ArticlePDF AvailableLiterature Review

Trastuzumab emtansine: Mechanisms of action and drug resistance

Authors:

Abstract and Figures

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that is effective and generally well tolerated when administered as a single agent to treat advanced breast cancer. Efficacy has now been demonstrated in randomized trials as first line, second line, and later than the second line treatment of advanced breast cancer. T-DM1 is currently being evaluated as adjuvant treatment for early breast cancer. It has several mechanisms of action consisting of the anti-tumor effects of trastuzumab and those of DM1, a cytotoxic anti-microtubule agent released within the target cells upon degradation of the human epidermal growth factor receptor-2 (HER2)-T-DM1 complex in lysosomes. The cytotoxic effect of T-DM1 likely varies depending on the intracellular concentration of DM1 accumulated in cancer cells, high intracellular levels resulting in rapid apoptosis, somewhat lower levels in impaired cellular trafficking and mitotic catastrophe, while the lowest levels lead to poor response to T-DM1. Primary resistance of HER2-positive metastatic breast cancer to T-DM1 appears to be relatively infrequent, but most patients treated with T-DM1 develop acquired drug resistance. The mechanisms of resistance are incompletely understood, but mechanisms limiting the binding of trastuzumab to cancer cells may be involved. The cytotoxic effect of T-DM1 may be impaired by inefficient internalization or enhanced recycling of the HER2-T-DM1 complex in cancer cells, or impaired lysosomal degradation of trastuzumab or intracellular trafficking of HER2. The effect of T-DM1 may also be compromised by multidrug resistance proteins that pump DM1 out of cancer cells. In this review we discuss the mechanism of action of T-DM1 and the key clinical results obtained with it, the combinations of T-DM1 with other cytotoxic agents and anti-HER drugs, and the potential resistance mechanisms and the strategies to overcome resistance to T-DM1.
Content may be subject to copyright.
REVIEW
Trastuzumab emtansine: mechanisms of action
and drug resistance
Mark Barok
1*
, Heikki Joensuu
2
and Jorma Isola
3
Abstract
Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that is effective and generally well tolerated when
administered as a single agent to treat advanced breast cancer. Efficacy has now been demonstrated in randomized
trials as first line, second line, and later than the second line treatment of advanced breast cancer. T-DM1 is
currently being evaluated as adjuvant treatment for early breast cancer. It has several mechanisms of action consisting
of the anti-tumor effects of trastuzumab and those of DM1, a cytotoxic anti-microtubule agent released within the
target cells upon degradation of the human epidermal growth factor receptor-2 (HER2)-T-DM1 complex in
lysosomes. The cytotoxic effect of T-DM1 likely varies depending on the intracellular concentration of DM1
accumulated in cancer cells, high intracellular levels resulting in rapid apoptosis, somewhat lower levels in
impaired cellular trafficking and mitotic catastrophe, while the lowest levels lead to poor response to T-DM1.
Primary resistance of HER2-positive metastatic breast cancer to T-DM1 appears to be relatively infrequent, but most
patientstreatedwithT-DM1developacquireddrugresistance. The mechanisms of resistance are incompletely
understood, but mechanisms limiting the binding of trastuzumab to cancer cells may be involved. The cytotoxic
effect of T-DM1 may be impaired by inefficient internalization or enhanced recycling of the HER2-T-DM1 complex
in cancer cells, or impaired lysosomal degradation of trastuzumab or intracellular trafficking of HER2. The effect of
T-DM1 may also be compromised by multidrug resistance proteins that pump DM1 out of cancer cells. In this
review we discuss the mechanism of action of T-DM1 and the key clinical results obtained with it, the combinations of
T-DM1 with other cytotoxic agents and anti-HER drugs, and the potential resistance mechanisms and the strategies to
overcome resistance to T-DM1.
Introduction
Overexpression and amplification of human epidermal
growth factor receptor-2 (HER2, ErbB2) is present in 15
to 20% of primary human breast cancers [1]. In the past,
patients with HER2-positive breast cancer generally had
unfavorable outcome [2], but this changed radically after
discovery of trastuzumab, a recombinant humanized
monoclonal antibody that binds to the extracellular sub-
domain IV of HER2. Trastuzumab showed substantial
anti-tumor efficacy in both preclinical and clinical trials
[3,4], and introduction of trastuzumab for the treatment
of HER2-positive breast cancer can be considered a mile-
stone in medical oncology [4,5]. However, resistance to
trastuzumab eventually emerges in the great majority of
patients treated [6].
Several other HER2-targeted agents have been evaluated
in clinical trials since the introduction of trastuzumab in
1998. Lapatinib, an orally administered small molecule
inhibitor of the HER1 and HER2 tyrosine kinases, was
found to be superior in combination with capecitabine
compared with capecitabine alone in the treatment of
metastatic breast cancer (MBC) that had progressed after
trastuzumab-based therapy [7]. As to trastuzumab, resis-
tance to lapatinib develops frequently among patients who
initially respond [8]. Recently, pertuzumab, a recombinant
humanized monoclonal antibody that binds to subdomain
II of the extracellular portion of HER2 and inhibits recep-
tor dimerization, was found to be more effective in com-
bination with trastuzumab and docetaxel compared with
placebo, trastuzumab and docetaxel as first-line treatment
of HER2-positive MBC [9].
Despite these new therapeutic options, HER2-positive
MBC still remains an incurable disease. In this review we
discuss the mechanisms of action of trastuzumab
* Correspondence: barok.mark@gmail.com
1
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum,
Haartmaninkatu 8, Helsinki FIN-00290, Finland
Full list of author information is available at the end of the article
© 2014 Barok et al.; licensee BioMed Central Ltd. The licensee has exclusive rights to distribute this article, in any medium, for
6 months following its publication. After this time, the article is available under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution , and
reproduction in any medium, provided the original work is properly cited.
Barok et al. Breast Cancer Research 2014, 16:209
http://breast-cancer-research.com/content/16/2/209
emtansine (T-DM1), a novel agent that has challenged in
efficacy and safety all existing systemic therapies for
HER2-positive MBC, and the resistance mechanisms to it.
T-DM1 is an excellent example of a principle suggested
already in the 1970s to use antibodies as carriers of drugs
to highly specific targets [10].
Trastuzumab emtansine, a HER2-targeted
antibody-drug conjugate
Antibody-drug conjugates (ADCs) are a means to deliver
cytotoxic drugs specifically to cancer cells. The delivery is
followed by internalization of the ADC and release of free,
highly active cytotoxic agents within cancer cells, leading
eventually to cell death. The components of an effective
ADC typically consist of: (i) a humanized or human
monoclonal antibody that selectively and specifically
delivers a cytotoxic agent to cancer cells by evoking
receptor-mediated endocytosis; (ii) a cytotoxic agent that
will kill the cell; and (iii) a linker that binds the cytotoxic
agent to the antibody.
The first ADC targeting the HER2 receptor is T-DM1
(ado-trastuzumab emtansine; T-MCC-DM1; Kadcyla®),
which is a conjugate of trastuzumab and a cytotoxic
moiety (DM1, derivative of maytansine). T-DM1 carries
an average of 3.5 DM1 molecules per one molecule of
trastuzumab. Each DM1 molecule is conjugated to
Figure 1 Intracellular trafficking of trastuzumab emtansine (T-DM1). Binding of T-DM1 onto human epidermal growth factor receptor-2
(HER2) on the plasma membrane is followed by entry of the HER2-T-DM1 complex into the cell via receptor-mediated endocytosis. Internalized
endocytic vesicles form early endosomes. The load of early endosomes can be recycled back to the cell membrane or the early endosome can
mature to a lysosome. Release of DM1 occurs as a result of proteolytic degradation of the antibody part of T-DM1 in the lysosomes. Intracellular
lysine (lys)-MCC-DM1 inhibits microtubule assembly, causing mitotic arrest, apoptosis, mitotic catastrophe, and disrupted intracellular trafficking.
MCC, non-reducible thioether linker.
Barok et al. Breast Cancer Research 2014, 16:209 Page 2 of 12
http://breast-cancer-research.com/content/16/2/209
trastuzumab via a non-reducible thioether linker (N-succi-
nimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate;
SMCC, MCC after conjugation) [11].
Mechanisms of action of T-DM1
Binding of T-DM1 to HER2 triggers entry of the HER2-
T-DM1 complex into the cell via receptor-mediated
endocytosis [12,13]. Since the non-reducible linker is
stable in both the circulation and the tumor microenvi-
ronment, active DM1 release occurs only as a result of
proteolytic degradation of the antibody part of T-DM1
in the lysosome [11,14]. Following release from the lyso-
some, DM1-containing metabolites inhibit microtubule
assembly, eventually causing cell death [15] (Figure 1).
Linkage of DM1 to trastuzumab does not affect the
binding affinity of trastuzumab to HER2 [16,17], nor does
it reduce the inherent anti-tumor effects of trastuzumab
[16,18]. Consequently, T-DM1 has mechanisms of action
consisting of the anti-tumor effects related to trastuzumab
and those associated with intracellular DM1 metabolites
(Table 1).
Trastuzumab-mediated effects
Both trastuzumab and T-DM1 inhibit HER2 receptor
signaling, mediate antibody-dependent cell-mediated
cytotoxicity, and inhibit shedding of the extracellular
domain of HER2 [16,18]. Although the anti-tumor
effects of DM1 are more pronounced than those of tras-
tuzumab [16], trastuzumab-mediated effects should not
be underestimated and might be particularly important
when the target cells do not undergo rapid apoptotic
death caused by DM1. This may be common in the
clinic, where trastuzumab therapy of MBC often lasts
for several months or years, and continuation of trastu-
zumab therapy beyond breast cancer progression on
trastuzumab-containing systemic therapy may still be
beneficial [32,33].
DM1-mediated effects
At least four molecular mechanisms have been suggested
for DM1 anti-tumor activity. First, active DM1 metabo-
lites disrupt the microtubule networks of the target cells,
which causes cell cycle arrest at the G
2
-M phase and
apoptotic cell death [11,18]. Second, prolonged treatment
of breast cancer xenografts with T-DM1 caused both
apoptosis and mitotic catastrophe, the latter being identi-
fied as presence of cells with aberrant mitotic figures and
a giant multinucleated structure (Figure 2) [18]. Third,
disruption of microtubule network-mediated intracellular
trafficking may occur. Microtubule targeting agents often
disrupt intracellular trafficking via microtubules [34,35],
and prolonged treatment with T-DM1, but not with tras-
tuzumab, caused defective intracellular trafficking of
HER2 in a preclinical breast cancer model [18]. Impaired
intracellular trafficking may be an important mechanism
of action of T-DM1, particularly in non-dividing cells.
Finally, as we discuss below, free intracellular DM1 may
lead to cell death in a concentration-dependent manner.
Table 1 Mechanisms of action of trastuzumab and trastuzumab emtansine
Mechanism of action Mechanism causing trastuzumab resistance
Trastuzumab
Fab-mediated Down-regulation of HER2 on the plasma membrane [19] Masking of trastuzumab binding epitope of HER2 [20,21]
Inhibition of HER2 ectodomain shedding [22] Expression of p95HER2 [23]
HLA-I-restricted antigen presentation of HER2 [24] Activation of the IGF-IR pathway [25]
Inactivation of the PTEN-PI3K/AKT pathway [26] Defects in the PTEN-PI3K/AKT pathway [26]
Induction of apoptosis [19] Overexpression of cyclin E [27]
Inhibition of angiogenesis [28] Autocrine production of EGF-related ligands [29]
Fc-mediated ADCC [30] Impaired ADCC [31]
T-DM1
Trastuzumab part
Fab-mediated Inhibition of HER2 ectodomain shedding [16]
Inhibition of PI3K/AKT signaling pathway [16]
Fc-mediated ADCC [16,18]
DM1 part Mitotic arrest [11]
Apoptosis [11,17,18]
Mitotic catastrophe [18]
Disruption of intracellular trafficking [18]
ADCC, antibody-dependent cell-mediated cytotoxicity; AKT, protein kinase B; EGF, epidermal growth factor; HER2, human epidermal growth factor receptor-2; HLA,
human leukocyte antigen; IGF-IR, insulin-like growth factor-I receptor; PI3K, phosphatidylinositol 3-kinase; PTEN, phosphatase and tensin homolog; T-DM1,
trastuzumab emtansine.
Barok et al. Breast Cancer Research 2014, 16:209 Page 3 of 12
http://breast-cancer-research.com/content/16/2/209
Activity of T-DM1 in preclinical models and
clinical trials
A comprehensive review of the efficacy and safety results
obtained with T-DM1 is beyond the scope of the current
review but, in brief, T-DM1 has shown substantial anti-
tumor efficacy in preclinical studies and clinical trials.
T-DM1 has superior activity compared with trastuzumab
on trastuzumab-sensitive breast cancer cell cultures and
tumor xenografts (Additional file 1) [11,18]. Importantly,
T-DM1 is effective in in vitro and in vivo models of
trastuzumab-resistant breast cancer, and in trastuzumab
and lapatinib cross-resistant breast cancer models
(Additional file 2) [11,18].
A key clinical trial to investigate the efficacy and safety
of T-DM1 in the treatment of breast cancer was the
EMILIA study, where 991 patients previously treated for
locally advanced or metastatic breast cancer with trastuzu-
mab and a taxane were randomly assigned to receive
either single-agent T-DM1 3.6 mg per kilogram of body
weight intravenously 3-weekly or lapatinib plus capecita-
bine. The median progression-free survival (PFS) was
9.6 months with T-DM1 versus 6.4 months with the con-
trol regimen, and a hazard ratio for progression or death
was 0.65 in favor of T-DM1 (95% CI 0.55 to 0.77). Import-
antly, patients assigned to T-DM1 lived longer (30.9
versus 25.1 months, respectively) and had fewer serious
adverse events recorded. T-DM1 was associated with
higher rates of thrombocytopenia and serum aminotrans-
ferase level elevations, whereas lapatinib and capecitabine
were associated with more frequent diarrhea, nausea and
palmar-plantar erythrodysesthesia [36]. These data led to
approval of T-DM1 by the US Food and Drug Administra-
tion (FDA) in February 2013 for the treatment of patients
with HER2-positive MBC who had previously received
trastuzumab and a taxane.
In another randomized study (TDM4450g), where 137
patients with HER2-positive MBC or recurrent locally
advanced breast cancer were assigned to either T-DM1 or
trastuzumab plus docetaxel as first-line treatment, the
median PFS was 14.2 months with T-DM1 and 9.2 months
with trastuzumab plus docetaxel (hazard ratio 0.59; 95%
CI 0.36 to 0.97) [37]. T-DM1 was associated with a more
favorable safety profile with fewer serious adverse effects.
In the TH3RESA study, 602 patients with unresectable
HER2-positive locally advanced breast cancer or MBC
who had progressed on at least two prior HER2-
directed regimens were randomly assigned to receive
either T-DM1 or therapy chosen by the physician. Patients
treated with T-DM1 achieved longer PFS (6.2 versus
3.3 months, respectively; hazard ratio 0.53, 95% CI 0.42 to
0.66) and longer survival (not reached versus 14.9 months),
and had fewer severe (grade 3 or higher) adverse effects
compared with a regimen chosen by the physician [38].
Resistance to T-DM1
Despite these favorable efficacy results, most patients
treated with T-DM1 eventually progress [36-38], and
some HER2-positive breast cancers are primarily non-
responsive or are only minimally responsive to T-DM1.
Understanding of the resistance mechanisms is important
for further development of T-DM1-directed therapies.
T-DM1 resistance in preclinical models
Both primary and acquired resistance to T-DM1 have
been observed in in vitro models of HER2-positive breast
cancer and gastric cancer (Additional file 3) [17,39,40]. In
Figure 2 Histological findings in a human epidermal growth factor receptor-2-positive, trastuzumab and lapatinib-resistant breast can-
cer (JIMT-1) xenograft following trastuzumab emtansine treatment. Numerous apoptotic cells are present (stained brown with CytoDeath
staining). Hematoxylin counterstain reveals multinucleated giant cells and pathological mitoses (arrows), which are hallmarks of mitotic
catastrophe. Mitotic catastrophes were absent in trastuzumab-treated tumors.
Barok et al. Breast Cancer Research 2014, 16:209 Page 4 of 12
http://breast-cancer-research.com/content/16/2/209
in vivo preclinical models, efficacy of T-DM1 varied
depending on the tumor mass in a trastuzumab- and
lapatinib-resistant human breast cancer xenograft model
(JIMT-1). While large (approximately 350 mm
3
)xeno-
grafts were resistant to T-DM1, small ones (approximately
70 mm
3
) were partially sensitive. T-DM1 inhibited re-
markably well growth of very small JIMT-1 xenografts
with no macroscopic tumor detected until resistance to T-
DM1 emerged after prolonged treatment (16 weeks) with
T-DM1 [18]. In another preclinical study, large HER2-
positive human gastric xenografts (N-87) disappeared
macroscopically totally with T-DM1, but microscopic de-
posits of residual tumor cells remained at the tumor
inoculation sites. The residual cells had a low cell prolifer-
ation rate when stained for Ki-67, and survived T-DM1
treatment despite maintaining high HER2 protein expres-
sion [17]. These findings suggest that cancer relapse may
occur after a long latency period despite macroscopically
complete response to T-DM1.
Primary and acquired resistance to T-DM1 in clinical trials
In a phase II study (TDM4558g) conducted in a cohort of
112 patients with HER2-positive MBC who had received
prior chemotherapy and who had progressed on prior
HER2-directed therapy or within 60 days after the last dose
of trastuzumab, 29 (26%, 95% CI 18% to 34%) patients
achieved objective response with single-agent T-DM1 (none
hadcompleteresponse)and55(49%)hadstabledisease
[41]. In this study only 22 (20%) patients had disease
progression as their best response, suggesting that most
patients with HER2-positive MBC are not primarily resistant
to T-DM1 despite prior exposure to HER2-directed therapy.
Primary resistance to T-DM1 may be more infrequent
when the patients are naive to trastuzumab, although only
indirect data are currently available to support this hy-
pothesis. In the TDM4450g trial carried out in the first-
line setting with most patients not previously treated with
trastuzumab, 43 (64%, 95% CI 52% to 75%) out of the 67
patients with MBC treated with T-DM1 achieved objective
response, including seven (10%) complete responders, and
the median duration of response was not reached [37],
whereas in the EMILIA trial conducted in the second-line
setting in a patient population who had previously been
treated with trastuzumab and a taxane, 169 (44%, 95% CI
39% to 49%) out of the 397 patients treated with T-DM1
had objective response, including four (1%) complete
responders, and the median duration of response was
12.6 months [36].
While primary resistance to T-DM1 may be relatively
infrequent, particularly in patients who have no prior ex-
posure to trastuzumab, most initially responding patients
eventually cease to respond despite continued treatment
with T-DM1 [36-38], suggesting that acquired resistance
to T-DM1 is a common problem.
Potential factors that cause resistance to T-DM1
Except for low HER2 expression in cancer, the clinical,
biological and pharmacological factors that are related to
poor efficacy of T-DM1 are incompletely understood.
Yet, factors that are strongly implicated in the biological
mechanism of action of T-DM1 are good candidates for
having a role in resistance to T-DM1.
DM1 and its metabolites (lysine-MCC-DM1) need to
accumulate in cancer cells to reach a concentration that
exceeds the threshold to evoke cell death [12]. Here we
summarize the factors that may influence the intracellular
DM1 concentration and thus cause resistance to T-DM1
(Figure 3, Table 2).
Low tumor HER2 expression
Expression of HER2 on cancer cells is essential for T-DM1
efficacy. Not surprisingly, retrospective analyses of two
phase II trials (TDM4258g and TDM4374g) carried out in
advanced breast cancer revealed that patients with HER2-
positive cancer (defined either as immunohistochemistry
(IHC) 3+ or fluorescence in situ hybridization +) had more
frequent responses to T-DM1 than patients who had
HER2-normal cancer; in TDM4258g the objective
response rates were 34% and 5%, respectively, and in
TDM4374g, 41% and 20%, respectively [41-43]. When
cancer HER2 mRNA levels were quantified by quantitative
reverse transcriptase polymerase chain reaction in the sub-
group of HER2 IHC 3+ disease, patients with the median
or higher HER2 mRNA concentration responded more
often to T-DM1 than those with a lower concentration
(in TDM4374g, the response rates were 50% and 33%,
and in TDM4258g, 36% and 28%, respectively) [41-43].
Quantitative HER2 assays should probably be performed
from the most recent cancer biopsy tissue material rather
than the primary tumor, since the primary tumor HER2
content may sometimes be discordant with that of most
metastatic lesions [44,45].
Poor internalization of the HER2-T-DM1complexes
Binding of T-DM1 to the extracellular domain of HER2
triggers entry of the HER2-T-DM1 complex into cancer
cells via receptor-mediated endocytosis [12,13]. A high
rate of complex internalization may result in high intracel-
lular concentrations of DM1, and deceleration of the
endocytosis rate might cause loss of sensitivity to T-DM1.
However, it is unknown whether the rate of internalization
differs between cancers, and the factors affecting the rate
have not been identified.
Defective intracellular and endosomal trafficking of the
HER2-T-DM1 complex
The internalized endocytotic vesicles containing HER2-
T-DM1 complexes fuse and form early endosomes. The
contents of early endosomes can be recycled back to the
Barok et al. Breast Cancer Research 2014, 16:209 Page 5 of 12
http://breast-cancer-research.com/content/16/2/209
cell membrane, or the early endosome can mature into a
lysosome [13] where proteolytic degradation of the anti-
body part of T-DM1 occurs (Figure 1). The dynamics of
loading of the lysosomes with the HER2-T-DM1 cargo
may influence the intracellular DM1 levels. T-DM1
treatment results in defective intracellular trafficking of
the HER2 protein [18], which is not in disagreement
with a hypothesis that mitosis is not the only target of
anti-microtubule agents, but rather trafficking on the
microtubules [34].
Defective lysosomal degradation of T-DM1
Since DM1 release in the cytosol occurs only following
proteolytic degradation of the trastuzumab part of the T-
DM1 complex in the lysosomes, efficient lysosomal deg-
radation is essential. Expression and activity of lysosomal
enzymes may vary between tumors and even cancer cells,
and is influenced by several factors such as tumor necrosis
factor-α, lysosomal vacuolar H
+
-ATPase (V-ATPase), and
Bax inhibitor-1 [46-48]. All of these factors may thus affect
cancer sensitivity to T-DM1. For example, V-ATPase
threshold
Mitotic arrest
Rapid apoptosis
Resistance
Mitotic catastrophe
Disrupted intracellular
trafficking
Intracellular concentration of DM1
Availability of T-DM1 to cancer cells
(e.g. pharmacokinetics, tumor vascularization)
Potential resistance mechanisms
to T-DM1
Low HER2 expression in cancer
Masking of the HER2 epitope
High p95HER2 expression
Poor HER2-T-DM1 complex
internalization
Defective intracellular and
endosomal trafficking of the
HER2-T-DM1 complex
Defective lysosomal degradation
of T-DM1
A high rate of HER2-T-DM1
recycling
Drug efflux proteins
-
Cancer cell with HER2
overexpression
+
Figure 3 Factors influencing the intracellular DM1 level. DM1 may evoke cell death in a concentration-dependent manner, where a threshold
concentration of intracellular DM1 and its metabolites needs to be exceeded for cell kill. At high DM1 concentrations mitotic arrest and rapid
apoptotic death follow, whereas at lower levels mitotic catastrophe and disrupted intracellular trafficking occur, and at the lowest levels of DM1
cells show resistance. HER2, human epidermal growth factor receptor-2; T-DM1, trastuzumab emtansine.
Table 2 Potential factors that may cause resistance to trastuzumab emtansine
Factors decreasing intracellular DM1 level
T-DM1 binding to HER2 Low cancer HER2 expression
HER2 down-regulation
Shedding of HER2 ectodomain
Masking of the trastuzumab binding epitope on HER2 p95HER2 expression
Intracellular trafficking and lysosomal degradation Poor HER2-T-DM1complex internalization
HER2-T-DM1 recycling to plasma membrane
Failure of HER2 intracellular trafficking
Inefficient lysosomal degradation of T-DM1
Drug efflux MDR1 expression
Other factors
Altered DM1 target Beta1-tubulin mutation
Autocrine or stromal growth factors Overexpression of a beta3-tubulin isoform
Modulators of the apoptotic pathway Microtubule-associated proteins
Activation of cell survival pathways
HER2, human epidermal growth factor receptor-2; T-DM1, trastuzumab emtansine.
Barok et al. Breast Cancer Research 2014, 16:209 Page 6 of 12
http://breast-cancer-research.com/content/16/2/209
inhibition using archazolid, an inhibitor of myxobacterial
origin, results in apoptosis, growth inhibition, and im-
paired HER2 signaling in the trastuzumab-resistant cell
line JIMT-1 [49].
Masking of the HER2 epitope
The trastuzumab binding epitope of HER2 can be masked
at least partly by MUC4 or hyaluronan inhibiting the
binding of trastuzumab to HER2 [20,21]. Although no
similar data are available regarding T-DM1, masking of
the epitope may also decrease the binding of T-DM1 to
HER2.
High p95HER2 expression
p95HER2 is an amino-terminally truncated form of
HER2 that lacks most of the extracellular domain of the
protein, including subdomain IV recognized by trastuzu-
mab. Therefore, trastuzumab or T-DM1 cannot bind to
p95HER2 [23]. No studies have thus far correlated breast
cancer p95HER2 expression with sensitivity to T-DM1.
A high rate of HER2-T-DM1 recycling
After internalization, trastuzumab-HER2 complexes can
evade degradation and undergo rapid and efficient recyc-
ling to the cell membrane. About 50% of internalized
HER2-bound trastuzumab is recycled back to the cell
membrane within 5 minutes and 85% within 30 minutes
in in vitro breast cancer cell culture [50]. It is currently
unknown whether cytoplasmic recycling of T-DM1 differs
from that of trastuzumab. Extensive recycling of T-DM1
could yet lead to decreased efficacy, since in the absence
of proteolytic degradation of trastuzumab no release of
intracellular DM1 can occur.
Drug efflux pumps
MDR1 (also known as P-glycoprotein) is an ATP-
dependent transporter that mediates efflux of drugs and
toxins from the cell. Tumor MDR1 expression is associ-
ated with poor response to chemotherapy in many types
of cancer [51,52]. DM1 and other maytansinoids are sub-
strates of MDR1, and MDR1 expression is linked with a
maytansine-resistant cancer phenotype [53]. In one study,
one out of three T-DM1-resistant breast cancer cell
lines showed upregulation of multi-drug resistance
transporters [40], but the role of drug efflux proteins in
resistance to T-DM1 may be complex and requires
further study [39].
Neuregulin-HER3 signaling
Presence of the HER3 ligand neuregulin-1β(NRG-1β,
heregulin) suppressed the cytotoxic activity of T-DM1 in
four out of the six breast cancer cell lines tested, this effect
being reversed by pertuzumab [54]. Activating PIK3CA
mutations were present in the two breast cancer cell lines
where NRG-1βdid not inhibit T-DM1 activity, while the
four cell lines where T-DM1 activity was reduced did not
harbor PIK3CA mutations [54]. As trastuzumab, T-DM1
suppresses the phosphatidylinositol 3-kinase (PI3K)
signaling pathway [40]. The potential association between
PIK3CA mutational status and T-DM1 efficacy remains
unknown, but the results from clinical breast cancer series
suggest that trastuzumab benefit does not depend on the
mutational status of PIK3CA [55,56] or tumor PTEN
expression [57].
Altered tubulins
Since DM1 binds to tubulin, altered or mutant tubulins
[58,59] or altered modulators of the microtubule dynamics
might also impact on the response to T-DM1 [39,47].
Concentration-dependent mechanism of action of free
intracellular DM1
A high intracellular concentration of DNA damaging
agents often leads to terminal mitotic arrest and apop-
tosis [60,61]. Besides apoptosis, aberrant cytokinesis
(pathological mitoses) and multinucleation may take
place at low concentrations of DNA damaging agents
[60-62], which is called mitotic catastrophe [60,63].
T-DM1 caused rapid tumor shrinkage of human gastric
cancer xenografts with high HER2 expression (IHC 3+),
the type of cell death being predominantly apoptosis [17],
whereas T-DM1 was less effective on human breast cancer
xenografts expressing moderate HER2 levels (IHC 2+),
but prolonged treatment times eventually evoked apop-
tosis and mitotic catastrophe in these xenografts [18]. T-
DM1 may thus cause cell death through two molecular
mechanisms depending on the intracellular DM1 concen-
tration, high concentrations of DM1 causing mitotic arrest
with no or few mitotic catastrophes followed by apoptosis,
whereas cell exposure to low DM1 concentrations of long
duration may lead to mitotic catastrophes and cell death.
Prolonged T-DM1 treatment led to disruption of intracel-
lular trafficking of HER2 in the breast cancer xenografts
with moderate HER2 expression (IHC 2+) [18].
Based on these findings, we hypothesize that the anti-
cancer effects of T-DM1 depend on the intracellular
concentration of DM1 and the duration of exposure.
When the intracellular concentration of DM1 exceeds a
critical threshold level, mitotic arrest and rapid apoptotic
death follows, whereas mitotic catastrophe and disrupted
intracellular trafficking occur at lower DM1 levels pro-
vided that the exposure time is long enough (Figure 3).
This hypothesis requires further research in preclinical
models, but it could support carrying out clinical trials
evaluating prolonged administration of T-DM1 in cancer
patient populations with low to moderate tumor HER2
expression levels.
Barok et al. Breast Cancer Research 2014, 16:209 Page 7 of 12
http://breast-cancer-research.com/content/16/2/209
Strategies to improve T-DM1 efficacy and
circumvent resistance
Here we summarize the potential strategies to improve
efficacy of T-DM1 and to prevent drug resistance. Some
of these strategies are already being tested in clinical trials.
T-DM1 in the adjuvant and neoadjuvant setting
At present T-DM1 has been approved by the FDA for
second-line treatment of HER2-positive MBC. Ongoing
clinical trials are evaluating the potential role of T-DM1
as first-line treatment of MBC and in the adjuvant and
neoadjuvant settings [64]. The trials to be carried out in
patient populations with a small or minimal tumor bulk
are clearly of great importance, since T-DM1 has substan-
tial efficacy and a favorable safety profile as a single agent
in advanced breast cancer, and T-DM1 may be particularly
effective in eradication of cancer when the tumor mass is
small [65].
Combination therapies with T-DM1
There is substantial interest in investigating the efficacy and
safety of T-DM1 in combination with other anti-cancer
agents, particularly with those that have proved effective in
combination with trastuzumab. Both paclitaxel and doce-
taxel are approved for the treatment of HER2-positive
MBC in combination with trastuzumab [4,66]. Since DM1
and taxanes bind to tubulins at different sites [12,67], a
combination of taxanes and T-DM1 could have synergistic
effects. Two ongoing clinical trials are evaluating such
combinations (NCT00951665 and NCT00934856).
An ongoing clinical trial (NCT01702558) evaluates effi-
cacy and safety of capecitabine plus T-DM1 in MBC. This
trial is built on the clinical activity observed in a phase II
single cohort study that evaluated the combination of cap-
ecitabine and trastuzumab in HER2-positive MBC [68],
and a randomized phase II trial that compared a combin-
ation of capecitabine, trastuzumab and docetaxel to tras-
tuzumab plus docetaxel, the triple combination resulting
in significantly improved PFS [69].
Patients with HER2-positive MBC treated with pertuzu-
mab in combination with trastuzumab and docetaxel had
longer PFS and overall survival compared with patients
who received placebo, trastuzumab and docetaxel in a
large randomized trial (CLEOPATRA) [70]. The on-
going trials evaluating the combinations of pertuzumab
plus T-DM1 and the triple combination of pertuzumab
plus T-DM1 plus a taxane are thus well founded [64].
MARIANNE (NCT01120184) is an ongoing trial with a
planned target population of over 1,000 patients with
HER2-positive MBC. In this study, patients who have
not received prior chemotherapy for MBC are randomly
assigned to receive T-DM1 plus placebo, T-DM1 plus
pertuzumab, or trastuzumab plus paclitaxel or doce-
taxel. The combination of T-DM1 and lapatinib also
deserves clinical evaluation considering the superior
efficacy of lapatinib and trastuzumab in HER2-positive
MBC over lapatinib alone [71].
Trastuzumab has been approved for the treatment of
patients with HER2-positive and hormone receptor-
positive postmenopausal MBC in combination with an
aromatase inhibitor [72,73]. The efficacy and safety of T-
DM1 is being investigated in combination with endocrine
therapy (with tamoxifen in premenopausal women and
aromatase inhibitor in postmenopausal women) as neoad-
juvant treatment of HER2-positive and hormone receptor-
positive operable breast cancer (NCT01745965).
GDC-0941, a selective and potent PI3K inhibitor, was
effective in preclinical models of trastuzumab-resistant
breast cancer, where administration of GDC-0941 in
combination with HER2-directed treatment (trastuzumab,
pertuzumab, or lapatinib) inhibited in a synergistic fashion
growth of breast cancer cells [74,75]. In an ongoing dose
escalation phase Ib study (NCT00928330), the safety,
tolerability, pharmacokinetics, and efficacy of T-DM1 and
GDC-0941 are being investigated in patients with HER2-
positive MBC who have progressed on prior trastuzumab
therapy.
Circumventing MDR1-mediated resistance by a modified
linker
Since the maytansinoids are substrates for the MDR1
transporters [53], drug efflux by MDR1 may decrease
the intracellular DM1 concentration, resulting in a
decline in efficacy. Kovtun and colleagues [53] developed
a potential strategy to circumvent MDR1-mediated re-
sistance to T-DM1 by attaching DM1 to an antibody
using a hydrophilic linker, PEG
4
Mal. The degradation of
such conjugates in cancer cells resulted in the release of
lysine-PEG
4
Mal-DM1 instead of lysine-MCC-DM1 (the
active metabolite of T-DM1). Lysine-PEG
4
Mal-DM1 is a
poor substrate of MDR1, and the conjugates with the
PEG
4
Mal linker evaded MDR1-mediated resistance both
in MDR1-expressing cells in vitro and in MDR1-
expressing xenografts in vivo [53]. Therefore, MDR1
drug transporter-mediated resistance to T-DM1 might
be overcome by replacing the SMCC linker with the
PEG
4
Mal linker.
Modulation of HER2 recycling
When intracellular HER2 is recycled to the plasma mem-
brane, trastuzumab recycles as a part of the HER2-
trastuzumab complex [50]. Heat shock protein (Hsp)90 is
a molecular chaperone that participates in the regulation
of HER2 recycling. Geldanamycin, an inhibitor of Hsp90,
reduces HER2 recycling and results in an over three-fold
increase in the concentration of the HER2-trastuzumab
complex being retained in tumor cells [50]. Geldanamycin
redistributes cell surface HER2 into the internal vesicles of
Barok et al. Breast Cancer Research 2014, 16:209 Page 8 of 12
http://breast-cancer-research.com/content/16/2/209
the endosomes, enhancing proteolytic degradation of
HER2 [50,76].
It has currently not been established whether intracellular
T-DM1 is also recycled, but inhibition of recycling is of
potential interest from the therapeutic point of view. Hypo-
thetically, sequential administration of T-DM1 followed by
geldanamycin (or one of its derivatives [77]) might inhibit
recycling of T-DM1 and direct the HER2-T-DM1
complexes for lysosomal degradation, thus increasing the
intracellular levels of DM1 and cytotoxicity. Sequential
administration of the two drugs in this order could be
important, since Hsp90 inhibitors decrease cell surface
HER2 and might reduce T-DM1 binding to cells [50].
Modification of the cytotoxic drug moiety
Since the intracellular DM1 concentration is crucial for
the cell-killing potency of T-DM1, delivery of greater
quantities of DM1 into the tumor cells would likely im-
prove efficacy. This could be achieved using more heavily
loaded T-DM1 that delivers more cytotoxic drug to the
target cells. However, increasing the drug-antibody ratio
(DAR) of an ADC usually results in a faster clearance of
the ADC. ADCs with a DAR of 2 to 4 have a more favor-
able pharmacokinetic profile than those with a higher
DAR [78,79]. Therefore, increasing the number of DM1
molecules from the average of 3.5 per one trastuzumab
might result in a shorter half-life and destabilization of the
complex, and decreased efficacy. Alternative strategies in-
clude binding of another cytotoxic drug in addition to
DM1 to trastuzumab, or administering another ADC in
combination with T-DM1, such as a cytotoxic drug linked
to pertuzumab. The second cytotoxic drug could have an
alternative (non-tubulin) mechanism of action [39].
Improving the Fc part of trastuzumab
Trastuzumab retains its anti-cancer activity when conju-
gated to DM1, and improving the antibody component of
the conjugate could thus result in a more efficient ADC.
Afucosylated trastuzumab has superior efficacy compared
with trastuzumab in some preclinical breast cancer
models [80], and amino acid modifications of the Fc part
of trastuzumab may also improve efficacy [81]. Yet, DM1
remains a key component regarding the overall anti-
tumor activity of T-DM1.
Radioimmunotherapy conjugates
Auger electron emitting
111
In-NLS-trastuzumab is effective
in the treatment of trastuzumab-resistant breast cancer cells
[82]. Radioimmunotherapy conjugatesmightfindarolein
the treatment of patients who have failed T-DM1 therapy.
Conclusion
T-DM1 is a valuable new agent for the treatment of
HER2-positive breast cancer. T-DM1 has several
mechanisms of action consisting of the anti-tumor effects
associated with its key components, trastuzumab and the
cytotoxic drug DM1. Clinical research carried out suggests
superior efficacy of T-DM1 compared with trastuzumab
or trastuzumab plus chemotherapy in the treatment of
MBC. However, both primary and secondary resistance to
T-DM1 occurs. In addition to the identified resistance
mechanisms related to trastuzumab, several factors that
influence the intracellular DM1 concentration may confer
resistance to T-DM1. Understanding of these factors may
lead to the development of strategies that improve efficacy
of T-DM1 and may circumvent drug resistance.
Additional files
Additional file 1: Time-lapse microscopy of SKBR-3 breast cancer
cells grown with 1 μg/mL trastuzumab or T-DM1. SKBR-3 is a HER2-
positive, trastuzumab-sensitive breast cancer cell line. SKBR-3 is much
more sensitive to T-DM1 than to trastuzumab. Images were taken by
Cell-IQ
W
system (Chip-Man Technologies Ltd, Tampere, Finland).
Additional file 2: Time-lapse microscopy of JIMT-1 breast cancer
cells grown with 1 μg/mL trastuzumab or T-DM1. JIMT-1 is a HER2-
positive, trastuzumab and lapatinib cross-resistant breast cancer cell line.
JIMT-1 is sensitive to T-DM1. Images were taken by Cell-IQ
W
system
(Chip-Man Technologies Ltd, Tampere, Finland).
Additional file 3: Time-lapse microscopy of SNU-216 gastric cancer
cells grown with 1 μg/mL trastuzumab or T-DM1. SNU-216 is a HER2-
positive, trastuzumab-resistant gastric cancer cell line. SNU-216 is resistant
to trastuzumab and T-DM1. Images were taken by Cell-IQ
W
system (Chip-
Man Technologies Ltd, Tampere, Finland).
Abbreviations
ADC: Antibody-drug conjugate; DAR: Drug-antibody ratio; DM1: Derivative of
maytansine 1; FDA: Food and drug administration; HER2: Human epidermal
growth factor receptor-2; Hsp: Heat shock protein;
IHC: Immunohistochemistry; MBC: Metastatic breast cancer; PFS: Progression-
free survival; PI3K: Phosphatidylinositol 3-kinase; SMCC: N-succinimidyl-4-
(N-maleimidomethyl)cyclohexane-1-carboxylate; T-DM1: Trastuzumab-emtansine;
V-ATPase: Vacuolar H
+
-ATPase.
Competing interests
The authors declare that they have no competing interests.
Author details
1
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum,
Haartmaninkatu 8, Helsinki FIN-00290, Finland.
2
Department of Oncology,
Helsinki University Central Hospital Haartmaninkatu 4, Helsinki FIN-00029,
Finland.
3
MioMediTech, University of Tampere, Biokatu 6, Tampere 33014,
Finland.
Published: 5 March 2014
Note: This article is part of a series on Recent advances in
breast cancer treatment,edited by Jenny Chang. Other
articles in this series can be found at http://breast-cancer-
research.com/series/treatment.
Barok et al. Breast Cancer Research 2014, 16:209 Page 9 of 12
http://breast-cancer-research.com/content/16/2/209
References
1. Pauletti G, Dandekar S, Rong H, Ramos L, Peng H, Seshadri R, Slamon DJ:
Assessment of methods for tissue-based detection of the HER-2/neu alter-
ation in human breast cancer: a direct comparison of fluorescence in situ
hybridization and immunohistochemistry. JClinOncol2000, 18:36513664.
2. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL: Human
breast cancer: correlation of relapse and survival with amplification of
the HER-2/neu oncogene. Science 1987, 235:177182.
3. Sliwkowski MX, Lofgren JA, Lewis GD, Hotaling TE, Fendly BM, Fox JA:
Nonclinical studies addressing the mechanism of action of trastuzumab
(Herceptin). Semin Oncol 1999, 26:6070.
4. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Flem-
ing T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L: Use of chemo-
therapy plus a monoclonal antibody against HER2 for metastatic breast
cancer that overexpresses HER2. N Engl J Med 2001, 344:783792.
5. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M,
Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Bar-
rios CH, Steger G, Huang CS, Andersson M, Inbar M, Lichinitser M, Lang I,
Nitz U, Iwata H, Thomssen C, Lohrisch C, Suter TM, Ruschoff J, Suto T, Great-
orex V, Ward C, Straehle C, McFadden E, Dolci MS, Gelber R, et al:Trastuzu-
mab after adjuvant chemotherapy in HER2-positive breast cancer.
N Engl J Med 2005, 353:16591672.
6. Nahta R, Yu D, Hung MC, Hortobagyi GN, Esteva FJ: Mechanisms of
disease: understanding resistance to HER2-targeted therapy in human
breast cancer. Nat Clin Pract Oncol 2006, 3:269280.
7. Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-
Gruszfeld A, Crown J, Chan A, Kaufman B, Skarlos D, Campone M, Davidson
N, Berger M, Oliva C, Rubin SD, Stein S, Cameron D: Lapatinib plus capecit-
abine for HER2-positive advanced breast cancer. N Engl J Med 2006,
355:27332743.
8. Nahta R, Shabaya S, Ozbay T, Rowe DL: Personalizing HER2-targeted therapy
in metastatic breast cancer beyond HER2 status: what we have learned
from clinical specimens. Curr Pharmacogenomics Person Med 2009, 7:263274.
9. Baselga J, Cortes J, Kim SB, Im SA, Hegg R, Im YH, Roman L, Pedrini JL,
Pienkowski T, Knott A, Clark E, Benyunes MC, Ross G, Swain SM:
Pertuzumab plus trastuzumab plus docetaxel for metastatic breast
cancer. N Engl J Med 2012, 366:109119.
10. ONeill GJ: The use of antibodies as drug carriers.InDrug Carriers in
Biology and Medicine. Edited by Gregoriadis G. London: Academic Press;
1979:2343.
11. Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E, Blattler
WA, Lambert JM, Chari RV, Lutz RJ, Wong WL, Jacobson FS, Koeppen H,
Schwall RH, Kenkare-Mitra SR, Spencer SD, Sliwkowski MX: Targeting HER2-
positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic
drug conjugate. Cancer Res 2008, 68:92809290.
12. Kovtun YV, Goldmacher VS: Cell killing by antibody-drug conjugates.
Cancer Lett 2007, 255:232240.
13. Ritchie M, Tchistiakova L, Scott N: Implications of receptor-mediated endo-
cytosis and intracellular trafficking dynamics in the development of anti-
body drug conjugates. MAbs 2013, 5:1321.
14. Erickson HK, Park PU, Widdison WC, Kovtun YV, Garrett LM, Hoffman K, Lutz
RJ, Goldmacher VS, Blattler WA: Antibody-maytansinoid conjugates are
activated in targeted cancer cells by lysosomal degradation and linker-
dependent intracellular processing. Cancer Res 2006, 66:44264433.
15. Chari RV: Targeted cancer therapy: conferring specificity to cytotoxic
drugs. Acc Chem Res 2008, 41:98107.
16. Junttila TT, Li G, Parsons K, Phillips GL, Sliwkowski MX: Trastuzumab-DM1
(T-DM1) retains all the mechanisms of action of trastuzumab and
efficiently inhibits growth of lapatinib insensitive breast cancer. Breast
Cancer Res Treat 2010, 128:347356.
17. Barok M, Tanner M, Koninki K, Isola J: Trastuzumab-DM1 is highly effective
in preclinical models of HER2-positive gastric cancer. Cancer Lett 2011,
306:171179.
18. Barok M, Tanner M, Koninki K, Isola J: Trastuzumab-DM1 causes tumour
growth inhibition by mitotic catastrophe in trastuzumab-resistant breast
cancer cells in vivo. Breast Cancer Res 2011, 13:R46.
19. Cuello M, Ettenberg SA, Clark AS, Keane MM, Posner RH, Nau MM, Dennis
PA, Lipkowitz S: Down-regulation of the erbB-2 receptor by trastuzumab
(herceptin) enhances tumor necrosis factor-related apoptosis-inducing
ligand-mediated apoptosis in breast and ovarian cancer cell lines that
overexpress erbB-2. Cancer Res 2001, 61:48924900.
20. Nagy P, Friedlander E, Tanner M, Kapanen AI, Carraway KL, Isola J, Jovin TM:
Decreased accessibility and lack of activation of ErbB2 in JIMT-1, a
herceptin-resistant, MUC4-expressing breast cancer cell line. Cancer Res
2005, 65:473482.
21. Palyi-Krekk Z, Barok M, Isola J, Tammi M, Szollosi J, Nagy P: Hyaluronan-
induced masking of ErbB2 and CD44-enhanced trastuzumab internalisation
in trastuzumab resistant breast cancer. Eur J Cancer 2007, 43:24232433.
22. Molina MA, Codony-Servat J, Albanell J, Rojo F, Arribas J, Baselga J: Trastu-
zumab (herceptin), a humanized anti-Her2 receptor monoclonal anti-
body, inhibits basal and activated Her2 ectodomain cleavage in breast
cancer cells. Cancer Res 2001, 61:47444749.
23. Arribas J, Baselga J, Pedersen K, Parra-Palau JL: p95HER2 and breast cancer.
Cancer Res 2011, 71:15151519.
24. Kono K, Sato E, Naganuma H, Takahashi A, Mimura K, Nukui H, Fujii H:
Trastuzumab (Herceptin) enhances class I-restricted antigen presentation
recognized by HER-2/neu-specific T cytotoxic lymphocytes. Clin Cancer
Res 2004, 10:25382544.
25. Lu Y, Zi X, Zhao Y, Mascarenhas D, Pollak M: Insulin-like growth factor-I
receptor signaling and resistance to trastuzumab (Herceptin).
J Natl Cancer Inst 2001, 93:18521857.
26. Nagata Y, Lan KH, Zhou X, Tan M, Esteva FJ, Sahin AA, Klos KS, Li P, Monia
BP, Nguyen NT, Hortobagyi GN, Hung MC, Yu D: PTEN activation
contributes to tumor inhibition by trastuzumab, and loss of PTEN
predicts trastuzumab resistance in patients. Cancer Cell 2004, 6:117127.
27. Scaltriti M, Eichhorn PJ, Cortes J, Prudkin L, Aura C, Jimenez J, Chandarlapaty S,
Serra V, Prat A, Ibrahim YH, Guzman M, Gili M, Rodriguez O, Rodriguez S, Perez
J, Green SR, Mai S, Rosen N, Hudis C, Baselga J: Cyclin E amplification/
overexpression is a mechanism of trastuzumab resistance in HER2+ breast
cancer patients. Proc Natl Acad Sci U S A 2011, 108:37613766.
28. Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK: Tumour biology:
herceptin acts as an anti-angiogenic cocktail. Nature 2002, 416:279280.
29. Motoyama AB, Hynes NE, Lane HA: The efficacy of ErbB receptor-targeted
anticancer therapeutics is influenced by the availability of epidermal
growth factor-related peptides. Cancer Res 2002, 62:31513158.
30. Barok M, Isola J, Palyi-Krekk Z, Nagy P, Juhasz I, Vereb G, Kauraniemi P, Kapa-
nen A, Tanner M, Szollosi J: Trastuzumab causes antibody-dependent cel-
lular cytotoxicity-mediated growth inhibition of submacroscopic JIMT-1
breast cancer xenografts despite intrinsic drug resistance. Mol Cancer
Ther 2007, 6:20652072.
31. Kono K, Takahashi A, Ichihara F, Sugai H, Fujii H, Matsumoto Y: Impaired
antibody-dependent cellular cytotoxicity mediated by herceptin in pa-
tients with gastric cancer. Cancer Res 2002, 62:58135817.
32. von Minckwitz G, du Bois A, Schmidt M, Maass N, Cufer T, de Jongh FE,
Maartense E, Zielinski C, Kaufmann M, Bauer W, Baumann KH, Clemens MR,
Duerr R, Uleer C, Andersson M, Stein RC, Nekljudova V, Loibl S:
Trastuzumab beyond progression in human epidermal growth factor
receptor 2-positive advanced breast cancer: a german breast group 26/
breast international group 0305 study. J Clin Oncol 2009, 27:19992006.
33. Blackwell KL, Burstein HJ, Storniolo AM, Rugo H, Sledge G, Koehler M, Ellis C,
Casey M, Vukelja S, Bischoff J, Baselga J, OShaughnessy J: Randomized
study of lapatinib alone or in combination with trastuzumab in women
with ErbB2-positive, trastuzumab-refractory metastatic breast cancer.
J Clin Oncol 2010, 28:11241130.
34. Komlodi-Pasztor E, Sackett D, Wilkerson J, Fojo T: Mitosis is not a key target of
microtubule agents in patient tumors. Nat Rev Clin Oncol 2011, 8:244250.
35. Komlodi-Pasztor E, Sackett DL, Fojo AT: Inhibitors targeting mitosis: tales
of how great drugs against a promising target were brought down by a
flawed rationale. Clin Cancer Res 2012, 18:5163.
36. Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh DY,
Dieras V, Guardino E, Fang L, Lu MW, Olsen S, Blackwell K: Trastuzumab
emtansine for HER2-positive advanced breast cancer. N Engl J Med 2012,
367:17831791.
37. Hurvitz SA, Dirix L, Kocsis J, Bianchi GV, Lu J, Vinholes J, Guardino E, Song C,
Tong B, Ng V, Chu YW, Perez EA: Phase II randomized study of
trastuzumab emtansine versus trastuzumab plus docetaxel in patients
with human epidermal growth factor receptor 2-positive metastatic
breast cancer. J Clin Oncol 2013, 31:11571163.
38. Wildiers H, Kim SB, Gonzalez-Martin A, LoRusso PM, Ferrero JM, Smitt M, Yu
R, Leung A, Krop IE: T-DM1 for HER2-positive metastatic breast cancer:
primary results of TH3RESA, a phase 3 study of T-DM1 vs treatment of
physicians choice [abstract LBA15]. Eur J Cancer 2013:S7S8.
Barok et al. Breast Cancer Research 2014, 16:209 Page 10 of 12
http://breast-cancer-research.com/content/16/2/209
39. Tan X, Jin G, Myers J, Diesl V, Follettie M, Lam M-H, Musto S, Khandke K,
Charati M, Graziani E, Maderna A, Subramanyam C, Koehn F, Dushin R, Arndt
K, ODonnell CJ, Gerber HP, Loganzo F: Tumor cells selected for resistance
to an antibody-drug conjugate retain sensitivity to ADCs with modified
linkers and payloads [abstract]. Cancer Res 2013, 73:Abstract 4629.
40. Li G, Fields CT, Parsons KL, Guo J, Lewis Phillips GD: Trastuzumab-DM1:
mechanisms of action and mechanisms of resistance [abstract].
Eur J Cancer Supplements 2010, 8:73.
41. Burris HA 3rd, Rugo HS, Vukelja SJ, Vogel CL, Borson RA, Limentani S, Tan-
Chiu E, Krop IE, Michaelson RA, Girish S, Amler L, Zheng M, Chu YW, Klencke
B, OShaughnessy JA: Phase II study of the antibody drug conjugate
trastuzumab-DM1 for the treatment of human epidermal growth factor
receptor 2 (HER2)-positive breast cancer after prior HER2-directed ther-
apy. J Clin Oncol 2011, 29:398405.
42. LoRusso P, Krop IE, Burris HA III, Vukelja SJ, Miller K, Zheng M, Chu Y, Lu M, Amler
LC, Rugo HS: Quantitative assessment of diagnostic markers and correlations
with efficacy in two phase II studies of trastuzumab-DM1 (T-DM1) for
patients (pts) with metastatic breast cancer (MBC) who had progressed on
prior HER2-directed therapy [abstract 1016]. JClinOncol2010, 28:15 s.
43. LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX: Trastuzumab
emtansine: a unique antibody-drug conjugate in development for hu-
man epidermal growth factor receptor 2-positive cancer. Clin Cancer Res
2011, 17:64376447.
44. Lower EE, Glass E, Blau R, Harman S: HER-2/neu expression in primary and
metastatic breast cancer. Breast Cancer Res Treat 2009, 113:301306.
45. Niikura N, Liu J, Hayashi N, Mittendorf EA, Gong Y, Palla SL, Tokuda Y,
Gonzalez-Angulo AM, Hortobagyi GN, Ueno NT: Loss of human epidermal
growth factor receptor 2 (HER2) expression in metastatic sites of HER2-
overexpressing primary breast tumors. J Clin Oncol 2012, 30:593599.
46. Lee GH, Kim DS, Kim HT, Lee JW, Chung CH, Ahn T, Lim JM, Kim IK, Chae
HJ, Kim HR: Enhanced lysosomal activity is involved in Bax inhibitor-1-
induced regulation of the endoplasmic reticulum (ER) stress response
and cell death against ER stress: involvement of vacuolar H + ATPase
(V-ATPase). J Biol Chem 2011, 286:2474324753.
47. Dornan D, Settleman J: Predictive biomarkers for antibody-drug conju-
gates.InAntibody-Drug Conjugates and Immunotoxins, From Pre-Clinical De-
velopment to Therapeutic Application. Edited by Lewis Phillips GD. New York:
Humana Press; 2013:7792.
48. Liddil JD, Dorr RT, Scuderi P: Association of lysosomal activity with
sensitivity and resistance to tumor necrosis factor in murine L929 cells.
Cancer Res 1989, 49:27222728.
49. Von Schwarzenberg K, Lajtos T, Simon L, Muller R, Vereb G, Vollmar AM: V-
ATPase inhibition overcomes trastuzumab resistance in breast cancer.
Mol Oncol 2013:. in press.
50. Austin CD, De Maziere AM, Pisacane PI, van Dijk SM, Eigenbrot C, Sliwkowski
MX, Klumperman J, Scheller RH: Endocytosis and sorting of ErbB2 and the
site of action of cancer therapeutics trastuzumab and geldanamycin. Mol
Biol Cell 2004, 15:52685282.
51. Leonard GD, Fojo T, Bates SE: The role of ABC transporters in clinical
practice. Oncologist 2003, 8:411424.
52. Takara K, Sakaeda T, Okumura K: An update on overcoming MDR1-
mediated multidrug resistance in cancer chemotherapy. Curr Pharm Des
2006, 12:273286.
53. Kovtun YV, Audette CA, Mayo MF, Jones GE, Doherty H, Maloney EK,
Erickson HK, Sun X, Wilhelm S, Ab O, Lai KC, Widdison WC, Kellogg B,
Johnson H, Pinkas J, Lutz RJ, Singh R, Goldmacher VS, Chari RV: Antibody-
maytansinoid conjugates designed to bypass multidrug resistance.
Cancer Res 2010, 70:25282537.
54. Lewis Phillips GD, Fields CT, Li G, Dowbenko D, Schaefer G, Miller K, Andre
F, Burris HA III, Albain KS, Harbeck N, Dieras V, Crivellari D, Fang L, Guardino
E, Olsen SR, Crocker LM, Sliwkowski MX: Dual targeting of HER2-positive
cancer with trastuzumab emtansine and pertuzumab: critical role for
neuregulin blockade in antitumor response to combination therapy.
Clin Cancer Res 2014, 20:456468.
55. Barbareschi M, Cuorvo LV, Girlando S, Bragantini E, Eccher C, Leonardi E,
Ferro A, Caldara A, Triolo R, Cantaloni C, Decarli N, Galligioni E, Dalla Palma
P: PI3KCA mutations and/or PTEN loss in Her2-positive breast carcinomas
treated with trastuzumab are not related to resistance to anti-Her2 ther-
apy. Virchows Arch 2012, 461:129139.
56. Loi S, Michiels S, Lambrechts D, Fumagalli D, Claes B, Kellokumpu-Lehtinen
PL, Bono P, Kataja V, Piccart MJ, Joensuu H, Sotiriou C: Somatic mutation
profiling and associations with prognosis and trastuzumab benefit in
early breast cancer. J Natl Cancer Inst 2013, 105:960967.
57. Perez EA, Dueck AC, McCullough AE, Chen B, Geiger XJ, Jenkins RB, Lingle
WL, Davidson NE, Martino S, Kaufman PA, Kutteh LA, Sledge GW, Harris LN,
Gralow JR, Reinholz MM: Impact of PTEN protein expression on benefit
from adjuvant trastuzumab in early-stage human epidermal growth
factor receptor 2-positive breast cancer in the North Central Cancer
Treatment Group N9831 trial. J Clin Oncol 2013, 31:21152122.
58. Cabral F, Sobel ME, Gottesman MM: CHO mutants resistant to colchicine,
colcemid or griseofulvin have an altered beta-tubulin. Cell 1980, 20:2936.
59. Kavallaris M: Microtubules and resistance to tubulin-binding agents. Nat
Rev Cancer 2010, 10:194204.
60. Morse DL, Gray H, Payne CM, Gillies RJ: Docetaxel induces cell death
through mitotic catastrophe in human breast cancer cells. Mol Cancer
Ther 2005, 4:14951504.
61. Torres K, Horwitz SB: Mechanisms of Taxol-induced cell death are concen-
tration dependent. Cancer Res 1998, 58:36203626.
62. Hernandez-Vargas H, Palacios J, Moreno-Bueno G: Molecular profiling of
docetaxel cytotoxicity in breast cancer cells: uncoupling of aberrant
mitosis and apoptosis. Oncogene 2007, 26:29022913.
63. Vakifahmetoglu H, Olsson M, Zhivotovsky B: Death through a tragedy:
mitotic catastrophe. Cell Death Differ 2008, 15:11531162.
64. ClinicalTrials.gov. : [http://clinicaltrials.gov].
65. Mohsin SK, Weiss HL, Gutierrez MC, Chamness GC, Schiff R, Digiovanna MP,
Wang CX, Hilsenbeck SG, Osborne CK, Allred DC, Elledge R, Chang JC:
Neoadjuvant trastuzumab induces apoptosis in primary breast cancers.
J Clin Oncol 2005, 23:24602468.
66. Marty M, Cognetti F, Maraninchi D, Snyder R, Mauriac L, Tubiana-Hulin M,
Chan S, Grimes D, Anton A, Lluch A, Kennedy J, OByrne K, Conte P, Green
M, Ward C, Mayne K, Extra JM: Randomized phase II trial of the efficacy
and safety of trastuzumab combined with docetaxel in patients with hu-
man epidermal growth factor receptor 2-positive metastatic breast
cancer administered as first-line treatment: the M77001 study group.
J Clin Oncol 2005, 23:42654274.
67. Bhattacharyya B, Wolff J: Maytansine binding to the vinblastine sites of
tubulin. FEBS Lett 1977, 75:159162.
68. Schaller G, Fuchs I, Gonsch T, Weber J, Kleine-Tebbe A, Klare P, Hindenburg
HJ, Lakner V, Hinke A, Bangemann N: Phase II study of capecitabine plus
trastuzumab in human epidermal growth factor receptor 2 overexpress-
ing metastatic breast cancer pretreated with anthracyclines or taxanes.
J Clin Oncol 2007, 25:32463250.
69. Wardley AM, Pivot X, Morales-Vasquez F, Zetina LM, de Fatima Dias Gaui M,
Reyes DO, Jassem J, Barton C, Button P, Hersberger V, Torres AA: Random-
ized phase II trial of first-line trastuzumab plus docetaxel and capecita-
bine compared with trastuzumab plus docetaxel in HER2-positive
metastatic breast cancer. J Clin Oncol 2010, 28:976983.
70. Swain SM, Kim SB, Cortes J, Ro J, Semiglazov V, Campone M, Ciruelos E, Ferrero
JM,SchneeweissA,KnottA,ClarkE,RossG,BenyunesMC,BaselgaJ:Pertuzumab,
trastuzumab, and docetaxel for HER2-positive metastatic breast cancer
(CLEOPATRA study): overall survival results from a randomised, double-
blind, placebo-controlled, phase 3 study. Lancet Oncol 2013, 14:461471.
71. Blackwell KL, Burstein HJ, Storniolo AM, Rugo HS, Sledge G, Aktan G, Ellis C,
Florance A, Vukelja S, Bischoff J, Baselga J, OShaughnessy J: Overall survival
benefit with lapatinib in combination with trastuzumab for patients with
human epidermal growth factor receptor 2-positive metastatic breast can-
cer: final results from the EGF104900 Study. JClinOncol2012, 30:25852592.
72. Kaufman B, Mackey JR, Clemens MR, Bapsy PP, Vaid A, Wardley A, Tjulandin S,
Jahn M, Lehle M, Feyereislova A, Revil C, Jones A: 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. JClinOncol2009, 27:55295537.
73. Huober J, Fasching PA, Barsoum M, Petruzelka L, Wallwiener D, Thomssen C,
Reimer T, Paepke S, Azim HA, Ragosch V, Kubista E, Baumgartner AK,
Beckmann MW, May C, Nimmrich I, Harbeck N: Higher efficacy of letrozole
in combination with trastuzumab compared to letrozole monotherapy
as first-line treatment in patients with HER2-positive, hormone-receptor-
positive metastatic breast cancer - results of the eLEcTRA trial.
Breast 2012, 21:2733.
74. Yao E, Zhou W, Lee-Hoeflich ST, Truong T, Haverty PM, Eastham-Anderson J,
Lewin-Koh N, Gunter B, Belvin M, Murray LJ, Friedman LS, Sliwkowski MX,
Barok et al. Breast Cancer Research 2014, 16:209 Page 11 of 12
http://breast-cancer-research.com/content/16/2/209
Hoeflich KP: Suppression of HER2/HER3-mediated growth of breast
cancer cells with combinations of GDC-0941 PI3K inhibitor, trastuzumab,
and pertuzumab. Clin Cancer Res 2009, 15:41474156.
75. Junttila TT, Akita RW, Parsons K, Fields C, Lewis Phillips GD, Friedman LS,
Sampath D, Sliwkowski MX: Ligand-independent HER2/HER3/PI3K
complex is disrupted by trastuzumab and is effectively inhibited by the
PI3K inhibitor GDC-0941. Cancer Cell 2009, 15:429440.
76. Tikhomirov O, Carpenter G: Geldanamycin induces ErbB-2 degradation by
proteolytic fragmentation. J Biol Chem 2000, 275:2662526631.
77. Isaacs JS, Xu W, Neckers L: Heat shock protein 90 as a molecular target
for cancer therapeutics. Cancer Cell 2003, 3:213217.
78. Khandelwal A, Saber H, Shapiro MA, Zhao H: Antibody-drug conjugate
development.InAntibody-Drug Conjugates and Immunotoxins, From Pre-
Clinical Development to Therapeutic Application. Edited by Lewis Phillips GD.
New York: Humana Press; 2013:2340.
79. Hamblett KJ, Senter PD, Chace DF, Sun MM, Lenox J, Cerveny CG, Kissler
KM, Bernhardt SX, Kopcha AK, Zabinski RF, Meyer DL, Francisco JA: Effects
of drug loading on the antitumor activity of a monoclonal antibody
drug conjugate. Clin Cancer Res 2004, 10:70637070.
80. Junttila TT, Parsons K, Olsson C, Lu Y, Xin Y, Theriault J, Crocker L, Pabonan
O, Baginski T, Meng G, Totpal K, Kelley RF, Sliwkowski MX: Superior in vivo
efficacy of afucosylated trastuzumab in the treatment of HER2-amplified
breast cancer. Cancer Res 2010, 70:44814489.
81. Lazar GA, Dang W, Karki S, Vafa O, Peng JS, Hyun L, Chan C, Chung HS,
Eivazi A, Yoder SC, Vielmetter J, Carmichael DF, Hayes RJ, Dahiyat BI:
Engineered antibody Fc variants with enhanced effector function. Proc
Natl Acad Sci U S A 2006, 103:40054010.
82. Costantini DL, Bateman K, McLarty K, Vallis KA, Reilly RM: Trastuzumab-
resistant breast cancer cells remain sensitive to the auger electron-
emitting radiotherapeutic agent 111In-NLS-trastuzumab and are radio-
sensitized by methotrexate. J Nucl Med 2008, 49:14981505.
doi:10.1186/bcr3621
Cite this article as: Barok et al.:Trastuzumab emtansine: mechanisms of
action and drug resistance. Breast Cancer Research 2014 16:209.
Barok et al. Breast Cancer Research 2014, 16:209 Page 12 of 12
http://breast-cancer-research.com/content/16/2/209
... T-DXd and T-DM1 are both antibody-drug conjugates composed of humanized monoclonal antibodies targeting HER2, linked to a cytotoxic payload [17][18][19] . T-DM1 incorporates a microtubule-disrupting agent, which is tethered by a durable thioether bond, whereas T-DXd employs a topoisomerase I inhibitor connected through a tetrapeptide-based cleavable linker, which enables greater specificity in targeting cancer cells, thereby diminishing unintended toxicity 17,19,20 . ...
... T-DXd and T-DM1 are both antibody-drug conjugates composed of humanized monoclonal antibodies targeting HER2, linked to a cytotoxic payload [17][18][19] . T-DM1 incorporates a microtubule-disrupting agent, which is tethered by a durable thioether bond, whereas T-DXd employs a topoisomerase I inhibitor connected through a tetrapeptide-based cleavable linker, which enables greater specificity in targeting cancer cells, thereby diminishing unintended toxicity 17,19,20 . T-DXd has a high, homogeneous drug-to-antibody ratio of approximately 8, while T-DM1 has a drug-to-antibody ratio of approximately 3.5 (refs. ...
Article
Full-text available
Trastuzumab deruxtecan (T-DXd) demonstrated significantly improved efficacy over trastuzumab emtansine (T-DM1) in DESTINY-Breast03 (median follow-up, 28 months). We report updated efficacy and safety analyses, including secondary and exploratory efficacy endpoints (median follow-up, 41 months) of DESTINY-Breast03. Patients with advanced HER2-positive metastatic breast cancer previously treated with taxane and trastuzumab were randomized to T-DXd (5.4 mg per kg (261 patients)) or T-DM1 (3.6 mg per kg (263 patients)). The primary endpoint was progression-free survival (PFS) by blinded independent central review and was previously reported. The key secondary endpoint was overall survival (OS). Other secondary endpoints included objective response rate, duration of response and PFS (all by investigator assessment) and safety. At data cutoff, 20 November 2023, median PFS by investigator assessment was 29.0 versus 7.2 months (hazard ratio (HR), 0.30; 95% confidence interval (CI), 0.24–0.38), the 36-month PFS rate was 45.7% versus 12.4% and median OS was 52.6 versus 42.7 months (HR, 0.73; 95% CI, 0.56–0.94) with T-DXd versus T-DM1, respectively. Treatment-emergent adverse events were consistent with the previous analyses. No new instances of grade ≥3 interstitial lung disease or pneumonitis occurred (all grade rate, 16.7% (T-DXd) versus 3.4% (T-DM1)). With longer follow-up, T-DXd continued to demonstrate superior efficacy over T-DM1 with a manageable safety profile. ClinicalTrials.gov registration: NCT03529110.
... Additionally, T-DM1 showed had a substantial impact in the adjuvant setting after insufficient pathological response to neo-adjuvant treatment in 2019 [86]. T-DM1 exerts its effect through the same mechanisms as trastuzumab with the addition of the chemotherapeutic payload [87]. ...
... In parallel to the quantification of HER2 receptors, we performed viability assays on cells harvested on the same day to determine the response to two different HER2-targeting ADCs with either monomethyl auristatin E (MMAE), a tubulin polymerization inhibitor [5], or emtansine (DM1), a maytansine derivative and a second microtubule inhibitor [19], to produce trastuzumab-monomethyl auristatin E (T-MMAE) and trastuzumab-emtansine (T-DM1), respectively. In ADC synthesis, chemical linkers are added to small-molecule drugs to facilitate conjugation to the antibody and to provide a mechanism for drug release after cellular internalization. ...
Article
Full-text available
HER2-targeting therapies have advanced breast cancer treatment over the past decade. Clinically, eligibility for HER2 therapies is determined by assessing HER2 levels on tumor cell surfaces through immunohistochemistry or by gene regulation through fluorescence in situ hybridization. HER2 therapies are not always effective in patients with elevated levels of HER2, questioning whether the amount of HER2 is sufficiently predictive of patient outcomes. Additionally, the HER2-targeting antibody–drug conjugate (ADC) Enhertu® was recently approved for metastasized HER2-low cancers, confirming the benefits of HER2 treatment for patients with low HER2 levels. To evaluate the correlation between HER2 levels and treatment efficacy, we quantified HER2 on eight cell lines using flow cytometry while simultaneously determining the toxicity of two HER2-targeting ADCs. Both HER2-high cell lines and HER2-low cell lines had significant toxicity responses to ADCs. We quantified HER2 internalization and found no correlation between HER2 levels and the percentage of internalization. We found a useful metric suggesting that a minimum number of HER2 receptors trafficked to lysosomes is sufficient to provide effective treatment. Our results indicate that the current standards of determining eligibility for HER2 therapy could limit patients’ access to effective treatment. In conclusion, HER2 levels are not wholly adequate to determine the response to ADC treatment.
... Upon administration, T-DM1 selectively binds exquisitely to subdomain IV of the HER2 receptor on the surface of antigen-positive cells via the natural tropism of TRZ [17]. The interaction of TRZ with HER2 receptor leads to endocytosis [102], degradation of endosomes on fusion with lysosomes [103] and liberation of MCC-DM1 which navigates through the nuclear membrane to interfere with the polymerization of tubulin [104]. Specifically, inhibition of tubulin assembly into a functional mitotic spindle prevents the important nexus between kinetochores on chromatids and the spindles, hence inducing cell-cycle arrest in metaphase culminating in ultimate mitotic disaster [105]. ...
Article
Full-text available
The HER2 receptor tyrosine kinase is a member of the epidermal growth factor receptor family which includes EGFR, HER3 and HER4. They are known to play critical roles in both normal development and cancer. A subset of breast cancers is associated with the HER2 gene, which is amplified and/or overexpressed in 20-25% of invasive breast cancers and is correlated with tumor resistance to chemotherapy, Metastatic Breast Cancer (MBC) and poor patient survival. The advent of receptor tyrosine kinase inhibitors has improved the prognosis of HER2-postive breast cancers; however, HER2+MBC invariably progresses (acquired resistance or de novo resistance). The monoclonal antibody-based drugs (large molecule TKIs) target the extracellular binding domain of HER2; while the small molecule TKIs act intracellularly to inhibit proliferation and survival signals. We reviewed the modes of action of the TKIs with a view to showing which of the TKIs could be combined in nanoparticles to benefit from the power of nanotechnology (reduced toxicity, improved solubility of hydrophobic drugs, long circulation half-lives, circumventing efflux pumps and preventing capture by the reticuloendothelial system (mononuclear phagocyte system). Nanotherapeutics also mediate the synchronization of the pharmacokinetics and biodistribution of multiple drugs incorporated in the nanoparticles. Novel TKIs that are currently under investigation with or without nanoparticle delivery are mentioned, and nano-based strategies to improve their delivery are suggested. Immunotherapies currently in clinical practice, clinical trials or at the preclinical stage are discussed. However, immunotherapy only works well in relatively small subsets of patients. Combining nanomedicine with immunotherapy can boost therapeutic outcomes, by turning “cold” nonimmunoresponsive tumors and metastases into “hot” immunoresponsive lesions.
Article
Introduction: Urothelial carcinoma (UC) is a refractory disease for which achieving satisfactory outcomes remains challenging with current surgical interventions. Antibody–drug conjugates (ADCs) are a novel class of targeted therapeutics that have demonstrated encouraging results for UC. Although there is a limited number of high-quality randomized control trials (RCTs) examining the use of ADCs in patients with UC, some prospective non-randomized studies of interventions (NRSIs) provide valuable insights and pertinent information. We aim to assess the efficacy and safety of ADCs in patients with UC, particularly those with locally advanced and metastatic diseases. Methods: A systematic search was conducted across PubMed, Embase, the Cochrane Library, and Web of Science databases to identify pertinent studies. Outcomes, such as the overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), adverse events (AEs), and treatment-related adverse events (TRAEs), were extracted for further analyses. Results: Twelve studies involving 1,311 patients were included in this meta-analysis. In terms of tumor responses, the pooled ORR and DCR were 40% and 74%, respectively. Regarding survival analysis, the pooled median PFS and OS were 5.66 months and 12.63 months, respectively. The pooled 6-month PFS and OS were 47% and 80%, while the pooled 1-year PFS and OS were 22% and 55%, respectively. The most common TRAEs of the ADCs were alopecia (all grades: 45%, grades ≥ III: 0%), decreased appetite (all grades: 34%, grades ≥ III: 3%), dysgeusia (all grades: 40%, grades ≥ III: 0%), fatigue (all grades: 39%, grades ≥ III: 5%), nausea (all grades: 45%, grades ≥ III: 2%), peripheral sensory neuropathy (all grades: 37%, grades ≥ III: 2%), and pruritus (all grades: 32%, grades ≥ III: 1%). Conclusion: The meta-analysis in this study demonstrates that ADCs have promising efficacies and safety for patients with advanced or metastatic UC. Systematic review registration: https://www.crd.york.ac.uk/prospero/, identifier: CRD42023460232
Article
We aimed to evaluate the efficacy and safety of trastuzumab emtansine in patients with metastatic breast cancer previously treated with pertuzumab plus trastuzumab and taxane. We reviewed the medical records of patients who were diagnosed with Human Epidermal Growth Factor Receptor 2 (HER-2) positive metastatic breast cancer and received pertuzumab and then TDM-1 between January 2014 and January 2021 from twenty- five cancer centers. The Kaplan- Meier method estimated progression-free survival (PFS) and overall survival (OS). Additionally, objective response rate (ORR), clinical benefit rate (CBR), and safety were evaluated. One hundred fifty-three patients were included,79.1% of the patients received TDM-1 in the second line, 90.8% had visceral metastasis, and 30.7% had central nervous system involvement. The PFS and OS of TDM-1 were evaluated according to the number of previous lines (on the 2nd line or more than two lines) metastatic sites (visceral and non-visceral) and the presence of central nervous metastasis. In TDM-1 therapy, PFS in second line therapy was ten months (95% CI: 7.7 − 12.2); this was statistically higher than later-line PFS, which was six months (95% CI: 3.3 to 8.6) (p = 0.004). The median OS time was 25 months (95% CI: 21.0 to 28.9) in patients treated with TDM-1 in the second line and 19 months (95% CI: 12.3 to 25.6) in patients who received later than the second line(p = 0.175). There were no significant differences in PFS time of patients with and without visceral and central nervous metastases. Our study showed that TDM-1 was also effective in patients using pertuzumab, contributes significantly to PFS when used in the second line compared to its use in the later line, and does not make any difference in OS.
Article
This meta-analysis aims to comprehensively evaluate the efficacy and safety of T-DM1 in treating HER2-positive breast cancer, providing insights for clinical practice. We conducted a literature search in PubMed, Cochrane Library, and Embase databases up to September 2023, collecting randomized controlled trials and cohort studies on T-DM1 for HER2-positive breast cancer. Out of 316 initially retrieved articles, 12 studies meeting the quality and inclusion criteria were included after a rigorous screening process. We used RevMan 5.3 software for the meta-analysis, employing fixed or random-effect models. Odds ratios (RRs) and 95% confidence intervals (CIs) were calculated as effect size measures. We conducted sensitivity analyses and assessed publication bias to ensure the results’ stability and reliability. In seven studies, T-DM1 treatment significantly prolonged OS in patients with HER2-positive breast cancer [hazard ratio (HR) = 0.70, 95% CI: 0.64–0.77, P < 0.01], and the effect was especially pronounced in patients with advanced disease (HR = 0.64, 95% CI: 0.54–0.76, P < 0.001). Analysis of pCR rates did not show a significant difference (OR = 0.91, 95% CI: 0.48–1.73, P = 0.77). In five studies, ORR improved, but the difference between the two groups was not significant (OR = 1.16, 95% CI: 0.66–2.05, P = 0.61). Analysis of progression-free survival (PFS) showed a significant improvement in the experimental group relative to the control group (HR = 0.69, 95% CI: 0.57–0.84, P = 0.0003). Regarding the incidence of total adverse events, no significant difference was seen between the two groups (OR = 2.16, 95% CI: 0.98–4.79, P = 0.06), but for specific adverse events, such as leukopenia and neutropenia, the T-DM1 group demonstrated a significant reduction relative to the other treatment regimens. The results underscore the potential of T-DM1 in enhancing survival among patients with advanced HER2-positive breast cancer, yet they also highlight variability in effectiveness concerning pCR rate and ORR. The findings on adverse effects underscore the necessity of a balanced consideration of T-DM1’s risks and benefits. Future research should focus on a more detailed examination of responses in varied patient populations, long-term outcomes, and a thorough economic evaluation of T-DM1, along with an exploration into treatment resistance. This will provide a more nuanced understanding of T-DM1’s role in the treatment landscape of HER2-positive breast cancer.
Article
In the ever-evolving landscape of cancer research, immuno-oncology stands as a beacon of hope, offering novel avenues for treatment. This study capitalizes on the vast repository of immuno-oncology-related scientific documents within the CAS Content Collection, totaling over 350,000, encompassing journals and patents. Through a pioneering approach melding natural language processing with the CAS indexing system, we unveil over 300 emerging concepts, depicted in a comprehensive “Trend Landscape Map”. These concepts, spanning therapeutic targets, biomarkers, and types of cancers among others, are hierarchically organized into eight major categories. Delving deeper, our analysis furnishes detailed quantitative metrics showcasing growth trends over the past three years. Our findings not only provide valuable insights for guiding future research endeavors but also underscore the merit of tapping the vast and unparalleled breadth of existing scientific information to derive profound insights.
Chapter
Directing chemotherapy to cancers utilizing antibody–drug conjugate technology is likely to be a “game changing” development in the management of cancer patient care. With promising data already emerging from clinical testing of trastuzumab-DM1 and brentuximab vedotin, it is becoming clear that ADCs are likely to define a promising new treatment option for patients. However, in the short term, the onslaught of ADCs being evaluated in clinical trials highlights the need to delineate any heterogeneity in drug response by the use of predictive biomarkers in order to maximize clinical benefit. In this chapter, we discuss the importance of understanding ADC mechanism of action in the context of predictive biomarkers for testing these experimental therapeutics in clinical trials. We also discuss the challenges associated with implementation of a successful biomarker testing strategy for the ADCs.
Article
Antibody–drug conjugate (ADC) development started about three decades ago with the hypothesis that toxins conjugated to antibodies would enhance antitumor activity and reduce toxicity by delivering toxins to specific tumor sites. Since then, the field has evolved to include potent small molecule drugs (SMD) and radiolabeled drugs conjugated to antibodies targeting both solid tumors and hematologic malignancies. Improved ADC technology has paved the way for increased drug delivery to the target tumors and decreased normal tissue exposure to cytotoxic agents. Radio-immunoconjugates (RICs) present a different set of challenges leading to unique development pathways. Several factors need to be taken into consideration when developing RICs, such as decay of radioactivity, potentially higher exposure to normal tissue caused by lower specificity, and dehalogenation leading to a loss of signal. This chapter focuses on antibodies conjugated to SMDs.
Article
Antibody-drug conjugates (ADCs) enable targeted delivery of therapeutics to cancer cells and offer potential for more selective therapy. Several ADCs are demonstrating promising clinical efficacy, however due to the complexity of human cancer, tumors become refractory to most drug treatments. We hypothesized that cultured tumor cells chronically treated with an ADC would acquire mechanisms of resistance unique to ADC-based therapy. Human breast cancer cell lines were exposed to multiple cycles of an anti-Her2 trastuzumab-maytansinoid conjugate (TM) at IC80 concentrations for 3 days followed by ∼1 week without treatment to simulate a maximally tolerated dose followed by recovery. After ∼2 months, significant resistance developed in JIMT1 and MDA-MB-361 cell lines. The potency of TM conjugate on drug-selected cell lines was reduced to the activity observed on Her2-negative cells (>20 & >270X in JIMT1 & 361 cell models, respectively). Flow cytometry revealed 58% and 25% decreases in Her2 receptor number in JIMT-TM & 361-TM, respectively. Proteomic profiling of surface proteins in JIMT-TM cells demonstrated significant increases in proteins involved in post-translational modification (e.g., ubiquitinating enzymes, kinases, and phosphatases), as well as elevated levels of endosomal and vesicle proteins (e.g., RAB family members), and proteins mediating microtubule and actin dynamics. Notably, ABC drug transporters were not altered in JIMT-TM cells. In 361-TM cells, an increase in ABCC1 (MRP1) was observed, but no changes in ABCB1 (MDR1) which typically effluxes tubulin inhibitors. These data suggest the acquisition of complex resistance mechanisms upon ADC treatment. The cross-resistance profile of these ADC refractory models was evaluated. Minimal or no resistance (1 - 6X) was observed to free drugs, including maytansine or other standard-of-care tubulin or DNA targeted therapeutics. In JIMT-TM cells, cross-resistance was observed to other trastuzumab-based ADCs, including those containing either non-cleavable or cleavable linkers, and delivering payloads with various biological mechanisms of action. In contrast, 361-TM cells (which were made resistant to an ADC delivering a tubulin inhibitor via a non-cleavable linker) retained significant sensitivity to ADCs containing cleavable linkers, even those containing other tubulin inhibitor-based payloads. Moreover, both 361-TM and JIMT-TM resistant cell lines retained sensitivity to ADCs delivering payloads with alternative (non-tubulin) mechanisms of action. Therefore, despite the reduction in antigen levels observed in both cell lines, modification of the linker and/or the payload was able to overcome resistance mediated by the initial ADC therapy. These data offer the potential to treat refractory tumors with ADCs containing the same antibody vehicle, but delivering alternative linkers or payloads. Citation Format: Xingzhi Tan, Guixian Jin, Jeremy Myers, Veronica Diesl, Max Follettie, My-Hanh Lam, Sylvia Musto, Kiran Khandke, Manoj Charati, Edmund Graziani, Andreas Maderna, Chakrapani Subramanyam, Frank Koehn, Russell Dushin, Kim Arndt, Christopher J. O'Donnell, Hans-Peter Gerber, Frank Loganzo. Tumor cells selected for resistance to an antibody-drug conjugate retain sensitivity to ADCs with modified linkers and payloads. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4629. doi:10.1158/1538-7445.AM2013-4629
Article
Targeting HER2 with multiple HER2-directed therapies represents a promising area of treatment for HER2-positive cancers. We investigated combining the HER2-directed antibody-drug conjugate trastuzumab emtansine (T-DM1) with the HER2 dimerization inhibitor pertuzumab (Perjeta™). Drug combination studies with T-DM1 and pertuzumab were performed on cultured tumor cells and in mouse xenograft models of HER2-amplified cancer. In patients with HER2-positive locally advanced or metastatic breast cancer, T-DM1 was dose-escalated with a fixed standard pertuzumab dose in a 3+3 Phase Ib/II study design. Treatment of HER2-overexpressing tumor cells in vitro with T-DM1 plus pertuzumab resulted in synergistic inhibition of cell proliferation and induction of apoptotic cell death. The presence of the HER3 ligand, heregulin (NRG-1β) reduced the cytotoxic activity of T-DM1 in a subset of breast cancer lines; this effect was reversed by the addition of pertuzumab. Results from mouse xenograft models showed enhanced anti-tumor efficacy with T-DM1 and pertuzumab resulting from the unique anti-tumor activities of each agent. In patients with metastatic breast cancer previously treated with trastuzumab, lapatinib, and chemotherapy, T-DM1 could be dosed at the maximum tolerated dose (MTD, 3.6 mg/kg every 3 weeks) with standard-dose pertuzumab. Adverse events were mostly Grade 1 and 2, with indications of clinical activity. Dual-targeting of HER2 with the combination of T-DM1 and pertuzumab in cell culture and mouse xenograft models resulted in enhanced anti-tumor activity. In patients, this combination showed an encouraging safety and tolerability profile with preliminary evidence of efficacy.
Article
The HER2 oncogene targeting drug trastuzumab shows remarkable efficacy in patients overexpressing HER2. However acquired or primary resistance develops in most of the treated patients why alternative treatment strategies are strongly needed. As endosomal sorting and recycling are crucial steps for HER2 activity and the vacuolar H(+)-ATPase (V-ATPase) is an important regulator of endocytotic trafficking, we proposed that targeting V-ATPase opens a new therapeutic strategy against trastuzumab-resistant tumor cells in vitro and in vivo. V-ATPase inhibition with archazolid, a novel inhibitor of myxobacterial origin, results in growth inhibition, apoptosis and impaired HER2 pro-survival signaling of the trastuzumab-resistant cell line JIMT-1. This is accompanied by a decreased expression on the plasma membrane and accumulation of HER2 in the cytosol, where it colocalizes with endosomes, lysosomes and autophagosomes. Importantly, microscopic analysis of JIMT-1 xenograft tumor tissue of archazolid treated mice confirms the defect in HER2-recycling which leads to reduced tumor growth. These results suggest that V-ATPase inhibition by archazolid induces apoptosis and inhibits growth of trastuzumab-resistant tumor cells by retaining HER2 in dysfunctional vesicles of the recycling pathway and consequently abrogates HER2-signaling in vitro as well as in vivo. V-ATPase inhibition is thus suggested as a promising strategy for treatment of trastuzumab-resistant tumors.