ArticlePDF AvailableLiterature Review

HER2-positive breast cancer: new therapeutic frontiers and overcoming resistance

SAGE Publications Inc
Therapeutic Advances in Medical Oncology
Authors:
  • Catalan Institute of Oncology- Bellvitge Institute for Biomedical Research (IDIBELL)

Abstract and Figures

The introduction of anti-HER2 therapies to the treatment of patients with HER2-positive breast cancer has led to dramatic improvements in survival in both early and advanced settings. Despite this breakthrough, nearly all patients with metastatic HER2-positive breast cancer eventually progress on anti-HER2 therapy due to de novo or acquired resistance. A better understanding not only of the underlying mechanisms of HER2 therapy resistance but of tumor heterogeneity as well as the host and tumor microenvironment is essential for the development of new strategies to further improve patient outcomes. One strategy has focused on inhibiting the HER2 signaling pathway more effectively with dual-blockade approaches and developing improved anti-HER2 therapies like antibody–drug conjugates, new anti-HER2 antibodies, bispecific antibodies, or novel tyrosine kinase inhibitors that might replace or be used in addition to some of the current anti-HER2 treatments. Combinations of anti-HER2 therapy with other agents like immune checkpoint inhibitors, CDK4/6 inhibitors, and PI3K/AKT/mTOR inhibitors are also being extensively evaluated in clinical trials. These add-on strategies of combining optimized targeted therapies could potentially improve outcomes for patients with HER2-positive breast cancer but may also allow de-escalation of treatment in some patients, potentially sparing some from unnecessary treatments, and their related toxicities and costs.
This content is subject to copyright.
https://doi.org/10.1177/1758835919833519
https://doi.org/10.1177/1758835919833519
Therapeutic Advances in Medical Oncology
journals.sagepub.com/home/tam 1
Ther Adv Med Oncol
2019, Vol. 11: 1 –16
DOI: 10.1177/
1758835919833519
© The Author(s), 2019.
Article reuse guidelines:
sagepub.com/journals-
permissions
Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License
(http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission
provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
Introduction
A better understanding of tumor biology and
HER2 signaling has led to the development and
approval of new HER2-targeted agents that,
together with the use of continued anti-HER2
therapy beyond progression, have resulted in
unpreceded survival outcomes in patients with
advanced HER2-positive breast cancer.1
The addition of trastuzumab to standard therapy
dramatically improved prognosis for patients with
HER2-positive breast cancer, and became a land-
mark in the treatment of these patients.2,3 The
second anti-HER2 agent that was incorporated
into routine practice of advanced HER2-positive
disease was lapatinib, an oral tyrosine kinase
inhibitor (TKI) that reversibly inhibits HER1 or
epidermal growth factor receptor (EGFR)
and HER2 kinases. The approval of lapatinib was
based on the improvement in progression-free
survival (PFS) found in a phase III trial when
combined with capecitabine versus capecitabine
alone though no improvement in overall survival
(OS) was observed.4 Pertuzumab is a humanized
monoclonal antibody that binds to HER2 on
extracellular domain II, a different domain than
trastuzumab, preventing homo- and heterodimer
formations and blocking one of the most powerful
heterodimers, HER2/HER3, that activates sev-
eral intracellular signaling cascades including cell
proliferation and survival. The addition of pertu-
zumab to a taxane and trastuzumab combination
HER2-positive breast cancer: new
therapeutic frontiers and overcoming
resistance
Sonia Pernas and Sara M. Tolaney
Abstract: The introduction of anti-HER2 therapies to the treatment of patients with HER2-
positive breast cancer has led to dramatic improvements in survival in both early and
advanced settings. Despite this breakthrough, nearly all patients with metastatic HER2-
positive breast cancer eventually progress on anti-HER2 therapy due to de novo or acquired
resistance. A better understanding not only of the underlying mechanisms of HER2 therapy
resistance but of tumor heterogeneity as well as the host and tumor microenvironment is
essential for the development of new strategies to further improve patient outcomes. One
strategy has focused on inhibiting the HER2 signaling pathway more effectively with dual-
blockade approaches and developing improved anti-HER2 therapies like antibody–drug
conjugates, new anti-HER2 antibodies, bispecific antibodies, or novel tyrosine kinase inhibitors
that might replace or be used in addition to some of the current anti-HER2 treatments.
Combinations of anti-HER2 therapy with other agents like immune checkpoint inhibitors,
CDK4/6 inhibitors, and PI3K/AKT/mTOR inhibitors are also being extensively evaluated in
clinical trials. These add-on strategies of combining optimized targeted therapies could
potentially improve outcomes for patients with HER2-positive breast cancer but may also
allow de-escalation of treatment in some patients, potentially sparing some from unnecessary
treatments, and their related toxicities and costs.
Keywords:
breast cancer, drug–antibody conjugates, HER2-positive, new anti-HER2 therapies,
novel combinations, resistance, tyrosine kinase inhibitors
Received: 25 October 2018; revised manuscript accepted: 23 January 2019.
Correspondence to:
Sara M. Tolaney
Department of Medical
Oncology, Dana-Farber
Cancer Institute, 450
Brookline Avenue, Boston,
MA 02215, USA
Sara_Tolaney@DFCI.
HARVARD.EDU
Sonia Pernas
Department of Medical
Oncology, Dana-Farber
Cancer Institute, Boston,
MA, USA; Department of
Medical Oncology-Breast
Cancer Unit, Institut
Catala d’Oncologia (ICO)-
H.U. Bellvitge-IDIBELL,
Barcelona, Spain
833519TAM0010.1177/1758835919833519Therapeutic Advances in Medical OncologyS Pernas and SM Tolaney
review-article20192019
Review
Therapeutic Advances in Medical Oncology 11
2 journals.sagepub.com/home/tam
compared with taxane and trastuzumab therapy
alone as a first-line treatment in advanced HER2-
positive breast cancer resulted in an improvement
not only in PFS but also in OS by almost 16
months, reaching a median survival of nearly 5
years and establishing this regimen as the pre-
ferred regimen in the first-line setting.1 Finally,
trastuzumab emtansine (T-DM1) is an antibody–
drug conjugate (ADC) comprised of trastuzumab
covalently linked to a maytansine derivate (DM1),
a potent antimitotic agent that binds microtu-
bules.5 After selectively binding to HER2, the
conjugate is internalized within endocytic vesicles
and degraded in the lysosomes, releasing the
active payload within the cell. This results in cell
death by mitotic catastrophe.6 T-DM1 signifi-
cantly improved both PFS and OS compared
with lapatinib plus capecitabine as a second-line
treatment7 and as a later line in patients with
advanced HER2-positive breast cancer previously
treated with trastuzumab.8 Based on those results,
T-DM1 is currently the only ADC approved to
treat breast cancer and the standard second-line
therapy for advanced HER2-positive disease. To
date, there is no standard of care treatment for
patients with advanced HER2-positive tumors
following treatment with trastuzumab, pertu-
zumab and T-DM1. Treatment options at this
point include lapatinib plus capecitabine, combi-
nations of trastuzumab with other chemothera-
pies (such as vinorelbine or gemcitabine), or
dual-blockade combinations without chemother-
apy, such as trastuzumab with lapatinib or endo-
crine therapy with either trastuzumab or lapatinib
in patients with hormone receptor (HR)-positive
disease.
Despite the outstanding improvement in survival
with the introduction of anti-HER2 therapies
alone or as dual HER2-blockade in the standard
treatment of advanced disease, most patients ulti-
mately develop progressive disease and die.
Furthermore, up to 40–50% of patients with
advanced HER2-positive breast cancer will
develop brain metastases during their disease
course. Better options for the prevention and
treatment of brain metastases are clearly needed.9
A growing understanding of the underlying mech-
anisms of primary and acquired resistance to anti-
HER2 therapies and compensatory pathways as
well as tumor heterogeneity and the tumor micro-
environment is essential for the development of
novel therapeutic strategies. A substantial num-
ber of novel anti-HER2 treatments are being
investigated extensively in the preclinical and
clinical settings to further improve patient out-
comes. Here, we review the rationale and latest
evidence of those novel treatments and approaches
to overcome resistance in advanced HER2-
positive breast cancer.
Mechanisms of resistance and response
heterogeneity to anti-HER2 therapy
Many potential resistance mechanisms to anti-
HER2 therapy have been described that ultimately
lead to reactivation of the HER2 pathway or its
downstream signaling, through pathway redun-
dancy or stimulation of alternative survival path-
ways.10 Some of these mechanisms include
incomplete blockade of the HER2 receptor that
activates compensatory mechanisms within the
HER family (such as HER3), activation of alterna-
tive receptor tyrosine kinases (RTKs) or other
membrane receptors outside of the HER family
[such as insulin-like growth factor 1 receptor
(IGF-1R)11 and MET12], and alterations in down-
stream signaling pathways, such as hyperactivation
of the PI3K/AKT/mTOR pathway13,14 by reduced
levels of tumor suppressor genes (like PTEN and
INPP4-B), or by activating mutations in PIK3CA
(phosphatidylinositol-4,5 bisphosphate 3-kinase
catalytic subunit).15 Several other biologic features
have been associated with response heterogeneity
to anti-HER2 therapy, including HER2 mRNA or
protein levels,16 tumor intrinsic subtype,17 altera-
tions in the HER2-receptor (such as p95HER2),18
and host and tumor microenvironment compo-
nents, such as tumor infiltrating lymphocytes
(TILs)19 and FCγR polymorphisms.20 In the
CLEOPATRA trial for instance, high HER2 pro-
tein and high HER2 and HER3 mRNA levels were
associated with a significantly better outcome (p <
0.05). In contrast, PIK3CA mutation was identi-
fied as a strong negative prognostic biomarker,
despite deriving benefit from pertuzumab and tras-
tuzumab treatment.21 In the EMILIA trial, a
greater benefit in OS was also observed in patients
treated with T-DM1 and high HER2 mRNA
expression.22 Notably, PIK3CA mutations were
associated with significantly shorter PFS and OS
in patients treated with capecitabine plus lapatinib,
but not in T-DM1 treated patients (median PFS
10.9 vs. 9.8 months; OS, not reached in mutant or
wild type).22 Regarding TILs, an increased quan-
tity of stromal TILs was significantly associated
with improved OS in patients with advanced
HER2-positive breast cancer treated with doc-
etaxel, trastuzumab, and pertuzumab or placebo in
the CLEOPATRA trial.19
S Pernas and SM Tolaney
journals.sagepub.com/home/tam 3
It has also been demonstrated that the cyclin
D1-CDK4 pathway can mediate resistance to
HER2-targeting therapies in vitro and in vivo and
that targeting resistant tumor cells with CDK 4/6
inhibitors re-sensitizes them to anti-HER2 ther-
apy and delays tumor recurrence in HER2-driven
breast cancers in vivo in patient-derived xenograft
tumors.23 As discussed below, trials are currently
underway to evaluate the efficacy of combined
HER2 and CDK4/6 inhibition in HER2-positive
breast cancer.
Substantial preclinical and clinical studies support
the bidirectional cross-talk between HER2 and
estrogen receptor (ER) signaling when both recep-
tors are expressed in breast cancer cells.24 Tumors
that express both ER and HER2 are less sensitive
to endocrine therapy than ER-positive and HER2-
negative tumors, and ER can act as an escape
pathway to HER2 inhibition.25,26 Concurrent
inhibition of ER together with dual anti-HER2
therapy can improve outcomes, as demonstrated
in several trials in early and advanced HER2-
positive breast cancer.27–29
The HER2Δ16 splice variant is a major onco-
genic driver that promotes trastuzumab resist-
ance. Preclinical data suggest trastuzumab-resistant
HER2Δ16 cells are sensitive to the SRC kinase
inhibitor dasatinib and data from a phase I/II
(GEICAM/2010-04) study suggest there may be
a signal for activity when dasatinib is combined
with trastuzumab and paclitaxel in the first-line
treatment for patients with advanced HER2-
positive breast cancer.30,31 In addition, SRC acti-
vation by itself has been associated with
trastuzumab resistance.32
The mechanisms that contribute to T-DM1
resistance are not fully understood. There are
multiple components to consider when identify-
ing mechanisms of resistance for ADCs, such as
the ones related to the antibody, the linker or
the payload. Preclinical studies have shown that
CDK1/cyclin B1 activity is needed for T-DM1
action. Silencing cyclin B1 induces resistance to
T-DM1 while increasing the levels of cyclin B1
in resistant cells partially restores sensitivity.33
Other potential mechanisms of T-DM1 resist-
ance have been proposed including the reduc-
tion of the intracellular DM1 payload due to
upregulation of multidrug resistance proteins
(e.g. MDR1),34 impaired lysosomal proteolytic
activity35 or lysosomal transporter loss (e.g.
SLC46A3).34
Interestingly, molecular imaging seems promising
not only to further our understanding of tumor
heterogeneity in advanced HER2-positive breast
cancer but also to identify patients who will
unlikely benefit from T-DM1.36 In the prospec-
tive ZEPHIR trial, striking levels of inter- and
intrapatient heterogeneity in HER2 expression
were observed, with one-third of patients having
little or no trastuzumab-zirconium uptake
(HER2-Positron emission tomography (PET)/
computed tomography (CT) scan [PET/CT
scan]) across their metastatic sites. Moreover, the
combined use of HER2-PET/CT scan and early
fluorodeoxyglucose-PET/CT scan discriminated
patients treated with T-DM1 with a median time
to treatment failure (TTF) of 2.8 months from
those with 15 months of TTF.36 Despite the
extensive translational research being conducted,
most of the mechanisms of HER2 resistance and
potential biomarkers of response or resistance
either have not been clinically validated, or the
results are contradictory.37 To date, no biomarker
beyond HER2 exists for patient selection for anti-
HER2 therapy in HER2-positive breast cancer.
Of note, the interpretation of mechanisms of
resistance based solely in preclinical models can
be challenging due to tumor heterogeneity, the
complex nature of drug resistance and compensa-
tory pathways, and the use of different tumor cell
lines. Moreover, multiple mechanisms of resist-
ance may coexist in the same cell.
Novel strategies to overcome resistance to
HER2-targeted therapy
Replacement of current anti-HER2 therapies
for improved anti-HER2 drugs
ADCs. ADCs are a therapeutic class that provide
wider therapeutic window by more efficient and
specific drug delivery. ADCs exploit target selectiv-
ity of monoclonal antibodies (MAbs) to deliver
cytotoxic drugs to antigen-expressing cells to
improve tumor selectivity and reduce damage to
normal cells.38 The success observed with the first-
in-class T-DM1 has led to a rapid and extensive
development of new ADCs. Table 1 lists several
anti-HER2 ADCs in clinical development.7,39–41
Trastuzumab deruxtecan (DS-8201, Daiichi
Sankyo, Inc.) is an ADC comprising trastuzumab,
a cleavable drug linker, and a topoisomerase I pay-
load that has a high drug to antibody ratio (7–8).
In preclinical studies, DS-8201a showed a broader
anti-tumor activity than T-DM1, including
Therapeutic Advances in Medical Oncology 11
4 journals.sagepub.com/home/tam
efficacy against low HER2-expressing tumors. In
an updated subgroup analysis from a phase I study
with multiple expansion cohorts,42 DS-8201a
demonstrated an overall response rate (ORR) of
54.5% (54/99 patients) in patients with HER2-
positive metastatic breast cancer pretreated with
T-DM1 (as well as trastuzumab and pertuzumab
in the majority of cases). Median duration of
response and median PFS had not yet been
reached.39 Interestingly, in patients with HER2
low-expressing metastatic breast cancer [defined
as immunohistochemistry (IHC) 1+ or 2+ and in
situ hybridization (ISH)-negative], the ORR was
50% (17/34 patients). Trastuzumab deruxtecan
was relatively well tolerated; common adverse
events (AEs) included nausea (73.5%; 3.5% grade
3), decreased appetite (59.5%; 4.5% grade 3)
and vomiting (39.5%; 1.5% grade 3). In August
2017, trastuzumab deruxtecan received a United
States Food and Drug Administration (US FDA)
Breakthrough Therapy designation for the treat-
ment of patients with HER2-positive, locally
advanced, or metastatic breast cancer who have
been treated with trastuzumab and pertuzumab
and, have progressed to T-DM1. DS-8201a is
being evaluated in numerous trials, including two
phase III studies: the DESTINY-Breast02
(ClinicalTrials.gov identifier: NCT03523585) is
evaluating DS-8201a versus investigator’s choice
(capecitabine with trastuzumab or lapatinib) for
patients with HER2-positive, unresectable or met-
astatic breast cancer pretreated with prior T-DM1,
and the DESTINY-Breast03 (ClinicalTrials.gov
identifier: NCT03529110) is a randomized, open-
label study of DS-8201a versus T-DM1 for patients
with HER2-positive, metastatic breast cancer pre-
viously treated with trastuzumab and taxane.
SYD985 (Synthon Biopharmaceuticals BV) is a
third generation ADC based on trastuzumab and
a ‘cleavable’ linker – duocarmycin payload, which
is present as an inactive prodrug (valine-citrulline-
seco – DUocarmycin – hydroxyBenzamide –
Azaindole -vc-seco-DUBA). Proteases present in
endosomes result in the linker cleavage in SYD985
and the release of the membrane-permeable active
toxin. It then binds to the minor groove of DNA,
causing irreversible DNA alkylation. This results in
cell death in both dividing and nondividing cells in
the tumor microenvironment but also in neighbor-
ing tumor cells due to the bystander effect. SYD985
has shown impressive preclinical results in breast
cancer (even more potent than T-DM1) and
encouraging clinical activity. Results from a dose-
escalation phase I trial showed encouraging activity
in heavily pretreated patients43,44 and led the US
Table 1. HER2-directed ADCs in clinical development.
Agent Anti-HER2 MAb/payload
(target)
Drug to
antibody
ratio
Linker drug Phase of
development
ORR in
HER2-
positive
ORR in
HER2 low
(IHC1+/2+/ISH-)
Trastuzumab-
DM1 (T-DM1)7
Trastuzumab/
DM1 (anti-tubulin)
3.5 Noncleavable US FDA
Approved
43.6% ———
Trastuzumab
duruxtecan
(DS-8201a)39
Trastuzumab/ exatecan
derivative (topoisomerase I
inhibitor)
8 Cleavable II/III
NCT03248492
NCT03529110
NCT03523585
54.5% 50%
SYD98540 Duocarmycin derivative
(alkylating agent)
2.8 Cleavable III
NCT03262935
33% HR + 27%
HR − 40%
XMT-152241 XMT-1519/ monomethyl
auristatin (anti-tubulin)
12 Cleavable I
NCT02952729
unknown unknown
ARX788 Anti-HER2 MAb/ auristatin
analog 269 (AS269) (anti-
tubulin)
1.9 Non-
cleavable
I
NCT03255070
unknown unknown
DHES0815A Trastuzumab/ alkylator 2 Cleavable I
NCT03451162
unknown unknown
ADC, antibody–drug conjugate; HR+, hormone receptor positive; HR−, hormone receptor negative; IHC, immunohistochemistry; ISH, In Situ
Hybridization; MAb, monoclonal antibody; NCT, ClinicalTrials.gov identifier; ORR, overall response rate; US FDA, United States Food and Drug
Administration.
S Pernas and SM Tolaney
journals.sagepub.com/home/tam 5
FDA to grant the agent Fast Track designation in
January 2018. Results from the expansion cohorts
recently reported an ORR of 33% and a median
PFS of 9.4 months40 in a cohort of patients with
advanced HER2-positive disease, previously
treated with a median of six lines of therapy for
metastatic disease (n = 50). Interestingly, SYD985
was also effective in patients with HER2-low meta-
static breast cancer, with an ORR of 27% and 40%
in the HR-positive, HER2-low, and in the
HR-negative, HER2-low cohorts, respectively.
Enrollment was based on central HER2 analysis by
IHC and ISH and HER2 low was defined as
IHC 1+/2+ and fluorescence in situ hybridization
(FISH) negative. Most of the adverse drug reac-
tions were mild or moderate, with ocular toxicity
and fatigue being most frequently reported. A
phase III pivotal study (TULIP) is ongoing
(ClinicalTrials.gov identifier: NCT03262935)
which compares SYD985 with the treatment of the
physician’s choice in patients with HER2-positive
metastatic breast cancer in the third line and
beyond.
MEDI4276 (Medimmune) is a bispecific ADC
that targets two different domains of the HER2
receptor, resulting in crosslinking followed by
internalization of the complex, cleavage of the
linker, and release of the payload. MEDI-4276
comprises the single-chain variable fragment
(scFv) of trastuzumab, which binds to domain IV
of HER2, and the anti-HER2 Mab 39S, which
binds to domain II of HER2. The bispecific anti-
body is then conjugated, via a cleavable linker, to
the cytotoxic anti-microtubule agent tubulysin.
Results from the phase I study (ClinicalTrials.
gov identifier: NCT02576548) in patients with
advanced HER2-positive breast or gastric can-
cer45 showed clinical activity, but also considera-
ble toxicity with 28% of patients (12/43) having
drug-related AEs of grade 3–4 severity; most
common were grade 3 elevated aspartate
transaminase (AST; 19%) and grade 3 elevated
alanine transaminase (ALT; 12%). Given the
challenges with toxicity, clinical testing with this
agent has been discontinued for breast and gastric
cancers.
ADCT-502 (ADC Therapeutics) is also a novel
pyrrolobenzodiazepine (PBD)-based ADC that
targets HER2-expressing solid tumors, including
breast cancers.46 However, based on data from
the phase I study (ClinicalTrials.gov identifier:
NCT03125200) that showed that ADCT-502
did not meet the necessary efficacy and safety
profile required for patient benefit, clinical testing
of this drug was recently halted.
In preclinical models, another bivalent bipara-
topic HER2-targeting ADC that targets two non-
overlapping epitopes on HER2 and is conjugated
with microtubule inhibitor demonstrated supe-
rior activity than T-DM1 in breast cancer models
and was able to overcome T-DM1 resistance.
This biparatopic ADC also demonstrated
bystander killing activity.47
In contrast with T-DM1, most of these new
ADCs have a cleavable drug linker (see Table 1)
that mediates the bystander killing effect. This is
the passive diffusion of the free cytotoxin from
target-positive cancer cells into the tumor micro-
environment, killing neighboring cancer cells that
are insensitive to the ADC because of the lack or
limited target expression. This desired feature of
those novel HER2-targeting ADCs, given that
heterogeneity is frequent in HER2-positive breast
cancer, may be however, a double-edged sword
with an increased toxicity.
Novel anti-HER2 antibodies. Margetuximab
(MGAH22, MacroGenics) is an Fc-optimized
chimeric monoclonal antibody that binds to the
same epitope as trastuzumab. Margetuximab has
enhanced Fcγ receptor-binding properties with
an increased affinity for CD16A polymorphisms
and a decreased affinity for FcγRIIB (CD16B),
an inhibitory receptor, which allows it to bind
more tightly to effector cells and increase anti-
body-dependent cell-mediated cytotoxicity
(ADCC); it also preserves the antiproliferative
properties of trastuzumab.48 A first-in-human
phase I study demonstrated promising single-
agent activity of margetuximab in heavily pre-
treated patients with HER2-positive solid tumors.
Among 24 patients with metastatic breast cancer,
the ORR was 17% and 3 out of the 4 responders
remained on treatment for 39–54 months.49 The
most common AEs were grade 1–2 constitutional
symptoms and no cardiotoxicity was observed.
Margetuximab is currently being evaluated in the
randomized phase III SOPHIA trial (ClinicalTri-
als.gov identifier: NCT02492711) that compares
margetuximab plus chemotherapy with trastu-
zumab plus chemotherapy as a third-line therapy
in patients with HER2-positive breast cancer after
prior treatment with trastuzumab, pertuzumab,
and T-DM1. The US FDA has granted Fast Track
designation for the investigation of margetuximab
for the treatment of patients with metastatic or
Therapeutic Advances in Medical Oncology 11
6 journals.sagepub.com/home/tam
locally advanced HER-positive breast cancer pre-
viously treated with anti-HER2-targeted therapy.
Bispecific antibodies. Bispecific antibodies
(BsAbs) combine the functionality of two MAbs
that target two different targets or epitopes, either
in the same or in different receptors. BsAbs can
interfere with two or more RTK signaling path-
ways, by inactivating either the RTKs or their
ligand. Several are currently being studied in
patients with advanced HER2-positive disease.
MCLA-128 (Merus) is a full-length immuno-
globulin (Ig)G1 BsAb with enhanced ADCC
activity targeting both HER2 and HER3.
Preliminary results from a phase I/II study in
solid tumors (ClinicalTrials.gov identifier: NCT
02912949) that include eight patients with heav-
ily pretreated (median of five prior lines in the
metastatic setting) HER2-positive metastatic
breast cancer showed an encouraging clinical
benefit rate of 70%.50 A phase II study
(ClinicalTrials.gov identifier: NCT03321981) is
ongoing to evaluate the activity of MCLA-128
in two metastatic breast cancer populations: in
HER2-positive/amplified patients (cohort 1) in
combination with trastuzumab ± chemotherapy,
and in ER-positive/low-HER2 expression (cohort
2) in which MCLA-128 is administered in combi-
nation with endocrine therapy.
ZW25 (Zymeworks Inc.) is a novel Azymetric
bispecific antibody biparatopic that binds to two
different epitopes on the extracellular domain of
HER2 ECD2 and ECD4. This results in increased
tumor cell binding, blockade of ligand-dependent
and independent growth, and improved receptor
internalization and downregulation relative to tras-
tuzumab. Also, in vivo studies demonstrate anti-
tumor activity in HER2-low to high expressing
models. Results from the phase I study
(ClinicalTrials.gov identifier: NCT02892123)
evaluating the safety and efficacy of single-agent
ZW25 in separate expansion cohorts, including
HER2-high (IHC 3+ or 2+/FISH+) breast, gas-
tric/esophageal, and other cancers, demonstrated a
promising anti-tumor activity and no dose-limiting
toxicities were observed. In patients with heavily
pretreated HER2-expressing breast cancers that
had progressed to a median of five HER2-targeted
regimens for metastatic disease, a partial response
rate of 33% (6/18 patients) was observed with a dis-
ease control rate of 50%. The most common AEs
were diarrhea and infusion reaction, all grade 1 or
2, with no treatment-related discontinuations.51
T-cell bispecific antibodies (TCBs) are engi-
neered molecules that include, within a single
entity, binding sites to the invariant CD3 chain of
the T-cell receptor (TCR) and to tumor-associ-
ated or tumor-specific antigens. Binding to the
tumor antigen results in crosslinking of the TCR
and subsequent lymphocyte activation and tumor
cell killing. However, on-target off-tumor effects
caused by redirected lymphocytes can result in
severe toxicities. Several are currently in clinical
development:
GBR1302 (Glenmark Pharmaceuticals) is a
HER2xCD3 bsAb developed to direct T-cells to
HER2-expressing tumor cells. Preclinically,
GBR1302 has demonstrated potent killing of
HER2-positive human cancer cells, as well as
growth suppression of the trastuzumab-resistant
cell line JIMT-1. In contrast, the GBR1302 con-
centration required to kill primary cardiomyo-
cytes with normal HER2 levels was up to 1000
times greater than the concentration needed to
kill HER2 3+ tumor cell lines. A first-in-human
phase I study of single-agent GBR1302 in pro-
gressive HER2-positive solid tumors is ongoing
(ClinicalTrials.gov identifier: NCT02829372).
Preliminary biomarker and pharmacodynamic
data from this study were recently presented,
demonstrating modulation of peripheral T-cell
populations and cytokines.52
PRS-343 (Pieris Pharmaceuticals, Inc.) is a mon-
oclonal antibody-bispecific protein targeting
HER2 and the immune receptor CD137. CD137
is a key costimulatory immunoreceptor and a
member of the tumor necrosis factor receptor
superfamily. PRS-343 is designed to promote
CD137 clustering by bridging CD137-positive
T-cells with HER2-positive tumor cells, thereby
providing a potent costimulatory signal to tumor
antigen-specific T-cells that has demonstrated
tumor inhibition and TIL expansion in a human-
ized mouse model.53 The two clinical studies
evaluating PRS-343 in HER2-positive solid
tumors are ongoing, either as a single-drug agent
(ClinicalTrials.gov identifier: NCT03330561) or
in combination with atezolizumab (ClinicalTrials.
gov identifier: NCT03650348).
Expression of the tumor-specific antigen
p95HER2, a truncated form of HER2, occurs in
about 40% of HER2-positive tumors. Rius Ruiz
and colleagues have developed a TCB against
p95HER2 (p95HER2-TCB) that has a potent
anti-tumor effect on breast tumors expressing
S Pernas and SM Tolaney
journals.sagepub.com/home/tam 7
p95HER2, both in vitro and in vivo.54 In contrast
with HER2, p95HER2 is not expressed in nor-
mal tissues, therefore, it has no effect on nontu-
mor cells that do not overexpress HER2. Those
findings support further investigation with this
compound.
Novel TKIs. TKIs are orally bioavailable small
molecules developed to further block the HER
receptor family, acting on the intracellular
domain. Those HER-directed TKIs have a lower
molecular weight compared with MAbs, allowing
them a more efficacious penetration through the
blood–brain barrier and therefore, theoretically
may be more effective for the treatment of HER2
brain metastases. Lapatinib was the first TKI
approved in HER2-positive advanced breast can-
cer (and to date, remains the only one) based on
the results described above. Dual blockade with
lapatinib plus trastuzumab without chemother-
apy also demonstrated benefit in OS in heavily
pretreated patients with advanced disease, when
compared with lapatinib alone.55 Initial results
with these molecules, however, have not been the
ones initially expected, even in the treatment of
brain metastases.56,57As a first-line therapy, lapa-
tinib was found to be inferior to trastuzumab
when combined with paclitaxel.58 In the same
way, afatinib was found to be not as effective as
trastuzumab and less tolerated when each was
combined with vinorelbine in a phase III trial
(LUX-Breast1 study).59 No difference was
detected between lapatinib-capecitabine and
trastuzumab-capecitabine for the incidence of
brain metastases in the phase III CEREBEL
(EGF111438) study.56 In the phase II LUX-
Breast3 trial, patients with HER2-positive breast
cancer and progressive brain metastasis previ-
ously treatment with trastuzumab, lapatinib or
both, were randomized to afatinib alone, afatinib
plus vinorelbine or the investigator’s choice of
treatment. Similarly, afatinib-containing regimens
not only did not show better activity than investi-
gator-selected treatments but also seemed to be
less tolerated. No further development of afatinib
for HER2-positive breast cancer is currently
planned. There are, however, several novel TKIs
in clinical development (Table 2).60–66
Neratinib is an irreversible pan-HER TKI that has
been recently approved as an extended adjuvant
therapy in early HER2-positive breast cancer based
on the results of the phase III ExteNET study.67 In
previously untreated patients with HER2-positive
advanced breast cancer, neratinib-paclitaxel was
not superior to trastuzumab-paclitaxel in terms of
PFS in the randomized phase II NEfERT-T trial
that included 479 women.68 Median PFS was
12.9 months in both arms [hazard ratio, 1.02;
95% confidence interval (CI), 0.81–1.27; p =
0.89]. However, the incidence of central nervous
system (CNS) recurrences was significantly lower
(relative risk, 0.48; 95% CI, 0.29–0.79; p = 0.002)
and time to CNS metastases significantly delayed
with neratinib-paclitaxel (hazard ratio, 0.45; 95%
CI, 0.26–0.78; p = 0.004). Diarrhea and gastroin-
testinal toxicity (i.e. nausea, vomiting) were more
common with neratinib-paclitaxel (30.4% versus
3.8% of diarrhea grade 3); however, primary
prophylaxis for diarrhea was not mandatory in this
trial. In patients with HER2-positive breast cancer
brain metastases, the combination of neratinib
and capecitabine demonstrated a reduction of
CNS lesions in 49% of patients in the phase II
trial TBCRC 022.60 This regimen has now been
endorsed by the National Comprehensive Cancer
Network for the treatment of HER2-positive brain
metastases.69 The phase III NALA study (Clinical
Trials.gov identifier: NCT01808573) is directly
comparing the combination of neratinib plus
capecitabine to lapatinib plus capecitabine in
patients with HER2-positive metastatic breast
cancer who have received at least two prior HER2-
directed regimens in the metastatic setting. This
study has already completed recruitment and it
will provide valuable data on the real efficacy of
lapatinib and neratinib after the current standard
treatments of pertuzumab and T-DM1.
Tucatinib (ONT-380) is an oral HER2-
selective small molecule TKI with nanomolar
potency and is approximately1000-fold more
potent for HER2 than EGFR. Because of its
selectivity for HER2, there are fewer EGFR-
related toxicities, such as rash and diarrhea,
which are common with many of the other anti-
HER TKIs. In a phase Ib study, in which
tucatinib was combined with capecitabine or
trastuzumab in heavily pretreated patients with
HER2-positive metastatic breast cancer (includ-
ing patients with brain metastases), the triple
combination demonstrated an ORR of 61% (in
14 patients out of 23 with measurable disease)
and a median duration of response of 10 months
(95% CI: 2.8–19.3). In patients with measurable
brain metastases at baseline, an encouraging
ORR of 42% (5/12) in the brain was observed.62
Median PFS and median duration of response
were 7.8 months, and 10 months, respectively.
In June 2017, tucatinib was granted US FDA
Therapeutic Advances in Medical Oncology 11
8 journals.sagepub.com/home/tam
Orphan Drug status for HER2-positive patients
with brain metastases. An ongoing phase II trial,
HER2CLIMB (ClinicalTrials.gov identifier:
NCT02614794), is randomizing patients with
HER2-positive breast cancer with or without
brain metastases to capecitabine and trastu-
zumab with or without tucatinib. The combina-
tion of tucatinib and T-DM1 was evaluated in
another phase Ib study (ClinicalTrials.gov iden-
tifier: NCT01983501), showing promising
activity in patients with HER2-positive meta-
static breast cancer who had undergone a median
of two prior HER2 therapies. ORR was 48% and
the median PFS of 8.2 months (95% CI, 4.8–
10.3 months). Among the 30 patients with brain
metastases included in this study, the median
PFS was 6.7 months (95% CI, 4.1–10.2 months)
and the brain-specific ORR among patients with
measurable disease was 36% (including 2
patients with complete response).63 Importantly,
the combination of tucatinib with T-DM1
appeared to have an acceptable toxicity, with
most AEs attributable to T-DM1 and consistent
with those observed in other studies that used
T-DM1 as a single agent.
Pyrotinib is a novel irreversible pan-HER TKI
that has demonstrated promising activity and
acceptable tolerability in a phase II trial in patients
with advanced HER2-positive breast cancer
treated with at least two previous lines of ther-
apy.65,70 Patients were randomized to receive
pyrotinib plus capecitabine or lapatinib plus
capecitabine. ORR was 78.5% in the pyrotinib/
capecitabine arm and 57.1% in the capecitabine/
lapatinib arm, with a median PFS of 18 months
and 7 months, respectively (hazard ratio = 0.36; p
< 0.001). In contrast, pyrotinib was associated
with higher rates of AEs, which included hand-foot
syndrome (25 versus 21%), diarrhea (15 versus 5%)
and neutropenia (9 versus 3%). Biomarker analyses
suggest that PIK3CA and TP53 mutations in
Table 2. HER2-directed TKIs in clinical development.
Agent Target Reported results of
efficacy in HER2-positive
advanced disease
CNS ORR
(monotherapy)
CNS ORR in
combination with
capecitabine
Phase of
development
Neratinib60,61 Irreversible
pan-HER
Single-agent
ORR 56% (phase II)
8% 49% (phase II) US FDA approved
only in the adjuvant
setting
III (metastatic)
NALA-NCT01808573
Tucatinib 62–64 Selectively
inhibits HER2
relative to EGFR
In combination with
capecitabine and
trastuzumab:
ORR 61%
PFS of 7.8m
In combination with
T-DM1:
ORR 48%
PFS 8.2 m
(phase Ib)
5–9%
(+trastuzumab)
42%
(+trastuzumab)
II
HER2CLIMB-
NCT02614794
Pyrotinib65 Irreversible
pan-HER
Single-agent ORR 50%,
CBR 61%, PFS 35.4 w
(phase I)
In combination with
capecitabine ORR 78.5%
PFS 18 m (phase II)
NA NA III
NCT003080805
Poziotinib66 Irreversible
pan-HER
Single-agent
DCR 75%
PFS 4 m (phase II)
NA NA II
CBR, clinical benefit rate; CNS, central nervous system; DCR, disease control rate; EGFR, epidermal growth factor receptor; m, months; NA, not
applicable; NCT, ClinicalTrials.gov identifier; ORR, overall response rate; PFS, progression-free survival; TKI, tyrosine kinase inhibitor; US FDA,
United States Food and Drug Administration; w, weeks.
S Pernas and SM Tolaney
journals.sagepub.com/home/tam 9
circulating tumor DNA rather than in archival
tumor samples may predict response to pyro-
tinib.65,70 An ongoing phase III trial (ClinicalTrials.
gov identifier: NCT03080805) is comparing
pyrotinib plus capecitabine versus lapatinib plus
capecitabine in patients with HER2-positive
breast cancer previously treated with trastuzumab
and taxane.
Poziotinib is also an oral pan-HER kinase show-
ing potent activity through irreversible inhibition
of these kinases. Results from a single-arm, phase
II trial (ClinicalTrials.gov identifier: NCT0241
8689) evaluating the efficacy and safety of pozio-
tinib as monotherapy in heavily pretreated
patients with HER2-positive metastatic breast
cancer showed a disease control rate of 75.5%
(77/102), including 20 patients with confirmed
partial response and a median PFS of 4 months
(95% CI, 2.9–4.4 months). The most common
AEs were diarrhea and stomatitis (being grade 3
15% and 26%, respectively).66
Combinations of anti-HER2 agents with other
drugs
Immunotherapy. Preclinical and clinical data
suggest that HER2-positive breast cancer is
immunogenic.71 In contrast with luminal tumors,
HER2-positive tumors have a higher mutational
burden, and harbor higher numbers of TILs and
programmed cell death protein 1 ligand (PD-L1)
positivity.72 In addition, mechanisms of action of
anti-HER2 MAbs include not only ADCC but
also the generation of adaptive immunity.73
Together, these data support the rationale of
combining anti-HER2 therapies with immune
checkpoint blockade (anti-PD-1 or anti-PD-L1
agents). Results from the JAVELIN phase I
study,74 however, were disappointing and no
responses were seen with single-agent avelumab
in the subgroup of patients with advanced pre-
treated HER2-positive breast cancer. The first
study to evaluate the addition of pembrolizumab
to trastuzumab in patients with trastuzumab-
resistant HER2-positive breast cancer was the
PANACEA (IBCSG 45-13/BIG 4-13/KEY-
NOTE-014) study. This phase Ib/II study dem-
onstrated that the combination was associated
with an ORR of 15.2% and a median of PFS and
OS of 2.7 months and 16 months, respectively, in
PD-L1 positive patients.75 However, these data
also highlight the limitations of this combination,
as no responses were seen in the PD-L1 negative
cohort and most of the PD-L1 positive patients
who initially responded eventually developed
resistant disease. Several randomized studies are
ongoing to further evaluate the role of immune
checkpoints inhibitors in HER2-positive meta-
static breast cancer. Results from the phase II
KATE2 trial (ClinicalTrials.gov identifier:
NCT02924883) assessing the efficacy and safety
of T-DM1 in combination with atezolizumab or
placebo in pretreated patients with HER2-posi-
tive advanced breast cancer were recently pre-
sented.76 In this trial, the addition of atezolizumab
to T-DM1 did not demonstrate a significant PFS
benefit in the ITT population (8.2 versus 6.8
months; hazard ratio 0.82; 95% CI 0.55–1.23).
However, an exploratory endpoint demonstrated
promising PFS in the PD-L1 positive (PD-L1
IHC expression >1%) and stromal TIL sub-
groups.76 The phase III NRG-BR004 trial
(ClinicalTrials.gov identifier: NCT03199885) is
investigating the combination of paclitaxel, trastu-
zumab, pertuzumab with or without atezolizumab
as a first-line treatment. In addition, trastuzumab
deruxtecan (DS-8201a) is also being evaluated in
combination with nivolumab in a phase Ib study
(ClinicalTrials.gov identifier: NCT03523572) in
patients with advanced breast (with high and low
HER2 expression) and urothelial cancers.
Immunotherapy with HER2-targeting vaccines
are also being currently investigated in clinical tri-
als. The HER2 vaccine NeuVaxTM (Nelipepimut-S
or E75 peptide combined with granulocyte mac-
rophage-colony stimulating factor)77 is being
evaluated in two phase II clinical trials in combi-
nation with trastuzumab in breast cancer patients
with HER2–3+ (ClinicalTrials.gov identifier:
NCT02297698) and in HER2–1+/2+
(ClinicalTrials.gov identifier: NCT02297698),
respectively. ETBX-021 is another HER2-
targeting vaccine comprising an Ad5 vector and a
modified HER2 gene insert that is being evalu-
ated in a phase I clinical trial with locally advanced
or metastatic HER2-low-expressing (IHC
1+/2+) breast cancer.
CDK4/6 inhibitors
There is a strong rationale to evaluate CDK4/6
inhibitors in HER2-positive breast cancer.
Activity of CDK4/6 is regulated by several mech-
anisms that include mitogenic signaling pathways
(such as HER2) by increasing CCND1 expression
or increasing cyclin D1 protein stability.23,78 In
addition, mouse models have shown that cyclin
D1/CDK4 plays an important role in the
Therapeutic Advances in Medical Oncology 11
10 journals.sagepub.com/home/tam
formation and growth of breast tumors driven by
ERBB2.23,79,80 Preclinical studies demonstrate a
clear synergy between anti-HER2 therapy and
CDK4/6 inhibitors,81,82 and that CDK4/6 inhibi-
tion can specifically overcome acquired resistance
to anti-HER2 therapy.23 Moreover, CDK4/6
inhibition delayed recurrence of HER2-driven
breast cancers in mouse models.23 Early clinical
data also support the use of CDK4/6 inhibitors in
HER2-driven breast cancers, especially in the
subset of patients with ER-positive, HER2-
positive disease.83,84 Currently, there are many
clinical trials evaluating the role of CDK4/6
inhibitors in advanced HER2-positive breast can-
cer, including two global, randomized trials: the
MonarcHER study (ClinicalTrials.gov identifier:
NCT02675231), which evaluates the role of abe-
maciclib with trastuzumab in pretreated meta-
static disease, and the PATINA study
(ClinicalTrials.gov identifier: NCT02947685),
which explores the benefits of adding palbociclib
to trastuzumab, pertuzumab and an aromatase
inhibitor after an induction of standard first-line
therapy. Initial reports from the phase II SOLTI-
1303 PATRICIA trial (ClinicalTrials.gov identi-
fier: NCT02448420), which evaluates the
combination of palbociclib, trastuzumab ± letro-
zole in heavily pretreated (up to 2–4 prior lines in
the metastatic setting) patients with HER2-
positive breast cancer, suggest that the combina-
tion is active particularly in the luminal subtype
by PAM50, with a better PFS compared with
nonluminal (12.4 versus 4.1 months, hazard ratio
0.30, 95% CI;0.11–0.86 p = 0.025).85,86 Thus,
identification of the nonluminal subtypes by
PAM50 might help to identify those patients who
might not derive a great benefit from this treat-
ment strategy, regardless of HR status. Other
nonrandomized phase Ib/II studies in advanced
HER2-positive breast cancer are ongoing, includ-
ing those combining palbociclib and T-DM1
(ClinicalTrials.gov identifier: NCT01976169);
palbociclib, trastuzumab, pertuzumab and anas-
trozole (ClinicalTrials.gov identifier: NCT033
04080); ribociclib with trastuzumab or T-DM1
(ClinicalTrials.gov identifier: NCT02657343);
and palbociclib with tucatinib and letrozole
(ClinicalTrials.gov identifier: NCT03054363).
The JPBO trial (ClinicalTrials.gov identifier:
NCT02308020) is testing abemaciclib as a single
agent in patients with brain metastasis secondary
to HR-positive breast cancer, non-small cell lung
cancer, or melanoma, including a cohort of
patients with HR+, HER2-positive breast cancer.
However, within the cohort of HR-positive,
HER2-positive patients, there were no objective
responses seen at the time of the interim analysis,
and the cohort was not able to move to the second
stage.87
PI3K/Akt/mTOR inhibitors
As mentioned previously, dysregulations in the
PI3K/AKT/mTOR pathway seem to play an
important role in trastuzumab resistance. PI3K
inhibition results in an enhanced HER2 signaling
in HER2-overexpressing breast cancer, especially
in an increased expression of HER2 and HER3.88
Targeting both pathways could prevent the devel-
opment of resistance. However, results of two
phase III trials evaluating the role of everolimus,
an mTOR inhibitor, in combination with either
trastuzumab plus paclitaxel as first-line treatment
(BOLERO-1)89 or in combination with trastu-
zumab plus vinorelbine in trastuzumab-resistant
(BOLERO-3)90 advanced HER2-positive breast
cancer were quite disappointing with a significant
increase in toxicity. Although in the PFS sub-
group analysis of both studies, the benefit of add-
ing everolimus to the standard therapy seemed
greater in patients who had HR-negative dis-
ease.89,90 Moreover, the combined biomarker
analyses of the BOLERO-1 and BOLERO-3 tri-
als demonstrate an improved PFS in patients har-
boring PIK3CA mutations or PTEN loss when
treated with everolimus.91 Current efforts have
focused on evaluating α-specific PI3K inhibitors,
the isoform encoded by the PIK3CA gene, such
as alpelisib (BYL719) in combination with anti-
HER2 therapies. Alpelisib was combined with
LJM716 (a HER3 inhibitor) and trastuzumab in
patients with HER2-positive advanced breast
cancer with a PIK3CA mutation and prior pertu-
zumab and T-DM1 (ClinicalTrials.gov identifier:
NCT02167854). Preliminary results of this com-
bination showed a high toxicity profile including
diarrhea, hyperglycemia, hypokalemia, mucositis
and transaminitis, and limited activity (best
response was stable disease (SD) in five of six
evaluable patients).92 A phase I study of alpelisib
and T-DM1 in heavily pretreated HER2-positive
patients showed significant activity, with an ORR
of 43% and median PFS of 8.1 months (95% CI
3.9–10.8). Furthermore, activity was observed in
T-DM1-resistant patients with an ORR and clini-
cal benefit rate of 30% and 60%, respectively,
and median PFS of 6.3 months (95% CI 1.6–
10.5). The dose-limiting toxicity (DLT) was a
maculopapular rash. Most frequently reported
toxicities included fatigue, rash, gastrointestinal
S Pernas and SM Tolaney
journals.sagepub.com/home/tam 11
side effects, thrombocytopenia, anemia, elevated
liver enzymes, and hyperglycemia.93 Taselisib
(GDC-0032), a β-sparing PI3K inhibitor, is
being evaluated in an ongoing phase Ib dose-
escalation trial in combination with different anti-
HER2 therapies in patients with advanced
HER2-positive breast cancer (ClinicalTrials.gov
identifier: NCT02390427). Copanlisib is a pan-
class I PI3K inhibitor with particular activity
against PI3Kα that is being evaluated in combi-
nation with trastuzumab. Results from a phase Ib
(ClinicalTrials.gov identifier: NCT02705859;
PantHER trial) in pretreated metastatic HER2-
positive breast cancer (with a median of four prior
lines) showed no DLTs but grade 3 hypertension
was reported in 33% (n = 4) of patients. The best
response was stable disease in 9/12 patients and 6
patients continued treatment 16 weeks. The
PIK3CA mutation was present in 6/12 (50%) of
tumors.94 MEN1611 is a potent, selective, orally
available class I PI3k inhibitor showing high activ-
ity against p110α mutant isoforms, and minimal
inhibition of the δ isoform that is going to be eval-
uated in a phase Ib study in combination with
trastuzumab with or without fulvestrant (B-PRE
CISE-01 study). The study will enroll patients
with PIK3CA-mutated, HER2-positive, advan-
ced breast cancer pretreated with anti-HER2
based therapy.
Conclusion
Although the use of anti-HER2-targeted therapy
has dramatically changed the outlook for patients
with advanced HER2-positive breast cancer,
almost all patients ultimately experience disease
progression. Most of them advance to the point
where no approved HER2-targeting treatment
controls their disease. This might change in the
near future as many promising anti-HER2 thera-
pies are being developed in this setting. The newer
HER2 ADCs such as DS-8201a and SYD985
may replace T-DM1 in the second-line treatment
space or may be utilized in the third-line and
beyond setting after progression on T-DM1.
Moreover, these newer ADCs, as opposed to
T-DM1, are active not only in patients with
HER2-positive breast cancer but also in patients
with HER2 low-expressing tumors (IHC1+ or
2+/FISH) in whom to date, there are no current
anti-HER2 therapies specifically indicated. This
desired feature of its bystander effect may be par-
ticularly useful in heterogeneous cancer cell popu-
lations among HER2-positive disease. On the
other hand, the new TKIs, such as neratinib or
tucatinib, are being explored with capecitabine
and may demonstrate activity in the third-line
setting, though may be associated with increased
toxicity relative to trastuzumab. And finally, mar-
getuximab plus chemotherapy is being compared
with trastuzumab plus chemotherapy also in the
third-line setting in a registrational study.
These strategies of combining optimized HER2-
targeted therapies could potentially improve out-
comes for HER2-positive breast cancer patients
but may also allow de-escalation of treatment in
selected patients, potentially sparing some from
unnecessary treatments and their related toxicities.
Hence, potential biomarkers of response or resist-
ance such as intrinsic subtypes, might be helpful to
better select patients for these strategies. Moreover,
specific strategies for HR-positive, HER2-positive
breast cancer are needed, such as the current stud-
ies exploring the use of CDK 4/6 inhibitors in the
first and later line setting with anti-HER2 therapy
and specific trials allowing patients with progres-
sive brain metastases (who are generally excluded
from clinical trials) should be enhanced. The
underlying mechanisms of resistance to anti-HER2
therapies and compensatory pathways are indeed
complex and a wide range of mechanisms of resist-
ance may coexist in the same cell. Therefore, com-
bining clinical strategies and strengthening
international collaborations in the translational
setting might be needed to validate predictive bio-
markers beyond HER2, which will help us to bet-
ter select patients and improve their outcomes.
Acknowledgements
We thank Kaitlyn T. Bifolck, BA, for her editorial
support. Fundación AECC (Asociación Española
Contra el Cáncer) and the Spanish Society of
Medical Oncology (SEOM) grants (to S. Pernas)
Funding
This manuscript did not receive specific funding.
Conflict of interest statement
S. Pernas has received honoraria for talks and
travel grants from Roche, outside of the submit-
ted work and has served on advisory boards for
Polyphor. S. Tolaney receives institutional
research funding from Eli Lilly, Pfizer, Novartis,
Exelixis, Eisai, Merck, Bristol Meyers Squibb,
AstraZeneca, Nektar, Nanostring, Cyclacel, and
Immunomedics. S. Tolaney has served on advi-
sory boards or as a consultant for Eli Lilly, Pfizer,
Novartis, Eisai, Merck, AstraZeneca, Nektar,
Nanostring, Immunomedics, and Puma.
Therapeutic Advances in Medical Oncology 11
12 journals.sagepub.com/home/tam
ORCID iD
Sonia Pernas https://orcid.org/0000-0002-14
85-5080
References
1. Swain SM, Baselga J, Kim SB, etal. Pertuzumab,
trastuzumab, and docetaxel in HER2-positive
metastatic breast cancer. N Engl J Med 2015;
372: 724–734.
2. Slamon DJ, Clark GM, Wong SG, etal. Human
breast cancer: correlation of relapse and survival
with amplification of the HER-2/neu oncogene.
Science 1987; 235: 177–182.
3. Slamon DJ, Leyland-Jones B, Shak S, etal. Use
of chemotherapy plus a monoclonal antibody
against HER2 for metastatic breast cancer that
overexpresses HER2. N Engl J Med 2001; 344:
783–792.
4. Geyer CE, Forster J, Lindquist D, etal. Lapatinib
plus capecitabine for HER2-positive advanced
breast cancer. N Engl J Med 2006; 355: 2733–
2743.
5. Lewis Phillips GD, Li G, Dugger DL, etal.
Targeting HER2-positive breast cancer with
trastuzumab-DM1, an antibody-cytotoxic drug
conjugate. Cancer Res 2008; 68: 9280–9290.
6. Barok M, Tanner M, Koninki K, etal.
Trastuzumab-DM1 causes tumour growth
inhibition by mitotic catastrophe in trastuzumab-
resistant breast cancer cells in vivo. Breast Cancer
Res 2011; 13: R46.
7. Verma S, Miles D, Gianni L, etal. Trastuzumab
emtansine for HER2-positive advanced breast
cancer. N Engl J Med 2012; 367: 1783–1791.
8. Krop IE, Kim SB, Martin AG, etal.
Trastuzumab emtansine versus treatment of
physician’s choice in patients with previously
treated HER2-positive metastatic breast cancer
(TH3RESA): final overall survival results from a
randomised open-label phase 3 trial. Lancet Oncol
2017; 18: 743–754.
9. Lin NU. Better treatments needed for breast
cancer brain metastases. Lancet Oncol 2015; 16:
1583–1584.
10. Rimawi MF, Schiff R and Osborne CK.
Targeting HER2 for the treatment of breast
cancer. Annu Rev Med 2015; 66: 111–128.
11. Nahta R, Yuan LX, Zhang B, etal. Insulin-like
growth factor-I receptor/human epidermal growth
factor receptor 2 heterodimerization contributes
to trastuzumab resistance of breast cancer cells.
Cancer Res 2005; 65: 11118–11128.
12. Minuti G, Cappuzzo F, Duchnowska R, etal.
Increased MET and HGF gene copy numbers are
associated with trastuzumab failure in HER2-
positive metastatic breast cancer. Br J Cancer
2012; 107: 793–799.
13. Nagata Y, Lan KH, Zhou X, etal. PTEN
activation contributes to tumor inhibition
by trastuzumab, and loss of PTEN predicts
trastuzumab resistance in patients. Cancer Cell
2004; 6: 117–127.
14. Berns K, Horlings HM, Hennessy BT, etal. A
functional genetic approach identifies the PI3K
pathway as a major determinant of trastuzumab
resistance in breast cancer. Cancer Cell 2007; 12:
395–402.
15. Chandarlapaty S, Sakr RA, Giri D, etal.
Frequent mutational activation of the PI3K-AKT
pathway in trastuzumab-resistant breast cancer.
Clin Cancer Res 2012; 18: 6784–6791.
16. Nuciforo P, Thyparambil S, Aura C, etal. High
HER2 protein levels correlate with increased
survival in breast cancer patients treated with anti-
HER2 therapy. Mol Oncol 2016; 10: 138–147.
17. Carey LA, Berry DA, Cirrincione CT, etal.
Molecular heterogeneity and response to
neoadjuvant human epidermal growth factor
receptor 2 targeting in CALGB 40601, a
randomized phase III trial of paclitaxel plus
trastuzumab with or without lapatinib. J Clin
Oncol 2016; 34: 542–549.
18. Scaltriti M, Rojo F, Ocana A, etal. Expression of
p95HER2, a truncated form of the HER2 receptor,
and response to anti-HER2 therapies in breast
cancer. J Natl Cancer Inst 2007; 99: 628–638.
19. Luen SJ, Salgado R, Fox S, etal. Tumour-
infiltrating lymphocytes in advanced HER2-
positive breast cancer treated with pertuzumab or
placebo in addition to trastuzumab and docetaxel:
a retrospective analysis of the CLEOPATRA
study. Lancet Oncol 2017; 18: 52–62.
20. Musolino A, Naldi N, Bortesi B, etal.
Immunoglobulin G fragment C receptor
polymorphisms and clinical efficacy of
trastuzumab-based therapy in patients with
HER-2/neu-positive metastatic breast cancer. J
Clin Oncol 2008; 26: 1789–1796.
21. Baselga J, Cortes J, Im SA, etal. Biomarker
analyses in CLEOPATRA: a phase III, placebo-
controlled study of pertuzumab in human
epidermal growth factor receptor 2-positive, first-
line metastatic breast cancer. J Clin Oncol 2014;
32: 3753–3761.
22. Baselga J, Lewis Phillips GD, Verma S, etal.
Relationship between tumor biomarkers
S Pernas and SM Tolaney
journals.sagepub.com/home/tam 13
and efficacy in EMILIA, a phase III study
of trastuzumab emtansine in HER2-positive
metastatic breast cancer. Clin Cancer Res 2016;
22: 3755–3763.
23. Goel S, Wang Q, Watt AC, etal. Overcoming
therapeutic resistance in HER2-positive breast
cancers with CDK4/6 inhibitors. Cancer Cell
2016; 29: 255–269.
24. Giuliano M, Hu H, Wang YC, etal. Upregulation
of ER signaling as an adaptive mechanism of cell
survival in HER2-positive breast tumors treated
with anti-HER2 therapy. Clin Cancer Res 2015;
21: 3995–4003.
25. 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: 5529–5537.
26. Johnston S, Pippen J Jr., 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: 5538–5546.
27. Rimawi MF, Mayer IA, Forero A, etal.
Multicenter phase II study of neoadjuvant
lapatinib and trastuzumab with hormonal
therapy and without chemotherapy in patients
with human epidermal growth factor receptor
2-overexpressing breast cancer: TBCRC 006. J
Clin Oncol 2013; 31: 1726–1731.
28. Llombart-Cussac A, Cortes J, Pare L, etal.
HER2-enriched subtype as a predictor of
pathological complete response following
trastuzumab and lapatinib without chemotherapy
in early-stage HER2-positive breast cancer
(PAMELA): an open-label, single-group,
multicentre, phase 2 trial. Lancet Oncol 2017; 18:
545–554.
29. Rimawi M, Ferrero JM, de la Haba-Rodriguez
J, etal. First-line trastuzumab plus an aromatase
inhibitor, with or without pertuzumab, in human
epidermal growth factor receptor 2-positive
and hormone receptor-positive metastatic or
locally advanced breast cancer (PERTAIN):
a randomized, open-label phase II trial. J Clin
Oncol 2018; 36: 2826–2835.
30. Ocana A, Gil-Martin M, Martin M, etal. A phase
I study of the SRC kinase inhibitor dasatinib with
trastuzumab and paclitaxel as first line therapy
for patients with HER2-overexpressing advanced
breast cancer. GEICAM/2010-04 study.
Oncotarget 2017; 8: 73144–73153.
31. Ocana Fernandez A, Ruiz Borrego M, Gil Martin
M, etal. A phase II trial of dasatinib (D) in
combination with trastuzumab (T) and paclitaxel
(P) in the first line treatment of HER2 positive
metastatic breast cancer (MBC) patients (pts):
GEICAM/2010-04 [abstract]. Ann Oncol 2017;
28(Suppl. 5): abstract 239PD.
32. Zhang S, Huang WC, Li P, etal. Combating
trastuzumab resistance by targeting SRC, a
common node downstream of multiple resistance
pathways. Nat Med 2011; 17: 461–469.
33. Sabbaghi M, Gil-Gomez G, Guardia C, etal.
Defective cyclin B1 induction in trastuzumab-
emtansine (T-DM1) acquired resistance in
HER2-positive breast cancer. Clin Cancer Res
2017; 23: 7006–7019.
34. Li G, Guo J, Shen BQ, etal. Mechanisms of
acquired resistance to trastuzumab emtansine in
breast cancer cells. Mol Cancer Ther 2018; 17:
1441–1453.
35. Rios-Luci C, Garcia-Alonso S, Diaz-Rodriguez
E, etal. Resistance to the antibody-drug
conjugate T-DM1 is based in a reduction in
lysosomal proteolytic activity. Cancer Res 2017;
77: 4639–4651.
36. Gebhart G, Lamberts LE, Wimana Z, etal.
Molecular imaging as a tool to investigate
heterogeneity of advanced HER2-positive breast
cancer and to predict patient outcome under
trastuzumab emtansine (T-DM1): the ZEPHIR
trial. Ann Oncol 2016; 27: 619–624.
37. Gingras I, Gebhart G, de Azambuja E, etal.
HER2-positive breast cancer is lost in translation:
time for patient-centered research. Nat Rev Clin
Oncol 2017; 14: 669–681.
38. Trail PA, Dubowchik GM and Lowinger TB.
Antibody drug conjugates for treatment of breast
cancer: Novel targets and diverse approaches
in ADC design. Pharmacol Ther 2018; 181:
126–142.
39. Iwata H, Tamura K, Doi T, etal. Trastuzumab
deruxtecan (DS-8201a) in subjects with HER2-
expressing solid tumors: long-term results of
a large phase 1 study with multiple expansion
cohorts [abstract]. J Clin Oncol 2018; 36(Suppl.):
abstract 2501.
40. Saura C, Thistlethwaite F, Banerji U, etal. A
phase I expansion cohorts study of SYD985 in
heavily pretreated patients with HER2-positive or
HER2-low metastatic breast cancer [abstract]. J
Clin Oncol 2018; 36(Suppl.): abstract 1014.
41. Yurkovetskiy A, Gumerov D, Ter-Ovanesyan E,
etal. Non-clinical pharmacokinetics of XMT-
1522, a HER2 targeting auristatin-based antibody
Therapeutic Advances in Medical Oncology 11
14 journals.sagepub.com/home/tam
drug conjugate [abstract]. Proc Am Assoc Cancer
Res 2017; 58: abstract 48.
42. Doi T, Shitara K, Naito Y, etal. Safety,
pharmacokinetics, and antitumour activity of
trastuzumab deruxtecan (DS-8201), a HER2-
targeting antibody-drug conjugate, in patients
with advanced breast and gastric or gastro-
oesophageal tumours: a phase 1 dose-escalation
study. Lancet Oncol 2017; 18: 1512–1522.
43. van Herpen CM, Banerji U, Mommers EC,
etal. Phase I dose-escalation trial with the DNA-
alkylating anti-HER2 antibody-drug conjugate
SYD985 [abstract]. Eur J Cancer 2015;
51(Suppl. 3): S65.
44. Aftimos PG, van Herpen CM, Mommers EC,
etal. SYD985, a novel anti-HER2 ADC, shows
promising activity in patients with HER2-positive
and HER2-negative metastatic breast cancer
[abstract]. San Antonio, TX: San Antonio Breast
Cancer Symposium, 2016.
45. Pegram M, Hamilton E, Tan AR, etal. Phase
1 study of bispecific HER2 antibody-drug
conjugate MEDI4276 in patients with advanced
HER2-positive breast or gastric cancer [abstract].
Ann Oncol 2018; 29: mdy048.05.
46. Zammarchi F, Chivers S, Williams DG, etal.
ADCT-502, a novel pyrrolobenzodiazepine
(PBD)-based antibody–drug conjugate (ADC)
targeting low HER2-expressing solid cancers
[abstract]. Eur J Cancer 2016; 69: S28.
47. Li JY, Perry SR, Muniz-Medina V, etal. A
biparatopic HER2-targeting antibody-drug
conjugate induces tumor regression in
primary models refractory to or ineligible for
HER2-targeted therapy. Cancer Cell 2016; 29:
117–129.
48. Nordstrom JL, Gorlatov S, Zhang W, etal.
Anti-tumor activity and toxicokinetics analysis of
MGAH22, an anti-HER2 monoclonal antibody
with enhanced Fcγ receptor binding properties.
Breast Cancer Res 2011; 13: R123.
49. Bang YJ, Giaccone G, Im SA, etal. First-
in-human phase 1 study of margetuximab
(MGAH22), an Fc-modified chimeric
monoclonal antibody, in patients with HER2-
positive advanced solid tumors. Ann Oncol 2017;
28: 855–861.
50. Alsina M, Boni V, Schellens JH, etal. First-in-
human phase 1/2 study of MCLA-128, a full
length IgG1 bispecific antibody targeting HER2
and HER3: final phase 1 data and preliminary
activity in HER2+ metastatic breast cancer
(MBC) [abstract]. J Clin Oncol 2017; 35
(Suppl.): abstract 2522.
51. Meric-Bernstam F, Beeram M, Ignacio
Mayordomo J, etal. Single agent activity of
ZW25, a HER2-targeted bispecific antibody,
in heavily pretreated HER2-expressing cancers
[abstract]. J Clin Oncol 2018; 36 (Suppl.):
abstract 2500.
52. Wermke M, Alt J, Kauh JS, etal. Preliminary
biomarker and pharmacodynamic data from a
phase I study of single-agent bispecific antibody
T-cell engager GBR 1302 in subjects with HER2-
positive cancers [abstract]. J Clin Oncol 2018; 36:
69.
53. Hinner MJ, Bel Aiba R, Wiedenmann A, etal.
Costimulatory T cell engagement via a novel
bispecific anti-CD137/anti-HER2 protein
[abstract]. J Immunother Cancer 2015; 3: P187.
54. Rius Ruiz I, Vicario R, Morancho B, etal.
p95HER2-T cell bispecific antibody for breast
cancer treatment. Sci Transl Med 2018; 10.
55. 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: 2585–2592.
56. Pivot X, Manikhas A, Zurawski B, etal.
CEREBEL (EGF111438): a phase III,
randomized, open-label study of lapatinib
plus capecitabine versus trastuzumab plus
capecitabine in patients with human epidermal
growth factor receptor 2-positive metastatic
breast cancer. J Clin Oncol 2015; 33: 1564–1573.
57. Cortes J, Dieras V, Ro J, etal. Afatinib alone or
afatinib plus vinorelbine versus investigator’s
choice of treatment for HER2-positive breast
cancer with progressive brain metastases after
trastuzumab, lapatinib, or both (LUX-Breast 3):
a randomised, open-label, multicentre, phase 2
trial. Lancet Oncol 2015; 16: 1700–1710.
58. Gelmon KA, Boyle FM, Kaufman B, etal.
Lapatinib or trastuzumab plus taxane therapy
for human epidermal growth factor receptor
2-positive advanced breast cancer: final results
of NCIC CTG MA.31. J Clin Oncol 2015; 33:
1574–1583.
59. Harbeck N, Huang CS, Hurvitz S, etal. Afatinib
plus vinorelbine versus trastuzumab plus
vinorelbine in patients with HER2-overexpressing
metastatic breast cancer who had progressed
on one previous trastuzumab treatment (LUX-
Breast 1): an open-label, randomised, phase 3
trial. Lancet Oncol 2016; 17: 357–366.
60. Freedman RA, Gelman RS, Melisko M, etal.
TBCRC 022: phase II trial of neratinib +
S Pernas and SM Tolaney
journals.sagepub.com/home/tam 15
capecitabine for patients (Pts) with human
epidermal growth factor receptor 2 (HER2+)
breast cancer brain metastases (BCBM)
[abstract]. J Clin Oncol 2017; 35: 1005.
61. Freedman RA, Gelman RS, Wefel JS, etal.
Translational breast cancer research consortium
(TBCRC) 022: a phase II trial of neratinib for
patients with human epidermal growth factor
receptor 2-positive breast cancer and brain
metastases. J Clin Oncol 2016; 34: 945–952.
62. Hamilton E, Borges V, Conlin A, etal. Efficacy
results of a phase 1b study of ONT-380, an oral
HER2-specic inhibitor, in combination with
capecitabine (C) and trastuzumab (T) in HER2+
metastatic breast cancer (MBC), including patients
(pts) with brain metastases (mets) [abstract].
San Antonio, TX: San Antonio Breast Cancer
Symposium, 2016.
63. Borges VF, Ferrario C, Aucoin N, etal.
Tucatinib combined with ado-trastuzumab
emtansine in advanced ERBB2/HER2-positive
metastatic breast cancer: a phase 1b clinical trial.
JAMA Oncol 2018; 4: 1214–1220.
64. Metzger O, Barry W, Krop I, etal. Phase I dose-
escalation trial of ONT-380 in combination with
trastuzumab in patients (pts) with HER2+ breast
cancer brain metastases [abstract]. San Antonio,
TX: San Antonio Breast Cancer Symposium,
2016.
65. Ma F, Li Q, Chen S, etal. Phase I study
and biomarker analysis of pyrotinib, a novel
irreversible pan-ErbB receptor tyrosine kinase
inhibitor, in patients with human epidermal
growth factor receptor 2-positive metastatic
breast cancer. J Clin Oncol 2017; 35: 3105–3112.
66. Park YH, Lee KH, Sohn JH, etal. A phase II trial
of the pan-HER inhibitor poziotinib, in patients
with HER2-positive metastatic breast cancer who
had received at least two prior HER2-directed
regimens: results of the NOV120101-203 trial.
Int J Cancer 2018; 143: 3240–3247.
67. Martin M, Holmes FA, Ejlertsen B, etal.
Neratinib after trastuzumab-based adjuvant
therapy in HER2-positive breast cancer
(ExteNET): 5-year analysis of a randomised,
double-blind, placebo-controlled, phase 3 trial.
Lancet Oncol 2017; 18: 1688–1700.
68. Awada A, Colomer R, Inoue K, etal. Neratinib
plus paclitaxel vs trastuzumab plus paclitaxel in
previously untreated metastatic ERBB2-positive
breast cancer: the NEfERT-T randomized
clinical trial. JAMA Oncol 2016; 2: 1557–1564.
69. National Comprehensive Cancer Network.
NCCN Guidelines for Patients: Breast Cancer
Metastatic. Plymouth Meeting, PA: National
Comprehensive Cancer Network, 2018.
70. Xu B, Ma F, Ouyang Q, etal. Abstract PD3-08:
A randomized phase II trial of pyrotinib plus
capecitabine versus lapatinib plus capecitabine
in patients with HER2-positive metastatic
breast cancer previously treated with taxanes,
anthracyclines and/or trastuzumab. Cancer Res
2018; 78: Abstract nr PD3-08.
71. Bianchini G and Gianni L. The immune
system and response to HER2-targeted
treatment in breast cancer. Lancet Oncol 2014;
15: e58–e68.
72. Kim A, Lee SJ, Kim YK, etal. Programmed
death-ligand 1 (PD-L1) expression in tumour cell
and tumour infiltrating lymphocytes of HER2-
positive breast cancer and its prognostic value. Sci
Rep 2017; 7: 11671.
73. Stagg J, Loi S, Divisekera U, etal. Anti-ErbB-2
mAb therapy requires type I and II interferons
and synergizes with anti-PD-1 or anti-CD137
mAb therapy. Proc Natl Acad Sci U S A 2011;
108: 7142–7147.
74. Dirix LY, Takacs I, Jerusalem G, etal. Avelumab,
an anti-PD-L1 antibody, in patients with locally
advanced or metastatic breast cancer: a phase 1b
JAVELIN Solid Tumor study. Breast Cancer Res
Treat 2018; 167: 671–686.
75. Loi S, Giobbie-Hurder A, Gombos A, etal.
Phase Ib/II study evaluating safety and efficacy of
pembrolizumab and trastuzumab in patients with
trastuzumab-resistant HER2-positive metastatic
breast cancer: results from the PANACEA (IBCSG
45-13/BIG 4-13/KEYNOTE-014) study [abstract].
San Antonio, TX: San Antonio Breast Cancer
Symposium, 2017.
76. Emens LA, Esteva F, Beresford M, etal. Results
from KATE2, a randomized phase 2 study of
atezolizumab (atezo)+trastuzumab emtansine
(T-DM1) vs placebo (pbo)+T-DM1 in previously
treated HER2+ advanced breast cancer (BC)
[abstract]. San Antonio, TX: San Antonio Breast
Cancer Symposium, 2018.
77. Mittendorf EA, Clifton GT, Holmes JP, etal.
Final report of the phase I/II clinical trial of
the E75 (nelipepimut-S) vaccine with booster
inoculations to prevent disease recurrence in
high-risk breast cancer patients. Ann Oncol 2014;
25: 1735–1742.
78. Diehl JA, Cheng M, Roussel MF, etal. Glycogen
synthase kinase-3beta regulates cyclin D1
proteolysis and subcellular localization. Genes &
Dev 1998; 12: 3499–3511.
Therapeutic Advances in Medical Oncology 11
16 journals.sagepub.com/home/tam
79. Yu Q, Geng Y and Sicinski P. Specific protection
against breast cancers by cyclin D1 ablation.
Nature 2001; 411: 1017–1021.
80. Choi YJ, Li X, Hydbring P, etal. The
requirement for cyclin D function in tumor
maintenance. Cancer Cell 2012; 22: 438–451.
81. 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.
82. Witkiewicz AK, Cox D and Knudsen ES.
CDK4/6 inhibition provides a potent adjunct
to Her2-targeted therapies in preclinical breast
cancer models. Genes Cancer 2014; 5: 261-72.
83. DeMichele A, Clark AS, Tan KS, etal. CDK
4/6 inhibitor palbociclib (PD0332991) in Rb+
advanced breast cancer: phase II activity, safety,
and predictive biomarker assessment. Clin Cancer
Res 2015; 21: 995–1001.
84. Patnaik A, Rosen LS, Tolaney SM, etal. Efficacy
and safety of abemaciclib, an inhibitor of CDK4
and CDK6, for patients with breast cancer, non-
small cell lung cancer, and other solid tumors.
Cancer Discov 2016; 6: 740–753.
85. Ciruelos E, Villagrasa P, Pare L, etal. PAM50
intrinsic subtype predicts survival outcome in HER2-
positive/hormone receptor-positive metastatic breast
cancer treated with palbociclib and trastuzumab:
a correlative analysis of the PATRICIA (SOLTI
13-03) trial [abstract]. San Antonio, TX: San
Antonio Breast Cancer Symposium, 2017.
86. Ciruelos E, Villagrasa P, Paré L, etal. SOLTI-
1303 PATRICIA phase II trial (STAGE 1) —
Palbociclib and trastuzumab in postmenopausal
patients with HER2-positive metastatic breast cancer
[abstract]. San Antonio, TX: San Antonio Breast
Cancer Symposium, 2018.
87. Bachelot T, Kabos P, Yardley D, etal.
Abemaciclib for the treatment of brain metastases
secondary to hormone receptor positive breast cancer
[abstract]. San Antonio, TX: San Antonio Breast
Cancer Symposium, 2017.
88. Serra V, Scaltriti M, Prudkin L, etal. PI3K
inhibition results in enhanced HER signaling
and acquired ERK dependency in HER2-
overexpressing breast cancer. Oncogene 2011; 30:
2547–2557.
89. Hurvitz SA, Andre F, Jiang Z, etal. Combination
of everolimus with trastuzumab plus paclitaxel
as first-line treatment for patients with HER2-
positive advanced breast cancer (BOLERO-1): a
phase 3, randomised, double-blind, multicentre
trial. Lancet Oncol 2015; 16: 816–829.
90. Andre F, O’Regan R, Ozguroglu M, etal.
Everolimus for women with trastuzumab-
resistant, HER2-positive, advanced breast cancer
(BOLERO-3): a randomised, double-blind,
placebo-controlled phase 3 trial. Lancet Oncol
2014; 15: 580–591.
91. Andre F, Hurvitz S, Fasolo A, etal. Molecular
alterations and everolimus efficacy in
human epidermal growth factor receptor
2-overexpressing metastatic breast cancers:
combined exploratory biomarker analysis from
BOLERO-1 and BOLERO-3. J Clin Oncol 2016;
34: 2115–2124.
92. Shah PD, Chandarlapaty S, Dickler MN,
etal. Phase I study of LJM716, BYL719, and
trastuzumab in patients (pts) with HER2-
amplified (HER2+) metastatic breast cancer
(MBC) [abstract]. J Clin Oncol 2015; 33: 590.
93. Jain S, Shah AN, Santa-Maria CA, etal. Phase
I study of alpelisib (BYL-719) and trastuzumab
emtansine (T-DM1) in HER2-positive metastatic
breast cancer (MBC) after trastuzumab and
taxane therapy. Breast Cancer Res Treat 2018;
171: 371–381.
94. Keegan NM, Walshe JM, Toomey S, etal. A
phase Ib trial of copanlisib and tratuzumab in
pretreated recurrent or metastatic HER2-positive
breast cancer “PantHER” [abstract]. J Clin Oncol
2018; 36: 1036.
Visit SAGE journals online
journals.sagepub.com/
home/tam
SAGE journals
... A common feature of DTBC tumors is that they do not respond well to existing therapies. For example, despite the clinical benefits of HER2-targeted therapies, many HER2 + tumors develop resistance to targeted therapy [20] and will eventually develop progressive disease. LumB BC is defined by aggressive clinical behavior and has a prognosis similar to that of other DTBC [21]. ...
Article
Full-text available
Introduction: Breast cancer (BC) is classified into four widely-accepted intrinsic subtypes based on PAM (Prediction Analysis of Microarray) 50 gene expression profiles: Luminal A (LumA), Luminal B (LumB), Her2-enriched (Her2) and basal-like (Basal). Recent multi-omic studies of human BC have identified many potential therapeutic biomarkers for these subtypes and have also revealed the extensive molecular heterogeneity of the disease. Patients with LumA tumors generally have better outcomes because these tumors are typically slower-growing and responsive to hormone therapy. However, there is still considerable variation in the clinical outcomes of patients with non-LumA tumors. Here, we strive to understand the proteogenomic characteristics of intrinsically-defined non-LumA tumors in reference to LumA tumors. Methods: A total of 117 retrospectively-collected, untreated primary breast tumor specimens, with a focus on non-LumA subtypes, were selected from the Clinical Breast Care Project and consented using a HIPAA-compliant, IRB-approved protocol. The study cohort had a median patient follow-up of 9.3 years, enabling clinical outcome-related analyses. Immunohistochemistry (IHC) subtyping was used to enrich the cohort with non-LumA subtypes. Breast tumors were embedded in OCT (Optimal Cutting Temperature) compound and processed by laser microdissection (LMD) to enrich for tumor cells. DNA, RNA, and protein were simultaneously extracted from each tumor using the illustra triplePrep kit. Paired-end RNA sequencing and whole genome sequencing (WGS) were performed for 117 and 99 tumors, respectively, using the Illumina HiSeq platform. Quantitative global proteomics and phosphoproteomic analyses were performed on 112 and 50 tumors, respectively, using isobaric TMT 6-plex labeling with the “universal reference” strategy. Results: We observed significantly lower stromal, immune and microenvironment scores in non-basal-like tumors, especially the LumA subtype from LMD-processed samples, compared to that of the same tumors but bulk-processed in TCGA-breast cancer study. There was also significantly lower stromal gene expression in LMD LumA compared to TCGA LumA. Unlike a recent report on proteomics clustering of bulk-processed tumors, we did not observe a stromal-enriched cluster, probably because the use of LMD minimized stromal components. Many common patterns of somatic mutations were observed among non-LumA tumors, such as dominant TP53 mutations and 5q deletion, suggesting a potentially common cell-of-origin for non-LumA tumors, such as an ER negative cancer stem cell or progenitor cell for these tumor subtypes. Cell proliferation-associated genes were amplified and proliferation pathways strongly enriched in non-LumA tumors. In addition to this, we identified two distinct phosphoproteomic profiles for relapsed and relapse-free basal-like BC. Moreover, we also identified 17 differentially expressed phosphopeptides that could identify cases of relapsed and relapse-free basal-like BC with a significant difference in progression-free interval of surviving cases. Conclusion: This study provides an integrated proteogenomic characterization of non-LumA vs LumA tumor specimens enriched for cancer cells. We identified many common features of non-LumA tumors and also identified phosphopeptides that could serve as potential biomarkers for less aggressive basal-like BC and possibly guide treatment selections. Disclaimer: The contents of this publication are the sole responsibility of the author(s) and do not necessarily reflect the views, opinions or policies of USUHS, HJF, the DoD or the Departments of the Army, Navy or Air Force or the DOE or PNNL. Mention of trade names, commercial products, or organizations does not imply endorsement by the U.S. Government. Citation Format: Praveen Kumar Raj Kumar, Xiaoying Lin, Tao Liu, Lori Sturtz, Marina Gritsenko, Vladislav Petyuk, Tyler Sagendorf, Brenda Deyarmin, Jianfang Liu, Anupama Praveen-Kumar, Guisong Wang, Jason McDermott, Anil Shukla, Ronald Moore, Matthew Monroe, Bobbie-Jo Webb-Robertson, Jeffrey Hooke, Leigh Fantacone-Campbell, Brad Mostoller, Leonid Kvecher, Jennifer Kane, Jennifer Melley, Stella Somiari, Patrick Soon-Shiong, Richard Smith, Richard Mural, Karin Rodland, Craig Shriver, Albert Kovatich, Hai Hu. Proteogenomic characterization of non-luminal A versus luminal A breast cancer tumor cells enriched by laser microdissection [abstract]. In: Proceedings of the 2023 San Antonio Breast Cancer Symposium; 2023 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2024;84(9 Suppl):Abstract nr PO2-16-04.
... In the case of ovarian and breast cancer, overexpression of the transmembrane glycoprotein HER2 has been associated with malignant transformation as well as carcinogenesis and decreased survival. It can also be found in osteosarcomas, GBM, and medulloblastomas [77,78]. Currently, multiple clinical trials using this target are under development, yet drugs such as lapatinib or trastuzumab have generally yielded unfavorable outcomes [79]. ...
Article
Full-text available
Chimeric antigen receptor T cell (CAR T cell) therapy has emerged as a prominent adoptive cell therapy and a therapeutic approach of great interest in the fight against cancer. This approach has shown notorious efficacy in refractory hematological neoplasm, which has bolstered its exploration in the field of solid cancers. However, successfully managing solid tumors presents considerable intrinsic challenges, which include the necessity of guiding the modified cells toward the tumoral region, assuring their penetration and survival in adverse microenvironments, and addressing the complexity of identifying the specific antigens for each type of cancer. This review focuses on outlining the challenges faced by CAR T cell therapy when used in the treatment of solid tumors, as well as presenting optimizations and emergent approaches directed at improving its efficacy in this particular context. From precise localization to the modulation of the tumoral microenvironment and the adaptation of antigen recognition strategies, diverse pathways will be examined to overcome the current limitations and buttress the therapeutic potential of CAR T cells in the fight against solid tumors.
... The US Food and Drug Administration's (FDA) 1998 approval of the anti-HER2 monoclonal antibody trastuzumab [6], and subsequent approvals of other HER2targeted therapies have redefined the treatment landscape for patients with HER2 + breast cancer, as these therapies offered improved progression-free survival (PFS) and overall survival (OS) compared to chemotherapy alone [7][8][9][10][11][12]. The median survival of patients diagnosed after the introduction of HER2-targeted therapies is about 34 months from metastatic diagnosis [13]. ...
Article
Full-text available
Purpose Standard-of-care for HER2-positive metastatic breast cancer (HER2 + mBC) patients consists of trastuzumab ± pertuzumab with chemotherapy in first-line (1L), and ado-trastuzumab emtansine (T-DM1) or the more recently approved trastuzumab deruxtecan (T-DXd) in second-line (2L). Contemporary data on treatment sequencing and real-world effectiveness is limited. This study aims to report 2L treatments and outcomes among HER2 + mBC patients in the United States (US). Methods HER2 + mBC patients initiating 2L treatment (index date) between January 2014 and February 2021 were identified from the Syapse Learning Health Network (LHN) database. Summary statistics for patient characteristics, treatment received, reasons for 2L discontinuation and time to 2L-clinical outcomes are reported. Results Of the 312 patients initiating 2L treatment, had a median age of 59 years (interquartile range [IQR], 50–66) at the start of 2L. The majority were white (69%) and had de novo mBC (62%). Top three 2L regimens included T-DM1 ± endocrine therapy (29%), trastuzumab/pertuzumab/taxane (10%) and T-DM1/trastuzumab (8%). Around 88% discontinued 2L and 63% received subsequent treatment. Median time-to-next-treatment was 10.6 months (95% CI, 8.8–13.3) and real-world progression-free-survival was 7.9 months (95% CI, 7.0–9.9). Among 274 patients who discontinued 2L, 47% discontinued due to progression and 17% because of intolerance/toxicity, respectively. Conclusion This real-world US study showed that approximately two-thirds of 2L patients received subsequent therapy and disease progression was the most common reason for 2L discontinuation highlighting the need for timely 2L treatment with the most efficacious drug to allow patients to achieve longer treatment duration and delayed progression.
Article
Breast cancer is currently the most commonly occurring cancer globally. Among breast cancer cases, the human epidermal growth factor receptor 2 (HER2)-positive breast cancer accounts for 15% to 20% and is a crucial focus in the treatment of breast cancer. Common HER2-targeted drugs approved for treating early and/or advanced breast cancer include trastuzumab and pertuzumab, which effectively improve patient prognosis. However, despite treatment, most patients with terminal HER2-positive breast cancer ultimately suffer death from the disease due to primary or acquired drug resistance. The prevalence of aberrantly activated the protein kinase B (AKT) signaling in HER2-positive breast cancer was already observed in previous studies. It is well known that p-AKT expression is linked to an unfavorable prognosis, and the phosphatidylinositol-3-kinase (PI3K)/AKT pathway, as the most common mutated pathway in breast cancer, plays a major role in the mechanism of drug resistance. Therefore, in the current review, we summarize the molecular alterations present in HER2-positive breast cancer, elucidate the relationships between HER2 overexpression and alterations in the PI3K/AKT signaling pathway and the pathways of the alterations in breast cancer, and summarize the resistant mechanism of drugs targeting the HER2–AKT pathway, which will provide an adjunctive therapeutic rationale for subsequent resistance to directed therapy in the future.
Chapter
The discoveries of breast cancer molecular subtypes and their deregulated pathways have given rise to the emergence of potential therapeutic targets and treatment options with excellent clinical outcomes. The HER2-amplified breast cancer remains the prototype in which the tyrosine kinase inhibitors are still being used clinically for molecular-targeted breast cancer treatment. Targeted therapy resistance in breast cancer (BC) can result from the proliferation of intrinsic and/or extrinsic instigators leading to relapse after a short remission. Intrinsic mechanisms (acquired before treatment) are a result of genetic mutations that result in a lack of target sensitivity in cancer cells. Extrinsic (acquired) resistance can result from; the emergence of the second proto-oncogene acting as the new driver gene, dysregulated expression of the drug targets as a result of new mutation, and alternations in tumor microenvironment (TME) post-treatment. To overcome the mutations and/or compensatory pathways that impart treatment resistance, often, combinational approaches to treatment are often required. Careful patient selection should be done before the start of targeted therapy to confirm the predictive response beforehand. Newer treatment agents are not without undesired side effects.
Article
Full-text available
Background Breast cancer (BC) is the most commonly diagnosed cancer and the leading cause of cancer death among women globally. Despite advances, there is considerable variation in clinical outcomes for patients with non-luminal A tumors, classified as difficult-to-treat breast cancers (DTBC). This study aims to delineate the proteogenomic landscape of DTBC tumors compared to luminal A (LumA) tumors. Methods We retrospectively collected a total of 117 untreated primary breast tumor specimens, focusing on DTBC subtypes. Breast tumors were processed by laser microdissection (LMD) to enrich tumor cells. DNA, RNA, and protein were simultaneously extracted from each tumor preparation, followed by whole genome sequencing, paired-end RNA sequencing, global proteomics and phosphoproteomics. Differential feature analysis, pathway analysis and survival analysis were performed to better understand DTBC and investigate biomarkers. Results We observed distinct variations in gene mutations, structural variations, and chromosomal alterations between DTBC and LumA breast tumors. DTBC tumors predominantly had more mutations in TP53, PLXNB3, Zinc finger genes, and fewer mutations in SDC2, CDH1, PIK3CA, SVIL, and PTEN. Notably, Cytoband 1q21, which contains numerous cell proliferation-related genes, was significantly amplified in the DTBC tumors. LMD successfully minimized stromal components and increased RNA–protein concordance, as evidenced by stromal score comparisons and proteomic analysis. Distinct DTBC and LumA-enriched clusters were observed by proteomic and phosphoproteomic clustering analysis, some with survival differences. Phosphoproteomics identified two distinct phosphoproteomic profiles for high relapse-risk and low relapse-risk basal-like tumors, involving several genes known to be associated with breast cancer oncogenesis and progression, including KIAA1522, DCK, FOXO3, MYO9B, ARID1A, EPRS, ZC3HAV1, and RBM14. Lastly, an integrated pathway analysis of multi-omics data highlighted a robust enrichment of proliferation pathways in DTBC tumors. Conclusions This study provides an integrated proteogenomic characterization of DTBC vs LumA with tumor cells enriched through laser microdissection. We identified many common features of DTBC tumors and the phosphopeptides that could serve as potential biomarkers for high/low relapse-risk basal-like BC and possibly guide treatment selections.
Article
In this study, we measured the kinase activity profiles of 32 pre-treatment tumor biopsies of HER2-positive breast cancer patients. The aim of this study was to assess the prognostic potential of kinase activity levels, to identify potential mechanisms of resistance and to predict treatment success of HER2-targeted therapy combined with chemotherapy. Indeed, our system-wide kinase activity analysis allowed us to link kinase activity to treatment response. Overall, high kinase activity in the HER2-pathway was associated with good treatment outcome. We found eleven kinases differentially regulated between treatment outcome groups, including well-known players in therapy resistance, such as p38a, ERK, and FAK, and an unreported one, namely MARK1. Lastly, we defined an optimal signature of four kinases in a multiple logistic regression diagnostic test for prediction of treatment outcome (AUC = 0.926). This kinase signature showed high sensitivity and specificity, indicating its potential as predictive biomarker for treatment success of HER2-targeted therapy.
Article
Full-text available
Background: CDK4/6 inhibition combined with anti-HER2 therapy is currently being explored in HER2-positive (HER2+) BC. Here, we report the efficacy, safety and genomic analysis of STAGE 1 of the PATRICIA phase II trial evaluating palbociclib in combination with trastuzumab (TTZ) in advanced HER2+ BC. Methods: PATRICIA is an exploratory, prospective, open-label, multicenter phase II trial. Patients (pts) had received 2-4 prior lines of anti-HER2-based regimens. Treatment consisted of TTZ 6 mg/kg every 3w and palbociclib 200 mg daily for 2w and 1w off. The study was based on a Simon 2-stage design comprising 3 cohorts: estrogen receptor (ER)-negative (cohort A), ER+ (cohort B1) and ER+ with letrozole (cohort B2). Pts ER+ were randomized to cohorts B1 or B2. The trial included a safety run-in phase of the first 12 pts. For part 1 to be successful, at least 6 pts of 15 had to be progression-free at 6 months (PFS6R of 40%) in each cohort. Secondary objectives included safety and the association of the research-based PAM50 intrinsic subtyping with PFS. PAM50 was performed from FFPE samples using the nCounter platform. Multivariable Cox regression analyses evaluating PAM50 subtypes, age, performance status, treatment line, type of biopsy and endocrine treatment were performed. Results: A total of 45 pts were recruited (n=15 in each cohort). Median age was 59.5y, and median number of prior lines was 3.0. The PFS6R in cohorts A, B1 and B2 were 33.3% (5/15), 40.0% (6/15) and 53.3% (8/15), respectively. Regarding safety, grade 1-2 and 3-4 toxicities occurred in 97.7% and 84.4% of pts. The most common grade 3-4 toxicities were neutropenia (80%) and thrombocytopenia (17%). Dose reductions were required in 60% of pts. Regarding PAM50, a total of 40 (83.9%) tumors samples (22 primary and 18 metastasis) were profiled. Subtype distribution was as follows: 92.9% HER2-enriched (HER2-E) and 7.1% Basal-like in cohort A, and 46.2% HER2-E, 23.1% Luminal B, 19.2% Luminal A and 11.5% Normal-like in cohorts B1+2. No significant differences in PFS were observed across the 3 cohorts. In cohorts A+B1+B2, Luminal disease defined by PAM50 showed a higher median PFS compared to non-luminal disease (12.4 vs. 4.1 months; adjusted hazard ratio=0.37; p-value=0.052). Clinical Benefit Rate (CBR6) was 73% in Luminal Vs. 31% in non-luminals (p=0.031). In cohorts B1+B2, Luminal disease defined by PAM50 showed a higher median PFS compared to non-luminal disease (12.4 vs. 4.1 months; adjusted hazard ratio=0.30; p-value=0.025). CBR6 was 73% in Luminal Vs. 25% in non-luminals (p=0.022). No clinical-pathological variable was found associated with PFS or CBR6 in all pts or in cohorts B1+B2. Conclusion: Palbociclib in combination with TTZ is safe and active in TTZ pre-treated HER2+ advanced BC, specially within ER+ disease. Identification of the non-luminal subtypes by PAM50 might help identify pts who might not derive a large benefit from this treatment strategy regardless ER status. Our results might have important implications for current and future clinical trials evaluating CDK4/6 inhibitors in HER2+ disease. In Part 2, a total of 92 pts in cohorts B1 and B2 will be included to better assess the efficacy of this treatment strategy. Citation Format: Ciruelos E, Villagrasa P, Paré L, Oliveira M, Pernas S, Cortés J, Soberino J, Adamo B, Vazquez S, Martínez N, Perelló A, Bermejo B, Martínez E, Garau I, Melé M, Morales S, Galván P, Pascual T, Canes J, Nuciforo P, Gonzalez X, Prat A. SOLTI-1303 PATRICIA phase II trial (STAGE 1) -- Palbociclib and trastuzumab in postmenopausal patients with HER2-positive metastatic breast cancer [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr PD3-03.
Article
Background T-DM1 has imroved PFS and OS in HER2+ advanced BC atients (ts) who reviously rogressed on trastuzumab and a taxane (Verma NEJM 2012). Preclinical data show that atezo (anti–PD-L1) inhibits PD-L1 binding to PD-1 and B7.1 to restore antitumor immunity. PD-L1 is often exressed on tumor-infiltrating immune cells (IC) in BC. KATE2 (NCT02924883) is the first study to assess atezo combined with T-DM1 in reviously treated HER2+ advanced BC. Method Eligible ts had HER2+ advanced BC reviously treated with trastuzumab and a taxane and rogressed on treatment for metastatic disease or within 6 mo of adjuvant theray. Pts were randomized 2:1 to atezo 1200 mg or bo+T-DM1 3.6 mg/kg IV q3w (crossover not ermitted). The rimary endoint was investigator-assessed PFS er RECIST v1.1. Additional endoints included OS, ORR, DoR (secondary), PFS in the PD-L1+ subgrou (exloratory) and safety. Data cutoff: 11 Dec 2017. Result 133 ts were randomized to atezo+T-DM1 and 69 ts to bo+T-DM1. 49% and 46% received rior ertuzumab for metastatic BC; median duration of ertuzumab treatment was 10 and 13 mo, resectively. Median follow-u was 8.5 and 8.4 mo, resectively. Efficacy in the ITT oulation and biomarker subgrous are shown. PFS HR was 0.82 (95% CI: 0.55, 1.23); =0.3332. At data cutoff, 13 OS events (10%) in the atezo+T-DM1 arm and 8 (12%) in the bo+T-DM1 arm had occurred. mDoR was not reached. 44% and 41% of safety-evaluable ts had an AE of Gr ≥3 with atezo+T-DM1 and bo+T-DM1, resectively. The most common was thrombocytoenia (13% and 4%). The incidence of serious AEs (SAEs) was 33% with atezo+T-DM1 and 19% with bo+T-DM1, with the most common being Gr 1-2 yrexia with atezo+T-DM1 (5%) and abdominal ain (3%) and seizure (3%) with bo+T-DM1. AE rates leading to atezo/bo or T-DM1 discontinuation were 25% and 15%, resectively, with atezo+T-DM1 and 15% and 13% with bo+T-DM1. 1 atient in the atezo+T-DM1 arm had a drug-related Gr 5 AE (hemohagocytic syndrome). View this table: • View inline • View ou ITT and Biomarker Subgrous: Efficacy Conclusion Atezo+T-DM1 did not demonstrate a clinically significant PFS benefit vs bo+T-DM1; OS and DoR data are not yet mature. Numerically higher PFS and ORR were seen with atezo+T-DM1 in PD-L1+ ts. T-DM1 safety in both arms was consistent with its known rofile. Although the combination of atezo+T-DM1 showed a numerically higher incidence of SAEs and discontinuation of atezo due to an AE, rates of Gr 3-5 AEs were similar between arms. Additional biomarker data, including gene exression and mutation data, will be resented. Citation Format: Emens LA, Esteva F, Beresford M, Saura C, De Laurentiis M, Kim S-B, Im S-A, Patre M, Wang Y, Mani A, Liu H, de Haas S, Loi S. Results from KATE2, a randomized hase 2 study of atezolizumab (atezo)+trastuzumab emtansine (T-DM1) vs lacebo (bo)+T-DM1 in reviously treated HER2+ advanced breast cancer (BC) [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symosium; 2018 Dec 4-8; San Antonio, TX. Philadelhia (PA): AACR; Cancer Res 2019;79(4 Sul):Abstract nr PD3-01.
Article
Purpose: Evidence-based treatments for metastatic, human epidermal growth factor receptor 2 (HER2)-positive breast cancer to the CNS are limited. We previously reported modest activity of neratinib monotherapy for HER2-positive breast cancer brain metastases. Here we report the results from additional study cohorts. Patients and methods: Patients with measurable, progressive, HER2-positive brain metastases (92% after receiving CNS surgery and/or radiotherapy) received neratinib 240 mg orally once per day plus capecitabine 750 mg/m2 twice per day for 14 days, then 7 days off. Lapatinib-naïve (cohort 3A) and lapatinib-treated (cohort 3B) patients were enrolled. If nine or more of 35 (cohort 3A) or three or more of 25 (cohort 3B) had CNS objective response rates (ORR), the drug combination would be deemed promising. The primary end point was composite CNS ORR in each cohort separately, requiring a reduction of 50% or more in the sum of target CNS lesion volumes without progression of nontarget lesions, new lesions, escalating steroids, progressive neurologic signs or symptoms, or non-CNS progression. Results: Forty-nine patients enrolled in cohorts 3A (n = 37) and 3B (n = 12; cohort closed for slow accrual). In cohort 3A, the composite CNS ORR = 49% (95% CI, 32% to 66%), and the CNS ORR in cohort 3B = 33% (95% CI, 10% to 65%). Median progression-free survival was 5.5 and 3.1 months in cohorts 3A and 3B, respectively; median survival was 13.3 and 15.1 months. Diarrhea was the most common grade 3 toxicity (29% in cohorts 3A and 3B). Neratinib plus capecitabine is active against refractory, HER2-positive breast cancer brain metastases, adding additional evidence that the efficacy of HER2-directed therapy in the brain is enhanced by chemotherapy. For optimal tolerance, efforts to minimize diarrhea are warranted.
Article
Background: ExteNET showed that 1 year of neratinib, an irreversible pan-HER tyrosine kinase inhibitor, significantly improves 2-year invasive disease-free survival after trastuzumab-based adjuvant therapy in women with HER2-positive breast cancer. We report updated efficacy outcomes from a protocol-defined 5-year follow-up sensitivity analysis and long-term toxicity findings. Methods: In this ongoing randomised, double-blind, placebo-controlled, phase 3 trial, eligible women aged 18 years or older (≥20 years in Japan) with stage 1–3c (modified to stage 2–3c in February, 2010) operable breast cancer, who had completed neoadjuvant and adjuvant chemotherapy plus trastuzumab with no evidence of disease recurrence or metastatic disease at study entry. Patients who were eligible patients were randomly assigned (1:1) via permuted blocks stratified according to hormone receptor status (hormone receptor-positive vs hormone receptor-negative), nodal status (0 vs 1–3 vs or ≥4 positive nodes), and trastuzumab adjuvant regimen (given sequentially vs concurrently with chemotherapy), then implemented centrally via an interactive voice and web-response system, to receive 1 year of oral neratinib 240 mg/day or matching placebo. Treatment was given continuously for 1 year, unless disease recurrence or new breast cancer, intolerable adverse events, or consent withdrawal occurred. Patients, investigators, and trial funder were masked to treatment allocation. The predefined endpoint of the 5-year analysis was invasive disease-free survival, analysed by intention to treat. ExteNET is registered with ClinicalTrials.gov, number NCT00878709, and is closed to new participants. Findings: Between July 9, 2009, and Oct 24, 2011, 2840 eligible women with early HER2-positive breast cancer were recruited from community-based and academic institutions in 40 countries and randomly assigned to receive neratinib (n=1420) or placebo (n=1420). After a median follow-up of 5·2 years (IQR 2·1–5·3), patients in the neratinib group had significantly fewer invasive disease-free survival events than those in the placebo group (116 vs 163 events; stratified hazard ratio 0·73, 95% CI 0·57–0·92, p=0·0083). The 5-year invasive disease-free survival was 90·2% (95% CI 88·3–91·8) in the neratinib group and 87·7% (85·7–89·4) in the placebo group. Without diarrhoea prophylaxis, the most common grade 3–4 adverse events in the neratinib group, compared with the placebo group, were diarrhoea (561 [40%] grade 3 and one [<1%] grade 4 with neratinib vs 23 [2%] grade 3 with placebo), vomiting (grade 3: 47 [3%] vs five [<1%]), and nausea (grade 3: 26 [2%] vs two [<1%]). Treatment-emergent serious adverse events occurred in 103 (7%) women in the neratinib group and 85 (6%) women in the placebo group. No evidence of increased risk of long-term toxicity or long-term adverse consequences of neratinib-associated diarrhoea were identified with neratinib compared with placebo. Interpretation: At the 5-year follow-up, 1 year of extended adjuvant therapy with neratinib, administered after chemotherapy and trastuzumab, significantly reduced the proportion of clinically relevant breast cancer relapses—ie, those that might lead to death, such as distant and locoregional relapses outside the preserved breast—without increasing the risk of long-term toxicity. An analysis of overall survival is planned after 248 events.
Article
T cell bispecific antibodies (TCBs) are engineered molecules that include, within a single entity, binding sites to the T cell receptor and to tumor-associated or tumor-specific antigens. The receptor tyrosine kinase HER2 is a tumor-associated antigen in ~25% of breast cancers. TCBs targeting HER2 may result in severe toxicities, likely due to the expression of HER2 in normal epithelia. About 40% of HER2-positive tumors express p95HER2, a carboxyl-terminal fragment of HER2. Using specific antibodies, here, we show that p95HER2 is not expressed in normal tissues. We describe the development of p95HER2-TCB and show that it has a potent antitumor effect on p95HER2-expressing breast primary cancers and brain lesions. In contrast with a TCB targeting HER2, p95HER2-TCB has no effect on nontransformed cells that do not overexpress HER2. These data pave the way for the safe treatment of a subgroup of HER2-positive tumors by targeting a tumor-specific antigen.
Article
1005 Background: Evidence-based treatments (tx) for metastatic, HER2+ BCBM are limited. We previously found a central nervous system (CNS) objective response rate (ORR) of 8% (95% CI 2-22%) for the irreversible, EGFR/HER2-targeted kinase inhibitor, neratinib. To enhance CNS activity, we evaluated the combination of neratinib + capecitabine in a subsequent cohort, and report results here. Methods: Pts with measurable BCBM (≥ 1 cm in longest dimension) and no prior lapatinib or capecitabine were eligible. All but 3 had CNS progression after local CNS tx. During 21 day cycles, pts received capecitabine 750 mg/m2 twice daily x 14 days followed by 7 days off + neratinib 240 mg orally once daily. Loperamide prophylaxis (16 mg total daily) was recommended during cycle 1. Brain MRI and non-CNS imaging were repeated every 2 cycles until 18 wks, then every 3 cycles. The primary endpoint was composite CNS ORR, requiring all of the following: ≥50% reduction in volumetric sum of target CNS lesions (central review, VORR), no progression of non-target or non-CNS lesions, no new lesions, no escalating steroids, and no progressive neurologic signs/symptoms. We used a two-stage design with hypotheses ORR 15% and 35% (error rates 5% and 20%), responses in ≥5/19 pts to enter 2 nd stage; responses in ≥9/35 [26%] pts to be promising. Results: 39 pts enrolled between 4/2014-11/2016 (2 withdrew before tx, 37 analyzed); median age 51, median prior metastatic lines 2 (range 0-6), 65% had prior WBRT. As of 11/15/16, 23 (62%) patients are alive and 7 remain on protocol tx; median number of cycles initiated = 5 (range 1-26). 18 women (49%) had a VORR (95% CI 32-66%, neurologic exams not yet available on all pts). Overall 12-month survival is 63% (95% CI 43%-77%); 4/7 pts still on protocol therapy have not yet reached 6 cycles. No pts had grade 4 toxicity; 18 (49%) had grade 3 toxicity, with diarrhea most common (32%), and 6 pts discontinued tx for toxicity. Conclusions: The combination of neratinib and capecitabine is active for BCBM with VORR in nearly half of pts, supporting further development of the regimen for BCBM. Updated results will be presented at the meeting. Clinical trial information: NCT01494662.
Article
2522 Background: MCLA-128 is a novel IgG1 bispecific antibody with enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) activity targeting HER2 and HER3 receptors. We report final phase 1 single agent escalation data, and safety and preliminary activity at the recommended phase 2 dose (RP2D). Methods: In the phase 1 part, patients (pts) with advanced solid tumors received MCLA-128 every 3 weeks (q3w) IV over 1-2 hr from 40 to 900 mg. In the phase 2 part, pts with selected metastatic indications were treated at the RP2D. Antitumor activity was assessed as per RECIST 1.1. Clinical benefit rate (CBR) was defined as CR + PR + SD ≥12 weeks. Results: As of January 2017, 28 advanced solid tumor pts were treated in the escalation part. No dose limiting toxicities were seen. The RP2D was established as 750 mg q3w (flat dose, corticosteroid premedication) based on safety and PK data. Fifteen pts with HER2 amplified tumors were treated at the RP2D (8 MBC, 4 gastric, 2 ovarian, 1 colorectal). Median age was 52 years (range 33-71), ECOG PS 0/1: 3/12, all ≥2 metastatic sites. The safety profile at the RP2D confirmed dose escalation data; the most common AEs were infusion related reactions in 6 pts (40%; G1-2 in 5 pts, G4 in 1 pt), and G1-2 diarrhea, rash, fatigue in 2 pts each (13%). No congestive heart failure or significant LVEF decreases occurred. The 8 MBC pts had a median 5.5 prior lines of metastatic therapy (range 4-14), all had progressed on 3 prior Her2 inhibitor therapies and received a median of 4.5 MCLA-128 cycles (range 2-12); 1 had a confirmed PR, 5 had SD (including 2 sustained, 11 and 12 cycles). SD was also seen in 2 evaluable MBC pts treated at 480 mg in the phase 1 part (7 and 4 cycles). Overall, CBR in these 10 MBC pts was 70%. Evaluation of other indications is ongoing. Conclusions: MCLA-128 showed a well tolerated safety profile. Consistent antitumor activity was seen in heavily pretreated MBC patients progressing on HER2 therapies. Further exploration of MCLA-128 based combinations with chemotherapy or trastuzumab in less pretreated MBC patients progressing after ≥2 prior Her2 inhibitors including TDM-1 is planned. Clinical trial information: NCT02912949.