 2. Correlation Between Intrinsic Subtype Classification by PAM50 andOncotypeDX and MammaPrint Prognostic Signatures

PAM50Classification

TotalPatients

OncotypeDX MammaPrint

ClassificationNo. of

Patients ClassificationNo. of

Patients

Luminal A 121Low 68 Good 87Intermediate 13High 40 Poor 34

Luminal B 55Low 2 Good 9Intermediate 2High 51 Poor 46

HER2 positive/ER negative

18Low 1 Good 2Intermediate 0High 17 Poor 16

Basal like 7Low 1 Good 0Intermediate 1High 5 Poor 7

NOTE. Data adapted.40

Abbreviations: ER, estrogen receptor; HER2, human epidermal growth factorreceptor 2; PAM, prediction of microarray.

Luminal B Breast Cancer

www.jco.org © 2014 by American Society of Clinical Oncology 3

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 4: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

A revealing study compared 47 published breast cancer signa-tures with 1,000 randomly generated signatures composed of the samenumber of genes. Not only did � 90% of the random signaturespredict outcome, but 60% of the random signatures also performedbetter than the original ones.44 This strengthens the notion that prog-nostic signatures do not identify specific mechanisms of cancer aggres-siveness.9 Instead, because a large number of transcripts deregulated inbreast cancer correlate with proliferation genes, random signatures arealso proliferation related and able to discriminate prognosis.

CLINICAL BEHAVIOR

Luminal B breast cancer is recognized as having an aggressive clinicalbehavior, with prognosis similar to that of HER2-enriched and basal-like groups, whereas luminal A breast cancer is identified as having amore favorable clinical outcome.2-4,8 Luminal B cancers show in-creased relapse rates in the first 5 years after diagnosis, decreasing overtime, and a metastatic dissemination time pattern similar to basal-likeand HER2-enriched cancers.45 In a series of 831 untreated node-negative patients, the HR for early metastasis (� 5 years) was 2.86; forlate metastasis (� 5 years), it was 0.65 in comparison with luminalA.45,46 Moreover, luminal B cancers have a distinctive pattern of sitedissemination, with predilection for bone and to a lesser degree forlung. This is in contrast with luminal A breast cancer, for which boneis also the principal metastatic site, although it shows a low frequencyof metastasis to other sites.47

LUMINAL B AND BENEFIT OF CHEMOTHERAPY

Most luminal B cancers are classified as having a high OncotypeDXrecurrence score (RS).40 The NSABP (National Surgical AdjuvantBreast and Bowel Project) B-20 study assessed node-negative ER-positive patients randomly assigned to tamoxifen versus tamoxifen

plus chemotherapy. Patients with high RS (� 31) derived large benefitfrom chemotherapy (recurrence: HR, 0.26; 95% CI, 0.13 to 0.53), witha mean absolute decrease in distant recurrence rate of 27.6%.48 In theSWOG (Southwest Oncology Group) 8814 trial, which randomlyassigned node-positive ER-positive patients to tamoxifen versus ta-moxifen followed by chemotherapy, a chemotherapy benefit was alsoobserved in patients with high RS (� 31; disease-free survival [DFS]:HR, 0.59; 95% CI, 0.35 to 1.01), whereas no improvement was seen inpatients with low RS (� 18).49 However, patients with high expressionof ER did not benefit from addition of chemotherapy, irrespective ofOncotypeDX results. RS was also positively correlated with the likeli-hood of pathologic complete response (pCR; P � .005), indicatinggreater sensitivity to neoadjuvant chemotherapy.50

In neoadjuvant studies, luminal tumors exhibit lower pCR ratesthan HER2-enriched and triple-negative breast cancers51-58 (Table 3).When comparing the luminal tumors, results are conflicting; higherpCR rates in luminal B than in luminal A cancers were not homoge-neously observed.53,58 Nonetheless, neoadjuvant chemotherapyseems effective in reducing tumor volume in luminal B cancers (Table3). Tumors with high prognostic signature values, according to severalGEP classifiers, are associated with a higher probability of pCR.59

The latest update from the Early Breast Cancer Trialists’ Collab-orative Group (EBCTCG) was unable to ascertain chemosensitivity inluminal tumors or benefit from specific chemotherapies, because oflimited data to define luminal tumors. Poor differentiation, defined bylocal pathologists, was not predictive of chemosensitivity in patientswith ER-positive disease.60 The St Gallen consensus panel consideredthat both anthracyclines and taxanes should be included in the chem-otherapy regimens for luminal B breast cancer.12

Three prospective randomized trials—MINDACT (Microarray inNode-Negative Disease May Avoid Chemotherapy; NCT00433589),TAILORx (Trial Assigning Individualized Options for Treatment;NCT00310180), and RxPONDER (Treatment for Positive Node,

Table 3. pCR After Neoadjuvant Chemotherapy in Luminal B Breast Cancer

TrialNo. of

Patients ChemotherapyMethod of Luminal

B ClassificationTechnique Used for

Classification

pCR (%)

LuminalA

LuminalB

HER2Positive

BasalLike

Rouzier et al51 82 P-FAC Molecular Intrinsic gene set andhierarchic clustering

7� 7� 45 45

Carey et al52 107 AC plus taxane IHC ER or PgR positive, HER2positive

0 15 36 27

de Ronde et al53 191 Dose-dense AC or DX Molecular Intrinsic gene set andhierarchic clustering

1 4 54 67

Esserman et al54 144 AC-P Molecular Intrinsic gene set andhierarchic clustering

5 13 55 34

Bhargava et al55 359 Anthracycline or taxane IHC ER positive, PgR positive,HER2 negative

1.8 1.4 33.3 30.4

Lv et al56 104 FEC, DC, or DE IHC ER positive, PgR positive,HER2 negative, Ki67� 14%

0 8.7 22.2 24.4

Straver et al57 167 AC or AD IHC and molecular ER positive, HER2 negative,70-gene MammaPrint

3� 3� 32 34

Lips et al58 211 Dose-dense AC or DX Molecular PAM50 1.3 4.4 — —

Abbreviations: AC, doxorubicin plus cyclophosphamide; AC-P, doxorubicin, cyclophosphamide, and paclitaxel; AD, doxorubicin plus docetaxel; DC, docetaxel pluscarboplatin; DE, docetaxel plus epirubicin; DX, docetaxel plus capecitabine; ER, estrogen receptor; FEC, fluorouracil, epirubicin, and cyclophosphamide; HER2,human epidermal growth factor receptor 2; IHC, immunohistochemistry; PAM, prediction of microarray; pCR, pathologic complete response; P-FAC, paclitaxel,fluorouracil, doxorubicin, and cyclophosphamide; PgR, progesterone receptor.

�Luminal A and B not discriminated.

Ades et al

4 © 2014 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 5: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

Endocrine Responsive Breast Cancer; NCT01272037)—are testingthe usefulness of gene signatures in predicting benefit from adju-vant chemotherapy in patients with ER-positive breast cancer.61-63

Results are expected between 2015 and 2017.

LUMINAL B AND BENEFIT OF ENDOCRINE THERAPY

The supporting evidence for the relatively lower benefit of endocrinetherapy in the luminal B subtype comes from few studies, which, aswas the case for chemotherapy, classify luminal tumors through sur-rogate markers. An EBCTCG meta-analysis evaluating patients en-rolled onto endocrine therapy trials showed small benefits withtamoxifen in patients with low ER levels.64 In the BIG (Breast Interna-tional Group) 1-98 trial, which assigned 8,010 patients to four treat-ment arms comparing different sequential administrations ofletrozole and tamoxifen, patients with lower ER levels had worse DFSthan those with high ER levels.65 Patients receiving tamoxifen or noadjuvant systemic treatment with an ER-positive/PgR-negative phe-notype had better outcomes than their ER-negative/PgR-negativecounterparts but worse outcomes than those with the ER-positive/PgR-positive phenotype.36 Nevertheless, this observation was notconfirmed in either in the NSABP B-14 study66 or in the IES (Inter-group Exemestane Study) comparing exemestane versus tamoxifen.67

The ATAC (Arimidex, Tamoxifen, Alone or in Combination)trial evaluated 5 years of anastrozole, tamoxifen, or their combinationin postmenopausal women; high OncotypeDX RS (� 31) was associ-ated with disease relapse when compared with low RS (� 18).68 A78-gene signature69 showed results similar to RS in evaluating thetreatment prognosis in tamoxifen-naive patients with metastaticbreast cancer.70 However, in the BCIRG (Breast Cancer InternationalResearch Group) study, comparing the combination of docetaxel orfluorouracil with doxorubicin and cyclophosphamide, both the lumi-nal A and luminal B subtypes showed significant responses to tamox-ifen (luminal B: HR, 0.44; 95% CI, 0.32 to 0.61; P � .001; luminal A:HR, 0.15; 95% CI, 0.07 to 0.33; P � .001).71

In contrast to the previously discussed markers that provide onlyprognostic information, dynamic changes in the expression of Ki67were able to predict outcome in luminal breast cancer treated withendocrine therapy. In the IMPACT (Immediate Preoperative Anas-trozole, Tamoxifen, or Combined With Tamoxifen) trial, patientswith higher Ki67 expression after 2 weeks of endocrine therapy hadlower recurrence-free survival (P � .004).72 In the P024 neoadjuvantendocrine therapy trial, baseline Ki67 was associated with higher riskof relapse (HR, 1.3; 95% CI, 1.05 to 1.50; P � .01).73 Luminal B and Atumors showed similar proportional falls in Ki67 in a neoadjuvant trialwith anastrozole; however, patients with luminal B cancers had apoorer outcome, explained by the higher baseline levels of Ki67.74

Using clinicopathologic (tumor size, node status, treatment re-sponse) and IHC information (ER status, Ki67, histologic grade), apreoperative endocrine prognostic index (PEPI) was developed froma cohort of patients in the P024 trial. This score was independentlyvalidated in the IMPACT trial, being able to correctly predict relapse-free survival.75 The PEPI score was also evaluated in the ACOSOG(American College of Surgeons Oncology Group) Z1031 trial check-ing for the effect of neoadjuvant anastrozole, letrozole, and exemes-tane in postmenopausal patients. Luminal B cancers had higher Ki67at baseline (P � .001), post–aromatase inhibitor treatment (P � .001),

and greater change in Ki67 (P � .001). No clinically relevant responsedifferences were found between treatment results among luminaltumors (HR, 1.67; 95% CI, 0.96 to 2.9; P � .07), nor in the rate ofbreast-conserving surgery, suggesting similar clinical efficacy (HR,0.75; 95% CI, 0.29 to 1.93; P � .54) despite the better PEPI scoreamong the luminal A group (27.1% v 10.7%; P � .004).76 In conclu-sion, both luminal A and B tumors derive benefit from endocrinetreatment, although the magnitude of benefit is larger in luminal A.

In the ATAC trial, ER-positive/PgR-negative patients obtainedlarger relative benefit from anastrozole than from tamoxifen (HR,0.42; 95% CI, 0.31 to 0.61), whereas ER-positive/PgR-positive patientsshowed no differences (HR, 0.84; 95% CI, 0.69 to 1.02).77 The relativereduction of risk was similar for anastrozole and tamoxifen amongpatients with different OncotypeDX RS values.68 In BIG 1-98, highKi67 predicted greater efficacy of letrozole over tamoxifen (HR, 0.53;95% CI, 0.39 to 0.72), whereas low Ki67 indicated no difference (HR,0.81; 95% CI, 0.56 to 1.15; P � .09).78 Nevertheless, using the surro-gate luminal B definition proposed by Cheang et al,11 no differencewas seen between tamoxifen and letrozole.78

In a neoadjuvant study randomly assigning postmenopausal ER-positive patients to letrozole or tamoxifen, letrozole was more effectivein tumors overexpressing epidermal growth factor receptor or HER2(clinical response, 88% v 21%; P � .001).79 In IMPACT, the responserate for tamoxifen was 22%, whereas for anastrozole, it was 58% inpatients presenting HER2 amplification or overexpression (HR, 4.9;95% CI, 0.53 to 63.22; P � .18).80 The same trend was observed in theBIG 1-98 trial; patients receiving letrozole had better DFS comparedwith those receiving sequential administrations of tamoxifen andletrozole or tamoxifen alone (P � .58 and .87, respectively).78 In ameta-analysis in patients with advanced ER-positive breast cancer,HER2-positive overexpression was identified as increasing the riskof disease recurrence (relative risk [RR], 1.42; 95% CI, 1.32 to 1.52;P � .001), independently from the type of hormone treatment(tamoxifen: RR, 1.33; 95% CI, 1.20 to 1.48; P � .001; other agents:RR, 1.49, 95% CI; 1.36 to 1.64), suggesting no additional benefitfrom aromatase inhibitors.81

Although some studies reported better outcomes with aromataseinhibitors over tamoxifen, these results were not homogeneously ob-served. These divergences do not allow the drawing of definitive con-clusions about the best endocrine agent in patients with luminal Bcancer; both choices are acceptable.

DRUG DEVELOPMENT IN LUMINAL B BREAST CANCER

Many of the mutated genes found in breast cancer occur at a lowfrequency, rendering their therapeutic exploitation difficult. This re-quests extensive molecular screening efforts to identify these pa-tients.82 In addition, functional characterization of these molecularevents is needed before they are pursued as therapeutic targets. Find-ings from high-dimension, clinically and molecularly annotatedbreast cancer data sets and functional genomic preclinical studies canlead to the development of solid hypotheses on predictive biomarkers,which can be tested prospectively through genotype-driven clinicaltrials.83 Figure 2 shows an overview of the signaling pathways underblockade with targeted compounds in luminal breast cancers.

Luminal B Breast Cancer

www.jco.org © 2014 by American Society of Clinical Oncology 5

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 6: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

Phosphatidylinositol-3-Kinase/AKT/Mammalian Target

of Rapamycin Signaling Pathway

In luminal breast cancer, phosphatidylinositol-3-kinase (PI3K)activation is implicated in de novo and acquired endocrineresistance.75,84-88 Despite lower frequency of PIK3CA mutations,14

luminal B cancers have higher PI3K activation than luminal A.89

PIK3CA mutations do not seem to be associated with PI3K pathwayactivation in early-stage ER-positive breast cancer; a PIK3CA muta-tion gene signature was identified as a positive prognosticator in thissetting.90 This gene signature indicates a phenotype of low mamma-lian target of rapamycin (mTOR) C1 signaling, suggesting thatPIK3CA mutations might act as weak PI3K pathway activators.PIK3CA mutations were associated with sensitivity to tamoxifen invitro,90 but it remains to be shown whether the strategy of combiningPI3K blocking with endocrine therapy will improve clinical outcome.

The most advanced compounds in clinical development arerapamycin analogs called mTOR inhibitors. The combination ofeverolimus and letrozole has been tested in the metastatic91 and neo-adjuvant settings,92 achieving positive results. The combination ofeverolimus with tamoxifen led to prolongation of progression-freesurvival (PFS; 8.6 v 4.5 months; P � .002) and overall survival (OS; notreached v 24.4 months; P � .01) in patients with aromatase inhibitor–pretreated metastatic disease.93 The BOLERO-2 (Breast Cancer Trialsfor Oral Everolimus) study randomly assigned patients with meta-static disease, pretreated with nonsteroidal aromatase inhibitors, toreceive everolimus combined with exemestane versus exemestane andplacebo.94 The study showed prolongation of PFS for the everolimusplus exemestane arm (PFS, 10.6 v 4.1 months; HR, 0.36; 95% CI, 0.27to 0.47; P � .001); OS data are still immature.94

No predictive biomarkers were identified to guide patient selec-tion for PI3K-blocking agents.95 In BOLERO-2, the four most fre-

quently mutated genes were PIK3CA (48%), CCND1 (31%), TP53(23%), and FGFR1 (18%), with the benefit of everolimus being similaracross these subgroups. Patients with zero or one genetic alteration inPI3K or fibroblast growth factor receptor (FGFR) pathways, or withCCND1 mutations, had increased benefit from treatment witheverolimus (HR, 0.27; 95% CI, 0.18 to 0.41). However, it should benoted that to date, no analysis has been conducted discriminatingbetween luminal A and B subtypes.

In a recent study, a PIK3CA mutation gene signature was foundto be predictive for Ki67 reduction in patients receiving letrozole witheverolimus in the neoadjuvant setting.96 In the TAMRAD (TamoxifenPlus Everolimus) study, low expression of LKB1, an inhibitor ofmTOR activation, was associated with improved clinical outcomeunder everolimus plus tamoxifen treatment.97

Despite positive results, rapalogs provide suboptimal PI3K path-way inhibition, because they block only mTORC1,98 causing down-regulation of inhibitory feedback mechanisms.99 Emphasis is nowbeing placed on the development of specific PI3K-targeted agents100

(Appendix Table A1, online only).

FGF Signaling Pathway

FGFR1 amplification101 was identified in 16% to 27% of luminalB breast cancers as a mechanism of endocrine resistance102 and asbeing associated with negative prognosis.103 The administration ofE-3810, a nonselective FGF-blocking agent, to 10 patients with meta-static breast cancer resulted in seven partial responses.104 A phase IIclinical trial assessed dovitinib, another nonselective inhibitor, inheavily pretreated patients with FGFR1-amplified luminal metastaticbreast cancer; it produced three partial responses and resulted in stabledisease in nine patients.105 Other studies are assessing the addition of

p85 p110IRS1

PIP3PIP2

S6K

rpS6

MDM2

p53 BAD FasL,Bim

p27Kip1p21

Cyclin D1

GSK3 TSC1/2

Akt mTOR-rictor

PTEN

FOXO

Src

PI3K

4E-BP1

eIF4E

Cell Survival Proliferation Metabolism Protein Synthesis

PDK1

RTKs: FGF-R, IGF-R

RAS

RAF

MEK

ERK

SOS

Grb2

PI3K Inhibitors FGF InhibitorsIGF Inhibitors CDK Inhibitors

Dalotuzumab,BMS-754807,MEDI-573

BKM120, GDC0941,GDC0032, GDC0980, XL765, BYL719, MK2206, AZD2014

PD0332991, LEE011,LY2835219

Dovitinib,AZD4547

mTOR-raptor

Fig 2. Signaling pathways under block-ade with target compounds in luminalbreast cancer. CDK, cycle-dependent ki-nase; FGF, fibroblast growth factor;FGF-R, fibroblast growth factor receptor;IGF, insulin-like growth factor; IGF-R,insulin-like growth factor receptor; mTOR,mammalian target of rapamycin; PI3K,phosphatidylinositol-3-kinase; RTK, recep-tor tyrosine kinase.

Ades et al

6 © 2014 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 7: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

FGF-blocking agents to endocrine therapy (Appendix Table A1, on-line only).

Insulin-Like Growth Factor Signaling Pathway

The insulin-like growth factor (IGF) signaling pathway causesactivation of the PI3K/AKT/mTOR and Ras/Raf/MEK/ERK path-ways.106 In luminal cancers, there is IGF-ER crosstalk; estrogen up-regulates IGF-1 and IGF-1 receptor (IGF-1R),107 and IGF-1 signalingboosts the proliferative effect of estrogen108 and is implicated in endo-crine resistance.109 Tamoxifen resistance has been suppressed with ananti–IGF-1R monoclonal antibody,110 and IGF blocking has synergis-tic effect with endocrine treatment in luminal breast cancers.111,112

A randomized phase II study testing ganitumab or placebo withexemestane or fulvestrant in postmenopausal women with metastatichormone receptor–positive disease was unable to demonstrate benefitfrom this strategy (PFS: HR, 1.17; 95% CI, 0.91 to 1.50; P � .44).113

However, a promising approach is the combination of mTOR andIGF inhibitors,114 acting synergistically through the downregulationof IGF-1R–mediated AKT activation induced by mTOR inhibition115

(Appendix Table A1, online only).

Cyclin-Dependent Kinases

Cycle-dependent kinase (CDK) deregulation confers a highlyproliferative cellular phenotype and is also linked to endocrine resis-tance.116 High expression of cyclin D1117,118 and E1119 is associatedwith poor clinical outcome in tamoxifen-treated women. Cyclin E2expression is characteristic of luminal B cancer and correlated withshorter distant metastasis–free survival.120 CCND1 (encoding gene forcyclin D1) amplification is found at higher frequencies in luminal Bcompared with luminal A cancer (58% v 29%).21 Furthermore, theMETABRIC study identified a bad prognosis molecular subtype ofluminal breast cancer, where CCND1 is located.20

PD-0332991, a highly specific CDK4 and CDK6 inhibitor, wasassessed in a phase II study randomly assigning patients with hormonereceptor–positive disease to the combination of PD-0332991 andletrozole versus letrozole alone.121 The combination caused significantprolongation of PFS (median PFS, 18.2 v 5.7 months; HR, 0.35; 95%CI, 0.17 to 0.72; P � .006). A phase III trial is ongoing. The patientpopulation in this trial consisted of two groups: a cohort of 66 patientsselected according to ER and HER2 status, and a cohort of 99 patientsselected according to CCND1 amplification and/or loss of p16. Thebenefit of the addition of PD-0332991 to letrozole occurred mainly inthe biomarker-negative population, in contrast with initial assump-tions (HR, 0.37 and 0.19, respectively). This is partially explained bythe excessively negative prognosis associated with CCND1 amplifica-tion.20 However, it indicates that patients with both luminal A and Bcancers will probably benefit from this strategy.

ADDITIONAL THERAPEUTIC TARGETS

TP53 is one of the most commonly mutated breast cancer genes.Luminal B cancers exhibit higher mutation rates than luminal Acancers (32% v 12%).21 Higher rates of MDM2 gene amplification, aTP53 antagonist, have been observed in luminal B (32%) than inluminal A cancers (14%).21 Thus, luminal B cancers exhibit increasedrates of TP53 pathway loss of function, which has been identified as amediator of endocrine resistance.22 Currently, a new class of targeted

agents functioning as MDM2 antagonists is under clinical assess-ment for patients with advanced solid tumors (NCT01462175,NCT01877382, and NCT01664000), aiming to restore wild-typeTP53 function.

The mixed-lineage leukemia gene MLL3 is also a frequentlymutated gene in luminal cancers.21,22,123 MLL is a family of histonetrimethyltransferases that can regulate gene transcription, includingregulation of ESR1 expression.124 Currently, there is a group of tar-geted agents under clinical development blocking epigenetic regula-tion, with histone deacetylase (HDAC) inhibitors being the mostadvanced class of compounds.125 At the preclinical level, HDAC in-hibitors reactivate gene expression of ESR1 and PGR, whereas whenthey were combined with DNA methyltranferase inhibitors, enhancedESR1 gene expression was induced.126,127 At the clinical level, theresults of a randomized phase II trial assessing exemestane with orwithout entinostat (HDAC inhibitor) in patients with ER-positivemetastatic breast cancer show that the combination significantly pro-longed median PFS (4.3 v 2.3 months; HR, 0.73; P � .055) and OS(28.1 v 19.8 months; HR, 0.59; P � .036).126

DISCUSSION

GEP has changed the way breast cancer is perceived. Rather than anaggressive variant of hormone receptor–positive tumors, the lumi-nal B subtype is a distinct entity. However, the application of GEPof luminal B disease in clinical practice has been limited, largelybecause this classification coincides with traditional characteriza-tion by IHC.

So far, the molecular definition of luminal B disease has also beenof limited use in tailoring breast cancer treatment. Despite the lowersensitivity of luminal B cancers to endocrine treatment, benefit is stillderived from these drugs, and the simple identification of hormonereceptor positivity by IHC is enough to justify its use. Therefore, themain clinical challenge relates to the selection of patients who willbenefit from adjuvant chemotherapy. Proliferation was identified as amain predictive marker for cytotoxic chemotherapy, because it is alsoa major discriminant between luminal B and luminal A subtypes.However, with proliferation being a continuous process, defining lowand high cutoffs poses an important research question. To date, nochemotherapy predictive biomarker has been validated. However, it ishoped that three ongoing prospective randomized trials testing prog-nostic gene signatures—MINDACT, TAILORx, and RxPONDER—will shed light on this issue.

Finally, after little more than a decade since the discovery ofbreast cancer intrinsic subtypes, the characterization of importantpathways and molecular alterations is leading to a new portfolio oftargeted agents for luminal tumors. Genotype-driven trials are alreadya reality in breast cancer research and will continue to contribute to theimprovement of treatments for patients with luminal B breast cancer.

AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTSOF INTEREST

Although all authors completed the disclosure declaration, the followingauthor(s) and/or an author’s immediate family member(s) indicated afinancial or other interest that is relevant to the subject matter underconsideration in this article. Certain relationships marked with a “U” are

Luminal B Breast Cancer

www.jco.org © 2014 by American Society of Clinical Oncology 7

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 8: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

those for which no compensation was received; those relationships markedwith a “C” were compensated. For a detailed description of the disclosurecategories, or for more information about ASCO’s conflict of interest policy,please refer to the Author Disclosure Declaration and the Disclosures ofPotential Conflicts of Interest section in Information for Contributors.Employment or Leadership Position: None Consultant or AdvisoryRole: Martine Piccart, sanofi-aventis (C), Amgen (C), Roche (C),GlaxoSmithKline (C), PharmaMar (C) Stock Ownership: NoneHonoraria: Evandro de Azambuja, Roche, GlaxoSmithKline; MartinePiccart, sanofi-aventis, Amgen, Roche, GlaxoSmithKline, PharmaMarResearch Funding: None Expert Testimony: None Patents, Royalties,

and Licenses: Christos Sotiriou, Gene Expression Grade Index (GGI)Other Remuneration: Evandro de Azambuja, Roche, GlaxoSmithKline

AUTHOR CONTRIBUTIONS

Conception and design: All authorsCollection and assembly of data: All authorsData analysis and interpretation: All authorsManuscript writing: All authorsFinal approval of manuscript: All authors

REFERENCES

1. Perou CM, Sørlie T, Eisen MB, et al: Molec-ular portraits of human breast tumours. Nature 406:747-752, 2000

2. Sørlie T, Perou CM, Tibshirani R, et al: Geneexpression patterns of breast carcinomas distin-guish tumor subclasses with clinical implications.Proc Natl Acad Sci U S A 98:10869-10874, 2001

3. Sorlie T, Tibshirani R, Parker J, et al: Re-peated observation of breast tumor subtypes inindependent gene expression data sets. Proc NatlAcad Sci U S A 100:8418-8423, 2003

4. Sotiriou C, Neo SY, McShane LM, et al:Breast cancer classification and prognosis based ongene expression profiles from a population-basedstudy. Proc Natl Acad Sci U S A 100:10393-10398,2003

5. Brenton JD, Carey LA, Ahmed AA, et al:Molecular classification and molecular forecasting ofbreast cancer: Ready for clinical application? J ClinOncol 23:7350-7360, 2005

6. Prat A, Cheang MCU, Martın M, et al: Prog-nostic significance of progesterone receptor–positivetumor cells within immunohistochemically defined lu-minal A breast cancer. J Clin Oncol 31:203-209, 2013

7. Perou CM, Jeffrey SS, van de Rijn M, et al:Distinctive gene expression patterns in humanmammary epithelial cells and breast cancers. ProcNatl Acad Sci U S A 96:9212-9217, 1999

8. Hu Z, Fan C, Oh DS, et al: The molecularportraits of breast tumors are conserved acrossmicroarray platforms. BMC Genomics 7:96, 2006

9. Wirapati P, Sotiriou C, Kunkel S, et al: Meta-analysis of gene expression profiles in breast can-cer: Toward a unified understanding of breastcancer subtyping and prognosis signatures. BreastCancer Res 10:R65, 2008

10. Blows FM, Driver KE, Schmidt MK, et al:Subtyping of breast cancer by immunohistochemis-try to investigate a relationship between subtypeand short and long term survival: A collaborativeanalysis of data for 10,159 cases from 12 studies.PLoS Med 7:e1000279, 2010

11. Cheang MC, Chia SK, Voduc D, et al: Ki67index, HER2 status, and prognosis of patients withluminal B breast cancer. J Natl Cancer Inst 101:736-750, 2009

12. Goldhirsch A, Wood WC, Coates AS, et al:Strategies for subtypes:d Dealing with the diversityof breast cancer—Highlights of the St. Gallen Inter-national Expert Consensus on the Primary Therapyof Early Breast Cancer 2011. Ann Oncol 22:1736-1747, 2011

13. Harbeck N, Thomssen C, Gnant M: St. Gallen2013: Brief preliminary summary of the consensusdiscussion. Breast Care (Basel) 8:102-109, 2013

14. Weinstein JN, Collisson EA, Mills GB, et al:The Cancer Genome Atlas pan-cancer analysis proj-ect. Nat Genet 45:1113-1120, 2013

15. Bergamaschi A, Kim YH, Wang P, et al:Distinct patterns of DNA copy number alteration areassociated with different clinicopathological fea-tures and gene-expression subtypes of breast can-cer. Genes Chromosomes Cancer 45:1033-1040,2006

16. Chin K, DeVries S, Fridlyand J, et al: Genomicand transcriptional aberrations linked to breast can-cer pathophysiologies. Cancer Cell 10:529-541,2006

17. Sircoulomb F, Nicolas N, Ferrari A, et al:ZNF703 gene amplification at 8p12 specifies luminalB breast cancer. EMBO Mol Med 3:153-166, 2011

18. Holland DG, Burleigh A, Git A, et al: ZNF703is a common luminal B breast cancer oncogene thatdifferentially regulates luminal and basal progenitorsin human mammary epithelium. EMBO Mol Med3:167-180, 2011

19. Iwanaga R, Wang CA, Micalizzi DS, et al:Expression of Six1 in luminal breast cancers predictspoor prognosis and promotes increases in tumorinitiating cells by activation of extracellular signal-regulated kinase and transforming growth factor-beta signaling pathways. Breast Cancer Res 14:R100, 2012

20. Curtis C, Shah SP, Chin SF, et al: Thegenomic and transcriptomic architecture of 2,000breast tumours reveals novel subgroups. Nature486:346-352, 2012

21. Cancer Genome Atlas Network: Comprehen-sive molecular portraits of human breast tumours.Nature 490:61-70, 2012

22. Ellis MJ, Ding L, Shen D, et al: Whole-genome analysis informs breast cancer response toaromatase inhibition. Nature 486:353-360, 2012

23. Banerji S, Cibulskis K, Rangel-Escareno C, etal: Sequence analysis of mutations and transloca-tions across breast cancer subtypes. Nature 486:405-409, 2012

24. Stender JD, Kim K, Charn TH, et al: Genome-wide analysis of estrogen receptor alpha DNA bind-ing and tethering mechanisms identifies Runx1 as anovel tethering factor in receptor-mediated tran-scriptional activation. Mol Cell Biol 30:3943-3955,2010

25. Kadota M, Yang HH, Gomez B, et al: Delin-eating genetic alterations for tumor progression inthe MCF10A series of breast cancer cell lines. PLoSONE 5:e9201, 2010

26. Perreard L, Fan C, Quackenbush JF, et al:Classification and risk stratification of invasivebreast carcinomas using a real-time quantitativeRT-PCR assay. Breast Cancer Res 8:R23, 2006

27. Weigelt B, Mackay A, A’Hern R, et al: Breastcancer molecular profiling with single sample pre-dictors: A retrospective analysis. Lancet Oncol 11:339-349, 2010

28. Gnant M, Filipits M, Dubsky P: Predicting riskfor late metastasis: The PAM50risk of recurrence(ROR) score after 5 years of endocrine therapy inpostmenopausal women with HR� early breastcancer—A study on 1,478 patients from theABCSG-8 trial. Presented at the IMPAKT BreastCancer Conference, Brussels, Belgium, May 2-4,2013

29. Dowsett M, Sestak I, Lopez-Knowles E, et al:Comparison of PAM50 risk of recurrence score withOncotype DX and IHC4 for predicting risk of distantrecurrence after endocrine therapy. J Clin Oncol31:2783-2790, 2013

30. Sotiriou C, Pusztai L: Gene-expression signa-tures in breast cancer. N Engl J Med 360:790-800,2009

31. Collins LC, Botero ML, Schnitt SJ: Bimodalfrequency distribution of estrogen receptor immu-nohistochemical staining results in breast cancer:An analysis of 825 cases. Am J Clin Pathol 123:16-20, 2005

32. Paik S, Shak S, Tang G, et al: A multigeneassay to predict recurrence of tamoxifen-treated,node-negative breast cancer. N Engl J Med 351:2817-2826, 2004

33. Gerdes J, Lemke H, Baisch H, et al: Cellcycle analysis of a cell proliferation-associated hu-man nuclear antigen defined by the monoclonalantibody Ki-67. J Immunol 133:1710-1715, 1984

34. Dowsett M, Nielsen TO, A’Hern R, et al:Assessment of Ki67 in breast cancer: Recommen-dations from the International Ki67 in Breast Cancerworking group. J Natl Cancer Inst 103:1656-1664,2011

35. de Azambuja E, Cardoso F, de Castro G Jr, etal: Ki-67 as prognostic marker in early breast cancer:A meta-analysis of published studies involving12,155 patients. Br J Cancer 96:1504-1513, 2007

36. Bardou VJ, Arpino G, Elledge RM, et al:Progesterone receptor status significantly improvesoutcome prediction over estrogen receptor statusalone for adjuvant endocrine therapy in two largebreast cancer databases. J Clin Oncol 21:1973-1979, 2003

37. Rakha EA, El-Sayed ME, Green AR, et al:Biologic and clinical characteristics of breast cancerwith single hormone receptor positive phenotype.J Clin Oncol 25:4772-4778, 2007

38. Cancello G, Maisonneuve P, Rotmensz N, etal: Progesterone receptor loss identifies luminal Bbreast cancer subgroups at higher risk of relapse.Ann Oncol 24:661-668, 2013

39. Nielsen TO, Parker JS, Leung S, et al: Acomparison of PAM50 intrinsic subtyping with im-munohistochemistry and clinical prognostic factorsin tamoxifen-treated estrogen receptor-positivebreast cancer. Clin Cancer Res 16:5222-5232, 2010

40. Fan C, Oh DS, Wessels L, et al: Concordanceamong gene-expression-based predictors for breastcancer. N Engl J Med 355:560-569, 2006

Ades et al

8 © 2014 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 9: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

41. Sotiriou C, Wirapati P, Loi S, et al: Geneexpression profiling in breast cancer: Understandingthe molecular basis of histologic grade to improveprognosis. J Natl Cancer Inst 98:262-272, 2006

42. Loi S, Haibe-Kains B, Desmedt C, et al: Defini-tion of clinically distinct molecular subtypes in estro-gen receptor–positive breast carcinomas throughgenomic grade. J Clin Oncol 25:1239-1246, 2007

43. Fan C, Prat A, Parker JS, et al: Buildingprognostic models for breast cancer patients usingclinical variables and hundreds of gene expressionsignatures. BMC Med Genomics 4:3, 2011

44. Venet D, Dumont JE, Detours V: Most ran-dom gene expression signatures are significantlyassociated with breast cancer outcome. PLoS Com-put Biol 7:e1002240, 2011

45. Ignatiadis M, Bedard P, Haibe-Kains B, et al:A meta-analysis of gene expression profiling studiesidentifies clinically relevant oncogenic pathways inbasal-like breast cancer. Cancer Res 69, 2009 (suppl24; abstr 106)

46. Tran B, Bedard PL: Luminal-B breast cancerand novel therapeutic targets. Breast Cancer Res13:221, 2011

47. Smid M, Wang Y, Zhang Y, et al: Subtypes ofbreast cancer show preferential site of relapse.Cancer Res 68:3108-3114, 2008

48. Paik S, Tang G, Shak S, et al: Gene expres-sion and benefit of chemotherapy in women withnode-negative, estrogen receptor–positive breastcancer. J Clin Oncol 24:3726-3734, 2006

49. Albain KS, Barlow WE, Shak S, et al: Prog-nostic and predictive value of the 21-gene recur-rence score assay in postmenopausal women withnode-positive, oestrogen-receptor-positive breastcancer on chemotherapy: A retrospective analysis ofa randomised trial. Lancet Oncol 11:55-65, 2010

50. Gianni L, Zambetti M, Clark K, et al: Geneexpression profiles in paraffin-embedded core bi-opsy tissue predict response to chemotherapy inwomen with locally advanced breast cancer. J ClinOncol 23:7265-7277, 2005

51. Rouzier R, Perou CM, Symmans WF, et al:Breast cancer molecular subtypes respond differ-ently to preoperative chemotherapy. Clin CancerRes 11:5678-5685, 2005

52. Carey LA, Dees EC, Sawyer L, et al: Thetriple negative paradox: Primary tumor chemosensi-tivity of breast cancer subtypes. Clin Cancer Res13:2329-2334, 2007

53. de Ronde JJ, Hannemann J, Halfwerk H, etal: Concordance of clinical and molecular breastcancer subtyping in the context of preoperativechemotherapy response. Breast Cancer Res Treat119:119-126, 2010

54. Esserman LJ, Perou C, Cheang M, et al:Breast cancer molecular profiles and tumor re-sponse of neoadjuvant doxorubicin and paclitaxel:The I-SPY trial (CALGB 150007/150012, ACRIN6657). J Clin Oncol 27:10s, 2009 (suppl; abstrLBA515)

55. Bhargava R, Beriwal S, Dabbs DJ, et al:Immunohistochemical surrogate markers of breastcancer molecular classes predicts response to neo-adjuvant chemotherapy: A single institutional expe-rience with 359 cases. Cancer 116:1431-1439, 2010

56. Lv M, Li B, Li Y, et al: Predictive role ofmolecularsubtypesinresponsetoneoadjuvantchem-otherapy in breast cancer patients in NortheastChina. Asian Pac J Cancer Prev 12:2411-2417, 2011

57. Straver ME, Glas AM, Hannemann J, et al:The 70-gene signature as a response predictor forneoadjuvant chemotherapy in breast cancer. BreastCancer Res Treat 119:551-558, 2010

58. Lips EH, Mulder L, de Ronde JJ, et al:Neoadjuvant chemotherapy in ER� HER2� breastcancer: Response prediction based on immunohis-tochemical and molecular characteristics. BreastCancer Res Treat 131:827-836, 2012

59. Ignatiadis M, Singhal SK, Desmedt C, et al:Gene modules and response to neoadjuvant chem-otherapy in breast cancer subtypes: A pooled anal-ysis. J Clin Oncol 30:1996-2004, 2012

60. Peto R, Davies C, Godwin J, et al: Compari-sons between different polychemotherapy regi-mens for early breast cancer: Meta-analyses oflong-term outcome among 100,000 women in 123randomised trials. Lancet 379:432-444, 2012

61. Cardoso F, Piccart-Gebhart M, Van’t Veer L,et al: The MINDACT trial: The first prospectiveclinical validation of a genomic tool. Mol Oncol1:246-251, 2007

62. Sparano JA: TAILORx: Trial assigning individ-ualized options for treatment (Rx). Clin Breast Can-cer 7:347-350, 2006

63. Gonzalez-Angulo AM, Barlow WE, Gralow J,et al: SWOG S1007: A phase III, randomized clinicaltrial of standard adjuvant endocrine therapy with orwithout chemotherapy in patients with one to threepositive nodes, hormone receptor (HR)-positive, andHER2-negative breast cancer with recurrence score(RS) of 25 or less. J Clin Oncol 29:8s, 2011 (suppl;abstr TPS104)

64. Tamoxifen for early breast cancer: An over-view of the randomised trials—Early Breast CancerTrialists’ Collaborative Group. Lancet 351:1451-1467, 1998

65. Viale G, Regan MM, Maiorano E, et al: Prog-nostic and predictive value of centrally reviewedexpression of estrogen and progesterone receptorsin a randomized trial comparing letrozole and tamox-ifen adjuvant therapy for postmenopausal earlybreast cancer: BIG 1-98. J Clin Oncol 25:3846-3852,2007

66. Paik S, Shak S, Tang G, et al: Expression ofthe 21 genes in the recurrence score assay andtamoxifen clinical benefit in the NSABP study B-14of node negative, estrogen receptor positive breastcancer. J Clin Oncol 23, 2005 (suppl; abstr 510)

67. Coombes RC, Kilburn LS, Snowdon CF, et al:Survival and safety of exemestane versus tamoxifenafter 2-3 years’ tamoxifen treatment (IntergroupExemestane Study): A randomised controlled trial.Lancet 369:559-570, 2007

68. Dowsett M, Cuzick J, Wale C, et al: Predic-tion of risk of distant recurrence using the 21-generecurrence score in node-negative and node-positivepostmenopausal patients with breast cancer treatedwith anastrozole or tamoxifen: A TransATAC study.J Clin Oncol 28:1829-1834, 2010

69. Jansen MP, Foekens JA, van Staveren IL, etal: Molecular classification of tamoxifen-resistantbreast carcinomas by gene expression profiling.J Clin Oncol 23:732-740, 2005

70. Kok M, Linn SC, Van Laar RK, et al: Compar-ison of gene expression profiles predicting progres-sion in breast cancer patients treated withtamoxifen. Breast Cancer Res Treat 113:275-283,2009

71. Hugh J, Hanson J, Cheang MCU, et al:Breast cancer subtypes and response to docetaxelin node-positive breast cancer: Use of an immuno-histochemical definition in the BCIRG 001 trial. J ClinOncol 27:1168-1176, 2009

72. Dowsett M, Smith IE, Ebbs SR, et al: Prog-nostic value of Ki67 expression after short-termpresurgical endocrine therapy for primary breastcancer. J Natl Cancer Inst 99:167-170, 2007

73. Ellis MJ, Tao Y, Luo J, et al: Outcomeprediction for estrogen receptor-positive breast can-cer based on postneoadjuvant endocrine therapytumor characteristics. J Natl Cancer Inst 100:1380-1388, 2008

74. Dunbier AK, Anderson H, Ghazoui Z, et al:Association between breast cancer subtypes andresponse to neoadjuvant anastrozole. Steroids 76:736-740, 2011

75. Miller TW, Balko JM, Arteaga CL: Phospha-tidylinositol 3-kinase and antiestrogen resistance inbreast cancer. J Clin Oncol 29:4452-4461, 2011

76. Ellis MJ, Suman VJ, Hoog J, et al: Random-ized phase II neoadjuvant comparison betweenletrozole, anastrozole, and exemestane for post-menopausal women with estrogen receptor–richstage 2 to 3 breast cancer: Clinical and biomarkeroutcomes and predictive value of the baselinePAM50-based intrinsic subtype—ACOSOG Z1031.J Clin Oncol 29:2342-2349, 2011

77. Dowsett M, Cuzick J, Wale C, et al: Retro-spective analysis of time to recurrence in the ATACtrial according to hormone receptor status: Ahypothesis-generating study. J Clin Oncol 23:7512-7517, 2005

78. Viale G, Regan MM, Dell’Orto P, et al: Whichpatients benefit most from adjuvant aromatase in-hibitors? Results using a composite measure ofprognostic risk in the BIG 1-98 randomized trial. AnnOncol 22:2201-2207, 2011

79. Ellis MJ, Coop A, Singh B, et al: Letrozole ismore effective neoadjuvant endocrine therapy thantamoxifen for ErbB-1– and/or ErbB-2–positive, estro-gen receptor-positive primary breast cancer: Evi-dence from a phase III randomized trial. J Clin Oncol19:3808-3816, 2001

80. Smith IE, Dowsett M, Ebbs SR, et al: Neoad-juvant treatment of postmenopausal breast cancerwith anastrozole, tamoxifen, or both in combination:The Immediate Preoperative Anastrozole, Tamoxifen,or Combined with Tamoxifen (IMPACT) multicenterdouble-blind randomized trial. J Clin Oncol 23:5108-5116, 2005

81. De Laurentiis M, Arpino G, Massarelli E, etal: A meta-analysis on the interaction betweenHER-2 expression and response to endocrine treat-ment in advanced breast cancer. Clin Cancer Res11:4741-4748, 2005

82. Bose R, Kavuri SM, Searleman AC, et al:Activating HER2 mutations in HER2 gene amplifica-tion negative breast cancer. Cancer Discov 3:224-237, 2013

83. Sleijfer S, Bogaerts J, Siu LL: Designingtransformative clinical trials in the cancer genomeera. J Clin Oncol 31:1834-1841, 2013

84. Miller TW, Hennessy BT, Gonzalez-AnguloAM, et al: Hyperactivation of phosphatidylinositol-3kinase promotes escape from hormone depen-dence in estrogen receptor-positive human breastcancer. J Clin Invest 120:2406-2413, 2010

85. Miller TW, Perez-Torres M, Narasanna A, etal: Loss of phosphatase and tensin homologue de-leted on chromosome 10 engages ErbB3 andinsulin-like growth factor-I receptor signaling to pro-mote antiestrogen resistance in breast cancer. Can-cer Res 69:4192-4201, 2009

86. Massarweh S, Osborne CK, Creighton CJ, etal: Tamoxifen resistance in breast tumors is drivenby growth factor receptor signaling with repressionof classic estrogen receptor genomic function. Can-cer Res 68:826-833, 2008

87. Juncker-Jensen A, Lykkesfeldt AE, Worm J,et al: Insulin-like growth factor binding protein 2 is a

Luminal B Breast Cancer

www.jco.org © 2014 by American Society of Clinical Oncology 9

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 10: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

marker for antiestrogen resistant human breast can-cer cell lines but is not a major growth regulator.Growth Horm IGF Res 16:224-239, 2006

88. Zardavas D, Fumagalli D, Loi S: Phosphati-dylinositol 3-kinase/AKT/mammalian target of rapa-mycin pathway inhibition: A breakthrough in themanagement of luminal (ER�/HER2�) breast can-cers? Curr Opin Oncol 24:623-634, 2012

89. Loi S, Haibe-Kains B, Majjaj S, et al: PIK3CAmutations associated with gene signature of lowmTORC1 signaling and better outcomes in estrogenreceptor-positive breast cancer. Proc Natl Acad SciU S A 107:10208-10213, 2010

90. Awada A, Cardoso F, Fontaine C, et al: Theoral mTOR inhibitor RAD001 (everolimus) in combi-nation with letrozole in patients with advancedbreast cancer: Results of a phase I study withpharmacokinetics. Eur J Cancer 44:84-91, 2008

91. Baselga J, Semiglazov V, van Dam P, et al:Phase II randomized study of neoadjuvant everoli-mus plus letrozole compared with placebo plusletrozole in patients with estrogen receptor–positivebreast cancer. J Clin Oncol 27:2630-2637, 2009

92. Bachelot T, Bourgier C, Cropet C, et al:Randomized phase II trial of everolimus in combina-tion with tamoxifen in patients with hormone recep-tor–positive, human epidermal growth factorreceptor 2–negative metastatic breast cancer withprior exposure to aromatase inhibitors: A GINECOstudy. J Clin Oncol 30:2718-2724, 2012

93. Baselga J, Campone M, Piccart M, et al:Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med366:520-529, 2012

94. Hortobagyi GN, Piccart MJ, Rugo HS, et al:Correlation of molecular alterations with efficacy ofeverolimus in hormone receptor-positive, HER2-negative advanced breast cancer: Results fromBOLERO-2. J Clin Oncol 31, 2013 (suppl 15s; abstrLBA509)

95. Loi S, Michiels S, Baselga J, et al: PIK3CAgenotype and a PIK3CA mutation-related gene sig-nature and response to everolimus and letrozole inestrogen receptor positive breast cancer. PLoS ONE8:e53292, 2013

96. Bachelot T, Ferrero J, Cropet C, et al: Trans-lational studies within the TAMRAD randomizedGineco trial: Evidence for TORC1 activation markeras a predictive factor for everolimus efficacy inmetastatic breast cancer (MBC). Presented at theIMPAKT Breast Cancer Conference, Brussels, Bel-gium, May 3-5, 2012

97. Choo AY, Blenis J: Not all substrates aretreated equally: Implications for mTOR, rapamycin-resistance and cancer therapy. Cell Cycle 8:567-572,2009

98. Harrington LS, Findlay GM, Gray A, et al: TheTSC1-2 tumor suppressor controls insulin-PI3K sig-naling via regulation of IRS proteins. J Cell Biol166:213-223, 2004

99. Zardavas D, Baselga J, Piccart M: Emergingtargeted agents in metastatic breast cancer. NatRev Clin Oncol 10:191-210, 2013

100. Turner N, Grose R: Fibroblast growth factorsignalling: From development to cancer. Nat RevCancer 10:116-129, 2010

101. Turner N, Pearson A, Sharpe R, et al: FGFR1amplification drives endocrine therapy resistanceand is a therapeutic target in breast cancer. CancerRes 70:2085-2094, 2010

102. Elbauomy Elsheikh S, Green AR, LambrosMB, et al: FGFR1 amplification in breast carcinomas:A chromogenic in situ hybridisation analysis. BreastCancer Res 9:R23, 2007

103. Dienstmann R, Andre F, Soria J, et al: 319O:Significant antitumor activity of E-3810, a novelFGFR and VEGFR inhibitor, in patients with FGFR1amplified breast cancer. Presented at the EuropeanSociety for Medical Oncology Congress, Vienna,Austria, September 28-October 2, 2012

104. Andre F, Bachelot T, Campone M, et al: Amulticenter, open-label phase II trial of dovitinib, afibroblast growth factor receptor 1 (FGFR1) inhibitor,inFGFR1-amplified and nonamplified metastaticbreast cancer (BC). J Clin Oncol 29, 2011 (suppl;abstr 289)

105. Pollak M: The insulin and insulin-like growthfactor receptor family in neoplasia: An update. NatRev Cancer 12:159-169, 2012

106. Clarke RB, Howell A, Anderson E: Type Iinsulin-like growth factor receptor gene expressionin normal human breast tissue treated with oestro-gen and progesterone. Br J Cancer 75:251-257,1997

107. Hamelers IH, van Schaik RF, van TeeffelenHA, et al: Synergistic proliferative action of insulin-like growth factor I and 17 beta-estradiol in MCF-7Sbreast tumor cells. Exp Cell Res 273:107-117, 2002

108. Wiseman LR, Johnson MD, Wakeling AE, etal: Type I IGF receptor and acquired tamoxifenresistance in oestrogen-responsive human breastcancer cells. Eur J Cancer 29A:2256-2264, 1993

109. Parisot JP, Hu XF, DeLuise M, et al: Alteredexpression of the IGF-1 receptor in a tamoxifen-resistant human breast cancer cell line. Br J Cancer79:693-700, 1999

110. Lisztwan J, Pornon A, Chen B, et al: Thearomatase inhibitor letrozole and inhibitors ofinsulin-like growth factor I receptor synergisticallyinduce apoptosis in in vitro models of estrogen-dependent breast cancer. Breast Cancer Res 10:R56, 2008

111. Hou X, Huang F, Macedo LF, et al: Dual IGF-1R/InsR inhibitor BMS-754807 synergizes with hor-monal agents in treatment of estrogen-dependentbreast cancer. Cancer Res 71:7597-7607, 2011

112. Kaufman P, Ferrero J, Bourgeois H, et al: Arandomized, double-blind, placebo-controlled, phase2 study of AMG 479 with exemestane (E) or fulves-trant (F) in postmenopausal women with hormone-receptor positive (HR�) metastatic (M) or locallyadvanced (LA) breast cancer (BC). Cancer Res 70,2011 (abstr S1-4)

113. Di Cosimo S, Bendell JC, Cervantes-Ruiperez A, et al: A phase I study of the oral mTORinhibitor ridaforolimus (RIDA) in combination withthe IGF-1R antibody dalotozumab (DALO) in patients

(pts) with advanced solid tumors. J Clin Oncol28:235s, 2010 (suppl; abstr 3008)

114. O’Reilly KE, Rojo F, She QB, et al: MTORinhibition induces upstream receptor tyrosine kinasesignaling and activates Akt. Cancer Res 66:1500-1508, 2006

115. Musgrove EA, Caldon CE, Barraclough J, etal: Cyclin D as a therapeutic target in cancer. NatRev Cancer 11:558-572, 2011

116. Stendahl M, Kronblad A, Ryden L, et al:Cyclin D1 overexpression is a negative predictivefactor for tamoxifen response in postmenopausalbreast cancer patients. Br J Cancer 90:1942-1948,2004

117. Rudas M, Lehnert M, Huynh A, et al: CyclinD1 expression in breast cancer patients receivingadjuvant tamoxifen-based therapy. Clin Cancer Res14:1767-1774, 2008

118. Span PN, Tjan-Heijnen VC, Manders P, et al:Cyclin-E is a strong predictor of endocrine therapyfailure in human breast cancer. Oncogene 22:4898-4904, 2003

119. Caldon CE, Sergio CM, Kang J, et al: CyclinE2 overexpression is associated with endocrineresistance but not insensitivity to CDK2 inhibition inhuman breast cancer cells. Mol Cancer Ther 11:1488-1499, 2012

120. Finn R, Crown J, Boer K, et al: Results of arandomized phase 2 study of PD 0332991, a cyclin-dependent kinase (CDK) 4/6 inhibitor, in combinationwith letrozole vs letrozole alone for first-line treat-ment of ER�, HER2� advanced breast cancer(TRIO-18). Presented at the IMPAKT Breast CancerConference, Brussels, Belgium, May 3-5, 2012

121. Shangary S, Wang S: Targeting the MDM2–p53 interaction for cancer therapy. Clin Cancer Res14:5318-5324, 2008

122. Reference deleted123. Rabello Ddo A, de Moura CA, de Andrade

RV, et al: Altered expression of MLL methyltrans-ferase family genes in breast cancer. Int J Oncol43:653-660, 2013

124. Rodrıguez-Paredes M, Esteller M: Cancerepigenetics reaches mainstream oncology. Nat Med17:330-339, 2011

125. Zhou Q, Atadja P, Davidson NE: Histonedeacetylase inhibitor LBH589 reactivates silencedestrogen receptor alpha (ER) gene expression with-out loss of DNA hypermethylation. Cancer Biol Ther6:64-69, 2007

126. Hostetter CL, Licata LA, Keen JC: Timing iseverything: Order of administration of 5-aza 2� de-oxycytidine, trichostatin A and tamoxifen changesestrogen receptor mRNA expression and cell sensi-tivity. Cancer Lett 275:178-184, 2009

127. Yardley DA, Ismail-Khan RR, Melichar B, etal: Randomized phase II, double-blind, placebo-controlled study of exemestane with or withoutentinostat in postmenopausal women with locallyrecurrent or metastatic estrogen receptor–positivebreast cancer progressing on treatment with a non-steroidal aromatase inhibitor. J Clin Oncol 31:2128-2135, 2013

■ ■ ■

Ades et al

10 © 2014 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 11: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

Appendix

Table A1. Ongoing Clinical Trials With Targeted Agents in Luminal Breast Cancers

Trial PhaseNo. of

Patients Subtype Setting Treatment Primary End Point Secondary End Points

Pan-PI3K inhibitorsBKM120

NCT01339442 I 22 ER positive Metastatic, HTpretreatment

BKM120 plus fulvestrant MTD, safety PR, CR, SD, PDs, TR

NCT01610284(BELLE-2)

III 1,060 HR positive Metastatic, AIpretreatment

Fulvestrant plus BKM120or placebo

PFS OS, ORR, CBR, AEs,PKs, QOL

NCT01633060(BELLE-3)

III 615 HR positive, HER2negative

Metastatic, mTORinhibitorpretreatment

Fulvestrant plus BKM120or placebo

PFS OS, ORR, CBR, AEs,PKs, QOL

GDC-0941NCT01437566 II 270 ER positive, HER2

negative,PIK3CAmutation

Metastatic, AIpretreatment

GDC-0941 or GDC-0980plus fulvestrant vfulvestrant

PFS, AEs ORR, CBR, durationof OR, PKs

NCT01740336 II 180 ER positive, HER2negative

Metastatic Paclitaxel plus GDC-0941or placebo

PFS Safety, ORR, CBR,duration of OR

Dual PI3K/mTOR inhibitorsBEZ235

NCT01248494 I 72 HR positive Metastatic BEZ235 or BKM120 plusletrozole

MTD PFS, ORR

XL765NCT01082068 I/II 72 HR positive, HER2

negativeMetastatic, AI

pretreatmentXL765 or XL147 plus

letrozoleMTD, safety, PFS at

3 monthsPKs, PDs

GDC-0980NCT01437566 II 270 ER positive, HER2

negative,PIK3CAmutation

Metastatic, AIpretreatment

GDC-0941 or GDC-0980plus fulvestrant vfulvestrant

PFS, AEs ORR, CBR, DOR, PKs

Isoform-selective PI3Kinhibitors

BYL719NCT01791478 I 30 HR positive, HER2

negativeMetastatic, HT

pretreatmentBYL719 plus letrozole MTD CBR, PFS, ORR,

safetyAKT inhibitors

MK2206NCT01344031 I 54 ER positive Metastatic, no

progressionwith AIs orfulvestrant

MK2206 plus anastrozole,fulvestrant, oranastrozole plusfulvestrant

MTD, RPTD Safety, CBR

NCT01776008 II 87 ER positive, HER2negative,PIK3CAmutation

Neoadjuvant MK2206 plus anastrozoleor anastrozole andgoserelin

pCR CRR, RRR, AEs

mTORC1/2 inhibitorsAZD2014

NCT01597388 I 30 ER positive Metastatic AZD2014 plus fulvestrant Safety PKsFGF inhibitors

DovitinibNCT01484041 I/II 36 HR positive, HER2

negativeMetastatic Dovitinib plus AI CBR RPTD, toxicities, PDs,

PFSNCT01528345 II 150 HR positive, HER2

negativeMetastatic, HT

pretreatmentFulvestrant plus dovitinib

or placeboPFS ORR, DOR, OS,

safety, ECOG PSAZD4547

NCT01202591 (GLOW) I/II 120 ER positive,FGFR1polysomy orgeneamplification

Metastatic, HTpretreatment

Fulvestrant plus AZD4547or placebo

Safety, PFS PKs, PDs, ORR, DOR,PFS at 12 weeks,QOL

NCT01791985(RADICAL)

I/II 99 ER positive Metastatic, HTpretreatment

AZD4547 plus anastrozoleor letrozole vexemestane

Safety, tumor changeat 12 weeks

NR

(continued on following page)

Luminal B Breast Cancer

www.jco.org © 2014 by American Society of Clinical Oncology 11

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Page 12: Luminal B Breast Cancer: Molecular Characterization, Clinical ...news.medlive.cn/uploadfile/20140723/14060957108067.pdf · distinct profile of response to hormone therapy and chemotherapy

Table A1. Ongoing Clinical Trials With Targeted Agents in Luminal Breast Cancers (continued)

Trial PhaseNo. of

Patients Subtype Setting Treatment Primary End Point Secondary End Points

IGF inhibitorsDalotuzumab

NCT01605396 (MK-8669-064 AM3)

II 84 ER positive, Ki67� 15%

Metastatic Dalotuzumab,ridaforolimus, andexemestane vridaforolimus plusexemestane

PFS ORR, OS

NCT00903006 I/II 40 HR positive, HER2negative

Metastatic Fulvestrant v fulvestrant,dalotuzumab, anddasatinib

MTD, TTDP NR

BMS-754807NCT01225172 II 100 HR positive, HER2

negativeMetastatic, AI

pretreatmentBMS-754807 plus

letrozole v BMS-754807

PFS at 24 weeks ORR, DOR, safety,TFR, TR

MEDI-573NCT01446159 I 193 HR positive, HER2

negativeMetastatic, HT

pretreatmentMEDI-573 plus AI v AI Safety, PFS ORR, TTR, DOR, TTP,

change in tumorsize, OS, PKs, PDs,immunogenicity

CDK inhibitorsPD0332991

NCT01709370 II 45 ER positive, HER2negative

Neoadjuvant PD0332991 plus letrozole ORR AEs, PRR

NCT01723774 II 29 ER positive, HER2negative

Neoadjuvant PD0332991 plusanastrozole

pCR Safety, CRR, RRR,Ki67 at 2 weeks,PKs

NCT01740427 II/III 450 ER positive, HER2negative

Metastatic, firstline

PD0332991 plus letrozolev letrozole

PFS OS, ORR, DOR, DC,QTc interval, Cmin,QOL, TR

Abbreviations: AE, adverse event; BELLE, Buparlisib Breast Cancer Clinical Evaluation; CBR, clinical benefit rate; Cmin, minimum observed plasma troughconcentration; CR, complete response; CRR, clinical response rate; DOR, duration of response; ECOG PS, Eastern Cooperative Oncology Group performance status;ER, estrogen receptor; FGF, fibroblast growth factor; HER2, human epidermal growth factor receptor 2; HT, hormonal therapy; IGF, insulin growth factor; MTD,maximum-tolerated dose; mTOR, mammalian target of rapamycin; NR, not reported; OR, objective response; ORR, objective response rate; OS, overall survival;pCR, pathologic complete response; PD, pharmacodynamic; PI3K, phosphatidylinositol-3-kinase; PK, pharmacokinetic; PR, partial response; PRR, pathologicresponse rate; QOL, quality of life; RPTD, recommended phase II dose; RRR, radiologic response rate; SD, stable disease; TFR, treatment failure rate; TR,translational research; TTDP, time to disease progression; TTP, time to progression; TTR, time to response.

Ades et al

12 © 2014 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

129.94.53.232Information downloaded from jco.ascopubs.org and provided by at University of New South Wales on July 22, 2014 from

Copyright © 2014 American Society of Clinical Oncology. All rights reserved.