SlideShare a Scribd company logo
Review
Consequences of the Convergence of Multiple
Alternate Pathways on the Estrogen Receptor in
the Treatment of Metastatic Breast Cancer
Stefan Glück
Abstract
Endocrine therapy is the usual first-line therapy for patients with hormone receptor-positive metastatic breast cancer.
However, resistance to hormone therapies frequently occurs during the course of treatment. Growing understanding
of the signal cascade of estrogen receptors and the signaling pathways that interact with estrogen receptors has
revealed the complex role of these receptors in cell growth and proliferation, and on the mechanism in development of
resistance. These insights have led to the development of targeted therapies that may prove to be effective options for
the treatment of breast cancer and may overcome hormone therapy resistance. This article reviews current under-
standing of the cellular receptor signaling pathways that interact with estrogen receptors. It also reviews data from
recent ongoing clinical trials that examine the effects of targeted therapies, which might interfere with estrogen re-
ceptor pathways and might reduce or reverse resistance to traditional, sequential, single-agent endocrine therapy.
Clinical Breast Cancer, Vol. -, No. -, --- ª 2016 The Author. Published by Elsevier Inc. This is an open access article under
the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Keywords: Cyclin-dependent kinases, Endocrine therapy, Hormone receptor-positive, PI3K/Akt/mTOR pathway, Programmed
cell death inhibitor
Introduction
Breast cancer is the most common malignancy and the second
leading cause of cancer-related death among women in the United
States according to 2016 estimates.1
Approximately 6% of cases are
advanced or metastatic breast cancer (MBC) at diagnosis, and, for
those diagnosed at early stages, recurrence to distant sites occurs in
20% to 30%.2-4
Of patients with breast cancer, approximately 75%
have hormone receptor-positive (HRþ
) tumors.5
Therapies for MBC are aimed at palliation of symptoms with
improvement or conservation of quality of life and, potentially, pro-
longation of survival; unfortunately, prolonged survival is observed in
only a small percentage of patients.6,7
Both hormone receptor status
(eg, estrogen receptor [ER] and progesterone receptor [PgR]) and
human epidermal growth factor 2 (HER2) status are important pre-
dictive markers for treatment efficacy.7,8
Patients with HRþ
MBC are
candidates for initial endocrine therapy (ET).9
In premenopausal
women with MBC, various endocrine treatments are employed,
including ovarian suppression or oophorectomy, selective ER mod-
ulators such as tamoxifen, aromatase inhibitors (AIs) in conjunction
with ovarian suppression, and the selective ER downregulator, ful-
vestrant, all of which have different mechanisms of action. These
mechanisms have been reviewed extensively, demonstrating the
diversity of approaches that ultimately antagonize the growth-
promoting effects of estrogen on breast cancer cells. These mecha-
nistic differences are also linked to dissimilarities in resistance
mechanisms and thus influence treatment selection, particularly in the
context of sequencing and the use of combination regimens.10-17
In
the setting of postmenopausal HRþ
MBC, ET has a better tolerability
profile and longer time to progression (TTP) than chemotherapy.18,19
In clinical trials of AIs employed for first- or second-line treatment of
postmenopausal patients with MBC, AIs have greater efficacy than
either tamoxifen or megestrol acetate.20
Resistance to ET in HRþ
MBC is common, and given sufficient
time, most patients are faced with disease progression.21,22
The
mechanisms underlying disease progression and the development of
resistance to endocrine treatment are complex and not fully un-
derstood. Nevertheless, over the past several years, insights into
several pathways of resistance have grown and have led to increased
understanding of the clinical value of sequential lines of therapy and
co-targeting strategies. While receiving ET, up to 20% of patients
Department of Medicine, Division of Hematology Oncology, Sylvester Comprehensive
Cancer Center, Miami, FL
Submitted: Jun 17, 2016; Revised: Aug 1, 2016; Accepted: Aug 14, 2016
Address for correspondence: Stefan Glück, MD, PhD, FRCPC, Sylvester Professor of
Medicine, Department of Medicine, Division of Hematology Oncology, Sylvester
Comprehensive Cancer Center, Miami, FL 33136
E-mail contact: sgluck1@mac.com
1526-8209/ª 2016 The Author. Published by Elsevier Inc. This is an open access article
under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
http://dx.doi.org/10.1016/j.clbc.2016.08.004 Clinical Breast Cancer Month 2016 - 1
experience a loss of ERs with concomitant loss of response to es-
trogen.23
Amplified expression of HER2 may reduce ER levels and
serve as an alternative pathway for tumor cell survival.23,24
Cellular
signaling of growth-factor receptors interacts with ER nuclear and
nonnuclear pathways and may contribute to resistance through
posttranslational modifications of ER and/or coregulators.25-27
Mutations and/or aberrant forms of ER and their coregulators
may also be a primary cause or contribute to the development of
resistance to ET.21,22,25
The aim of this review is to describe emerging information on
cellular pathways that interact with ER and impact cell proliferation
and development of resistance to ET. We will discuss the biologic
rationale for combining ET with agents that target these pathways
and review the current status of clinical trials that are investigating
the effects of combination treatments (Table 1). We also illustrate
the cellular receptors and intracellular signaling pathways that
interact with ER (Figure 1).
Estrogen-Receptor Signaling
ER functions through 2 separate but interrelated pathways: nu-
clear (genomic) and nonnuclear (nongenomic) pathways. The nu-
clear pathway mediates the effects of ER on genomic activity,
thereby altering expression of many genes involved in physiologic
cell function and in abnormal cell proliferation, resulting in
tumorigenesis.28
The ER nuclear pathway is activated by estrogen
binding, receptor dimerization and translocation to the nucleus,
interaction with coregulator proteins (both coactivators and co-
repressors), and through estrogen response elements modulation of
gene transcription.29
ER coregulators have been implicated in the
pathogenesis of breast cancer. Most notable is amplified in breast
cancer 1, which, as its name implies, is overexpressed in breast
cancers. Moreover, amplified in breast cancer 1 overexpression may
play a role in resistance to ET.30
The nonnuclear pathway of ER
function (originating in cellular cytoplasm) is mediated by estrogen-
bound ER acting outside the cell nucleus through interaction with
multiple cellular signaling pathways, tyrosine kinases, and
membrane-bound growth factor-receptor pathways, including
HER2, insulin-like growth factor-1 receptor (IGF1R), and fibro-
blast growth factor receptor (FGFR).31,32
Multiple levels of inter-
action or crosstalk between ER and growth factor and tyrosine
kinase pathways may contribute to ER actions. These interactions
include modulation of ER activity, upregulation of competing
pathways and development of resistance to ET, and alternative or
escape pathways for cellular proliferation and tumorigenesis.
Estrogen can modify the activity of growth factor pathways by
increasing levels of growth factors such as transforming growth factor-
a and IGF1R and, alternatively, by altering the expression of
epidermal growth factor receptors (EGFR).27,32,33
Conversely, acti-
vation of growth factor-receptor signaling pathways may down-
regulate expression of ER, resulting in decreased estrogen effects.34,35
This crosstalk, which can up- or downregulate competing pathways
between ER and growth factor-receptor signaling, is recognized as a
major mechanism of ET resistance, and implies that ER-positive
(ERþ
)/HER2-positive (HER2þ
) breast cancer should be treated
with a combination of ET and HER2 inhibitors or antagonists.36
In addition to ER and HER2 status, assessment of PgR status is also
typically used to characterize MBC and shape treatment decisions. In
normal breast tissue, ER and PgR appear to be expressed in different
epithelial populations and regulate distinct pathways; estrogen is
involved in extracellular signaling and progesterone is associated with
cell growth.8
This independent activity is altered in breast cancer cells
where ER and PgR expression become correlated and converge on
pathways that promote tumor growth and metastasis, and both PgR
and ER become regulated by estrogen.8
Although PgR in ERþ
patients with MBC has been used as a predictor of response to ET, it is
controversial and not as well-accepted as the role of ER.37
Data indicate that ERþ
/PgR-negative (PgRÀ
) breast tumors are
not as responsive to selective ER modulator therapy as ERþ
/PgR-
positive (PgRþ
) tumors. One proposed mechanism for this anties-
trogen resistance involves aberrant growth factor signaling (a marker
for loss of PgR), but other unknown contributory mechanisms are
also likely to play a role. Further research is needed to identify such
mechanisms and thereby refine therapeutic strategies.38
In a recent study of MBC, PgR was found to associate with ER in
the presence of agonist ligands, and the ER-PgR ligand-activated
complex acted to modify chromatin-binding events and gene tran-
scription, leading to an antitumorigenic effect.39
Stimulation with
progesterone in an estrogen-rich context induced interactions be-
tween known ER cofactors and PgR, but did not alter the association
of those cofactors with ER, yielding an ER-PgR binding complex that
promoted cell death, apoptosis, and differentiation pathways.39
This
finding, coupled with the association between PgR gene (PgR) copy
number loss and poorer clinical outcome possibly owing to a reduc-
tion in ER-PgR binding complex formation, suggests a more intricate
relationship beyond PgR as a simple marker for ER pathway function
in the MBC setting.39
Although this contrasts with hormone
replacement studies in which synthetic progesterone (medrox-
yprogesterone) was associated with an increased breast cancer risk, it
was found that natural progesterone did not have the same effect,
which suggests different mechanisms of action for synthetic ago-
nists.40,41
Interestingly, semi-synthetic progestin, megestrol acetate,
provided clinical benefit to ERþ
patients with MBC who experienced
disease progression after estrogen suppression with a nonsteroidal
AI.42
Molecular subtyping of tumors may offer additional insight into
treatment of early-stage and locally advanced breast cancer. In a study
that evaluated molecular subtype and diagnostic classification of the
154 patients classified as Luminal type B (high risk), a group that is
typically sensitive to chemotherapy, 145 were ERþ
and 99 were PgRþ
tumors.43
Among the Luminal B patients, pathologic complete
response was 85%, suggesting a possible association between ER/PgR
status and the benefit of chemotherapy.43
Clearly the complex rela-
tionship between ER and PgR depends on many factors, including
genomic alteration, interaction of cofactors at a transcriptional level,
relative levels of estrogen and progesterone in the tumor cell envi-
ronment, and possibly variations in conformational changes related to
the form of the agonist.
Growth-Factor Receptors
Growth-factor receptors, particularly receptor tyrosine kinases,
play an integral role in growth promotion, cellular proliferation, and
tumorigenesis. Growth-factor pathways may act as ER-independent
drivers of tumor growth and survival, contributing to resistance to
all types of ET. HER2, IGF1R, and FGFR have been implicated in
resistance.26,44,45
2 - Clinical Breast Cancer Month 2016
Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer
Table 1 Clinical Trials Investigating the Effects of Combination Treatments in Breast Cancer (Available at https://clinicaltrials.gov/)
NCT Identifier (Acronym)
Study Agents Phase Study Design Selected Clinical Outcomes
Start Date/Estimated
Completion
Growth-factor receptor TK
inhibitors
NCT00634634
Sorafenib/letrozole
I/II  Open-label, single-arm
 Postmenopausal, HRþ advanced/MBC
Phase I
Primary: RPTD
Phase II
Primary: CBR
Phase I/II
Secondary: TTP, OS, safety
August 2008/December 2015
NCT00752986 (ZACFAST)
Vandetanib/fulvestrant
Placebo/fulvestrant
II  Randomized, double-blind
 Placebo-controlled
 Postmenopausal/advanced BC
Primary: Event-free survival
Secondary: TTP, PFS, OS, safety
December 2008/September 2013
NCT01528345
Dovitinib/fulvestrant
Placebo/fulvestrant
II  Randomized, double-blind
 Placebo-controlled
 Postmenopausal/HRþ
/HER2À
advanced/MBC
Primary: PFS
Secondary: ORR, DOR, OS, safety, ECOG performance
April 2012/October 2015
NCT02053636 (FINESSE)
Lucitanib
II  Open-label, single-arm
 HRþ
MBC
Primary: ORR December 2013/July 2016
NCT00305825
Bevacizumab/letrozole
II  Open-label, single-arm
 Postmenopausal/HRþ
 Unresectable advanced/MBC
Primary: Safety
Secondary: ORR, CBR
August 2004/November 2017
NCT00390455
Fulvestrant/lapatinib
Fulvestrant/placebo
III  Randomized, double-blind
 Placebo-controlled
 Postmenopausal/HRþ
advanced/MBC
Primary: PFS
Secondary: OS, ORR, DOR, OS, safety, QOL
September 2006/July 2014
NCT01160211
AI/trastuzumab/lapatinib
AI/trastuzumab AI/lapatinib
III  Randomized, open-label, multiple-arm
 Postmenopausal/HRþ
/HER2þ
, advanced/MBC; prior trastuzumab and ET
Primary: OS
Secondary: PFS, ORR, CBR, OS, safety, QOL, TTP, DOR
May 2011/December 2017
PI3K/Akt/mTOR inhibitors
NCT01248494
BEZ235/letrozole
BKM120/letrozole
BKM120 (intermittent)/letrozole
Ib  Open-label, multiple-arm
 Postmenopausal, HRþ
MBC
Primary: MTD BEZ235 or BKM120
Secondary: PFS, ORR
November 2010/October 2013
NCT01344031
MK-2206/anastrozole
MK-2206/fulvestrant
MK-2206/anastrozole/
fulvestrant
I  Open-label, multiple-arm
 Postmenopausal, HRþ
advanced/MBC
Primary: MTD MK-2206, RPTD for all agents
Secondary: Safety, CBR
April 2011/October 2013
NCT01339442
BKM120/fulvestrant
I  Open-label, multiple-arm
 Postmenopausal, HRþ
advanced/MBC
Primary: MTD BKM120 safety
Secondary: ORR, PK
November 2011/December 2014
NCT01791478
Letrozole/BYL719
Ib  Open-label, single-arm
 Postmenopausal HRþ
MBC
Primary: MTD BYL719
Secondary: CBR, PFS, ORR, Safety
April 2013/March 2016
NCT01797120
Fulvestrant/everolimus
Fulvestrant/placebo
II  Randomized, double-blind, placebo-controlled
 Postmenopausal HRþ MBC, AI refractory
Primary: PFS
Secondary: Safety, ORR, TTP, OS
May 2013/November 2015
StefanGlück
ClinicalBreastCancerMonth2016-3
Table 1 Continued
NCT Identifier (Acronym)
Study Agents Phase Study Design Selected Clinical Outcomes
Start Date/Estimated
Completion
NCT02049957
Exemestane/MLN0128
Fulvestrant/MLN0128
IIb/II  Open-label, multiple-arm
 Postmenopausal HRþ
/HER2À
, advanced/MBC, everolimus refractory
Primary: Safety, CBR
Secondary: ORR, tumor size, PK-PD, PFS, OS
February 2014/June 2016
NCT01992952 (FAKTION)
Fulvestrant/AZD5363
Fulvestrant/placebo
Ib/II  Randomized, double-blind, placebo-controlled
 Postmenopausal HRþ
, advanced/MBC
Phase Ib Primary: MTD AZD5363
Phase II Primary: PFS
Phase Ib/II Secondary: Safety, ORR, OS
May 2014/August 2016
NCT01698918 (BOLERO-4)
Letrozole/everolimus (1st line)
Exemestane/everolimus (2nd line)
II  Open-label, single-arm
 Postmenopausal HRþ
/HER2À
, advanced/MBC
Primary: PFS after 1st line
Secondary: ORR, OS, CBR, safety, PFS after 2nd line
March 2013/October 2016
NCT01296555
GDC0032 only
Letrozole/GDC0032
Fulvestrant/GDC0032
I/II  Open-label, multiple-arm
 Phase I advanced/metastatic solid tumors
 Phase II: Postmenopausal/HRþ
advanced/MBC
Phase I Primary: Safety
Phase II: Fulvestrant/GDC0032
Secondary: ORR, DOR, PFS
Phase II Primary: CBR, ORR
Secondary: DOR, PFS, OS
March 2011/July 2017
NCT01437566
Fulvestrant/GDC0941
Fulvestrant/GDC0980
Fulvestrant/placebo
II  Randomized, double-blind, placebo-controlled
 Postmenopausal HRþ
, MBC, AI refractory
Primary: PFS, safety
Secondary: ORR, CBR, DOR, PK
October 2011/November 2015
NCT02035813 (DETECT IV)
Everolimus/standard ET
II  Open-label, single-arm
 Postmenopausal HRþ
/HER2À
, advanced/MBC
Primary: PFS
Secondary: ORR, DCR, OS
January 2014/December 2019
NCT01610284 (BELLE-2)
Fulvestrant/BKM120
Fulvestrant/placebo
III  Randomized, double-blind, placebo-controlled
 Postmenopausal HRþ
, MBC, AI refractory
Primary: PFS
Secondary: OS, ORR, CBR, safety, PK
August 2012/March 2017
NCT01633060 (BELLE-3)
Fulvestrant/BKM120
Fulvestrant/placebo
III  Randomized, double-blind, placebo-controlled
 Postmenopausal HRþ
/HER2À
, advanced/MBC, AI treated, progressed
on/after mTOR
Primary: PFS
Secondary: OS, ORR, CBR, safety, PK
October 2012/March 2017
Src inhibitors
NCT00696072
Letrozole/dasatinib
Letrozole only
II  Open-label, multiple-arm
 Postmenopausal HRþ
/HER2À
, advanced/MBC
Primary: CBR
Secondary: ORR, PFS, TTF, safety
August 2008/February 2015
CDK 4/6 Inhibitors
NCT01857193
Exemestane
Everolimus/LEE011 (palbociclib)
Exemestane/everolimus
Exemestane/LEE011 (palbociclib)
Ib/II  Open-label, multiple arm
 Postmenopausal, HRþ
/HER2À
advanced/MBC
Phase I Primary: Safety
Phase II Primary: PFS
Phase Ib/II Secondary: Safety, PK, ORR, DOR, OS, DCR
September 2013/July 2016
NCT00721409
Letrozole/palbociclib
Letrozole only
I/II  Open-label, multiple-arm
 Postmenopausal HRþ
/HER2À
, advanced/MBC
Phase I Primary: Safety
Phase I Secondary: PK, QTc, antitumor activity
Phase II Primary: PFS
Phase II Secondary: Antitumor activity, OS, safety
September 2008/July 2018
AlternatePathwaysonEstrogenReceptorinMetastaticBreastCancer
4-ClinicalBreastCancerMonth2016
Table 1 Continued
NCT Identifier (Acronym)
Study Agents Phase Study Design Selected Clinical Outcomes
Start Date/Estimated
Completion
NCT02088684
LEE011/BKM120/
Fulvestrant LEE011/BYL719/
Fulvestrant LEE011/fulvestrant
IIb/II  Open-label, multiple-arm
 Postmenopausal HRþ
/HER2À
, advanced/MBC
Phase Ib Primary: Dose-limiting toxicity
Phase II Primary: PFS
Phase Ib/II Secondary: Safety, PK, ORR, DOR, PFS (Phase I
only), OS (Phase II only)
May 2014/February 2019
NCT01723774
Postmenopausal: Anastrozole/
palbociclib
Premenopausal: Goserelin/
palbociclib
II  Neoadjuvant, open-label, single-arm
 Pre/postmenopausal HRþ
/HER2À
, Stage 2/3 BC
Primary: Complete cell cycle arrest (Ki672.7%)
Secondary: PEPi 0 score, response- clinical, radiological,
Ki67 level, pCR rate, PK, safety
June 2013/August 2015
NCT02040857
Palbociclib/standard ET
II  Open-label, single-arm
 Pre/postmenopausal HRþ
/HER2À
, Stage 2/3 BC
Primary: Treatment DC rate January 2014/June 2019
NCT01942135 (PALOMA-3)
Fulvestrant/palbociclib
Fulvestrant/placebo
III  Randomized, double-blind, placebo-controlled
 Postmenopausal HRþ
/HER2À
, MBC, ET refractory
Primary: PFS
Secondary: OS, ORR, DOR, CBR, QOL, TTD
September 2013/January 2017
NCT01958021 (MONALEESA-2)
Letrozole/LEE011
Letrozole/placebo
III  Randomized, double-blind, placebo-controlled
 Postmenopausal HRþ
/HER2À
, advanced/MBC, therapy-naive
Primary: PFS
Secondary: OS, ORR, CBR, safety, QOL, QTc
December 2013/January 2017
PD-1 inhibitors
NCT02129556 (PANACEA)
MK-3475/trastuzumab
I/II  Open-label, single-arm
 HER2þ
, advanced/MBC, trastuzumab refractory
Primary: Dose-limiting toxicity, RPTD
Secondary: Safety, CBR, DOR, TTP, PFS, OS
July 2014/July 2023
Proteasome inhibitors
NCT01142401
Fulvestrant/bortezomib
Fulvestrant only
II  Randomized, double-blind
 Postmenopausal HRþ
, advanced/MBC
Primary: PFS
Secondary: CBR, OS, safety
May 2010/June 2014
Epigenetic modifiers
NCT02374099
Oral azacitidine/fulvestrant
Fulvestrant only
II  Randomized, open-label
 Postmenopausal ERþ
, HER2À
MBC who progressed on an AI
Primary: PFS
Secondary: ORR, CBR, OS, DOR, safety
March 2015/February 2018
NCT00676663 (ENCORE301)
Exemestane/entinostat
Exemestane only
II  Randomized, double-blind
 Postmenopausal ERþ
, locally recurrent or MBC who progressed on an AI
Primary: PFS
Secondary: ORR, CBR, safety
May 2008/October 2012
NCT01349959
Azacitidine/entinostat
II  Randomized, open-label
 TNBC or HRþ
/HER2À
, advanced/MBC
Primary: Confirmed response rate by RECIST
Secondary: CBR, OS, PFS, Safety
April 2011/March 2014
NCT01194908
Decitabine/panobinostat/tamoxifen
I/II  Randomized, open-label
 TNBC, MBC or locally advanced
Primary: MTD
Secondary: Safety
July 2010/January 2014
Abbreviations: AI ¼ aromatase inhibitor; BC ¼ breast cancer; CBR ¼ clinical benefit rate; DC ¼ discontinuation rate; DOR ¼ duration of response; ECOG ¼ Eastern Cooperative Oncology Group; ET ¼ endocrine therapy; HER2þ
/HER2À
¼ human epidermal growth factor
receptor-2 positive/negative; HRþ
¼ hormone receptor-positive; MBC ¼ metastatic breast cancer; MTD ¼ maximum tolerated dose; mTOR ¼ mammalian target of rapamycin inhibitor; ORR ¼ overall response rate; OS ¼ overall survival; pCR ¼ pathologic complete response;
PD ¼ pharmacodynamics; PD-1 ¼ programmed cell death protein 1; PFS ¼ progression-free survival; PK ¼ pharmacokinetics; QOL ¼ quality of life; QTc ¼ corrected QT interval; RPTD ¼ recommended phase II dose; TK ¼ tyrosine kinase; TNBC ¼ triple negative breast
cancer; TTD ¼ time to deterioration; TTF ¼ time to treatment failure; TTP ¼ time to progression.
StefanGlück
ClinicalBreastCancerMonth2016-5
In breast cancer, HER2, a member of the EGFR family, is an
important independent pathway for cell proliferation.46
HER2 is
constitutively in an active conformation that allows dimerization with
itself (homodimerization) or other members of the EGFR family
(heterodimerization). When HER2 is overexpressed, as it is in w15%
to 20% of all breast cancers, it mediates cell growth and survival
through activation of its downstream mediators, PI3K/Akt/mTOR
and Ras/Raf/MEK/MAP kinase.47
Overexpression of HER2
(HER2þ
) is an independent prognostic (and predictive for sensitivity
to its antagonists/inhibitors) factor indicating a more aggressive form
of cancer with a higher recurrence rate and higher mortality.46,48
Approximately 10% of patients with HRþ
MBC are also HER2þ
and, conversely, approximately 50% of HER2þ
breast cancers are also
HRþ
. Women with HRþ
and HER2þ
disease may not respond to ET
as well as women with HRþ
and HER2-negative (HER2À
) MBC.7,49
Reversing resistance to antiestrogen therapy in MBC is an important
tactic to avoid and delay the use of cytotoxic compounds. The
mammalian target of rapamycin (mTOR) has been associated with
in vitro reversal of drug resistance, including tamoxifen resistance.50
A number of compounds that interfere with HER2 are either
approvedfortherapyofHER2þ
breastcancerorareunderinvestigation
as treatments for HER2þ
MBC and HRþ
HER2þ
MBC. Trastuzu-
mab is a humanized monoclonal antibody directed against the extra-
cellular domain-2 of HER2, which prevents ligand-independent
signaling.51,52
Trastuzumab has been widely studied for treatment of
HER2þ
breast cancer and is approved by the US Food and Drug
Administration (FDA) for first-line treatment of HER2þ
MBC as well
as a component of adjuvant systemic therapy for HER2þ
breast can-
cer.53
Two large phase III trials have demonstrated the benefit of
trastuzumab treatment with ET in patients with HRþ
and HER2þ
MBC. In the Trastuzumab and Anastrozole Directed Against
ER-Positive HER2-Positive Mammary Carcinoma (TAnDEM) trial,
median progression-free survival (PFS) was significantly improved in
patients with HRþ
and HER2þ
MBC who received anastrozole and
trastuzumab (n ¼ 103) compared with anastrozole (n ¼ 104) alone
(4.8 vs. 2.4 months, respectively; P ¼ .0016).54
In the Efficacy of
letrozole in combination with trastuzumab compared to letrozole
monotherapy (eLEcTRA) trial, median time to progression was
numerically longer in HRþ
and HER2þ
MBC patients receiving
letrozoleandtrastuzumab(n¼26)versusletrozole(n¼ 31)alone(14.1
vs. 3.3 months, respectively; P ¼ .23). The failure to achieve statistical
significance was attributed to the small sample of patients evaluated.55
Another FDA-approved agent, lapatinib, is a dual EGFR/ErbB2
reversible tyrosine kinase inhibitor that competes with ATP for its
binding site on the tyrosine kinase domain. It blocks PI3K/Akt/
mTOR and Ras/Raf/MEK/MAP kinase pathways downstream of
HER2.56
Lapatinib is FDA-approved for systemic treatment of
HER2þ
advanced or MBC with systemic chemotherapy as second-
line treatment or in first-line combination with letrozole for HRþ
and HER2þ
MBC.57
In the EGF30008 trial, addition of lapatinib to
letrozole significantly reduced the risk of disease progression versus
letrozole plus placebo; median PFS was 8.2 versus 3.0 months,
respectively, (P ¼ .019) in patients with ERþ
and HER2þ
early breast
cancer. The clinical benefit rate was significantly greater for lapatinib-
letrozole versus letrozole-placebo (48% vs. 29%, respectively;
P ¼ .003).58,59
These findings led to FDA approval of first-line MBC
therapy in ERþ
and HER2þ
disease. Phase III trials of lapatinib for
postmenopausal advanced HRþ
MBC are also underway. For
example, one study is examining lapatinib plus fulvestrant compared
with lapatinib and placebo (NCT00390455). Another study is
comparing 3 arms: lapatinib plus trastuzumab plus an AI, trastuzu-
mab plus an AI, and lapatinib plus an AI (NCT01160211).
Combined targeting of HER2 with lapatinib and trastuzumab may
improve outcomes compared with single-agent targeting. Dual inhibi-
tion with these agents may also provide a regimen with greater tolera-
bility compared with alternative treatments that involve the use of
cytotoxic agents.60
A phase III trial demonstrated that lapatinib plus
trastuzumabsignificantlyimprovedPFSandoverallsurvival(OS)versus
lapatinib monotherapy in patients with HER2þ
MBC whose disease
had progressed during prior trastuzumab therapy.61
The ALTERNA-
TIVE (Safety and efficacy of lapatinib [L], trastuzumab [T], or both in
combination with an aromatase inhibitor [AI]) study is an ongoing
phase III, randomized, open-label, multicenter trial in HRþ
HERþ
MBC examining the safety and efficacy of lapatinib plus trastuzumab
plus AI versus trastuzumab plus AI.62
The rationale for this trial is to
evaluate dual anti-HER2 therapy with an AI compared with single anti-
HER2 therapy and an AI because HER2þ
MBC is associated with a
poor prognosis and increased resistance to ET in patients who are HRþ
.
Pertuzumab is a monoclonal antibody to the extracellular domain
4 of HER2 that blocks heterodimerization between HER2 and
HER3 as well as other HER family members, thereby inhibiting
signal transduction via MAPK and PI3K pathways, which can lead
to cell growth arrest and apoptosis.63
In a phase II trial, pertuzumab
plus trastuzumab showed some improvements in patients with
HER2þ
MBC with progression on trastuzumab.64
A phase III trial
of pertuzumab plus trastuzumab plus cytotoxic chemotherapy with
docetaxel as first-line treatment in HER2þ
MBC demonstrated
improved PFS compared with trastuzumab plus docetaxel alone,
and resulted in the FDA approval of this combination.65
IGF1R is a tyrosine kinase growth factor receptor that has been
shown to promote the growth of breast cancer cells.66
Similar to the
HER2/EGFR family of receptors, growth-promoting effects of
IGF1R are mediated by PI3K and Ras/Raf/Mek/MAPkinase intra-
cellular signaling. Inhibition of IGF1R function blocks the growth-
promoting effects of IGF1 on breast cancer.67,68
Early preclinical
studies were promising; however, trials with the monoclonal anti-
bodies figitumumab, AVE1642, ganitumab, and dalotuzumab tar-
geting IGF1R with ET in HRþ
MBC have not shown advantage over
ET alone.66
A potential reason for this lack of benefit may be related
to findings that breast cancer cells, which are resistant to tamoxifen,
are also refractory to IGF1R antibody treatment.69
Inhibition of
IGF1R function at different stages of HRþ
MBC or in combination
with inhibitors of other pathways may prove more effective.
GrowthfactorreceptorpathwayssuchasHER2,IGF1R,andFGFR,
which are independent of ER signaling, can subvert ET and lead to
tumor cell growth and proliferation in MBC. Combination strategies,
which include monoclonal antibodies that block growth factor re-
ceptors, coupled with tyrosine kinase inhibitors or other agents that act
to disrupt signaling of multiple downstream effector molecules may be
an effective approach to optimize treatment for the patient with MBC.
Intracellular Signaling Cascades
PI3K/Akt/mTOR Pathway. The PI3K signaling pathway plays a
vital role in cell proliferation, differentiation, and cell survival.70
The
Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer
6 - Clinical Breast Cancer Month 2016
cell cycle is often dysregulated in cancer cells, resulting in reduced
growth-factor dependency and increased replicative potential.
Approximately 8% of breast cancers have mutations in PI3K; gain-
of-function mutations in the PI3K alpha catalytic subunit
(PIK3CA) occur in approximately 30% of ERþ
breast cancers and
are a common genomic alteration in this subtype of breast can-
cer.71,72
PI3K is activated by growth-factor receptor tyrosine ki-
nases, HER2, EGFR, IGF1R, and FGFR, and through a series of
steps, activation of PI3K stimulates the downstream mediators Akt
and mTOR.73
The PI3K pathway interacts with ER directly and
indirectly in that PI3K and Akt can phosphorylate and thereby
activate ER in the absence of estrogen.74,75
This interaction of the
PI3K pathway with ER confers ET resistance.74,76
A phase II
clinical trial that studied the combination of ET with everolimus, an
inhibitor of mTOR, tested the rationale that inhibition of the PI3K
pathway might amplify response or reverse resistance to ET. In
patients with HRþ
breast cancer, the combination of everolimus
and letrozole resulted in increased clinical response and suppression
of tumor cell proliferation compared with letrozole and placebo.77
The Tamoxifen Plus Everolimus (TAMRAD) study in patients
with HRþ
HER2À
MBC who had progressed on an AI found that
the addition of everolimus to tamoxifen improved clinical benefit
rate, TTP, and OS compared with tamoxifen alone.78
The phase III
Combination of everolimus with trastuzumab plus paclitaxel as fi
rst-line treatment for patients with HER2-positive advanced breast
cancer (BOLERO-2) trial showed that treatment with everolimus
plus exemestane demonstrated a median TTP of 10.6 months
compared with 4.1 months with exemestane alone in post-
menopausal HRþ
MBC that recurred or progressed on previous
ET.79,80
However, in the phase III Placebo-controlled trial of
letrozole plus oral temsirolimus (HORIZON) trial, the addition of
the mTOR inhibitor temsirolimus to letrozole did not lead to
improvement in PFS in AI-naive advanced breast cancer.81
Several
differences between these trials may contribute to the varied results.
Figure 1 Cellular Signaling Pathways in Breast Cancer and Resistance to Endocrine Therapy. Estrogen Activates Cytoplasmic and
Nuclear ER. Cytoplasmic ER Binds to Growth Factor Signaling Components Such as PI3K, Activating Key Signaling Molecules
Such as Akt and RAS, and Downstream Molecules Such as mTOR, Raf, and MAPK, Which Promote Cell Proliferation and
Survival. In Addition, Many of These Signal-Transduction Molecules Can Phosphorylate and Activate ER and its Co-
Regulators to Enhance Nuclear Genomic ER-Mediated Responses. Src Can Also Interact With ER and Mediate Nongenomic
ER Activation of Signaling Pathways and Gene Transcription. Src May Be a Key Signaling Molecule in the Development of
Resistance to ET. ER Degradation by the 26S Proteasome is Depicted by the Schematic Model. Src Activation and ER
Proteolysis Support a Model Whereby Crosstalk Between Ligand-Bound ER and Src Drives ER Transcriptional Activity and
Targets ER for Ubiquitin-Dependent Proteolysis. Src Activation May Promote Not Only Proliferation, but Also Estrogen-
Activated ER Loss Through Proteolysis. Cyclin E1-CDK2 and Cyclin E2-CDK2 Are Activated During Estrogen-Mediated
Proliferation. Estrogen, via ER, Upregulates Cyclin D1. Cyclin D1 Upregulates E2F Transcription Factors, Leading to Active
Cyclin E2-CDK2 Complexes. Mitogenic Signals Converge at the Level of Cyclin D1 Upregulation and CDK4/6 Association,
Localization, and Kinase Activity
Abbreviations: AI ¼ Aromatase inhibitor; CDK ¼ Cyclin-dependent kinases; EGFR ¼ epidermal growth factor receptor; ER ¼ estrogen receptor; ERE ¼ estrogen response elements; FGFR ¼ fibroblast
growth factor receptor; HER2 ¼ human epidermal growth factor; IGF ¼ insulin-like growth factor; MAPK ¼ mitogen-activated protein kinases; mTOR ¼ mammalian target of rapamycin inhibitor; p ¼
phosphorylation sites; PI3K ¼ phosphatidylinositol 3-kinase; RTK ¼ receptor tyrosine kinase; Ub ¼ ubiquitin; TF ¼ transcription factor.
Stefan Glück
Clinical Breast Cancer Month 2016 - 7
First, patient populations differed, with AI-resistant patients in the
TAMRAD and BOLERO-2 trials, but AI-naive patients in the
HORIZON trial. With both mTOR inhibitors subject to meta-
bolism by the same CYP enzyme (CYP3A), population differences
in its expression might have resulted in variable metabolism across
diverse ethnic groups. Genotypic and phenotypic variation across
populations may have also accounted for variable response. Finally,
relative differences in drug effectiveness between everolimus and
temsirolimus cannot be ruled out.82
Ongoing studies of fulvestrant
with everolimus and standard ET with everolimus will provide
additional perspective on the potential combined benefit of mTOR
and ER inhibition (NCT01797120, NCT02035813).
Inhibitors of PI3K in preclinical and clinical trials for breast
cancer include BKM120, LY294002, GDC0941, BYL719, and
GDC0032.83
Additionally, combination PI3K/mTOR inhibitors,
GDC0980 and BEZ235, are also being studied. In preclinical
studies, GDC0980, GDC0032, and GDC0941, given in combi-
nation, enhanced activity of fulvestrant in MCF-7 xenografts,
resulting in tumor regression and tumor growth delay.84
A com-
bination of tamoxifen, everolimus, and LY294002 produced
increased antitumor effects compared with tamoxifen alone or
tamoxifen and everolimus in a breast cancer cell line.85
In addition,
treating breast cancer cells with BEZ235 induced apoptosis when
combined with estrogen deprivation, suggesting potential activity in
HRþ
breast cancer.86
In an initial phase I/II study, patients with
trastuzumab-resistant locally advanced or metastatic HER2þ
breast
cancer were treated with BKM120 (buparlisib) and trastuzumab.
Two patients (17%) had partial responses, and 7 (58%) had stable
disease (! 6 weeks) with a disease-control rate of 75%.87
Buparlisib
in combination with letrozole has also been used to treat ERþ
HER2À
MBC, and provided clinical benefit in both patients with
PI3K-wild type and mutant tumors, but patients with coexisting
PI3K and MAP3K mutations derived the greatest clinical benefit
from this regimen.88
The Akt inhibitor MK2206 is also being
studied in breast cancer. In MCF-7 cells, overexpression of consti-
tutively active Akt1 induced resistance to anastrozole; addition of
the Akt inhibitor MK2206 restored sensitivity to this AI.89
Taken together, the preclinical and clinical data suggest that ER
and PI3K pathways counter-regulate each other with low PI3K
activation resulting in high ER levels, and hyperactivation of
PI3K causing a reduction in ER levels.90
Further validation of this
notion comes from studies showing that inhibition of HER2 with the
antibody trastuzumab or the tyrosine kinase inhibitor lapatinib
restores or upregulates ER levels or transcriptional activity in breast
cancer cells and patient tumors.91,92
Also, AIs or fulvestrant inhibit
the growth of HER2þ
tumors that have progressed on trastuzumab or
lapatinib.92
Numerous ongoing phase I to III studies are examining
the effects of AIs or fulvestrant in combination with PI3K and
Akt inhibitors (NCT01610284, NCT01633060, NCT01791478,
NCT02088684, NCT01248494, NCT01339442, NCT01437566,
NCT01296555, and NCT01344031).
Src kinases, intracellular tyrosine kinases that contribute to
control of cell proliferation, have been implicated in oncogenic
activity in breast cancer, particularly HRþ
breast cancer.93
Dasatinib
is a small-molecule inhibitor of several Src family kinases approved
for use in chronic myelogenous leukemia.94
A recent phase II trial of
dasatinib in women with MBC showed modest activity with disease
control rates (ie, complete response, partial response, or stable dis-
ease for at least 16 weeks) for 8.3% of patients with HER2þ
MBC
and 15.6% of those with HRþ
MBC.95
Early results of a trial of
dasatinib in combination with fulvestrant as second-line treatment
of HRþ
MBC in postmenopausal women did not find improved
efficacy with the addition of dasatinib.96
With the variety of agents available that target the PI3K/Akt/
mTOR pathway, the challenge remains to identify which subset of
targets, and therefore, which combination of agents may be the
most impactful to maximize clinical benefit and adapt to ET
resistance in individual patients.
Cell Cycle Control and Proliferation
Cellular pathways involved in cell proliferation are targets for new
drugs that interfere with development of resistance to ET and that
treat HRþ
MBC. Cyclin-dependent kinases (CDKs) are a subgroup
of serine/threonine kinases that play a key role in regulating cell
cycle progression.97
The cell cycle G1 to S-phase transit is
controlled by CDK4 and CDK6, which are activated upon binding
to D-type cyclins leading to expression of genes required for S-phase
entry.98
Several studies have identified alterations of cell cycle reg-
ulators in human breast cancer and provide a rationale for the po-
tential therapeutic role for CDK4/6 inhibition in breast tumors.
Cyclin D1 is a direct transcriptional target of ER.99,100
Treatment
with antiestrogens induces growth arrest of HRþ
breast cancer cells
and decreases cyclin D1 expression.101
Conversely, microinjection
of antibodies into cyclin D1 inhibits estrogen-induced S-phase en-
try.102,103
Endocrine resistance is associated with persistent cyclin
D1 expression and Rb phosphorylation.104
Compared with other
subtypes of breast cancer, HRþ
breast cancer is commonly associ-
ated with hyperactivation of cyclin D1-CDK4/6.105
CDK4/6 inhibitors, including palbociclib, abemaciclib, and ribo-
ciclib, are being investigated in both the preclinical and clinical settings;
some are becoming available for treatment of HRþ
MBC. In 2015,
palbociclib, a highly selective inhibitor of CDK4/6 kinase, was
approved by the FDA in combination with letrozole for the treatment
of postmenopausal women with ERþ
HER2À
advanced breast cancer
as initial endocrine-based therapy for their metastatic disease.106,107
Approval for this indication was accelerated based on PFS data.
Continued approval for this indication may be contingent upon veri-
fication and description of clinical benefit in a confirmatory trial. In
preclinical studies, palbociclib inhibited growth of HRþ
cancer cells,
including those that are resistant to antiestrogen.105
Additionally, a
synergistic antitumor effect was observed when palbociclib was com-
bined with tamoxifen in both tamoxifen-sensitive and tamoxifen-
resistant cell lines. Palbociclib in combination with letrozole versus
letrozole alone (PALOMA-1), a randomized phase II study of the AI
letrozole with or without palbociclib as first-line therapy for ERþ
,
HER2À
MBC(n¼ 165),reportedastrikingimprovementinPFSfrom
7.5 to 26.2 months (hazard ratio [HR], 0.32; 95% confidence interval
[CI], 0.19-0.56; P  .001) without additional safety concerns.108
In
the final results analysis, the median PFS was 20.2 months with
letrozole plus palbociclib compared with 10.2 months for letrozole
alone (HR, 0.488; 95% CI, 0.319-0.748; 1-sided P ¼ .0004), and the
combination demonstrated significant clinical benefit as first-line
treatment in this patient population.109
Several phase I, II, and III
studies of palbociclib in combination with fulvestrant and AIs (eg,
Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer
8 - Clinical Breast Cancer Month 2016
PALOMA-2 and PALOMA-3) in HRþ
MBC are currently underway
or have recently concluded.110,111
Most recently, treatment with the
combination of palbociclib and fulvestrant in patients with advanced
HRþ
HER2À
breast cancer who had relapsed or progressed during
prior endocrine therapy, received FDA approval based on the findings
of the phase III study, PALOMA-3.110
The study demonstrated that
the combination of palbociclib and fulvestrant resulted in significantly
longer median PFS (9.5months [95%CI, 9.2-11.0 months] compared
with 4.6 months [95% CI, 3.5-5.6 months]) for fulvestrant alone (HR,
0.46; 95% CI, 0.36-0.59; P  .001).110
Other CDK inhibitors are in various stages of clinical develop-
ment (NCT00721409, NCT01723774, NCT01942135,
NCT02040857). Additionally, another CDK inhibitor, LEE001,
ribociclib, is being used in addition to everolimus and exemestane in
patients with advanced HRþ
, HER2À
breast cancer who have
anastrozole- or letrozole-resistant disease (NCT01857193). Interim
results from the phase Ib/II study showed a 70.9% disease control
rate.112
This agent is also being examined in combination with
fulvestrant and PI3K inhibitors (NCT02088684).
The CDK pathway regulates the cell cycle, with CDK4 and 6
directly controlling the gap phase 1 of mitosis (G1). Dysregulation of
these components may occur through multiple mechanisms
contributing to tumor growth and metastatic potential, and is a
common feature in many cancers, including breast cancer. Therefore,
therapeutic inhibition of CDK is a relevant treatment strategy, and a
number of agents are currently under investigation in clinical tri-
als.98,105
Ongoing clinical trials that combine CDK inhibitors with
inhibitors of other dysregulated pathways commonly found in breast
cancer may improve outcomes and act to overcome ET resistance.
Immune Response Modulation
Antitumor immunity in human cancer is often ineffective owing
to the tight regulation associated with the maintenance of immune
homeostasis. Previous attempts to develop effective immunother-
apies for cancer have been unsuccessful because there are significant
barriers that have been difficult to overcome. However, blockade of
the programmed death receptor (PD-1), which may play a role in
the tumor’s ability to bypass immune modulation and is overex-
pressed in certain tumors such as melanoma, has been proven
effective for treatment.113-115
Additionally, a monoclonal antibody
targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4), an immune
checkpoint molecule that downregulates pathways of T-cell activa-
tion, has also been proven effective for treatment of melanoma.116
The place of PD-1 and CTLA-4 inhibition in the treatment of
HRþ
MBC is not as clearly defined.
The extent of mutations and aberrant protein expression associ-
ated with cancer cells may also dictate the potential for efficacy of
immunotherapy.117
Foreign antigens expressed on the surface of
pathogens trigger the body’s immune system to target and eliminate
these invaders. However, tumor cells are often undetected by im-
mune surveillance owing to identification of tumor cells as “self.”
Although tumor cells may over express tumor-associated antigens,
these same antigens are expressed at low levels on normal cells,
leading to immune tolerance. In contrast, neoantigens that arise due
to mutations in a tumor cell, which are not expressed on any other
normal cells, represent prime targets for Tecell-mediated elimina-
tion with no risk to normal tissues.117
This has led to the hypothesis
that tumors with high mutation rates have increased neoepitopes,
and use of checkpoint blockade therapies (PD-1, PD-L1, and
CTLA-4) can reactivate tumor-infiltrating T cells in these cases,
leading to tumor regression.117
The heterogeneous nature of breast cancer has led to studies that
seek to identify biomarkers associated with drug response. Triple-
negative breast cancer (TNBC) lacks the molecular targets for
many treatment options and is associated with high genetic alter-
ation rates. In particular, one study of 2111 patients with TNBC
(ERÀ
/PgRÀ
/HER2À
) found that 91% of patients showed dimin-
ished androgen receptor expression and/or mutations in PI3KCA/
PTEN/AKT1, or loss of PTEN.118
In addition, expression of PD-L1
is more prevalent in TNBC, and this coupled with increased PD-
L1-mediated T-cell reactivation suggests that PD-L1 inhibitors may
be effective in this patient population.117,119
In fact, a preliminary
study that targeted PD-L1 showed promising objective clinical ac-
tivity in TNBC patients (NCT01375842).119
Estrogen-Receptor Degradation and Proteasome
Inhibitors (PIs)
Proteasomes are multicatalytic protease complexes in the cell,
involved in the non-lysosomal recycling of intracellular proteins. Pro-
teasomes play a critical role in regulation of cell division in both normal
and cancer cells.120-123
In cancer cells, this homeostatic function is
deregulated, leading to the hyperactivation of the proteasomes. PIs
interfere with the ubiquitin proteasome pathway and lead to the
accumulation of proteins engaged in cell cycle progression. They ulti-
mately put a halt to cancer cell division and induce apoptosis. Cell cycle
proteins are regulatory proteins whose temporal activity is tightly
controlled and are thus important targets of proteasome degradation.
Major proteins related to the cell cycle process are cyclins, CDKs, CDK
inhibitors (CKIs), and some transcription factors. Cyclin proteins are
found to be highly upregulated in cases of aberrant cell division in
cancer cells,particularly cyclins D and E.124,125
The precise mechanism
ofactionofPIs incancertreatment isnot well-understood.Intreatment
of breast cancer, 2 potential mechanisms have been described: the PI,
bortezomib, may inhibit cell proliferation and may decrease levels of
ER.126,127
Results from early preclinical and clinical trials have been
mixed, with 1 small study showing no effect of treatment with borte-
zomib and another showing enhanced MCF-7 tumor suppression with
bortezomib and trastuzumab.128
Data in MCF-7 breast cancer cells
suggest that fulvestrant’s antiestrogenic effects of induction of protea-
somal degradation of ER may involve c-SRC, a cellular kinase proto-
oncogene. In light of this potential interaction, a phase II study of
fulvestrant and bortezomib in women with locally advanced or MBC is
underway (NCT01142401).
Epigenetic Modifiers
In addition to genetic alterations such as the mutations and de-
letions that alter the composition of genomic DNA, epigenetic
changes including DNA methylation, histone modifications, and
chromatin remodeling can reversibly alter gene expression and play
a key role in tumorigenicity and metastatic potential of breast cancer
cells.129
Therapeutic interventions that target epigenetic modifica-
tions have been used to partially or completely revert the altered
gene expression, and several epigenetic agents that are approved for
use in other settings have potential as breast cancer therapies.
Stefan Glück
Clinical Breast Cancer Month 2016 - 9
Several clinical trials of these epigenetic agents are planned or
underway in the breast cancer setting. Azacitidine and decitabine are
hypomethylating agents that inhibit DNA methyltransferase,
thereby halting tumor cell growth and division through epigenetic
manipulation.130,131
Both agents have been approved for use in
myelodysplastic syndrome and are currently being investigated for
breast cancer treatment.130,131
In combination with fulvestrant,
azacitidine is being evaluated in a phase II trial in patients with
ERþ
, HER2À
MBC who have progressed on an AI
(NCT02374099). In addition, a class I HDAC inhibitor, entino-
stat, was assessed in a phase II trial (NCT00676663) with or
without the AI exemestane in patients with ERþ
breast cancer who
have progressed on an AI, and the combination was found to not
only improve PFS and OS, but restore sensitivity to hormone
therapy.132
Entinostat is currently being studied in combination
with azacitidine in a phase II trial of patients with either TNBC or
HRþ
HER2À
breast cancer (NCT01349959). Decitabine in
combination with the HDAC inhibitor panobinostat (LBH589)
was also being investigated in TNBC in an attempt to re-express
ER; however, the study was closed due to slow accrual
(NCT01194908).
Conclusions
ER-targeted therapy is important for many women with breast
cancer, but resistance to therapy inevitably occurs. The ER
signaling pathway is a complex network of extensive crosstalk with
growth-factor signaling pathways, cell cycle control pathways, and
protein degradation pathways. These pathways provide many
alternative targets for agents that may be useful in combination
with ET to decrease resistance to treatment and to extend benefit
to patients who do not achieve optimal benefit from ET alone. As
effective treatments are identified, an important area of focus must
be on the factors that determine optimal combination treatments
in subtypes of breast cancer. Such factors comprise not only the
responsiveness of the cells of a particular subgroup of tumors, but
also the characteristics of different treatment combinations with
respect to the temporal profile of effects, relative tolerability of
different combinations, and the compatibility of routes of
administration. Several therapies that are discussed in this review,
among them everolimus (mTOR inhibitor), palbociclib (CDK4/6
inhibitor), and MK-3475 (PD-1 inhibitor), are already proving to
be effective treatments for MBC and useful in reversing or pre-
venting the development of resistance to ET. Through inhibition
of central components of pathways for growth factor-promoted
cellular proliferation, cell cycle-mediated cellular proliferation,
and immunosuppressive programs that initiate and sustain tumor
growth, these agents are effective options that may extend survival
and maintain quality of life for women with MBC who have
relapsed on endocrine therapies.
Acknowledgments
Medical writing and editorial services were provided by SCI
Scientific Communications  Information, Parsippany, NJ, and
The Lockwood Group, Stamford, CT (funded by AstraZeneca LP).
Disclosure
This article was supported by AstraZeneca LP. Dr Glück has
received grant support and consulting fees from Agendia, Celgene,
Dara, Eisai, Genentech/Roche, GHI, Mylan, and Onyx/Amgen. At
the time of the initiation of manuscript development, Dr Glück was
the Sylvester Professor of Medicine, Department of Medicine, Di-
vision of Hematology Oncology, Sylvester Comprehensive Cancer
Center, Miami, FL. Dr Glück is a current employee and shareholder
of Celgene Corporation, Summit, NJ.
References
1. American Cancer Society. Cancer Facts  Figures 2016. Available at: http://
www.cancer.org/acs/groups/content/@research/documents/document/acspc-
047079.pdf. Accessed: May 24, 2016.
2. National Cancer Institute. Surveillance, Epidemiology, and End Results (SEER)
Fact Sheets: Breast. Available at: http://seer.cancer.gov/statfacts/html/breast.html.
Accessed: April 04, 2016.
3. Lu J, Steeg PS, Price JE, et al. Breast cancer metastasis: challenges and oppor-
tunities. Cancer Res 2009; 69:4951-3.
4. Brewster AM, Hortobagyi GN, Broglio KR, et al. Residual risk of breast cancer
recurrence 5 years after adjuvant therapy. J Natl Cancer Inst 2008; 100:1179-83.
5. Nadji M, Gomez-Fernandez C, Ganjei-Azar P, Morales AR. Immunohisto-
chemistry of estrogen and progesterone receptors reconsidered: experience with 5,
993 breast cancers. Am J Clin Pathol 2005; 123:21-7.
6. Chung CT, Carlson RW. Goals and objectives in the management of metastatic
breast cancer. Oncologist 2003; 8:514-20.
7. Gluck S, Arteaga CL, Osborne CK. Optimizing chemotherapy-free survival for
the ER/HER2-positive metastatic breast cancer patient. Clin Cancer Res 2011;
17:5559-61.
8. Hilton HN, Doan TB, Graham JD, et al. Acquired convergence of hormone
signaling in breast cancer: ER and PR transition from functionally distinct in
normal breast to predictors of metastatic disease. Oncotarget 2014; 5:8651-64.
9. National Comprehensive Cancer Network. Clinical Practice Guidelines in
Oncology. Breast Cancer.v.3.2015. Available at: http://www.nccn.org/
professionals/physician_gls/pdf/breast.pdf. Accessed: November 12, 2015.
10. Buzdar AU, Hortobagyi G. Update on endocrine therapy for breast cancer. Clin
Cancer Res 1998; 4:527-34.
11. Cardoso F, Fallowfield L, Costa A, Castiglione M, Senkus E, ESMO Guidelines
Working Group. Locally recurrent or metastatic breast cancer: ESMO Clinical
Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2011;
22(Suppl 6):vi25-30.
12. Nagaraj G, Ma C. Revisiting the estrogen receptor pathway and its role in
endocrine therapy for postmenopausal women with estrogen receptor-positive
metastatic breast cancer. Breast Cancer Res Treat 2015; 150:231-42.
13. Patani N, Martin LA. Understanding response and resistance to oestrogen
deprivation in ER-positive breast cancer. Mol Cell Endocrinol 2014; 382:683-94.
14. Miller WR, Bartlett JM, Canney P, Verrill M. Hormonal therapy for post-
menopausal breast cancer: the science of sequencing. Breast Cancer Res Treat
2007; 103:149-60.
15. Gluck S. Extending the clinical benefit of endocrine therapy for women with
hormone receptor-positive metastatic breast cancer: differentiating mechanisms of
action. Clin Breast Cancer 2014; 14:75-84.
16. Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer.
Annu Rev Med 2011; 62:233-47.
17. Pietras RJ. Biologic basis of sequential and combination therapies for hormone-
responsive breast cancer. Oncologist 2006; 11:704-17.
18. Colleoni M, Gelber S, Coates AS, et al. Influence of endocrine-related factors on
response to perioperative chemotherapy for patients with node-negative breast
cancer. J Clin Oncol 2001; 19:4141-9.
19. Wilcken N, Hornbuckle J, Ghersi D. Chemotherapy alone versus endocrine
therapy alone for metastatic breast cancer. Cochrane Database Syst Rev 2003; 2:
CD002747.
20. Gluck S, von Minckwitz G, Untch M. Aromatase inhibitors in the treatment of
elderly women with metastatic breast cancer. Breast 2013; 22:142-9.
21. Bedard PL, Freedman OC, Howell A, Clemons M. Overcoming endocrine
resistance in breast cancer: are signal transduction inhibitors the answer? Breast
Cancer Res Treat 2008; 108:307-17.
22. Miller WR, Larionov AA. Understanding the mechanisms of aromatase inhibitor
resistance. Breast Cancer Res 2012; 14:201.
23. Gutierrez MC, Detre S, Johnston S, et al. Molecular changes in tamoxifen-
resistant breast cancer: relationship between estrogen receptor, HER-2, and
p38 mitogen-activated protein kinase. J Clin Oncol 2005; 23:2469-76.
24. Arpino G, Green SJ, Allred DC, et al. HER-2 amplification, HER-1 expression,
and tamoxifen response in estrogen receptor-positive metastatic breast cancer: a
southwest oncology group study. Clin Cancer Res 2004; 10:5670-6.
Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer
10 - Clinical Breast Cancer Month 2016
25. Musgrove EA, Sutherland RL. Biological determinants of endocrine resistance in
breast cancer. Nat Rev Cancer 2009; 9:631-43.
26. Arpino G, Wiechmann L, Osborne CK, Schiff R. Crosstalk between the estrogen
receptor and the HER tyrosine kinase receptor family: molecular mechanism
and clinical implications for endocrine therapy resistance. Endocr Rev 2008; 29:
217-33.
27. Schiff R, Massarweh SA, Shou J, Bharwani L, Mohsin SK, Osborne CK. Cross-
talk between estrogen receptor and growth factor pathways as a molecular target
for overcoming endocrine resistance. Clin Cancer Res 2004; 10:331S-6S.
28. Frasor J, Stossi F, Danes JM, Komm B, Lyttle CR, Katzenellenbogen BS. Se-
lective estrogen receptor modulators: discrimination of agonistic versus antago-
nistic activities by gene expression profiling in breast cancer cells. Cancer Res
2004; 64:1522-33.
29. Klinge CM. Estrogen receptor interaction with estrogen response elements.
Nucleic Acids Res 2001; 29:2905-19.
30. Osborne CK, Bardou V, Hopp TA, et al. Role of the estrogen receptor coac-
tivator AIB1 (SRC-3) and HER-2/neu in tamoxifen resistance in breast cancer.
J Natl Cancer Inst 2003; 95:353-61.
31. Levin ER. Cell localization, physiology, and nongenomic actions of estrogen
receptors. J Appl Physiol (1985) 2001; 91:1860-7.
32. Lee AV, Cui X, Oesterreich S. Cross-talk among estrogen receptor, epidermal
growth factor, and insulin-like growth factor signaling in breast cancer. Clin
Cancer Res 2001; 7:4429s-35s, discussion: 4411s-4412s.
33. Yarden RI, Wilson MA, Chrysogelos SA. Estrogen suppression of EGFR
expression in breast cancer cells: a possible mechanism to modulate growth. J Cell
Biochem Suppl 2001; Suppl 36:232-46.
34. Guo S, Sonenshein GE. Forkhead box transcription factor FOXO3a regulates
estrogen receptor alpha expression and is repressed by the Her-2/neu/
phosphatidylinositol 3-kinase/Akt signaling pathway. Mol Cell Biol 2004; 24:
8681-90.
35. Lopez-Tarruella S, Schiff R. The dynamics of estrogen receptor status in breast
cancer: re-shaping the paradigm. Clin Cancer Res 2007; 13:6921-5.
36. Taneja P, Maglic D, Kai F, et al. Classical and novel prognostic markers for breast
cancer and their clinical significance. Clin Med Insights Oncol 2010; 4:15-34.
37. Bardou VJ, Arpino G, Elledge RM, Osborne CK, Clark GM. Progesterone re-
ceptor status significantly improves outcome prediction over estrogen receptor
status alone for adjuvant endocrine therapy in two large breast cancer databases.
J Clin Oncol 2003; 21:1973-9.
38. Cui X, Schiff R, Arpino G, Osborne CK, Lee AV. Biology of progesterone re-
ceptor loss in breast cancer and its implications for endocrine therapy. J Clin
Oncol 2005; 23:7721-35.
39. Mohammed H, Russell IA, Stark R, et al. Progesterone receptor modulates
ERalpha action in breast cancer. Nature 2015; 523:313-7.
40. Campagnoli C, Clavel-Chapelon F, Kaaks R, Peris C, Berrino F. Progestins and
progesterone in hormone replacement therapy and the risk of breast cancer.
J Steroid Biochem Mol Biol 2005; 96:95-108.
41. Farhat GN, Parimi N, Chlebowski RT, et al. Sex hormone levels and risk
of breast cancer with estrogen plus progestin. J Natl Cancer Inst 2013; 105:
1496-503.
42. Bines J, Dienstmann R, Obadia RM, et al. Activity of megestrol acetate in
postmenopausal women with advanced breast cancer after nonsteroidal aromatase
inhibitor failure: a phase II trial. Ann Oncol 2014; 25:831-6.
43. Gluck S, de Snoo F, Peeters J, Stork-Sloots L, Somlo G. Molecular subtyping of
early-stage breast cancer identifies a group of patients who do not benefit from
neoadjuvant chemotherapy. Breast Cancer Res Treat 2013; 139:759-67.
44. Chakraborty AK, Welsh A, Digiovanna MP. Co-targeting the insulin-like growth
factor I receptor enhances growth-inhibitory and pro-apoptotic effects of anti-
estrogens in human breast cancer cell lines. Breast Cancer Res Treat 2010; 120:
327-35.
45. Kern FG, McLeskey SW, Zhang L, et al. Transfected MCF-7 cells as a model for
breast-cancer progression. Breast Cancer Res Treat 1994; 31:153-65.
46. Ross JS, Fletcher JA. The HER-2/neu oncogene in breast cancer: prognostic
factor, predictive factor, and target for therapy. Stem Cells 1998; 16:413-28.
47. Arteaga CL, Sliwkowski MX, Osborne CK, Perez EA, Puglisi F, Gianni L.
Treatment of HER2-positive breast cancer: current status and future perspectives.
Nat Rev Clin Oncol 2012; 9:16-32.
48. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human
breast cancer: correlation of relapse and survival with amplification of the HER-2/
neu oncogene. Science 1987; 235:177-82.
49. De Laurentiis M, Arpino G, Massarelli E, et al. A meta-analysis on the interaction
between HER-2 expression and response to endocrine treatment in advanced
breast cancer. Clin Cancer Res 2005; 11:4741-8.
50. Westin GF, Perez CA, Wang E, Gluck S. Biologic impact and clinical implication
of mTOR inhibition in metastatic breast cancer. Int J Biol Markers 2013; 28:
233-41.
51. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a
monoclonal antibody against HER2 for metastatic breast cancer that over-
expresses HER2. N Engl J Med 2001; 344:783-92.
52. Incorvati JA, Shah S, Mu Y, Lu J. Targeted therapy for HER2 positive breast
cancer. J Hematol Oncol 2013; 6:38.
53. Herceptin (Trastuzumab) Intravenous Infusion [Package Insert]. South San
Francisco, CA: Genentech, Inc; 2010.
54. Kaufman B, Mackey JR, Clemens MR, et al. Trastuzumab plus anastrozole versus
anastrozole alone for the treatment of postmenopausal women with human
epidermal growth factor receptor 2-positive, hormone receptor-positive metastatic
breast cancer: results from the randomized phase III TAnDEM study. J Clin
Oncol 2009; 27:5529-37.
55. Huober J, Fasching PA, Barsoum M, et al. Higher efficacy of letrozole in com-
bination with trastuzumab compared to letrozole monotherapy as first-line
treatment in patients with HER2-positive, hormone-receptor-positive metasta-
tic breast cancer - results of the eLEcTRA trial. Breast 2012; 21:27-33.
56. Montemurro F, Valabrega G, Aglietta M. Lapatinib: a dual inhibitor of EGFR
and HER2 tyrosine kinase activity. Expert Opin Biol Ther 2007; 7:257-68.
57. Tykerb (Lapatinib) Tablets [Package Insert]. Research Triangle Park, NC:
GlaxoSmithKline; 2015.
58. Johnston S, Pippen J, Pivot X, et al. Lapatinib combined with letrozole versus
letrozole and placebo as first-line therapy for postmenopausal hormone receptor-
positive metastatic breast cancer. J Clin Oncol 2009; 27:5538-46.
59. Schwartzberg LS, Franco SX, Florance A, O’Rourke L, Maltzman J, Johnston S.
Lapatinib plus letrozole as first-line therapy for HER-2þ hormone receptor-
positive metastatic breast cancer. Oncologist 2010; 15:122-9.
60. Ahn ER, Wang E, Gluck S. Is the improved efficacy of trastuzumab and lapatinib
combination worth the added toxicity? A discussion of current evidence, rec-
ommendations, and ethical issues regarding dual HER2-targeted therapy. Breast
Cancer (Auckl) 2012; 6:191-207.
61. Blackwell KL, Burstein HJ, Storniolo AM, et al. Overall survival benefit with
lapatinib in combination with trastuzumab for patients with human epidermal
growth factor receptor 2-positive metastatic breast cancer: final results from the
EGF104900 Study. J Clin Oncol 2012; 30:2585-92.
62. Johnston S, Wroblewski S, Huang Y. ALTERNATIVE: safety and efficacy of
lapatinib (L), trastuzumab (T), or both in combination with an aromatase in-
hibitor (AI) for the treatment of hormone receptor positive (HRþ), human
epidermal growth factor receptor 2 positive (HER2þ) metastatic breast cancer
[abstract]. Cancer Res 2012; 72(24 Suppl):558s.
63. Perjeta (Pertuzumab) Injection [Package Insert]. South San Francisco, CA: Gen-
entech, Inc; 2016.
64. Baselga J, Gelmon KA, Verma S, et al. Phase II trial of pertuzumab and trastu-
zumab in patients with human epidermal growth factor receptor 2-positive
metastatic breast cancer that progressed during prior trastuzumab therapy.
J Clin Oncol 2010; 28:1138-44.
65. Swain SM, Baselga J, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel in
HER2-positive metastatic breast cancer. N Engl J Med 2015; 372:724-34.
66. Yang Y, Yee D. Targeting insulin and insulin-like growth factor signaling in
breast cancer. J Mammary Gland Biol Neoplasia 2012; 17:251-61.
67. Arteaga CL, Kitten LJ, Coronado EB, et al. Blockade of the type I somatomedin
receptor inhibits growth of human breast cancer cells in athymic mice. J Clin
Invest 1989; 84:1418-23.
68. Sachdev D, Li SL, Hartell JS, Fujita-Yamaguchi Y, Miller JS, Yee D. A chimeric
humanized single-chain antibody against the type I insulin-like growth factor
(IGF) receptor renders breast cancer cells refractory to the mitogenic effects of
IGF-I. Cancer Res 2003; 63:627-35.
69. Fagan DH, Uselman RR, Sachdev D, Yee D. Acquired resistance to tamoxifen is
associated with loss of the type I insulin-like growth factor receptor: implications
for breast cancer treatment. Cancer Res 2012; 72:3372-80.
70. Katso R, Okkenhaug K, Ahmadi K, White S, Timms J, Waterfield MD. Cellular
function of phosphoinositide 3-kinases: implications for development, homeo-
stasis, and cancer. Annu Rev Cell Dev Biol 2001; 17:615-75.
71. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the
PIK3CA gene in human cancers. Science 2004; 304:554.
72. Ellis MJ, Lin L, Crowder R, et al. Phosphatidyl-inositol-3-kinase alpha cata-
lytic subunit mutation and response to neoadjuvant endocrine therapy for
estrogen receptor positive breast cancer. Breast Cancer Res Treat 2010; 119:
379-90.
73. Miller TW, Rexer BN, Garrett JT, Arteaga CL. Mutations in the phosphatidy-
linositol 3-kinase pathway: role in tumor progression and therapeutic implications
in breast cancer. Breast Cancer Res 2011; 13:224.
74. Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S,
Nakshatri H. Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen
receptor alpha: a new model for anti-estrogen resistance. J Biol Chem 2001; 276:
9817-24.
75. Yamnik RL, Digilova A, Davis DC, Brodt ZN, Murphy CJ, Holz MK. S6 kinase
1 regulates estrogen receptor alpha in control of breast cancer cell proliferation.
J Biol Chem 2009; 284:6361-9.
76. Clark AS, West K, Streicher S, Dennis PA. Constitutive and inducible Akt ac-
tivity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast
cancer cells. Mol Cancer Ther 2002; 1:707-17.
77. Baselga J, Semiglazov V, van Dam P, et al. Phase II randomized study of neo-
adjuvant everolimus plus letrozole compared with placebo plus letrozole in
patients with estrogen receptor-positive breast cancer. J Clin Oncol 2009; 27:
2630-7.
78. Bachelot T, Bourgier C, Cropet C, et al. Randomized phase II trial of everolimus
in combination with tamoxifen in patients with hormone receptor-positive, hu-
man epidermal growth factor receptor 2-negative metastatic breast cancer with
prior exposure to aromatase inhibitors: a GINECO study. J Clin Oncol 2012; 30:
2718-24.
79. Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal
hormone-receptor-positive advanced breast cancer. N Engl J Med 2012; 366:
520-9.
Stefan Glück
Clinical Breast Cancer Month 2016 - 11
80. Yardley DA, Noguchi S, Pritchard KI, et al. Everolimus plus exemestane in
postmenopausal patients with HR(þ) breast cancer: BOLERO-2 final
progression-free survival analysis. Adv Ther 2013; 30:870-84.
81. Wolff AC, Lazar AA, Bondarenko I, et al. Randomized phase III placebo-
controlled trial of letrozole plus oral temsirolimus as first-line endocrine ther-
apy in postmenopausal women with locally advanced or metastatic breast cancer.
J Clin Oncol 2013; 31:195-202.
82. Dees EC, Carey LA. Improving endocrine therapy for breast cancer: it’s not that
simple. J Clin Oncol 2013; 31:171-3.
83. Khan KH, Yap TA, Yan L, Cunningham D. Targeting the PI3K-AKT-mTOR
signaling network in cancer. Chin J Cancer 2013; 32:253-65.
84. Friedman L, Ross L, Wallin J, et al. Selective PI3K and dual PI3K/mTOR in-
hibitors enhance the efficacy of endocrine therapies in breast cancer models
[abstract]. Cancer Res 2012; 72(24 suppl), P5-19-02.
85. Chen X, Zhao M, Hao M, et al. Dual inhibition of PI3K and mTOR mitigates
compensatory AKT activation and improves tamoxifen response in breast cancer.
Mol Cancer Res 2013; 11:1269-78.
86. Crowder RJ, Phommaly C, Tao Y, et al. PIK3CA and PIK3CB inhibition pro-
duce synthetic lethality when combined with estrogen deprivation in estrogen
receptor-positive breast cancer. Cancer Res 2009; 69:3955-62.
87. Saura C, Bendell J, Jerusalem G, et al. Phase Ib study of buparlisib plus trastu-
zumab in patients with HER2-positive advanced or metastatic breast cancer that
has progressed on Trastuzumab-based therapy. Clin Cancer Res 2014; 20:
1935-45.
88. Balko JM, Hicks M, Berger MF, et al. Genomic alterations indicative of a luminal
A subtype associate with clinical benefit to buparlisib and letrozole in endocrine-
resistant ERþ/HER2e metastatic breast cancer [abstract P3-07-41]. San Antonio
Breast Cancer Symposium. San Antonio, Texas; 2015.
89. Vilquin P, Villedieu M, Grisard E, et al. Molecular characterization of anastrozole
resistance in breast cancer: pivotal role of the Akt/mTOR pathway in the
emergence of de novo or acquired resistance and importance of combining the
allosteric Akt inhibitor MK-2206 with an aromatase inhibitor. Int J Cancer 2013;
133:1589-602.
90. Creighton CJ, Fu X, Hennessy BT, et al. Proteomic and transcriptomic profiling
reveals a link between the PI3K pathway and lower estrogen-receptor (ER) levels
and activity in ERþ breast cancer. Breast Cancer Res 2010; 12:R40.
91. Munzone E, Curigliano G, Rocca A, et al. Reverting estrogen-receptor-negative
phenotype in HER-2-overexpressing advanced breast cancer patients exposed to
trastuzumab plus chemotherapy. Breast Cancer Res 2006; 8:R4.
92. Xia W, Bacus S, Hegde P, et al. A model of acquired autoresistance to a potent
ErbB2 tyrosine kinase inhibitor and a therapeutic strategy to prevent its onset in
breast cancer. Proc Natl Acad Sci U S A 2006; 103:7795-800.
93. Finn RS. Targeting Src in breast cancer. Ann Oncol 2008; 19:1379-86.
94. Sprycel (Dasatinib) Tablets [Package Insert]. Princeton, NJ: Bristol-Myers Squibb
Company; 2014.
95. Mayer EL, Baurain JF, Sparano J, et al. A phase 2 trial of dasatinib in patients
with advanced HER2-positive and/or hormone receptor-positive breast cancer.
Clin Cancer Res 2011; 17:6897-904.
96. Wright GL, Blum J, Krekow LK, et al. Phase II trial of fulvestrant with or
without dasatinib in postmenopausal patients with hormone receptor-positive
metastatic breast cancer previously treated with an aromatase inhibitor [ab-
stract]. Clin Cancer Res 2011; 71(24 Suppl), PD01e01.
97. Caldon CE, Daly RJ, Sutherland RL, Musgrove EA. Cell cycle control in breast
cancer cells. J Cell Biochem 2006; 97:261-74.
98. Lundberg AS, Weinberg RA. Control of the cell cycle and apoptosis. Eur J Cancer
1999; 35:1886-94.
99. Altucci L, Addeo R, Cicatiello L, et al. Estrogen induces early and timed acti-
vation of cyclin-dependent kinases 4, 5, and 6 and increases cyclin messenger
ribonucleic acid expression in rat uterus. Endocrinology 1997; 138:978-84.
100. Said TK, Conneely OM, Medina D, O’Malley BW, Lydon JP. Progesterone, in
addition to estrogen, induces cyclin D1 expression in the murine mammary
epithelial cell, in vivo. Endocrinology 1997; 138:3933-9.
101. Watts CK, Brady A, Sarcevic B, deFazio A, Musgrove EA, Sutherland RL.
Antiestrogen inhibition of cell cycle progression in breast cancer cells in associated
with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma
protein phosphorylation. Mol Endocrinol 1995; 9:1804-13.
102. Lukas J, Bartkova J, Bartek J. Convergence of mitogenic signalling cascades from
diverse classes of receptors at the cyclin D-cyclin-dependent kinase-pRb-
controlled G1 checkpoint. Mol Cell Biol 1996; 16:6917-25.
103. Prall OW, Sarcevic B, Musgrove EA, Watts CK, Sutherland RL. Estrogen-
induced activation of Cdk4 and Cdk2 during G1-S phase progression is
accompanied by increased cyclin D1 expression and decreased cyclin-dependent
kinase inhibitor association with cyclin E-Cdk2. J Biol Chem 1997; 272:
10882-94.
104. Thangavel C, Dean JL, Ertel A, et al. Therapeutically activating RB: reestab-
lishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat
Cancer 2011; 18:333-45.
105. Finn RS, Dering J, Conklin D, et al. PD 0332991, a selective cyclin D kinase 4/6
inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive
human breast cancer cell lines in vitro. Breast Cancer Res 2009; 11:R77.
106. Ibrance (Palbociclib) Capsules [Package Insert]. New York, NY: Pfizer, Inc; 2015.
107. US Food and Drug Administration. FDA approves Ibrance for postmenopausal women
with advanced breast cancer 2015. Available at: http://www.fda.gov/NewsEvents/
Newsroom/PressAnnouncements/ucm432871.htm. Accessed: February 25, 2015.
108. Finn RS, Crown JP, Lang I, et al. Results of a randomized phase 2 study of PD
0332991, a cyclin-dependent kinase (CDK) 4/6 inhibitor, in combination with
letrozole vs letrozole alone for first-line treatment of ERþ/HER2- advanced
breast cancer (BC) [abstract S1e6]. Cancer Res 2012; 72(24 Suppl):91s.
109. Finn RS, Crown JP, Lang I, et al. Final results of a randomized phase II study of
PD 0332991, a cyclin-dependent kinase (CDK)-4/6 inhibitor, in combination
with letrozole vs letrozole alone for first-line treatment of ERþ/HER2- advanced
breast cancer (PALOMA-1; TRIO-18) [abstract CT101]. Cancer Res 2014; 74(19
Suppl):CT101.
110. Cristofanilli M, Turner NC, Bondarenko I, et al. Fulvestrant plus palbociclib
versus fulvestrant plus placebo for treatment of hormone-receptor-positive,
HER2-negative metastatic breast cancer that progressed on previous endocrine
therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3
randomised controlled trial. Lancet Oncol 2016; 17:425-39.
111. Finn RS, Martin M, Rugo HS, et al. PALOMA-2: Primary results from a phase
III trial of palbociclib (P) with letrozole (L) compared with letrozole alone in
postmenopausal women with ERþ/HER2e advanced breast cancer (ABC).
J Clin Oncol 2016; 34(Suppl), abstract 507.
112. Bardia A, Modi S, Oliveira M, et al. Triplet therapy with ribociclib, everolimus,
and exemestane in women with HRþ/HER2e advanced breast cancer [abstract
P6-13-01]. San Antonio Breast Cancer Symposium. San Antonio, Texas; 2015.
113. Blank C, Kuball J, Voelkl S, et al. Blockade of PD-L1 (B7-H1) augments human
tumor-specific T cell responses in vitro. Int J Cancer 2006; 119:317-27.
114. Yang W, Chen PW, Li H, Alizadeh H, Niederkorn JY. PD-L1: PD-1 interaction
contributes to the functional suppression of T-cell responses to human uveal
melanoma cells in vitro. Invest Ophthalmol Vis Sci 2008; 49:2518-25.
115. Hirano F, Kaneko K, Tamura H, et al. Blockade of B7-H1 and PD-1 by
monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res
2005; 65:1089-96.
116. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in
patients with metastatic melanoma. N Engl J Med 2010; 363:711-23.
117. Lu YC, Robbins PF. Cancer immunotherapy targeting neoantigens. Semin
Immunol 2016; 28:22-7.
118. Millis SZ, Gatalica Z, Winkler J, et al. Predictive biomarker profiling of  6000
breast cancer patients shows heterogeneity in TNBC, with treatment implica-
tions. Clin Breast Cancer 2015; 15:473-481e3.
119. Emens L, Adams S, Loi S, et al. A phase III randomized trial of atezolizumab in
combination with nab-paclitaxel as first line therapy for patients with metastatic
triple-negative breast cancer (mTNBC) [abstract OT1-01-06]. San Antonio
Breast Cancer Symposium. San Antonio, Texas; 2015.
120. Adams J. The proteasome: a suitable antineoplastic target. Nat Rev Cancer 2004;
4:349-60.
121. Adams J, Palombella VJ, Sausville EA, et al. Proteasome inhibitors: a novel class
of potent and effective antitumor agents. Cancer Res 1999; 59:2615-22.
122. Rivett AJ. Proteasomes: multicatalytic proteinase complexes. Biochem J 1993;
29(Pt 1):1-10.
123. Goldberg AL. Functions of the proteasome: the lysis at the end of the tunnel.
Science 1995; 268:522-3.
124. Diehl JA, Ponugoti B. Ubiquitin-dependent proteolysis in G1/S phase control
and its relationship with tumor susceptibility. Genes Cancer 2010; 1:717-24.
125. Masamha CP, Benbrook DM. Cyclin D1 degradation is sufficient to induce G1
cell cycle arrest despite constitutive expression of cyclin E2 in ovarian cancer cells.
Cancer Res 2009; 69:6565-72.
126. Jones MD, Liu JC, Barthel TK, et al. A proteasome inhibitor, bortezomib, in-
hibits breast cancer growth and reduces osteolysis by downregulating metastatic
genes. Clin Cancer Res 2010; 16:4978-89.
127. Powers GL, Ellison-Zelski SJ, Casa AJ, Lee AV, Alarid ET. Proteasome
inhibition represses ERalpha gene expression in ERþ cells: a new link be-
tween proteasome activity and estrogen signaling in breast cancer. Oncogene
2010; 29:1509-18.
128. Yeh WL, Shioda K, Coser KR, Rivizzigno D, McSweeney KR, Shioda T. Ful-
vestrant-induced cell death and proteasomal degradation of estrogen receptor
alpha protein in MCF-7 cells require the CSK c-Src tyrosine kinase. PLoS One
2013; 8:e60889.
129. Jeschke J, Collignon E, Fuks F. DNA methylome profiling beyond promoters -
taking an epigenetic snapshot of the breast tumor microenvironment. FEBS J
2015; 282:1801-14.
130. Dacogen (Decitabine) for Injection [Package Insert]. Rockville, MD: Otsuka
America Pharmaceutical, Inc; 2014.
131. Vidaza (Azacitidine) for Injection [Package Insert]. Summit, NJ: Celgene
Corporation; 2015.
132. Yardley DA, Ismail-Khan RR, Melichar B, et al. Randomized phase II, double-
blind, placebo-controlled study of exemestane with or without entinostat in
postmenopausal women with locally recurrent or metastatic estrogen receptor-
positive breast cancer progressing on treatment with a nonsteroidal aromatase
inhibitor. J Clin Oncol 2013; 31:2128-35.
Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer
12 - Clinical Breast Cancer Month 2016

More Related Content

What's hot

MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...
MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...
MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...European School of Oncology
 
Estrogen and breast cancer
Estrogen and breast cancerEstrogen and breast cancer
Estrogen and breast cancer
jamesnagel
 
DNA repair and prostate cancer
DNA repair and prostate cancerDNA repair and prostate cancer
DNA repair and prostate cancer
jamieritchey
 
Er pos and pr neg breast cancer
Er pos and pr neg breast cancerEr pos and pr neg breast cancer
Er pos and pr neg breast cancermadurai
 
Potential of Targeting Bone Metastases with Immunotherapies_Crimson Publishers
Potential of Targeting Bone Metastases with Immunotherapies_Crimson PublishersPotential of Targeting Bone Metastases with Immunotherapies_Crimson Publishers
Potential of Targeting Bone Metastases with Immunotherapies_Crimson Publishers
CrimsonpublishersCancer
 
Mol Cancer Ther-2015-Tonsing-Carter-2850-63
Mol Cancer Ther-2015-Tonsing-Carter-2850-63Mol Cancer Ther-2015-Tonsing-Carter-2850-63
Mol Cancer Ther-2015-Tonsing-Carter-2850-63Eva Tonsing-Carter
 
Triple-Negative Breast Cancer: 2018 Status Update
Triple-Negative Breast Cancer: 2018 Status UpdateTriple-Negative Breast Cancer: 2018 Status Update
Triple-Negative Breast Cancer: 2018 Status Update
Zeena Nackerdien
 
Molecular subtypes of breast cancer
Molecular subtypes of breast cancerMolecular subtypes of breast cancer
Molecular subtypes of breast cancerJoydeep Ghosh
 
Early breast updates
Early breast updatesEarly breast updates
Early breast updatesAhmed Allam
 
Gene Profiling in Clinical Oncology - Slide 8 - G. Viale - Classic pathology...
Gene Profiling in Clinical Oncology - Slide 8 -  G. Viale - Classic pathology...Gene Profiling in Clinical Oncology - Slide 8 -  G. Viale - Classic pathology...
Gene Profiling in Clinical Oncology - Slide 8 - G. Viale - Classic pathology...European School of Oncology
 
Adjuvant endocrine therapy in breast cancer
Adjuvant endocrine therapy in breast cancer   Adjuvant endocrine therapy in breast cancer
Adjuvant endocrine therapy in breast cancer
Mamdouh Sabry
 
Dr arun Triple Negative Breast cancer Presentation
Dr arun Triple Negative Breast cancer PresentationDr arun Triple Negative Breast cancer Presentation
Dr arun Triple Negative Breast cancer Presentation
Arun Shahi MD,MPH
 
A Medical Device Strategy to Inhibit HER2+ Breast Cancer Progression
A Medical Device Strategy to Inhibit HER2+ Breast Cancer ProgressionA Medical Device Strategy to Inhibit HER2+ Breast Cancer Progression
A Medical Device Strategy to Inhibit HER2+ Breast Cancer Progression
Aethlon Medical, Inc.
 
Journal club- breast ca basal like
Journal club- breast ca basal likeJournal club- breast ca basal like
Journal club- breast ca basal like
Sakshi Gupta
 
Advances in the management of breast cancer
Advances in the management of breast cancerAdvances in the management of breast cancer
Advances in the management of breast cancer
Mohamed Abdulla
 
Tnbc 2018 update
Tnbc 2018 updateTnbc 2018 update
Tnbc 2018 update
Pratik patil
 
Prognostic & predictive factors of breast cancer
Prognostic & predictive factors of breast cancerPrognostic & predictive factors of breast cancer
Prognostic & predictive factors of breast cancer
Mohammed Fathy
 

What's hot (20)

MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...
MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...
MCO 2011 - Slide 9 - G. Curigliano - Joint medics and nurses spotlight sessio...
 
Russell
RussellRussell
Russell
 
Estrogen and breast cancer
Estrogen and breast cancerEstrogen and breast cancer
Estrogen and breast cancer
 
DNA repair and prostate cancer
DNA repair and prostate cancerDNA repair and prostate cancer
DNA repair and prostate cancer
 
Er pos and pr neg breast cancer
Er pos and pr neg breast cancerEr pos and pr neg breast cancer
Er pos and pr neg breast cancer
 
Potential of Targeting Bone Metastases with Immunotherapies_Crimson Publishers
Potential of Targeting Bone Metastases with Immunotherapies_Crimson PublishersPotential of Targeting Bone Metastases with Immunotherapies_Crimson Publishers
Potential of Targeting Bone Metastases with Immunotherapies_Crimson Publishers
 
Mol Cancer Ther-2015-Tonsing-Carter-2850-63
Mol Cancer Ther-2015-Tonsing-Carter-2850-63Mol Cancer Ther-2015-Tonsing-Carter-2850-63
Mol Cancer Ther-2015-Tonsing-Carter-2850-63
 
Triple-Negative Breast Cancer: 2018 Status Update
Triple-Negative Breast Cancer: 2018 Status UpdateTriple-Negative Breast Cancer: 2018 Status Update
Triple-Negative Breast Cancer: 2018 Status Update
 
Molecular subtypes of breast cancer
Molecular subtypes of breast cancerMolecular subtypes of breast cancer
Molecular subtypes of breast cancer
 
Early breast updates
Early breast updatesEarly breast updates
Early breast updates
 
Gene Profiling in Clinical Oncology - Slide 8 - G. Viale - Classic pathology...
Gene Profiling in Clinical Oncology - Slide 8 -  G. Viale - Classic pathology...Gene Profiling in Clinical Oncology - Slide 8 -  G. Viale - Classic pathology...
Gene Profiling in Clinical Oncology - Slide 8 - G. Viale - Classic pathology...
 
Cancer lecture
Cancer lectureCancer lecture
Cancer lecture
 
journal.pone.0061537
journal.pone.0061537journal.pone.0061537
journal.pone.0061537
 
Adjuvant endocrine therapy in breast cancer
Adjuvant endocrine therapy in breast cancer   Adjuvant endocrine therapy in breast cancer
Adjuvant endocrine therapy in breast cancer
 
Dr arun Triple Negative Breast cancer Presentation
Dr arun Triple Negative Breast cancer PresentationDr arun Triple Negative Breast cancer Presentation
Dr arun Triple Negative Breast cancer Presentation
 
A Medical Device Strategy to Inhibit HER2+ Breast Cancer Progression
A Medical Device Strategy to Inhibit HER2+ Breast Cancer ProgressionA Medical Device Strategy to Inhibit HER2+ Breast Cancer Progression
A Medical Device Strategy to Inhibit HER2+ Breast Cancer Progression
 
Journal club- breast ca basal like
Journal club- breast ca basal likeJournal club- breast ca basal like
Journal club- breast ca basal like
 
Advances in the management of breast cancer
Advances in the management of breast cancerAdvances in the management of breast cancer
Advances in the management of breast cancer
 
Tnbc 2018 update
Tnbc 2018 updateTnbc 2018 update
Tnbc 2018 update
 
Prognostic & predictive factors of breast cancer
Prognostic & predictive factors of breast cancerPrognostic & predictive factors of breast cancer
Prognostic & predictive factors of breast cancer
 

Similar to Gluck-Clin Breast Cancer-2016

3478-43782-3-PB
3478-43782-3-PB3478-43782-3-PB
3478-43782-3-PB东妮 郑
 
DNA TRANSCRIPTION
DNA TRANSCRIPTION DNA TRANSCRIPTION
DNA TRANSCRIPTION
saratoro20
 
Clinical Utility of Pharmacogenetic Testing in Compounding Pharmacy
Clinical Utility of Pharmacogenetic Testing in Compounding PharmacyClinical Utility of Pharmacogenetic Testing in Compounding Pharmacy
Clinical Utility of Pharmacogenetic Testing in Compounding PharmacyBrian Fichter
 
Opportunities and challenges provided by crosstalk between signalling pathway...
Opportunities and challenges provided by crosstalk between signalling pathway...Opportunities and challenges provided by crosstalk between signalling pathway...
Opportunities and challenges provided by crosstalk between signalling pathway...Anirudh Prahallad
 
Unfortunately several cancers are not predictable with simple tests .pdf
Unfortunately several cancers are not predictable with simple tests .pdfUnfortunately several cancers are not predictable with simple tests .pdf
Unfortunately several cancers are not predictable with simple tests .pdf
aquastore223
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
JohnJulie1
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
EditorSara
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
NainaAnon
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
EditorSara
 
Triple_Negative_Breast_Cancer_Is_it_Poss.pdf
Triple_Negative_Breast_Cancer_Is_it_Poss.pdfTriple_Negative_Breast_Cancer_Is_it_Poss.pdf
Triple_Negative_Breast_Cancer_Is_it_Poss.pdf
semualkaira
 
Role of progesterone receptor in bc
Role of progesterone receptor in bcRole of progesterone receptor in bc
Role of progesterone receptor in bc
Aleena Haqqi
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
daranisaha
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
eshaasini
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
semualkaira
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
semualkaira
 

Similar to Gluck-Clin Breast Cancer-2016 (16)

3478-43782-3-PB
3478-43782-3-PB3478-43782-3-PB
3478-43782-3-PB
 
DNA TRANSCRIPTION
DNA TRANSCRIPTION DNA TRANSCRIPTION
DNA TRANSCRIPTION
 
Clinical Utility of Pharmacogenetic Testing in Compounding Pharmacy
Clinical Utility of Pharmacogenetic Testing in Compounding PharmacyClinical Utility of Pharmacogenetic Testing in Compounding Pharmacy
Clinical Utility of Pharmacogenetic Testing in Compounding Pharmacy
 
Opportunities and challenges provided by crosstalk between signalling pathway...
Opportunities and challenges provided by crosstalk between signalling pathway...Opportunities and challenges provided by crosstalk between signalling pathway...
Opportunities and challenges provided by crosstalk between signalling pathway...
 
Unfortunately several cancers are not predictable with simple tests .pdf
Unfortunately several cancers are not predictable with simple tests .pdfUnfortunately several cancers are not predictable with simple tests .pdf
Unfortunately several cancers are not predictable with simple tests .pdf
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 
Triple_Negative_Breast_Cancer_Is_it_Poss.pdf
Triple_Negative_Breast_Cancer_Is_it_Poss.pdfTriple_Negative_Breast_Cancer_Is_it_Poss.pdf
Triple_Negative_Breast_Cancer_Is_it_Poss.pdf
 
Role of progesterone receptor in bc
Role of progesterone receptor in bcRole of progesterone receptor in bc
Role of progesterone receptor in bc
 
Black cohosh research
Black cohosh researchBlack cohosh research
Black cohosh research
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
Clinic Correlation and Prognostic Value of P4HB and GRP78 Expression in Gastr...
 

Gluck-Clin Breast Cancer-2016

  • 1. Review Consequences of the Convergence of Multiple Alternate Pathways on the Estrogen Receptor in the Treatment of Metastatic Breast Cancer Stefan Glück Abstract Endocrine therapy is the usual first-line therapy for patients with hormone receptor-positive metastatic breast cancer. However, resistance to hormone therapies frequently occurs during the course of treatment. Growing understanding of the signal cascade of estrogen receptors and the signaling pathways that interact with estrogen receptors has revealed the complex role of these receptors in cell growth and proliferation, and on the mechanism in development of resistance. These insights have led to the development of targeted therapies that may prove to be effective options for the treatment of breast cancer and may overcome hormone therapy resistance. This article reviews current under- standing of the cellular receptor signaling pathways that interact with estrogen receptors. It also reviews data from recent ongoing clinical trials that examine the effects of targeted therapies, which might interfere with estrogen re- ceptor pathways and might reduce or reverse resistance to traditional, sequential, single-agent endocrine therapy. Clinical Breast Cancer, Vol. -, No. -, --- ª 2016 The Author. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Keywords: Cyclin-dependent kinases, Endocrine therapy, Hormone receptor-positive, PI3K/Akt/mTOR pathway, Programmed cell death inhibitor Introduction Breast cancer is the most common malignancy and the second leading cause of cancer-related death among women in the United States according to 2016 estimates.1 Approximately 6% of cases are advanced or metastatic breast cancer (MBC) at diagnosis, and, for those diagnosed at early stages, recurrence to distant sites occurs in 20% to 30%.2-4 Of patients with breast cancer, approximately 75% have hormone receptor-positive (HRþ ) tumors.5 Therapies for MBC are aimed at palliation of symptoms with improvement or conservation of quality of life and, potentially, pro- longation of survival; unfortunately, prolonged survival is observed in only a small percentage of patients.6,7 Both hormone receptor status (eg, estrogen receptor [ER] and progesterone receptor [PgR]) and human epidermal growth factor 2 (HER2) status are important pre- dictive markers for treatment efficacy.7,8 Patients with HRþ MBC are candidates for initial endocrine therapy (ET).9 In premenopausal women with MBC, various endocrine treatments are employed, including ovarian suppression or oophorectomy, selective ER mod- ulators such as tamoxifen, aromatase inhibitors (AIs) in conjunction with ovarian suppression, and the selective ER downregulator, ful- vestrant, all of which have different mechanisms of action. These mechanisms have been reviewed extensively, demonstrating the diversity of approaches that ultimately antagonize the growth- promoting effects of estrogen on breast cancer cells. These mecha- nistic differences are also linked to dissimilarities in resistance mechanisms and thus influence treatment selection, particularly in the context of sequencing and the use of combination regimens.10-17 In the setting of postmenopausal HRþ MBC, ET has a better tolerability profile and longer time to progression (TTP) than chemotherapy.18,19 In clinical trials of AIs employed for first- or second-line treatment of postmenopausal patients with MBC, AIs have greater efficacy than either tamoxifen or megestrol acetate.20 Resistance to ET in HRþ MBC is common, and given sufficient time, most patients are faced with disease progression.21,22 The mechanisms underlying disease progression and the development of resistance to endocrine treatment are complex and not fully un- derstood. Nevertheless, over the past several years, insights into several pathways of resistance have grown and have led to increased understanding of the clinical value of sequential lines of therapy and co-targeting strategies. While receiving ET, up to 20% of patients Department of Medicine, Division of Hematology Oncology, Sylvester Comprehensive Cancer Center, Miami, FL Submitted: Jun 17, 2016; Revised: Aug 1, 2016; Accepted: Aug 14, 2016 Address for correspondence: Stefan Glück, MD, PhD, FRCPC, Sylvester Professor of Medicine, Department of Medicine, Division of Hematology Oncology, Sylvester Comprehensive Cancer Center, Miami, FL 33136 E-mail contact: sgluck1@mac.com 1526-8209/ª 2016 The Author. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). http://dx.doi.org/10.1016/j.clbc.2016.08.004 Clinical Breast Cancer Month 2016 - 1
  • 2. experience a loss of ERs with concomitant loss of response to es- trogen.23 Amplified expression of HER2 may reduce ER levels and serve as an alternative pathway for tumor cell survival.23,24 Cellular signaling of growth-factor receptors interacts with ER nuclear and nonnuclear pathways and may contribute to resistance through posttranslational modifications of ER and/or coregulators.25-27 Mutations and/or aberrant forms of ER and their coregulators may also be a primary cause or contribute to the development of resistance to ET.21,22,25 The aim of this review is to describe emerging information on cellular pathways that interact with ER and impact cell proliferation and development of resistance to ET. We will discuss the biologic rationale for combining ET with agents that target these pathways and review the current status of clinical trials that are investigating the effects of combination treatments (Table 1). We also illustrate the cellular receptors and intracellular signaling pathways that interact with ER (Figure 1). Estrogen-Receptor Signaling ER functions through 2 separate but interrelated pathways: nu- clear (genomic) and nonnuclear (nongenomic) pathways. The nu- clear pathway mediates the effects of ER on genomic activity, thereby altering expression of many genes involved in physiologic cell function and in abnormal cell proliferation, resulting in tumorigenesis.28 The ER nuclear pathway is activated by estrogen binding, receptor dimerization and translocation to the nucleus, interaction with coregulator proteins (both coactivators and co- repressors), and through estrogen response elements modulation of gene transcription.29 ER coregulators have been implicated in the pathogenesis of breast cancer. Most notable is amplified in breast cancer 1, which, as its name implies, is overexpressed in breast cancers. Moreover, amplified in breast cancer 1 overexpression may play a role in resistance to ET.30 The nonnuclear pathway of ER function (originating in cellular cytoplasm) is mediated by estrogen- bound ER acting outside the cell nucleus through interaction with multiple cellular signaling pathways, tyrosine kinases, and membrane-bound growth factor-receptor pathways, including HER2, insulin-like growth factor-1 receptor (IGF1R), and fibro- blast growth factor receptor (FGFR).31,32 Multiple levels of inter- action or crosstalk between ER and growth factor and tyrosine kinase pathways may contribute to ER actions. These interactions include modulation of ER activity, upregulation of competing pathways and development of resistance to ET, and alternative or escape pathways for cellular proliferation and tumorigenesis. Estrogen can modify the activity of growth factor pathways by increasing levels of growth factors such as transforming growth factor- a and IGF1R and, alternatively, by altering the expression of epidermal growth factor receptors (EGFR).27,32,33 Conversely, acti- vation of growth factor-receptor signaling pathways may down- regulate expression of ER, resulting in decreased estrogen effects.34,35 This crosstalk, which can up- or downregulate competing pathways between ER and growth factor-receptor signaling, is recognized as a major mechanism of ET resistance, and implies that ER-positive (ERþ )/HER2-positive (HER2þ ) breast cancer should be treated with a combination of ET and HER2 inhibitors or antagonists.36 In addition to ER and HER2 status, assessment of PgR status is also typically used to characterize MBC and shape treatment decisions. In normal breast tissue, ER and PgR appear to be expressed in different epithelial populations and regulate distinct pathways; estrogen is involved in extracellular signaling and progesterone is associated with cell growth.8 This independent activity is altered in breast cancer cells where ER and PgR expression become correlated and converge on pathways that promote tumor growth and metastasis, and both PgR and ER become regulated by estrogen.8 Although PgR in ERþ patients with MBC has been used as a predictor of response to ET, it is controversial and not as well-accepted as the role of ER.37 Data indicate that ERþ /PgR-negative (PgRÀ ) breast tumors are not as responsive to selective ER modulator therapy as ERþ /PgR- positive (PgRþ ) tumors. One proposed mechanism for this anties- trogen resistance involves aberrant growth factor signaling (a marker for loss of PgR), but other unknown contributory mechanisms are also likely to play a role. Further research is needed to identify such mechanisms and thereby refine therapeutic strategies.38 In a recent study of MBC, PgR was found to associate with ER in the presence of agonist ligands, and the ER-PgR ligand-activated complex acted to modify chromatin-binding events and gene tran- scription, leading to an antitumorigenic effect.39 Stimulation with progesterone in an estrogen-rich context induced interactions be- tween known ER cofactors and PgR, but did not alter the association of those cofactors with ER, yielding an ER-PgR binding complex that promoted cell death, apoptosis, and differentiation pathways.39 This finding, coupled with the association between PgR gene (PgR) copy number loss and poorer clinical outcome possibly owing to a reduc- tion in ER-PgR binding complex formation, suggests a more intricate relationship beyond PgR as a simple marker for ER pathway function in the MBC setting.39 Although this contrasts with hormone replacement studies in which synthetic progesterone (medrox- yprogesterone) was associated with an increased breast cancer risk, it was found that natural progesterone did not have the same effect, which suggests different mechanisms of action for synthetic ago- nists.40,41 Interestingly, semi-synthetic progestin, megestrol acetate, provided clinical benefit to ERþ patients with MBC who experienced disease progression after estrogen suppression with a nonsteroidal AI.42 Molecular subtyping of tumors may offer additional insight into treatment of early-stage and locally advanced breast cancer. In a study that evaluated molecular subtype and diagnostic classification of the 154 patients classified as Luminal type B (high risk), a group that is typically sensitive to chemotherapy, 145 were ERþ and 99 were PgRþ tumors.43 Among the Luminal B patients, pathologic complete response was 85%, suggesting a possible association between ER/PgR status and the benefit of chemotherapy.43 Clearly the complex rela- tionship between ER and PgR depends on many factors, including genomic alteration, interaction of cofactors at a transcriptional level, relative levels of estrogen and progesterone in the tumor cell envi- ronment, and possibly variations in conformational changes related to the form of the agonist. Growth-Factor Receptors Growth-factor receptors, particularly receptor tyrosine kinases, play an integral role in growth promotion, cellular proliferation, and tumorigenesis. Growth-factor pathways may act as ER-independent drivers of tumor growth and survival, contributing to resistance to all types of ET. HER2, IGF1R, and FGFR have been implicated in resistance.26,44,45 2 - Clinical Breast Cancer Month 2016 Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer
  • 3. Table 1 Clinical Trials Investigating the Effects of Combination Treatments in Breast Cancer (Available at https://clinicaltrials.gov/) NCT Identifier (Acronym) Study Agents Phase Study Design Selected Clinical Outcomes Start Date/Estimated Completion Growth-factor receptor TK inhibitors NCT00634634 Sorafenib/letrozole I/II Open-label, single-arm Postmenopausal, HRþ advanced/MBC Phase I Primary: RPTD Phase II Primary: CBR Phase I/II Secondary: TTP, OS, safety August 2008/December 2015 NCT00752986 (ZACFAST) Vandetanib/fulvestrant Placebo/fulvestrant II Randomized, double-blind Placebo-controlled Postmenopausal/advanced BC Primary: Event-free survival Secondary: TTP, PFS, OS, safety December 2008/September 2013 NCT01528345 Dovitinib/fulvestrant Placebo/fulvestrant II Randomized, double-blind Placebo-controlled Postmenopausal/HRþ /HER2À advanced/MBC Primary: PFS Secondary: ORR, DOR, OS, safety, ECOG performance April 2012/October 2015 NCT02053636 (FINESSE) Lucitanib II Open-label, single-arm HRþ MBC Primary: ORR December 2013/July 2016 NCT00305825 Bevacizumab/letrozole II Open-label, single-arm Postmenopausal/HRþ Unresectable advanced/MBC Primary: Safety Secondary: ORR, CBR August 2004/November 2017 NCT00390455 Fulvestrant/lapatinib Fulvestrant/placebo III Randomized, double-blind Placebo-controlled Postmenopausal/HRþ advanced/MBC Primary: PFS Secondary: OS, ORR, DOR, OS, safety, QOL September 2006/July 2014 NCT01160211 AI/trastuzumab/lapatinib AI/trastuzumab AI/lapatinib III Randomized, open-label, multiple-arm Postmenopausal/HRþ /HER2þ , advanced/MBC; prior trastuzumab and ET Primary: OS Secondary: PFS, ORR, CBR, OS, safety, QOL, TTP, DOR May 2011/December 2017 PI3K/Akt/mTOR inhibitors NCT01248494 BEZ235/letrozole BKM120/letrozole BKM120 (intermittent)/letrozole Ib Open-label, multiple-arm Postmenopausal, HRþ MBC Primary: MTD BEZ235 or BKM120 Secondary: PFS, ORR November 2010/October 2013 NCT01344031 MK-2206/anastrozole MK-2206/fulvestrant MK-2206/anastrozole/ fulvestrant I Open-label, multiple-arm Postmenopausal, HRþ advanced/MBC Primary: MTD MK-2206, RPTD for all agents Secondary: Safety, CBR April 2011/October 2013 NCT01339442 BKM120/fulvestrant I Open-label, multiple-arm Postmenopausal, HRþ advanced/MBC Primary: MTD BKM120 safety Secondary: ORR, PK November 2011/December 2014 NCT01791478 Letrozole/BYL719 Ib Open-label, single-arm Postmenopausal HRþ MBC Primary: MTD BYL719 Secondary: CBR, PFS, ORR, Safety April 2013/March 2016 NCT01797120 Fulvestrant/everolimus Fulvestrant/placebo II Randomized, double-blind, placebo-controlled Postmenopausal HRþ MBC, AI refractory Primary: PFS Secondary: Safety, ORR, TTP, OS May 2013/November 2015 StefanGlück ClinicalBreastCancerMonth2016-3
  • 4. Table 1 Continued NCT Identifier (Acronym) Study Agents Phase Study Design Selected Clinical Outcomes Start Date/Estimated Completion NCT02049957 Exemestane/MLN0128 Fulvestrant/MLN0128 IIb/II Open-label, multiple-arm Postmenopausal HRþ /HER2À , advanced/MBC, everolimus refractory Primary: Safety, CBR Secondary: ORR, tumor size, PK-PD, PFS, OS February 2014/June 2016 NCT01992952 (FAKTION) Fulvestrant/AZD5363 Fulvestrant/placebo Ib/II Randomized, double-blind, placebo-controlled Postmenopausal HRþ , advanced/MBC Phase Ib Primary: MTD AZD5363 Phase II Primary: PFS Phase Ib/II Secondary: Safety, ORR, OS May 2014/August 2016 NCT01698918 (BOLERO-4) Letrozole/everolimus (1st line) Exemestane/everolimus (2nd line) II Open-label, single-arm Postmenopausal HRþ /HER2À , advanced/MBC Primary: PFS after 1st line Secondary: ORR, OS, CBR, safety, PFS after 2nd line March 2013/October 2016 NCT01296555 GDC0032 only Letrozole/GDC0032 Fulvestrant/GDC0032 I/II Open-label, multiple-arm Phase I advanced/metastatic solid tumors Phase II: Postmenopausal/HRþ advanced/MBC Phase I Primary: Safety Phase II: Fulvestrant/GDC0032 Secondary: ORR, DOR, PFS Phase II Primary: CBR, ORR Secondary: DOR, PFS, OS March 2011/July 2017 NCT01437566 Fulvestrant/GDC0941 Fulvestrant/GDC0980 Fulvestrant/placebo II Randomized, double-blind, placebo-controlled Postmenopausal HRþ , MBC, AI refractory Primary: PFS, safety Secondary: ORR, CBR, DOR, PK October 2011/November 2015 NCT02035813 (DETECT IV) Everolimus/standard ET II Open-label, single-arm Postmenopausal HRþ /HER2À , advanced/MBC Primary: PFS Secondary: ORR, DCR, OS January 2014/December 2019 NCT01610284 (BELLE-2) Fulvestrant/BKM120 Fulvestrant/placebo III Randomized, double-blind, placebo-controlled Postmenopausal HRþ , MBC, AI refractory Primary: PFS Secondary: OS, ORR, CBR, safety, PK August 2012/March 2017 NCT01633060 (BELLE-3) Fulvestrant/BKM120 Fulvestrant/placebo III Randomized, double-blind, placebo-controlled Postmenopausal HRþ /HER2À , advanced/MBC, AI treated, progressed on/after mTOR Primary: PFS Secondary: OS, ORR, CBR, safety, PK October 2012/March 2017 Src inhibitors NCT00696072 Letrozole/dasatinib Letrozole only II Open-label, multiple-arm Postmenopausal HRþ /HER2À , advanced/MBC Primary: CBR Secondary: ORR, PFS, TTF, safety August 2008/February 2015 CDK 4/6 Inhibitors NCT01857193 Exemestane Everolimus/LEE011 (palbociclib) Exemestane/everolimus Exemestane/LEE011 (palbociclib) Ib/II Open-label, multiple arm Postmenopausal, HRþ /HER2À advanced/MBC Phase I Primary: Safety Phase II Primary: PFS Phase Ib/II Secondary: Safety, PK, ORR, DOR, OS, DCR September 2013/July 2016 NCT00721409 Letrozole/palbociclib Letrozole only I/II Open-label, multiple-arm Postmenopausal HRþ /HER2À , advanced/MBC Phase I Primary: Safety Phase I Secondary: PK, QTc, antitumor activity Phase II Primary: PFS Phase II Secondary: Antitumor activity, OS, safety September 2008/July 2018 AlternatePathwaysonEstrogenReceptorinMetastaticBreastCancer 4-ClinicalBreastCancerMonth2016
  • 5. Table 1 Continued NCT Identifier (Acronym) Study Agents Phase Study Design Selected Clinical Outcomes Start Date/Estimated Completion NCT02088684 LEE011/BKM120/ Fulvestrant LEE011/BYL719/ Fulvestrant LEE011/fulvestrant IIb/II Open-label, multiple-arm Postmenopausal HRþ /HER2À , advanced/MBC Phase Ib Primary: Dose-limiting toxicity Phase II Primary: PFS Phase Ib/II Secondary: Safety, PK, ORR, DOR, PFS (Phase I only), OS (Phase II only) May 2014/February 2019 NCT01723774 Postmenopausal: Anastrozole/ palbociclib Premenopausal: Goserelin/ palbociclib II Neoadjuvant, open-label, single-arm Pre/postmenopausal HRþ /HER2À , Stage 2/3 BC Primary: Complete cell cycle arrest (Ki672.7%) Secondary: PEPi 0 score, response- clinical, radiological, Ki67 level, pCR rate, PK, safety June 2013/August 2015 NCT02040857 Palbociclib/standard ET II Open-label, single-arm Pre/postmenopausal HRþ /HER2À , Stage 2/3 BC Primary: Treatment DC rate January 2014/June 2019 NCT01942135 (PALOMA-3) Fulvestrant/palbociclib Fulvestrant/placebo III Randomized, double-blind, placebo-controlled Postmenopausal HRþ /HER2À , MBC, ET refractory Primary: PFS Secondary: OS, ORR, DOR, CBR, QOL, TTD September 2013/January 2017 NCT01958021 (MONALEESA-2) Letrozole/LEE011 Letrozole/placebo III Randomized, double-blind, placebo-controlled Postmenopausal HRþ /HER2À , advanced/MBC, therapy-naive Primary: PFS Secondary: OS, ORR, CBR, safety, QOL, QTc December 2013/January 2017 PD-1 inhibitors NCT02129556 (PANACEA) MK-3475/trastuzumab I/II Open-label, single-arm HER2þ , advanced/MBC, trastuzumab refractory Primary: Dose-limiting toxicity, RPTD Secondary: Safety, CBR, DOR, TTP, PFS, OS July 2014/July 2023 Proteasome inhibitors NCT01142401 Fulvestrant/bortezomib Fulvestrant only II Randomized, double-blind Postmenopausal HRþ , advanced/MBC Primary: PFS Secondary: CBR, OS, safety May 2010/June 2014 Epigenetic modifiers NCT02374099 Oral azacitidine/fulvestrant Fulvestrant only II Randomized, open-label Postmenopausal ERþ , HER2À MBC who progressed on an AI Primary: PFS Secondary: ORR, CBR, OS, DOR, safety March 2015/February 2018 NCT00676663 (ENCORE301) Exemestane/entinostat Exemestane only II Randomized, double-blind Postmenopausal ERþ , locally recurrent or MBC who progressed on an AI Primary: PFS Secondary: ORR, CBR, safety May 2008/October 2012 NCT01349959 Azacitidine/entinostat II Randomized, open-label TNBC or HRþ /HER2À , advanced/MBC Primary: Confirmed response rate by RECIST Secondary: CBR, OS, PFS, Safety April 2011/March 2014 NCT01194908 Decitabine/panobinostat/tamoxifen I/II Randomized, open-label TNBC, MBC or locally advanced Primary: MTD Secondary: Safety July 2010/January 2014 Abbreviations: AI ¼ aromatase inhibitor; BC ¼ breast cancer; CBR ¼ clinical benefit rate; DC ¼ discontinuation rate; DOR ¼ duration of response; ECOG ¼ Eastern Cooperative Oncology Group; ET ¼ endocrine therapy; HER2þ /HER2À ¼ human epidermal growth factor receptor-2 positive/negative; HRþ ¼ hormone receptor-positive; MBC ¼ metastatic breast cancer; MTD ¼ maximum tolerated dose; mTOR ¼ mammalian target of rapamycin inhibitor; ORR ¼ overall response rate; OS ¼ overall survival; pCR ¼ pathologic complete response; PD ¼ pharmacodynamics; PD-1 ¼ programmed cell death protein 1; PFS ¼ progression-free survival; PK ¼ pharmacokinetics; QOL ¼ quality of life; QTc ¼ corrected QT interval; RPTD ¼ recommended phase II dose; TK ¼ tyrosine kinase; TNBC ¼ triple negative breast cancer; TTD ¼ time to deterioration; TTF ¼ time to treatment failure; TTP ¼ time to progression. StefanGlück ClinicalBreastCancerMonth2016-5
  • 6. In breast cancer, HER2, a member of the EGFR family, is an important independent pathway for cell proliferation.46 HER2 is constitutively in an active conformation that allows dimerization with itself (homodimerization) or other members of the EGFR family (heterodimerization). When HER2 is overexpressed, as it is in w15% to 20% of all breast cancers, it mediates cell growth and survival through activation of its downstream mediators, PI3K/Akt/mTOR and Ras/Raf/MEK/MAP kinase.47 Overexpression of HER2 (HER2þ ) is an independent prognostic (and predictive for sensitivity to its antagonists/inhibitors) factor indicating a more aggressive form of cancer with a higher recurrence rate and higher mortality.46,48 Approximately 10% of patients with HRþ MBC are also HER2þ and, conversely, approximately 50% of HER2þ breast cancers are also HRþ . Women with HRþ and HER2þ disease may not respond to ET as well as women with HRþ and HER2-negative (HER2À ) MBC.7,49 Reversing resistance to antiestrogen therapy in MBC is an important tactic to avoid and delay the use of cytotoxic compounds. The mammalian target of rapamycin (mTOR) has been associated with in vitro reversal of drug resistance, including tamoxifen resistance.50 A number of compounds that interfere with HER2 are either approvedfortherapyofHER2þ breastcancerorareunderinvestigation as treatments for HER2þ MBC and HRþ HER2þ MBC. Trastuzu- mab is a humanized monoclonal antibody directed against the extra- cellular domain-2 of HER2, which prevents ligand-independent signaling.51,52 Trastuzumab has been widely studied for treatment of HER2þ breast cancer and is approved by the US Food and Drug Administration (FDA) for first-line treatment of HER2þ MBC as well as a component of adjuvant systemic therapy for HER2þ breast can- cer.53 Two large phase III trials have demonstrated the benefit of trastuzumab treatment with ET in patients with HRþ and HER2þ MBC. In the Trastuzumab and Anastrozole Directed Against ER-Positive HER2-Positive Mammary Carcinoma (TAnDEM) trial, median progression-free survival (PFS) was significantly improved in patients with HRþ and HER2þ MBC who received anastrozole and trastuzumab (n ¼ 103) compared with anastrozole (n ¼ 104) alone (4.8 vs. 2.4 months, respectively; P ¼ .0016).54 In the Efficacy of letrozole in combination with trastuzumab compared to letrozole monotherapy (eLEcTRA) trial, median time to progression was numerically longer in HRþ and HER2þ MBC patients receiving letrozoleandtrastuzumab(n¼26)versusletrozole(n¼ 31)alone(14.1 vs. 3.3 months, respectively; P ¼ .23). The failure to achieve statistical significance was attributed to the small sample of patients evaluated.55 Another FDA-approved agent, lapatinib, is a dual EGFR/ErbB2 reversible tyrosine kinase inhibitor that competes with ATP for its binding site on the tyrosine kinase domain. It blocks PI3K/Akt/ mTOR and Ras/Raf/MEK/MAP kinase pathways downstream of HER2.56 Lapatinib is FDA-approved for systemic treatment of HER2þ advanced or MBC with systemic chemotherapy as second- line treatment or in first-line combination with letrozole for HRþ and HER2þ MBC.57 In the EGF30008 trial, addition of lapatinib to letrozole significantly reduced the risk of disease progression versus letrozole plus placebo; median PFS was 8.2 versus 3.0 months, respectively, (P ¼ .019) in patients with ERþ and HER2þ early breast cancer. The clinical benefit rate was significantly greater for lapatinib- letrozole versus letrozole-placebo (48% vs. 29%, respectively; P ¼ .003).58,59 These findings led to FDA approval of first-line MBC therapy in ERþ and HER2þ disease. Phase III trials of lapatinib for postmenopausal advanced HRþ MBC are also underway. For example, one study is examining lapatinib plus fulvestrant compared with lapatinib and placebo (NCT00390455). Another study is comparing 3 arms: lapatinib plus trastuzumab plus an AI, trastuzu- mab plus an AI, and lapatinib plus an AI (NCT01160211). Combined targeting of HER2 with lapatinib and trastuzumab may improve outcomes compared with single-agent targeting. Dual inhibi- tion with these agents may also provide a regimen with greater tolera- bility compared with alternative treatments that involve the use of cytotoxic agents.60 A phase III trial demonstrated that lapatinib plus trastuzumabsignificantlyimprovedPFSandoverallsurvival(OS)versus lapatinib monotherapy in patients with HER2þ MBC whose disease had progressed during prior trastuzumab therapy.61 The ALTERNA- TIVE (Safety and efficacy of lapatinib [L], trastuzumab [T], or both in combination with an aromatase inhibitor [AI]) study is an ongoing phase III, randomized, open-label, multicenter trial in HRþ HERþ MBC examining the safety and efficacy of lapatinib plus trastuzumab plus AI versus trastuzumab plus AI.62 The rationale for this trial is to evaluate dual anti-HER2 therapy with an AI compared with single anti- HER2 therapy and an AI because HER2þ MBC is associated with a poor prognosis and increased resistance to ET in patients who are HRþ . Pertuzumab is a monoclonal antibody to the extracellular domain 4 of HER2 that blocks heterodimerization between HER2 and HER3 as well as other HER family members, thereby inhibiting signal transduction via MAPK and PI3K pathways, which can lead to cell growth arrest and apoptosis.63 In a phase II trial, pertuzumab plus trastuzumab showed some improvements in patients with HER2þ MBC with progression on trastuzumab.64 A phase III trial of pertuzumab plus trastuzumab plus cytotoxic chemotherapy with docetaxel as first-line treatment in HER2þ MBC demonstrated improved PFS compared with trastuzumab plus docetaxel alone, and resulted in the FDA approval of this combination.65 IGF1R is a tyrosine kinase growth factor receptor that has been shown to promote the growth of breast cancer cells.66 Similar to the HER2/EGFR family of receptors, growth-promoting effects of IGF1R are mediated by PI3K and Ras/Raf/Mek/MAPkinase intra- cellular signaling. Inhibition of IGF1R function blocks the growth- promoting effects of IGF1 on breast cancer.67,68 Early preclinical studies were promising; however, trials with the monoclonal anti- bodies figitumumab, AVE1642, ganitumab, and dalotuzumab tar- geting IGF1R with ET in HRþ MBC have not shown advantage over ET alone.66 A potential reason for this lack of benefit may be related to findings that breast cancer cells, which are resistant to tamoxifen, are also refractory to IGF1R antibody treatment.69 Inhibition of IGF1R function at different stages of HRþ MBC or in combination with inhibitors of other pathways may prove more effective. GrowthfactorreceptorpathwayssuchasHER2,IGF1R,andFGFR, which are independent of ER signaling, can subvert ET and lead to tumor cell growth and proliferation in MBC. Combination strategies, which include monoclonal antibodies that block growth factor re- ceptors, coupled with tyrosine kinase inhibitors or other agents that act to disrupt signaling of multiple downstream effector molecules may be an effective approach to optimize treatment for the patient with MBC. Intracellular Signaling Cascades PI3K/Akt/mTOR Pathway. The PI3K signaling pathway plays a vital role in cell proliferation, differentiation, and cell survival.70 The Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer 6 - Clinical Breast Cancer Month 2016
  • 7. cell cycle is often dysregulated in cancer cells, resulting in reduced growth-factor dependency and increased replicative potential. Approximately 8% of breast cancers have mutations in PI3K; gain- of-function mutations in the PI3K alpha catalytic subunit (PIK3CA) occur in approximately 30% of ERþ breast cancers and are a common genomic alteration in this subtype of breast can- cer.71,72 PI3K is activated by growth-factor receptor tyrosine ki- nases, HER2, EGFR, IGF1R, and FGFR, and through a series of steps, activation of PI3K stimulates the downstream mediators Akt and mTOR.73 The PI3K pathway interacts with ER directly and indirectly in that PI3K and Akt can phosphorylate and thereby activate ER in the absence of estrogen.74,75 This interaction of the PI3K pathway with ER confers ET resistance.74,76 A phase II clinical trial that studied the combination of ET with everolimus, an inhibitor of mTOR, tested the rationale that inhibition of the PI3K pathway might amplify response or reverse resistance to ET. In patients with HRþ breast cancer, the combination of everolimus and letrozole resulted in increased clinical response and suppression of tumor cell proliferation compared with letrozole and placebo.77 The Tamoxifen Plus Everolimus (TAMRAD) study in patients with HRþ HER2À MBC who had progressed on an AI found that the addition of everolimus to tamoxifen improved clinical benefit rate, TTP, and OS compared with tamoxifen alone.78 The phase III Combination of everolimus with trastuzumab plus paclitaxel as fi rst-line treatment for patients with HER2-positive advanced breast cancer (BOLERO-2) trial showed that treatment with everolimus plus exemestane demonstrated a median TTP of 10.6 months compared with 4.1 months with exemestane alone in post- menopausal HRþ MBC that recurred or progressed on previous ET.79,80 However, in the phase III Placebo-controlled trial of letrozole plus oral temsirolimus (HORIZON) trial, the addition of the mTOR inhibitor temsirolimus to letrozole did not lead to improvement in PFS in AI-naive advanced breast cancer.81 Several differences between these trials may contribute to the varied results. Figure 1 Cellular Signaling Pathways in Breast Cancer and Resistance to Endocrine Therapy. Estrogen Activates Cytoplasmic and Nuclear ER. Cytoplasmic ER Binds to Growth Factor Signaling Components Such as PI3K, Activating Key Signaling Molecules Such as Akt and RAS, and Downstream Molecules Such as mTOR, Raf, and MAPK, Which Promote Cell Proliferation and Survival. In Addition, Many of These Signal-Transduction Molecules Can Phosphorylate and Activate ER and its Co- Regulators to Enhance Nuclear Genomic ER-Mediated Responses. Src Can Also Interact With ER and Mediate Nongenomic ER Activation of Signaling Pathways and Gene Transcription. Src May Be a Key Signaling Molecule in the Development of Resistance to ET. ER Degradation by the 26S Proteasome is Depicted by the Schematic Model. Src Activation and ER Proteolysis Support a Model Whereby Crosstalk Between Ligand-Bound ER and Src Drives ER Transcriptional Activity and Targets ER for Ubiquitin-Dependent Proteolysis. Src Activation May Promote Not Only Proliferation, but Also Estrogen- Activated ER Loss Through Proteolysis. Cyclin E1-CDK2 and Cyclin E2-CDK2 Are Activated During Estrogen-Mediated Proliferation. Estrogen, via ER, Upregulates Cyclin D1. Cyclin D1 Upregulates E2F Transcription Factors, Leading to Active Cyclin E2-CDK2 Complexes. Mitogenic Signals Converge at the Level of Cyclin D1 Upregulation and CDK4/6 Association, Localization, and Kinase Activity Abbreviations: AI ¼ Aromatase inhibitor; CDK ¼ Cyclin-dependent kinases; EGFR ¼ epidermal growth factor receptor; ER ¼ estrogen receptor; ERE ¼ estrogen response elements; FGFR ¼ fibroblast growth factor receptor; HER2 ¼ human epidermal growth factor; IGF ¼ insulin-like growth factor; MAPK ¼ mitogen-activated protein kinases; mTOR ¼ mammalian target of rapamycin inhibitor; p ¼ phosphorylation sites; PI3K ¼ phosphatidylinositol 3-kinase; RTK ¼ receptor tyrosine kinase; Ub ¼ ubiquitin; TF ¼ transcription factor. Stefan Glück Clinical Breast Cancer Month 2016 - 7
  • 8. First, patient populations differed, with AI-resistant patients in the TAMRAD and BOLERO-2 trials, but AI-naive patients in the HORIZON trial. With both mTOR inhibitors subject to meta- bolism by the same CYP enzyme (CYP3A), population differences in its expression might have resulted in variable metabolism across diverse ethnic groups. Genotypic and phenotypic variation across populations may have also accounted for variable response. Finally, relative differences in drug effectiveness between everolimus and temsirolimus cannot be ruled out.82 Ongoing studies of fulvestrant with everolimus and standard ET with everolimus will provide additional perspective on the potential combined benefit of mTOR and ER inhibition (NCT01797120, NCT02035813). Inhibitors of PI3K in preclinical and clinical trials for breast cancer include BKM120, LY294002, GDC0941, BYL719, and GDC0032.83 Additionally, combination PI3K/mTOR inhibitors, GDC0980 and BEZ235, are also being studied. In preclinical studies, GDC0980, GDC0032, and GDC0941, given in combi- nation, enhanced activity of fulvestrant in MCF-7 xenografts, resulting in tumor regression and tumor growth delay.84 A com- bination of tamoxifen, everolimus, and LY294002 produced increased antitumor effects compared with tamoxifen alone or tamoxifen and everolimus in a breast cancer cell line.85 In addition, treating breast cancer cells with BEZ235 induced apoptosis when combined with estrogen deprivation, suggesting potential activity in HRþ breast cancer.86 In an initial phase I/II study, patients with trastuzumab-resistant locally advanced or metastatic HER2þ breast cancer were treated with BKM120 (buparlisib) and trastuzumab. Two patients (17%) had partial responses, and 7 (58%) had stable disease (! 6 weeks) with a disease-control rate of 75%.87 Buparlisib in combination with letrozole has also been used to treat ERþ HER2À MBC, and provided clinical benefit in both patients with PI3K-wild type and mutant tumors, but patients with coexisting PI3K and MAP3K mutations derived the greatest clinical benefit from this regimen.88 The Akt inhibitor MK2206 is also being studied in breast cancer. In MCF-7 cells, overexpression of consti- tutively active Akt1 induced resistance to anastrozole; addition of the Akt inhibitor MK2206 restored sensitivity to this AI.89 Taken together, the preclinical and clinical data suggest that ER and PI3K pathways counter-regulate each other with low PI3K activation resulting in high ER levels, and hyperactivation of PI3K causing a reduction in ER levels.90 Further validation of this notion comes from studies showing that inhibition of HER2 with the antibody trastuzumab or the tyrosine kinase inhibitor lapatinib restores or upregulates ER levels or transcriptional activity in breast cancer cells and patient tumors.91,92 Also, AIs or fulvestrant inhibit the growth of HER2þ tumors that have progressed on trastuzumab or lapatinib.92 Numerous ongoing phase I to III studies are examining the effects of AIs or fulvestrant in combination with PI3K and Akt inhibitors (NCT01610284, NCT01633060, NCT01791478, NCT02088684, NCT01248494, NCT01339442, NCT01437566, NCT01296555, and NCT01344031). Src kinases, intracellular tyrosine kinases that contribute to control of cell proliferation, have been implicated in oncogenic activity in breast cancer, particularly HRþ breast cancer.93 Dasatinib is a small-molecule inhibitor of several Src family kinases approved for use in chronic myelogenous leukemia.94 A recent phase II trial of dasatinib in women with MBC showed modest activity with disease control rates (ie, complete response, partial response, or stable dis- ease for at least 16 weeks) for 8.3% of patients with HER2þ MBC and 15.6% of those with HRþ MBC.95 Early results of a trial of dasatinib in combination with fulvestrant as second-line treatment of HRþ MBC in postmenopausal women did not find improved efficacy with the addition of dasatinib.96 With the variety of agents available that target the PI3K/Akt/ mTOR pathway, the challenge remains to identify which subset of targets, and therefore, which combination of agents may be the most impactful to maximize clinical benefit and adapt to ET resistance in individual patients. Cell Cycle Control and Proliferation Cellular pathways involved in cell proliferation are targets for new drugs that interfere with development of resistance to ET and that treat HRþ MBC. Cyclin-dependent kinases (CDKs) are a subgroup of serine/threonine kinases that play a key role in regulating cell cycle progression.97 The cell cycle G1 to S-phase transit is controlled by CDK4 and CDK6, which are activated upon binding to D-type cyclins leading to expression of genes required for S-phase entry.98 Several studies have identified alterations of cell cycle reg- ulators in human breast cancer and provide a rationale for the po- tential therapeutic role for CDK4/6 inhibition in breast tumors. Cyclin D1 is a direct transcriptional target of ER.99,100 Treatment with antiestrogens induces growth arrest of HRþ breast cancer cells and decreases cyclin D1 expression.101 Conversely, microinjection of antibodies into cyclin D1 inhibits estrogen-induced S-phase en- try.102,103 Endocrine resistance is associated with persistent cyclin D1 expression and Rb phosphorylation.104 Compared with other subtypes of breast cancer, HRþ breast cancer is commonly associ- ated with hyperactivation of cyclin D1-CDK4/6.105 CDK4/6 inhibitors, including palbociclib, abemaciclib, and ribo- ciclib, are being investigated in both the preclinical and clinical settings; some are becoming available for treatment of HRþ MBC. In 2015, palbociclib, a highly selective inhibitor of CDK4/6 kinase, was approved by the FDA in combination with letrozole for the treatment of postmenopausal women with ERþ HER2À advanced breast cancer as initial endocrine-based therapy for their metastatic disease.106,107 Approval for this indication was accelerated based on PFS data. Continued approval for this indication may be contingent upon veri- fication and description of clinical benefit in a confirmatory trial. In preclinical studies, palbociclib inhibited growth of HRþ cancer cells, including those that are resistant to antiestrogen.105 Additionally, a synergistic antitumor effect was observed when palbociclib was com- bined with tamoxifen in both tamoxifen-sensitive and tamoxifen- resistant cell lines. Palbociclib in combination with letrozole versus letrozole alone (PALOMA-1), a randomized phase II study of the AI letrozole with or without palbociclib as first-line therapy for ERþ , HER2À MBC(n¼ 165),reportedastrikingimprovementinPFSfrom 7.5 to 26.2 months (hazard ratio [HR], 0.32; 95% confidence interval [CI], 0.19-0.56; P .001) without additional safety concerns.108 In the final results analysis, the median PFS was 20.2 months with letrozole plus palbociclib compared with 10.2 months for letrozole alone (HR, 0.488; 95% CI, 0.319-0.748; 1-sided P ¼ .0004), and the combination demonstrated significant clinical benefit as first-line treatment in this patient population.109 Several phase I, II, and III studies of palbociclib in combination with fulvestrant and AIs (eg, Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer 8 - Clinical Breast Cancer Month 2016
  • 9. PALOMA-2 and PALOMA-3) in HRþ MBC are currently underway or have recently concluded.110,111 Most recently, treatment with the combination of palbociclib and fulvestrant in patients with advanced HRþ HER2À breast cancer who had relapsed or progressed during prior endocrine therapy, received FDA approval based on the findings of the phase III study, PALOMA-3.110 The study demonstrated that the combination of palbociclib and fulvestrant resulted in significantly longer median PFS (9.5months [95%CI, 9.2-11.0 months] compared with 4.6 months [95% CI, 3.5-5.6 months]) for fulvestrant alone (HR, 0.46; 95% CI, 0.36-0.59; P .001).110 Other CDK inhibitors are in various stages of clinical develop- ment (NCT00721409, NCT01723774, NCT01942135, NCT02040857). Additionally, another CDK inhibitor, LEE001, ribociclib, is being used in addition to everolimus and exemestane in patients with advanced HRþ , HER2À breast cancer who have anastrozole- or letrozole-resistant disease (NCT01857193). Interim results from the phase Ib/II study showed a 70.9% disease control rate.112 This agent is also being examined in combination with fulvestrant and PI3K inhibitors (NCT02088684). The CDK pathway regulates the cell cycle, with CDK4 and 6 directly controlling the gap phase 1 of mitosis (G1). Dysregulation of these components may occur through multiple mechanisms contributing to tumor growth and metastatic potential, and is a common feature in many cancers, including breast cancer. Therefore, therapeutic inhibition of CDK is a relevant treatment strategy, and a number of agents are currently under investigation in clinical tri- als.98,105 Ongoing clinical trials that combine CDK inhibitors with inhibitors of other dysregulated pathways commonly found in breast cancer may improve outcomes and act to overcome ET resistance. Immune Response Modulation Antitumor immunity in human cancer is often ineffective owing to the tight regulation associated with the maintenance of immune homeostasis. Previous attempts to develop effective immunother- apies for cancer have been unsuccessful because there are significant barriers that have been difficult to overcome. However, blockade of the programmed death receptor (PD-1), which may play a role in the tumor’s ability to bypass immune modulation and is overex- pressed in certain tumors such as melanoma, has been proven effective for treatment.113-115 Additionally, a monoclonal antibody targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4), an immune checkpoint molecule that downregulates pathways of T-cell activa- tion, has also been proven effective for treatment of melanoma.116 The place of PD-1 and CTLA-4 inhibition in the treatment of HRþ MBC is not as clearly defined. The extent of mutations and aberrant protein expression associ- ated with cancer cells may also dictate the potential for efficacy of immunotherapy.117 Foreign antigens expressed on the surface of pathogens trigger the body’s immune system to target and eliminate these invaders. However, tumor cells are often undetected by im- mune surveillance owing to identification of tumor cells as “self.” Although tumor cells may over express tumor-associated antigens, these same antigens are expressed at low levels on normal cells, leading to immune tolerance. In contrast, neoantigens that arise due to mutations in a tumor cell, which are not expressed on any other normal cells, represent prime targets for Tecell-mediated elimina- tion with no risk to normal tissues.117 This has led to the hypothesis that tumors with high mutation rates have increased neoepitopes, and use of checkpoint blockade therapies (PD-1, PD-L1, and CTLA-4) can reactivate tumor-infiltrating T cells in these cases, leading to tumor regression.117 The heterogeneous nature of breast cancer has led to studies that seek to identify biomarkers associated with drug response. Triple- negative breast cancer (TNBC) lacks the molecular targets for many treatment options and is associated with high genetic alter- ation rates. In particular, one study of 2111 patients with TNBC (ERÀ /PgRÀ /HER2À ) found that 91% of patients showed dimin- ished androgen receptor expression and/or mutations in PI3KCA/ PTEN/AKT1, or loss of PTEN.118 In addition, expression of PD-L1 is more prevalent in TNBC, and this coupled with increased PD- L1-mediated T-cell reactivation suggests that PD-L1 inhibitors may be effective in this patient population.117,119 In fact, a preliminary study that targeted PD-L1 showed promising objective clinical ac- tivity in TNBC patients (NCT01375842).119 Estrogen-Receptor Degradation and Proteasome Inhibitors (PIs) Proteasomes are multicatalytic protease complexes in the cell, involved in the non-lysosomal recycling of intracellular proteins. Pro- teasomes play a critical role in regulation of cell division in both normal and cancer cells.120-123 In cancer cells, this homeostatic function is deregulated, leading to the hyperactivation of the proteasomes. PIs interfere with the ubiquitin proteasome pathway and lead to the accumulation of proteins engaged in cell cycle progression. They ulti- mately put a halt to cancer cell division and induce apoptosis. Cell cycle proteins are regulatory proteins whose temporal activity is tightly controlled and are thus important targets of proteasome degradation. Major proteins related to the cell cycle process are cyclins, CDKs, CDK inhibitors (CKIs), and some transcription factors. Cyclin proteins are found to be highly upregulated in cases of aberrant cell division in cancer cells,particularly cyclins D and E.124,125 The precise mechanism ofactionofPIs incancertreatment isnot well-understood.Intreatment of breast cancer, 2 potential mechanisms have been described: the PI, bortezomib, may inhibit cell proliferation and may decrease levels of ER.126,127 Results from early preclinical and clinical trials have been mixed, with 1 small study showing no effect of treatment with borte- zomib and another showing enhanced MCF-7 tumor suppression with bortezomib and trastuzumab.128 Data in MCF-7 breast cancer cells suggest that fulvestrant’s antiestrogenic effects of induction of protea- somal degradation of ER may involve c-SRC, a cellular kinase proto- oncogene. In light of this potential interaction, a phase II study of fulvestrant and bortezomib in women with locally advanced or MBC is underway (NCT01142401). Epigenetic Modifiers In addition to genetic alterations such as the mutations and de- letions that alter the composition of genomic DNA, epigenetic changes including DNA methylation, histone modifications, and chromatin remodeling can reversibly alter gene expression and play a key role in tumorigenicity and metastatic potential of breast cancer cells.129 Therapeutic interventions that target epigenetic modifica- tions have been used to partially or completely revert the altered gene expression, and several epigenetic agents that are approved for use in other settings have potential as breast cancer therapies. Stefan Glück Clinical Breast Cancer Month 2016 - 9
  • 10. Several clinical trials of these epigenetic agents are planned or underway in the breast cancer setting. Azacitidine and decitabine are hypomethylating agents that inhibit DNA methyltransferase, thereby halting tumor cell growth and division through epigenetic manipulation.130,131 Both agents have been approved for use in myelodysplastic syndrome and are currently being investigated for breast cancer treatment.130,131 In combination with fulvestrant, azacitidine is being evaluated in a phase II trial in patients with ERþ , HER2À MBC who have progressed on an AI (NCT02374099). In addition, a class I HDAC inhibitor, entino- stat, was assessed in a phase II trial (NCT00676663) with or without the AI exemestane in patients with ERþ breast cancer who have progressed on an AI, and the combination was found to not only improve PFS and OS, but restore sensitivity to hormone therapy.132 Entinostat is currently being studied in combination with azacitidine in a phase II trial of patients with either TNBC or HRþ HER2À breast cancer (NCT01349959). Decitabine in combination with the HDAC inhibitor panobinostat (LBH589) was also being investigated in TNBC in an attempt to re-express ER; however, the study was closed due to slow accrual (NCT01194908). Conclusions ER-targeted therapy is important for many women with breast cancer, but resistance to therapy inevitably occurs. The ER signaling pathway is a complex network of extensive crosstalk with growth-factor signaling pathways, cell cycle control pathways, and protein degradation pathways. These pathways provide many alternative targets for agents that may be useful in combination with ET to decrease resistance to treatment and to extend benefit to patients who do not achieve optimal benefit from ET alone. As effective treatments are identified, an important area of focus must be on the factors that determine optimal combination treatments in subtypes of breast cancer. Such factors comprise not only the responsiveness of the cells of a particular subgroup of tumors, but also the characteristics of different treatment combinations with respect to the temporal profile of effects, relative tolerability of different combinations, and the compatibility of routes of administration. Several therapies that are discussed in this review, among them everolimus (mTOR inhibitor), palbociclib (CDK4/6 inhibitor), and MK-3475 (PD-1 inhibitor), are already proving to be effective treatments for MBC and useful in reversing or pre- venting the development of resistance to ET. Through inhibition of central components of pathways for growth factor-promoted cellular proliferation, cell cycle-mediated cellular proliferation, and immunosuppressive programs that initiate and sustain tumor growth, these agents are effective options that may extend survival and maintain quality of life for women with MBC who have relapsed on endocrine therapies. Acknowledgments Medical writing and editorial services were provided by SCI Scientific Communications Information, Parsippany, NJ, and The Lockwood Group, Stamford, CT (funded by AstraZeneca LP). Disclosure This article was supported by AstraZeneca LP. Dr Glück has received grant support and consulting fees from Agendia, Celgene, Dara, Eisai, Genentech/Roche, GHI, Mylan, and Onyx/Amgen. At the time of the initiation of manuscript development, Dr Glück was the Sylvester Professor of Medicine, Department of Medicine, Di- vision of Hematology Oncology, Sylvester Comprehensive Cancer Center, Miami, FL. Dr Glück is a current employee and shareholder of Celgene Corporation, Summit, NJ. References 1. American Cancer Society. Cancer Facts Figures 2016. Available at: http:// www.cancer.org/acs/groups/content/@research/documents/document/acspc- 047079.pdf. Accessed: May 24, 2016. 2. National Cancer Institute. Surveillance, Epidemiology, and End Results (SEER) Fact Sheets: Breast. Available at: http://seer.cancer.gov/statfacts/html/breast.html. Accessed: April 04, 2016. 3. Lu J, Steeg PS, Price JE, et al. Breast cancer metastasis: challenges and oppor- tunities. Cancer Res 2009; 69:4951-3. 4. Brewster AM, Hortobagyi GN, Broglio KR, et al. Residual risk of breast cancer recurrence 5 years after adjuvant therapy. J Natl Cancer Inst 2008; 100:1179-83. 5. Nadji M, Gomez-Fernandez C, Ganjei-Azar P, Morales AR. Immunohisto- chemistry of estrogen and progesterone receptors reconsidered: experience with 5, 993 breast cancers. Am J Clin Pathol 2005; 123:21-7. 6. Chung CT, Carlson RW. Goals and objectives in the management of metastatic breast cancer. Oncologist 2003; 8:514-20. 7. Gluck S, Arteaga CL, Osborne CK. Optimizing chemotherapy-free survival for the ER/HER2-positive metastatic breast cancer patient. Clin Cancer Res 2011; 17:5559-61. 8. Hilton HN, Doan TB, Graham JD, et al. Acquired convergence of hormone signaling in breast cancer: ER and PR transition from functionally distinct in normal breast to predictors of metastatic disease. Oncotarget 2014; 5:8651-64. 9. National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology. Breast Cancer.v.3.2015. Available at: http://www.nccn.org/ professionals/physician_gls/pdf/breast.pdf. Accessed: November 12, 2015. 10. Buzdar AU, Hortobagyi G. Update on endocrine therapy for breast cancer. Clin Cancer Res 1998; 4:527-34. 11. Cardoso F, Fallowfield L, Costa A, Castiglione M, Senkus E, ESMO Guidelines Working Group. Locally recurrent or metastatic breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2011; 22(Suppl 6):vi25-30. 12. Nagaraj G, Ma C. Revisiting the estrogen receptor pathway and its role in endocrine therapy for postmenopausal women with estrogen receptor-positive metastatic breast cancer. Breast Cancer Res Treat 2015; 150:231-42. 13. Patani N, Martin LA. Understanding response and resistance to oestrogen deprivation in ER-positive breast cancer. Mol Cell Endocrinol 2014; 382:683-94. 14. Miller WR, Bartlett JM, Canney P, Verrill M. Hormonal therapy for post- menopausal breast cancer: the science of sequencing. Breast Cancer Res Treat 2007; 103:149-60. 15. Gluck S. Extending the clinical benefit of endocrine therapy for women with hormone receptor-positive metastatic breast cancer: differentiating mechanisms of action. Clin Breast Cancer 2014; 14:75-84. 16. Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer. Annu Rev Med 2011; 62:233-47. 17. Pietras RJ. Biologic basis of sequential and combination therapies for hormone- responsive breast cancer. Oncologist 2006; 11:704-17. 18. Colleoni M, Gelber S, Coates AS, et al. Influence of endocrine-related factors on response to perioperative chemotherapy for patients with node-negative breast cancer. J Clin Oncol 2001; 19:4141-9. 19. Wilcken N, Hornbuckle J, Ghersi D. Chemotherapy alone versus endocrine therapy alone for metastatic breast cancer. Cochrane Database Syst Rev 2003; 2: CD002747. 20. Gluck S, von Minckwitz G, Untch M. Aromatase inhibitors in the treatment of elderly women with metastatic breast cancer. Breast 2013; 22:142-9. 21. Bedard PL, Freedman OC, Howell A, Clemons M. Overcoming endocrine resistance in breast cancer: are signal transduction inhibitors the answer? Breast Cancer Res Treat 2008; 108:307-17. 22. Miller WR, Larionov AA. Understanding the mechanisms of aromatase inhibitor resistance. Breast Cancer Res 2012; 14:201. 23. Gutierrez MC, Detre S, Johnston S, et al. Molecular changes in tamoxifen- resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. J Clin Oncol 2005; 23:2469-76. 24. Arpino G, Green SJ, Allred DC, et al. HER-2 amplification, HER-1 expression, and tamoxifen response in estrogen receptor-positive metastatic breast cancer: a southwest oncology group study. Clin Cancer Res 2004; 10:5670-6. Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer 10 - Clinical Breast Cancer Month 2016
  • 11. 25. Musgrove EA, Sutherland RL. Biological determinants of endocrine resistance in breast cancer. Nat Rev Cancer 2009; 9:631-43. 26. Arpino G, Wiechmann L, Osborne CK, Schiff R. Crosstalk between the estrogen receptor and the HER tyrosine kinase receptor family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocr Rev 2008; 29: 217-33. 27. Schiff R, Massarweh SA, Shou J, Bharwani L, Mohsin SK, Osborne CK. Cross- talk between estrogen receptor and growth factor pathways as a molecular target for overcoming endocrine resistance. Clin Cancer Res 2004; 10:331S-6S. 28. Frasor J, Stossi F, Danes JM, Komm B, Lyttle CR, Katzenellenbogen BS. Se- lective estrogen receptor modulators: discrimination of agonistic versus antago- nistic activities by gene expression profiling in breast cancer cells. Cancer Res 2004; 64:1522-33. 29. Klinge CM. Estrogen receptor interaction with estrogen response elements. Nucleic Acids Res 2001; 29:2905-19. 30. Osborne CK, Bardou V, Hopp TA, et al. Role of the estrogen receptor coac- tivator AIB1 (SRC-3) and HER-2/neu in tamoxifen resistance in breast cancer. J Natl Cancer Inst 2003; 95:353-61. 31. Levin ER. Cell localization, physiology, and nongenomic actions of estrogen receptors. J Appl Physiol (1985) 2001; 91:1860-7. 32. Lee AV, Cui X, Oesterreich S. Cross-talk among estrogen receptor, epidermal growth factor, and insulin-like growth factor signaling in breast cancer. Clin Cancer Res 2001; 7:4429s-35s, discussion: 4411s-4412s. 33. Yarden RI, Wilson MA, Chrysogelos SA. Estrogen suppression of EGFR expression in breast cancer cells: a possible mechanism to modulate growth. J Cell Biochem Suppl 2001; Suppl 36:232-46. 34. Guo S, Sonenshein GE. Forkhead box transcription factor FOXO3a regulates estrogen receptor alpha expression and is repressed by the Her-2/neu/ phosphatidylinositol 3-kinase/Akt signaling pathway. Mol Cell Biol 2004; 24: 8681-90. 35. Lopez-Tarruella S, Schiff R. The dynamics of estrogen receptor status in breast cancer: re-shaping the paradigm. Clin Cancer Res 2007; 13:6921-5. 36. Taneja P, Maglic D, Kai F, et al. Classical and novel prognostic markers for breast cancer and their clinical significance. Clin Med Insights Oncol 2010; 4:15-34. 37. Bardou VJ, Arpino G, Elledge RM, Osborne CK, Clark GM. Progesterone re- ceptor status significantly improves outcome prediction over estrogen receptor status alone for adjuvant endocrine therapy in two large breast cancer databases. J Clin Oncol 2003; 21:1973-9. 38. Cui X, Schiff R, Arpino G, Osborne CK, Lee AV. Biology of progesterone re- ceptor loss in breast cancer and its implications for endocrine therapy. J Clin Oncol 2005; 23:7721-35. 39. Mohammed H, Russell IA, Stark R, et al. Progesterone receptor modulates ERalpha action in breast cancer. Nature 2015; 523:313-7. 40. Campagnoli C, Clavel-Chapelon F, Kaaks R, Peris C, Berrino F. Progestins and progesterone in hormone replacement therapy and the risk of breast cancer. J Steroid Biochem Mol Biol 2005; 96:95-108. 41. Farhat GN, Parimi N, Chlebowski RT, et al. Sex hormone levels and risk of breast cancer with estrogen plus progestin. J Natl Cancer Inst 2013; 105: 1496-503. 42. Bines J, Dienstmann R, Obadia RM, et al. Activity of megestrol acetate in postmenopausal women with advanced breast cancer after nonsteroidal aromatase inhibitor failure: a phase II trial. Ann Oncol 2014; 25:831-6. 43. Gluck S, de Snoo F, Peeters J, Stork-Sloots L, Somlo G. Molecular subtyping of early-stage breast cancer identifies a group of patients who do not benefit from neoadjuvant chemotherapy. Breast Cancer Res Treat 2013; 139:759-67. 44. Chakraborty AK, Welsh A, Digiovanna MP. Co-targeting the insulin-like growth factor I receptor enhances growth-inhibitory and pro-apoptotic effects of anti- estrogens in human breast cancer cell lines. Breast Cancer Res Treat 2010; 120: 327-35. 45. Kern FG, McLeskey SW, Zhang L, et al. Transfected MCF-7 cells as a model for breast-cancer progression. Breast Cancer Res Treat 1994; 31:153-65. 46. Ross JS, Fletcher JA. The HER-2/neu oncogene in breast cancer: prognostic factor, predictive factor, and target for therapy. Stem Cells 1998; 16:413-28. 47. Arteaga CL, Sliwkowski MX, Osborne CK, Perez EA, Puglisi F, Gianni L. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat Rev Clin Oncol 2012; 9:16-32. 48. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/ neu oncogene. Science 1987; 235:177-82. 49. De Laurentiis M, Arpino G, Massarelli E, et al. A meta-analysis on the interaction between HER-2 expression and response to endocrine treatment in advanced breast cancer. Clin Cancer Res 2005; 11:4741-8. 50. Westin GF, Perez CA, Wang E, Gluck S. Biologic impact and clinical implication of mTOR inhibition in metastatic breast cancer. Int J Biol Markers 2013; 28: 233-41. 51. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that over- expresses HER2. N Engl J Med 2001; 344:783-92. 52. Incorvati JA, Shah S, Mu Y, Lu J. Targeted therapy for HER2 positive breast cancer. J Hematol Oncol 2013; 6:38. 53. Herceptin (Trastuzumab) Intravenous Infusion [Package Insert]. South San Francisco, CA: Genentech, Inc; 2010. 54. Kaufman B, Mackey JR, Clemens MR, et al. Trastuzumab plus anastrozole versus anastrozole alone for the treatment of postmenopausal women with human epidermal growth factor receptor 2-positive, hormone receptor-positive metastatic breast cancer: results from the randomized phase III TAnDEM study. J Clin Oncol 2009; 27:5529-37. 55. Huober J, Fasching PA, Barsoum M, et al. Higher efficacy of letrozole in com- bination with trastuzumab compared to letrozole monotherapy as first-line treatment in patients with HER2-positive, hormone-receptor-positive metasta- tic breast cancer - results of the eLEcTRA trial. Breast 2012; 21:27-33. 56. Montemurro F, Valabrega G, Aglietta M. Lapatinib: a dual inhibitor of EGFR and HER2 tyrosine kinase activity. Expert Opin Biol Ther 2007; 7:257-68. 57. Tykerb (Lapatinib) Tablets [Package Insert]. Research Triangle Park, NC: GlaxoSmithKline; 2015. 58. Johnston S, Pippen J, Pivot X, et al. Lapatinib combined with letrozole versus letrozole and placebo as first-line therapy for postmenopausal hormone receptor- positive metastatic breast cancer. J Clin Oncol 2009; 27:5538-46. 59. Schwartzberg LS, Franco SX, Florance A, O’Rourke L, Maltzman J, Johnston S. Lapatinib plus letrozole as first-line therapy for HER-2þ hormone receptor- positive metastatic breast cancer. Oncologist 2010; 15:122-9. 60. Ahn ER, Wang E, Gluck S. Is the improved efficacy of trastuzumab and lapatinib combination worth the added toxicity? A discussion of current evidence, rec- ommendations, and ethical issues regarding dual HER2-targeted therapy. Breast Cancer (Auckl) 2012; 6:191-207. 61. Blackwell KL, Burstein HJ, Storniolo AM, et al. Overall survival benefit with lapatinib in combination with trastuzumab for patients with human epidermal growth factor receptor 2-positive metastatic breast cancer: final results from the EGF104900 Study. J Clin Oncol 2012; 30:2585-92. 62. Johnston S, Wroblewski S, Huang Y. ALTERNATIVE: safety and efficacy of lapatinib (L), trastuzumab (T), or both in combination with an aromatase in- hibitor (AI) for the treatment of hormone receptor positive (HRþ), human epidermal growth factor receptor 2 positive (HER2þ) metastatic breast cancer [abstract]. Cancer Res 2012; 72(24 Suppl):558s. 63. Perjeta (Pertuzumab) Injection [Package Insert]. South San Francisco, CA: Gen- entech, Inc; 2016. 64. Baselga J, Gelmon KA, Verma S, et al. Phase II trial of pertuzumab and trastu- zumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol 2010; 28:1138-44. 65. Swain SM, Baselga J, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med 2015; 372:724-34. 66. Yang Y, Yee D. Targeting insulin and insulin-like growth factor signaling in breast cancer. J Mammary Gland Biol Neoplasia 2012; 17:251-61. 67. Arteaga CL, Kitten LJ, Coronado EB, et al. Blockade of the type I somatomedin receptor inhibits growth of human breast cancer cells in athymic mice. J Clin Invest 1989; 84:1418-23. 68. Sachdev D, Li SL, Hartell JS, Fujita-Yamaguchi Y, Miller JS, Yee D. A chimeric humanized single-chain antibody against the type I insulin-like growth factor (IGF) receptor renders breast cancer cells refractory to the mitogenic effects of IGF-I. Cancer Res 2003; 63:627-35. 69. Fagan DH, Uselman RR, Sachdev D, Yee D. Acquired resistance to tamoxifen is associated with loss of the type I insulin-like growth factor receptor: implications for breast cancer treatment. Cancer Res 2012; 72:3372-80. 70. Katso R, Okkenhaug K, Ahmadi K, White S, Timms J, Waterfield MD. Cellular function of phosphoinositide 3-kinases: implications for development, homeo- stasis, and cancer. Annu Rev Cell Dev Biol 2001; 17:615-75. 71. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004; 304:554. 72. Ellis MJ, Lin L, Crowder R, et al. Phosphatidyl-inositol-3-kinase alpha cata- lytic subunit mutation and response to neoadjuvant endocrine therapy for estrogen receptor positive breast cancer. Breast Cancer Res Treat 2010; 119: 379-90. 73. Miller TW, Rexer BN, Garrett JT, Arteaga CL. Mutations in the phosphatidy- linositol 3-kinase pathway: role in tumor progression and therapeutic implications in breast cancer. Breast Cancer Res 2011; 13:224. 74. Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S, Nakshatri H. Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. J Biol Chem 2001; 276: 9817-24. 75. Yamnik RL, Digilova A, Davis DC, Brodt ZN, Murphy CJ, Holz MK. S6 kinase 1 regulates estrogen receptor alpha in control of breast cancer cell proliferation. J Biol Chem 2009; 284:6361-9. 76. Clark AS, West K, Streicher S, Dennis PA. Constitutive and inducible Akt ac- tivity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Mol Cancer Ther 2002; 1:707-17. 77. Baselga J, Semiglazov V, van Dam P, et al. Phase II randomized study of neo- adjuvant everolimus plus letrozole compared with placebo plus letrozole in patients with estrogen receptor-positive breast cancer. J Clin Oncol 2009; 27: 2630-7. 78. Bachelot T, Bourgier C, Cropet C, et al. Randomized phase II trial of everolimus in combination with tamoxifen in patients with hormone receptor-positive, hu- man epidermal growth factor receptor 2-negative metastatic breast cancer with prior exposure to aromatase inhibitors: a GINECO study. J Clin Oncol 2012; 30: 2718-24. 79. Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med 2012; 366: 520-9. Stefan Glück Clinical Breast Cancer Month 2016 - 11
  • 12. 80. Yardley DA, Noguchi S, Pritchard KI, et al. Everolimus plus exemestane in postmenopausal patients with HR(þ) breast cancer: BOLERO-2 final progression-free survival analysis. Adv Ther 2013; 30:870-84. 81. Wolff AC, Lazar AA, Bondarenko I, et al. Randomized phase III placebo- controlled trial of letrozole plus oral temsirolimus as first-line endocrine ther- apy in postmenopausal women with locally advanced or metastatic breast cancer. J Clin Oncol 2013; 31:195-202. 82. Dees EC, Carey LA. Improving endocrine therapy for breast cancer: it’s not that simple. J Clin Oncol 2013; 31:171-3. 83. Khan KH, Yap TA, Yan L, Cunningham D. Targeting the PI3K-AKT-mTOR signaling network in cancer. Chin J Cancer 2013; 32:253-65. 84. Friedman L, Ross L, Wallin J, et al. Selective PI3K and dual PI3K/mTOR in- hibitors enhance the efficacy of endocrine therapies in breast cancer models [abstract]. Cancer Res 2012; 72(24 suppl), P5-19-02. 85. Chen X, Zhao M, Hao M, et al. Dual inhibition of PI3K and mTOR mitigates compensatory AKT activation and improves tamoxifen response in breast cancer. Mol Cancer Res 2013; 11:1269-78. 86. Crowder RJ, Phommaly C, Tao Y, et al. PIK3CA and PIK3CB inhibition pro- duce synthetic lethality when combined with estrogen deprivation in estrogen receptor-positive breast cancer. Cancer Res 2009; 69:3955-62. 87. Saura C, Bendell J, Jerusalem G, et al. Phase Ib study of buparlisib plus trastu- zumab in patients with HER2-positive advanced or metastatic breast cancer that has progressed on Trastuzumab-based therapy. Clin Cancer Res 2014; 20: 1935-45. 88. Balko JM, Hicks M, Berger MF, et al. Genomic alterations indicative of a luminal A subtype associate with clinical benefit to buparlisib and letrozole in endocrine- resistant ERþ/HER2e metastatic breast cancer [abstract P3-07-41]. San Antonio Breast Cancer Symposium. San Antonio, Texas; 2015. 89. Vilquin P, Villedieu M, Grisard E, et al. Molecular characterization of anastrozole resistance in breast cancer: pivotal role of the Akt/mTOR pathway in the emergence of de novo or acquired resistance and importance of combining the allosteric Akt inhibitor MK-2206 with an aromatase inhibitor. Int J Cancer 2013; 133:1589-602. 90. Creighton CJ, Fu X, Hennessy BT, et al. Proteomic and transcriptomic profiling reveals a link between the PI3K pathway and lower estrogen-receptor (ER) levels and activity in ERþ breast cancer. Breast Cancer Res 2010; 12:R40. 91. Munzone E, Curigliano G, Rocca A, et al. Reverting estrogen-receptor-negative phenotype in HER-2-overexpressing advanced breast cancer patients exposed to trastuzumab plus chemotherapy. Breast Cancer Res 2006; 8:R4. 92. Xia W, Bacus S, Hegde P, et al. A model of acquired autoresistance to a potent ErbB2 tyrosine kinase inhibitor and a therapeutic strategy to prevent its onset in breast cancer. Proc Natl Acad Sci U S A 2006; 103:7795-800. 93. Finn RS. Targeting Src in breast cancer. Ann Oncol 2008; 19:1379-86. 94. Sprycel (Dasatinib) Tablets [Package Insert]. Princeton, NJ: Bristol-Myers Squibb Company; 2014. 95. Mayer EL, Baurain JF, Sparano J, et al. A phase 2 trial of dasatinib in patients with advanced HER2-positive and/or hormone receptor-positive breast cancer. Clin Cancer Res 2011; 17:6897-904. 96. Wright GL, Blum J, Krekow LK, et al. Phase II trial of fulvestrant with or without dasatinib in postmenopausal patients with hormone receptor-positive metastatic breast cancer previously treated with an aromatase inhibitor [ab- stract]. Clin Cancer Res 2011; 71(24 Suppl), PD01e01. 97. Caldon CE, Daly RJ, Sutherland RL, Musgrove EA. Cell cycle control in breast cancer cells. J Cell Biochem 2006; 97:261-74. 98. Lundberg AS, Weinberg RA. Control of the cell cycle and apoptosis. Eur J Cancer 1999; 35:1886-94. 99. Altucci L, Addeo R, Cicatiello L, et al. Estrogen induces early and timed acti- vation of cyclin-dependent kinases 4, 5, and 6 and increases cyclin messenger ribonucleic acid expression in rat uterus. Endocrinology 1997; 138:978-84. 100. Said TK, Conneely OM, Medina D, O’Malley BW, Lydon JP. Progesterone, in addition to estrogen, induces cyclin D1 expression in the murine mammary epithelial cell, in vivo. Endocrinology 1997; 138:3933-9. 101. Watts CK, Brady A, Sarcevic B, deFazio A, Musgrove EA, Sutherland RL. Antiestrogen inhibition of cell cycle progression in breast cancer cells in associated with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma protein phosphorylation. Mol Endocrinol 1995; 9:1804-13. 102. Lukas J, Bartkova J, Bartek J. Convergence of mitogenic signalling cascades from diverse classes of receptors at the cyclin D-cyclin-dependent kinase-pRb- controlled G1 checkpoint. Mol Cell Biol 1996; 16:6917-25. 103. Prall OW, Sarcevic B, Musgrove EA, Watts CK, Sutherland RL. Estrogen- induced activation of Cdk4 and Cdk2 during G1-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E-Cdk2. J Biol Chem 1997; 272: 10882-94. 104. Thangavel C, Dean JL, Ertel A, et al. Therapeutically activating RB: reestab- lishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat Cancer 2011; 18:333-45. 105. Finn RS, Dering J, Conklin D, et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res 2009; 11:R77. 106. Ibrance (Palbociclib) Capsules [Package Insert]. New York, NY: Pfizer, Inc; 2015. 107. US Food and Drug Administration. FDA approves Ibrance for postmenopausal women with advanced breast cancer 2015. Available at: http://www.fda.gov/NewsEvents/ Newsroom/PressAnnouncements/ucm432871.htm. Accessed: February 25, 2015. 108. Finn RS, Crown JP, Lang I, et al. Results of a randomized phase 2 study of PD 0332991, a cyclin-dependent kinase (CDK) 4/6 inhibitor, in combination with letrozole vs letrozole alone for first-line treatment of ERþ/HER2- advanced breast cancer (BC) [abstract S1e6]. Cancer Res 2012; 72(24 Suppl):91s. 109. Finn RS, Crown JP, Lang I, et al. Final results of a randomized phase II study of PD 0332991, a cyclin-dependent kinase (CDK)-4/6 inhibitor, in combination with letrozole vs letrozole alone for first-line treatment of ERþ/HER2- advanced breast cancer (PALOMA-1; TRIO-18) [abstract CT101]. Cancer Res 2014; 74(19 Suppl):CT101. 110. Cristofanilli M, Turner NC, Bondarenko I, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol 2016; 17:425-39. 111. Finn RS, Martin M, Rugo HS, et al. PALOMA-2: Primary results from a phase III trial of palbociclib (P) with letrozole (L) compared with letrozole alone in postmenopausal women with ERþ/HER2e advanced breast cancer (ABC). J Clin Oncol 2016; 34(Suppl), abstract 507. 112. Bardia A, Modi S, Oliveira M, et al. Triplet therapy with ribociclib, everolimus, and exemestane in women with HRþ/HER2e advanced breast cancer [abstract P6-13-01]. San Antonio Breast Cancer Symposium. San Antonio, Texas; 2015. 113. Blank C, Kuball J, Voelkl S, et al. Blockade of PD-L1 (B7-H1) augments human tumor-specific T cell responses in vitro. Int J Cancer 2006; 119:317-27. 114. Yang W, Chen PW, Li H, Alizadeh H, Niederkorn JY. PD-L1: PD-1 interaction contributes to the functional suppression of T-cell responses to human uveal melanoma cells in vitro. Invest Ophthalmol Vis Sci 2008; 49:2518-25. 115. Hirano F, Kaneko K, Tamura H, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res 2005; 65:1089-96. 116. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363:711-23. 117. Lu YC, Robbins PF. Cancer immunotherapy targeting neoantigens. Semin Immunol 2016; 28:22-7. 118. Millis SZ, Gatalica Z, Winkler J, et al. Predictive biomarker profiling of 6000 breast cancer patients shows heterogeneity in TNBC, with treatment implica- tions. Clin Breast Cancer 2015; 15:473-481e3. 119. Emens L, Adams S, Loi S, et al. A phase III randomized trial of atezolizumab in combination with nab-paclitaxel as first line therapy for patients with metastatic triple-negative breast cancer (mTNBC) [abstract OT1-01-06]. San Antonio Breast Cancer Symposium. San Antonio, Texas; 2015. 120. Adams J. The proteasome: a suitable antineoplastic target. Nat Rev Cancer 2004; 4:349-60. 121. Adams J, Palombella VJ, Sausville EA, et al. Proteasome inhibitors: a novel class of potent and effective antitumor agents. Cancer Res 1999; 59:2615-22. 122. Rivett AJ. Proteasomes: multicatalytic proteinase complexes. Biochem J 1993; 29(Pt 1):1-10. 123. Goldberg AL. Functions of the proteasome: the lysis at the end of the tunnel. Science 1995; 268:522-3. 124. Diehl JA, Ponugoti B. Ubiquitin-dependent proteolysis in G1/S phase control and its relationship with tumor susceptibility. Genes Cancer 2010; 1:717-24. 125. Masamha CP, Benbrook DM. Cyclin D1 degradation is sufficient to induce G1 cell cycle arrest despite constitutive expression of cyclin E2 in ovarian cancer cells. Cancer Res 2009; 69:6565-72. 126. Jones MD, Liu JC, Barthel TK, et al. A proteasome inhibitor, bortezomib, in- hibits breast cancer growth and reduces osteolysis by downregulating metastatic genes. Clin Cancer Res 2010; 16:4978-89. 127. Powers GL, Ellison-Zelski SJ, Casa AJ, Lee AV, Alarid ET. Proteasome inhibition represses ERalpha gene expression in ERþ cells: a new link be- tween proteasome activity and estrogen signaling in breast cancer. Oncogene 2010; 29:1509-18. 128. Yeh WL, Shioda K, Coser KR, Rivizzigno D, McSweeney KR, Shioda T. Ful- vestrant-induced cell death and proteasomal degradation of estrogen receptor alpha protein in MCF-7 cells require the CSK c-Src tyrosine kinase. PLoS One 2013; 8:e60889. 129. Jeschke J, Collignon E, Fuks F. DNA methylome profiling beyond promoters - taking an epigenetic snapshot of the breast tumor microenvironment. FEBS J 2015; 282:1801-14. 130. Dacogen (Decitabine) for Injection [Package Insert]. Rockville, MD: Otsuka America Pharmaceutical, Inc; 2014. 131. Vidaza (Azacitidine) for Injection [Package Insert]. Summit, NJ: Celgene Corporation; 2015. 132. Yardley DA, Ismail-Khan RR, Melichar B, et al. Randomized phase II, double- blind, placebo-controlled study of exemestane with or without entinostat in postmenopausal women with locally recurrent or metastatic estrogen receptor- positive breast cancer progressing on treatment with a nonsteroidal aromatase inhibitor. J Clin Oncol 2013; 31:2128-35. Alternate Pathways on Estrogen Receptor in Metastatic Breast Cancer 12 - Clinical Breast Cancer Month 2016