Download Oncologist®

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
The
Oncologist
®
Epidermal Growth Factor Receptor Dependence
in Human Tumors: More Than Just Expression?
CARLOS L. ARTEAGA
Departments of Medicine and Cancer Biology, and Vanderbilt-Ingram Comprehensive Cancer Center,
Vanderbilt University School of Medicine, Nashville, Tennessee, USA
Key Words. EGFR-TKI · ZD1839 (Iressa™) · Prognostic factor · Cancer therapy · Predictive factor
After completing this course, the reader will be able to:
1. Describe the current limitations in measuring levels of EGR receptor (EGFR) expression in tissues.
2. Identify the molecular pathways for signal transduction induced by EGFR activation.
3. Identify the level of expression of EGF in different tumor types.
CME
Access and take the CME test online and receive one hour of AMA PRA category 1 credit at CME.TheOncologist.com
A BSTRACT
The epidermal growth factor receptor (EGFR) is a
rational target for antitumor strategies. EGFR signaling
causes increased proliferation, decreased apoptosis, and
enhanced tumor cell motility and neo-angiogenesis. The
EGFR is expressed or highly expressed in a variety of
human tumors of epithelial origin. ZD1839 (Iressa™) is
an orally active, selective EGFR tyrosine kinase
inhibitor, which blocks signal transduction pathways
implicated in proliferation and survival of cancer cells.
The lack of a consistent method of evaluating levels
of EGFR has caused a disparity in reports of the EGFR
as a prognostic factor; however, for some tumors,
EGFR is a strong prognostic indicator associated with
more aggressive disease and reduced survival. So far, no
clear association between EGFR levels and response to
EGFR-targeted agents has been found. Preclinical studies with ZD1839 have noted a relationship between the
two in some cases, but not others.
EGFR signaling may be increased by a number of
mechanisms in addition to high expression levels of
EGFR, including receptor mutations, heterodimerization with other members of this receptor family such as
HER2 (erbB2), increased expression of (autocrine/
paracrine) ligands, and alterations in molecules that
control receptor signaling output. Each of these components could be assessed to give an indication of the
magnitude of EGFR signal amplification. Evaluation of
signaling components downstream from EGFR should
provide information on the activation of the EGFR
pathway.
Until EGFR-based assays predictive of a response to
receptor-targeted therapies are available, there is no
clear justification for stratifying patients by EGFR status or excluding patients with low EGFR levels from trials with ZD1839 or other EGFR inhibitors. The
Oncologist 2002;7(suppl 4):31-39
Correspondence: Carlos L. Arteaga, M.D., Departments of Medicine and Cancer Biology, and Vanderbilt-Ingram Comprehensive
Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232-6307, USA. Telephone: 615-936-3524; Fax: 615936-1790; e-mail: [email protected] Received June 26, 2002; accepted for publication July 25, 2002.
©AlphaMed Press 1083-7159/2002/$5.00/0
The Oncologist 2002;7(suppl 4):31-39
www.TheOncologist.com
Downloaded from www.TheOncologist.com by on January 16, 2010
L EARNING O BJECTIVES
EGFR Dependence in Cancer: More Than Just Expression?
32
INTRODUCTION
EGFR Expression in Solid Tumors
The epidermal growth factor receptor (EGFR; HER1;
erbB1) is expressed or highly expressed in a variety of human
tumors including non-small cell lung cancer (NSCLC),
breast, head and neck, gastric, colorectal, esophageal,
prostate, bladder, renal, pancreatic, and ovarian cancers [1].
Activation of the EGFR signaling pathway has many
effects including increased proliferation and angiogenesis,
and decreased apoptosis. These effects are mediated by a
complex series of signaling mechanisms, such as engagement
of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3 kinase (PI3K) pathways (Fig. 1) [2]. High
EGFR expression has been associated with advanced tumor
stage, resistance to standard therapies (hormonal therapy,
chemotherapy, and radiation) [3-5] and, in some tumors, with
poor patient prognosis [6-8].
As a result, EGFR was proposed as a rational target for
antitumor strategies. EGFR-targeted therapies in development
include those that interact with the extracellular ligand-binding
A
Epiregulin β-cellulin
(1, 4)
(1)
TGF-α EGF
(1)
(1)
HB-EGF
(1, 4)
Amphiregulin
(1)
Ligands
Input
layer
1
3
1
2
1
1
1
4
Receptor
dimers
X
B
Src
Cbl
PLCγ
PI3K
Shp2
PKC
Sp1
C
Output
layer
Inhibition of
apoptosis
Akt
Bad
Jun
Fos
Elk
Proliferation Angiogenesis Maturation
Nck
Sos
RAF
MEK
MAPK
S6K
Myc
Grb2
GAP
Ras-GTP
Signal-processing
layer
Shc
Ras-GDP
Vav
Grb7
Adaptors
and
enzymes
Crk
Rac
PAK
JNKK
JNK
Transcription
factors
Egr1
Migration
Cascades
Abl
Adhesion
Invasion
Differentiation
Figure 1. The EGFR signaling network. Adapted from [2] by permission from Nature Reviews Molecular Cell Biology 2001;2:127-137. ©2001
Macmillan Magazines Ltd. A) Input layer. Ligands and dimeric receptor combinations for EGFR represent the input layer. Numbers in each ligand block indicate the respective high-affinity HER receptor among HER1 (1), HER2 (2), HER3 (3) and HER4 (4) (HB-EGF, heparin-binding
EGF). HER3 lacks intrinsic kinase activity (crossed kinase domains). B) Signal-processing layer. Signaling to the adaptor-enzyme layer is shown
only for the EGFR homodimer. Only selected pathways and transcription factors are represented. C) Output layer. Cellular processes accelerated or dysregulated by aberrant EGFR signaling output.
Downloaded from www.TheOncologist.com by on January 16, 2010
domain (monoclonal antibodies, bispecific and single chain
antibodies, immunotoxin conjugates), and those that act
intracellularly, such as the small-molecule EGFR tyrosine
kinase inhibitors (EGFR-TKIs) that compete with ATP to
bind to the receptor’s ATP site. In either case, the pathways
leading to tumor cell proliferation, angiogenesis, metastasis,
and cell survival are disrupted [9, 10]. Of these, one of the
most advanced in clinical development is ZD1839 (Iressa™),
an orally active, selective EGFR-TKI; with a mode of action
distinct from cytotoxic chemotherapy.
While there are increasing data to confirm the antitumor
activity of these agents in clinical trials, there are several
important issues still to be addressed, including how best to
evaluate EGFR expression, whether there is a correlation
between EGFR expression and patient prognosis, and whether
EGFR expression levels can predict response to therapy.
Furthermore, there are many components of the EGFR signaling network (Fig. 1), each of which can modulate EGFR signaling output and thus tumor dependence on it. How these
components relate to tumor progression and/or antitumor
activity of anti-EGFR therapies requires further investigation.
Arteaga
variety of methods, such as fluorescence in situ hybridization
or quantitative PCR, may enable detection of alterations to the
EGFR gene such as amplification, mutation or deletion,
which may, in turn, affect receptor signaling output.
EGFR as a Prognostic Factor
Although EGFR is generally considered to be predictive of poor prognosis in human cancers, conflicting results
have been reported [17]. EGFR has been identified as a
strong prognostic indicator in head and neck, breast, ovarian, cervical, bladder, and esophageal cancers. High EGFR
expression has been shown to correlate with poor survival
in a range of tumors including nasopharyngeal, NSCLC,
ovarian, and breast. In one of these studies, prognostic factors were evaluated in 77 patients with unresectable carcinoma of the pharynx [8]. In a multivariate analysis, EGFR
level was found to be a significant predictor for reduced
time to treatment failure (p = 0.0001) and overall survival
(p = 0.0001). In patients with nasopharyngeal carcinoma, a
significant correlation between high levels of EGFR and
poor survival has also been noted (p = 0.05) [18]. In 108
primary ovarian cancer specimens, 61% scored positive for
EGFR, and a significant correlation was observed between
EGFR expression and shorter overall and progression-free
survival [7]. This study also correlated EGFR status with
resistance to platinum-containing chemotherapy. In addition, several studies have reported that EGFR expression
predicts for a significantly shorter disease-free and overall
survival in patients with breast cancer [19, 20]. However,
its prognostic value in all patient subgroups has not been
consistent among these studies. For example, Tsutsui et al.
[20] demonstrated that EGFR was a significant prognostic
factor only for disease-free survival in lymph-node-negative breast cancer patients (p = 0.0241) and overall survival
in node-positive patients (p = 0.0333). Among NSCLCs,
squamous-cell carcinomas were found to be more likely
to be EGFR-positive than non-squamous-cell carcinomas
(p = 0.0121), and an absence of EGFR expression correlated with a longer survival (p = 0.024) [21]. This observation confirmed the results of another study showing that
patients with EGFR-positive NSCLC had shorter median
survival than patients with EGFR-negative tumors [22].
Potentially explaining the association with poor patient
outcome, the expression of EGFR has been linked with
resistance to both hormonal therapies and chemotherapeutic agents. However, the value of EGFR in predicting the
efficacy of cancer drugs is still being evaluated. In one
study of 155 breast cancer patients whose disease was progressing while they were receiving tamoxifen, EGFR
expression was examined by IHC in pretreatment biopsies.
The results of this study confirmed that pretreatment
Downloaded from www.TheOncologist.com by on January 16, 2010
Methods of Evaluating EGFR Expression
The causal role of high expression of HER2 in cancer
progression was the basis for the development of trastuzumab
(Herceptin®), a humanized monoclonal antibody against
HER2. This agent has demonstrated clinical benefits in
patients with HER2-positive breast cancer [11]. The U.S.
Food and Drug Administration has approved an immunohistochemical test for HER2 expression, which predicts for
clinical response to trastuzumab [12]. In contrast, the situation
with EGFR-targeted agents is less straightforward. No
method of analysis of EGFR is consistently employed in all
laboratories, making the comparison of results from different
studies difficult. A variety of techniques may be used to evaluate EGFR at the DNA, RNA, and protein levels, as well as
the level of receptor activation in situ.
Immunohistochemistry (IHC) is commonly used to
evaluate EGFR protein levels and is arguably the most convenient method for analysis of clinical samples. However,
there is no standard scoring system, with no consensus
available on the cut-off points between no, low, medium, or
high expression. The method is not strictly quantitative and
is prone to inter-observer scoring error, with the appraisal
of staining intensity being highly subjective. Furthermore,
the choice of antibodies and IHC protocol is not consistent
and may cause the sensitivity of these assays to vary.
However, an advantage of this method is that information
on the cellular distribution of EGFR is obtained. In addition
to total EGFR levels, activated (phosphorylated) EGFR has
been detected in human skin keratinocytes by IHC using
phosphospecific EGFR antibodies. Interestingly, the basal
level of phosphorylated EGFR was eliminated by treatment
with the EGFR-TKI ZD1839 [13]. Similar studies in human
tumors, either untreated or treated with EGFR inhibitors,
have not been reported.
EGFR protein levels may also be quantified by Western
analysis [14] or enzyme immunoassay (EIA) [15], which
measure total receptor protein in tumor specimens, regardless of the expressing cell type and cellular localization of
the receptor. EIA has been used to analyze EGFR in serum
samples from patients with breast cancer. The serum EGFR
level was 7-162 fmol/ml and 126-1,587 fmol/ml in healthy
controls and women with breast cancer, respectively. This
study noted that 67.5% of patients had elevated levels of circulating EGFR, using a cut-off value of 180 fmol/ml [15].
Assessment of the levels of EGF binding in tumors has also
been reported but considerable inter-assay variability has
been observed [16].
Levels of the EGFR RNA transcript, which do not necessarily reflect the levels of protein that will be produced, can be
assessed by Northern analysis and reverse transcriptase-polymerase chain reaction (RT-PCR). Analysis of DNA through a
33
34
Relationship Between EGFR Expression and Activity of Agents
Targeting EGFR
Sensitivity to anti-EGFR agents might not simply
depend on the number of EGFRs. Taking the EGFR-TKI
ZD1839 as an example, some studies have demonstrated a
relationship between relative EGFR expression and activity
of ZD1839 [29, 30], whereas others have reported no such
effect [31-33]. These studies involved cell lines derived
from a variety of carcinomas. Meye et al. [29] investigated
the effect of ZD1839 in four bladder cancer cell lines, each
expressing a different level of EGFR. The concentration
required to inhibit ligand-independent growth 50% (IC50)
ranged from 1.8 to 9.7 µM in these cell lines and correlated
with EGFR protein and transcript level. Similarly, Janmaat
et al. [30] found a correlation between EGFR expression
and ZD1839-induced growth inhibition in cell lines derived
from vulval squamous-cell carcinoma (A431) and NSCLC.
However, the linear correlation was less pronounced within
the series of NSCLC cell lines. Similar levels of growth
inhibition were achieved in two cell lines derived from head
and neck squamous-cell carcinoma and melanoma, despite
their different levels of EGFR expression [31]. In a study
using human tumor xenografts, ZD1839 caused growth
inhibition of tumors and markedly enhanced the activity of
a number of cytotoxic agents, but, interestingly, neither the
growth inhibition nor the degree of potentiation of
chemotherapy were dependent on high levels of EGFR
expression [32, 33].
Clinical studies have demonstrated activity of EGFRtargeted agents in patients who were not recruited on the
basis of their tumor EGFR expression. Patients in phase I
and II studies with ZD1839 were not selected on the basis
of EGFR levels [34]. Moasser et al. [35] reported in vitro
data with ZD1839 against a panel of breast cancer cell lines
with a wide range of EGFR levels, further suggesting that
high EGFR expression does not dictate sensitivity to
ZD1839 [35]. In contrast, trials of the small-molecule
EGFR-TKI OSI-774 and the monoclonal antibody C225
have been carried out in patients selected as EGFR-positive
by IHC [36, 37]. The phase II trial of OSI-774 found that
objective tumor response and stable disease were not associated with more EGFR-positive cells in tumor sections or
more intense EGFR staining [36].
Alternative Mechanisms by which EGFR Drive Is Increased
In addition to high expression of EGFR as a mechanism
for increased receptor signaling output, this transduction
pathway can be upregulated via alternative mechanisms
including activating EGFR mutations, increased coexpression of receptor ligands, and heterodimerization with HER2
as well as with heterologous receptor systems (Fig. 2) [38].
Increased (Autocrine) Receptor Ligands
EGFR signals may be enhanced by increased levels of
receptor ligands (such as EGF, amphiregulin, or transforming growth factor-α [TGF-α]). Coexpression of
EGFR and one or more of its ligands might result in activation of an autocrine system leading to dysregulated
EGFR action and uncontrolled tumor growth [1, 39-42].
Indeed, in a study of 173 patients with invasive ductal
breast carcinoma, a multivariate analysis found that coexpression of EGFR and TGF-α had the most significant
effect on survival compared, for example, with coexpression of EGFR and HER2 [43]. EGF expression has also
been correlated with poor prognosis in patients with breast
cancer [44]. Among EGFR-positive primary lung adenocarcinomas, overall survival was significantly worse for
patients with high levels of expression of EGF or TGF-α
compared with EGFR-positive cancers that were EGF- and
TGF-α negative [45].
Heterodimerization with HER2 and Cross-Talk with
Heterologous Receptors
Homodimers of EGFR signal weakly, in part due to
receptor downregulation and degradation after ligand-mediated activation [2]. The EGFR-homologous HER2 receptor,
which is highly expressed in several human cancers, can
potentiate EGFR function by increasing EGF binding affinity, stabilizing and recycling EGFR-HER2 heterodimers,
and expanding the repertoire of receptor-associated substrates and signaling responses [46, 47]. Cancers with high
expression of either EGFR or HER2 have a better prognosis
than cancers that have high expression of both receptors
Downloaded from www.TheOncologist.com by on January 16, 2010
expression of the receptor predicted for a lower response
rate to tamoxifen (p = 0.046) [4]. However, other reports,
such as the study of tamoxifen therapy in estrogen-receptor-positive breast cancer patients [23], have not supported
the predictive value of EGFR status for antiestrogen resistance. There is increasing evidence demonstrating that
growth factor pathways are highly interactive with estrogen
receptor signaling in the control of breast cancer growth
[24]. In tamoxifen-resistant breast cancer cell lines, antiestrogenic resistance is associated with upregulation of the
EGFR pathway [25]. Hence, EGFR-TKIs such as ZD1839
have the potential to treat endocrine-resistant tumors and
might abrogate acquired resistance when used early in
combination with antiestrogens [25, 26]. High expression
of EGFR has also been associated with resistance to radiotherapy [3], and recent studies have confirmed the capacity
of EGFR downregulation to modify the cellular response to
radiation [27, 28].
EGFR Dependence in Cancer: More Than Just Expression?
Arteaga
35
1
Increased expression
of EGFR protein
2
R
R
R
pY
pY
pY
pY
K
K
K
K
K
5
Ligand/
autocrine
loop
S
K
pY
S
pY
pY
pY
Mutant
EGFR
R
S
S
pY pY
K
R
Mitogenic
signals
pY
R
R
K
pY pY
pY
K
pY pY
pY
pY
pY
S
S
pY
TGF-α
EGF
R
R
pY
K
pY
pY
K
K
Phosphatase
K
K
K
R R
R
R
pY
S S
pY
pY
3
Heterodimerization
and cross-talk
Figure 2. Multiple mechanisms of increased EGFR activation. Abbreviations: EGF = epidermal growth factor; R = receptor; S = substrate;
TGF-α = transforming growth factor-α; K = tyrosine kinase; pY = phosphorylated tyrosine residue.
[48-51]. In 83 patients with resected NSCLC, coexpression
of both EGFR and HER2 was a better predictor of treatment
failure and poor survival than EGFR expression alone [52].
Furthermore, high expression of HER2 can counteract the
efficacy of EGFR-TKIs in EGFR-expressing tumor cells
[53]. Finally, EGFR-TKIs block HER2 phosphorylation by
inhibiting EGFR-mediated transactivation of HER2 in
tumor cells that highly express HER2 [35, 54, 55]. Taken
together, these studies strongly support EGFR-HER2 crosstalk in vivo, and high expression of HER2 as a mechanism
that can potentiate EGFR signals and EGFR-mediated
tumor progression.
In addition, the EGFR can cross-talk with heterologous
receptors activated by neurotransmitters, lymphokines, and
stress inducers [56-58]. G-protein-coupled receptors
(GPCRs) can exert positive effects on EGFR signaling in
several ways, including the activation of matrix metalloproteinases, which cleave membrane-tethered EGFR ligands that can then bind and activate EGFR [2]. In addition,
GPCRs indirectly activate Src, which can phosphorylate the
EGFR at tyrosines other than those autophosphorylated by
EGFR tyrosine kinase [57]. Steroid hormones can also influence EGFR signaling by activating the transcription of genes
encoding EGFR ligands (Fig. 3). Conversely, estrogen can
transactivate EGFR via the GPCR GPR30, potentially
explaining the EGF-like effect of estrogen [59].
EGFR Mutations
The best described and most common EGFR mutation,
EGFRvIII, involves deletion of exons 2 to 7 and loss of
residues 6 to 276 in the receptor’s ectodomain. This mutant
yields a constitutively active receptor that is not downregulated by endocytosis and is potently transforming [60]. This
mutant receptor is detected in 40% of high-grade gliomas,
where it frequently exhibits gene amplification, and less
frequently in NSCLC, breast, and ovarian cancers [61-63].
ZD1839 has been shown to inhibit autophosphorylation of
EGFRvIII in NR6M cells in culture [64]. In principle,
EGFR antibodies raised against extracellular receptor epitopes deleted in this mutant would not be expected to have
activity in this setting.
Assessment of Operative EGFR Signaling
Further research is required to assess the link between
EGFR signaling in situ (rather than EGFR expression
alone) and the prediction of both patient outcome and
response to anti-EGFR therapies. These investigations
should include the assessment of EGFR phosphorylation in
Downloaded from www.TheOncologist.com by on January 16, 2010
pY
pY K
K
pY
pY
pY
R R
4
K
pY
pY
S
R
K
R
S
K
EGFR Dependence in Cancer: More Than Just Expression?
36
LPA, thrombin,
ET, etc.
Steroid
hormone
HB-EGF
+
α
β γ
Inactive
erbB
MMP
G protein
P
P
+
+
Ca2+
Pyk2
Src
Ras
MAPK
Transcription
erbB ligand gene
Figure 3. Cross-talk of EGFR with G-protein coupled receptors and steroid receptors [2]. Reprinted by permission from Nature Reviews
Molecular Cell Biology 2001;2:127-137. ©2001 Macmillan Magazines Ltd.
vivo and its biochemical response as a function of treatment
with receptor-targeted therapies. Evaluation of markers
downstream from EGFR (e.g., MAPK, PI3K/Akt, the proliferation marker Ki67) might also give an indication of surrogate markers of EGFR inactivation and its association
with response or lack of response to treatment.
Studies of the pharmacodynamic effects of ZD1839 can be
used to determine optimal biologic doses to be used in clinical
trials. A study of ZD1839 given over a 28-day period to cancer
patients assessed a range of signaling, proliferation, and maturation markers in patients’ skin, a highly EGFR-expressing tissue. End points indicative of EGFR inactivation in the skin were
found at all dose levels in the range 150-1,000 mg/day [13]. In
part due to this observation, two doses of ZD1839 (250 and
500 mg/day) well below the maximum tolerated dose were chosen for phase II efficacy trials. These trials reported good tolerability and clinical activity of both doses of ZD1839 in patients
with NSCLC [65-66]. Preliminary results have shown that evaluation of these markers is feasible in serial tumor biopsies [67].
Measurement of proliferation (by Ki67 IHC) and levels of
apoptosis (by TUNEL) gave the most reproducible results and
the best indication of ZD1839 activity. Early evidence suggests
that ZD1839 can inhibit EGFR signaling in the vicinity of
tumors and may increase levels of tumor cell apoptosis [67].
CONCLUSIONS
The prognostic significance of EGFR expression in cancer
and, more importantly, its ability to predict response to antiEGFR therapies, are subjects of active research. Increased levels of receptor ligands, coexpression of EGFR mutants, and
cross-talk with HER2 or other receptors are mechanisms that
can enhance EGFR signaling output and potentially alter the
response to EGFR inhibitors. These factors will certainly modify the treatment-predictive value of future EGFR-based assays.
Clearly, the current methods to measure EGFR levels in tumors
are not quantitative and cannot be endorsed as predictive of
either patient prognosis or response to treatment. Thus, until
EGFR (or other) assays that can predict response to anti-EGFR
therapies are available, there is no compelling reason to exclude
patients with low or negative EGFR levels from trials with
EGFR inhibitors, as long as there is evidence for EGFR expression in the tumor type(s) tested by these trials. By the same
token, it is essential that tissues/blocks from all patients enrolled
in these trials are saved and retrospectively examined with
quantitative EGFR assays for correlation of these data with
clinical response to EGFR inhibitors.
ACKNOWLEDGMENT
At the time of publication, this paper discusses the unlabeled usage of ZD1839 (Iressa).
Downloaded from www.TheOncologist.com by on January 16, 2010
Steroid
hormone
receptor
Arteaga
37
R EFERENCES
1 Salomon DS, Brandt R, Ciardiello F et al. Epidermal growth
factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol 1995;19:183-232.
2 Yarden Y, Sliwkowski MX. Untangling the ErbB signalling
network. Nat Rev Mol Cell Biol 2001;2:127-137.
3 Sartor CI. Biological modifiers as potential radiosensitizers:
targeting the epidermal growth factor receptor family. Semin
Oncol 2000;27(suppl 11):15-20.
4 Newby JC, Johnston SR, Smith IE et al. Expression of epidermal
growth factor receptor and c-erbB2 during the development of
tamoxifen resistance in human breast cancer. Clin Cancer Res
1997;3:1643-1651.
6 Klijn JG, Look MP, Portengen H et al. The prognostic value of
epidermal growth factor receptor (EGF-R) in primary breast
cancer: results of a 10 year follow-up study. Breast Cancer Res
Treat 1994;29:73-83.
7 Fischer-Colbrie J, Witt A, Heinzl H et al. EGFR and steroid
receptors in ovarian carcinoma: comparison with prognostic
parameters and outcome of patients. Anticancer Res
1997;17:613-619.
8 Magne N, Pivot X, Bensadoun RJ et al. The relationship of
epidermal growth factor receptor levels to the prognosis of
unresectable pharyngeal cancer patients treated by chemoradiotherapy. Eur J Cancer 2001;37:2169-2177.
18 Poon TCW, Chan ATC, To KF et al. Expression and prognostic significance of epidermal growth factor receptor and
HER2 protein in nasopharyngeal carcinoma. Proc Am Soc
Clin Oncol 2001;20:229a (A913).
19 Neskovic-Konstantinovic Z, Nikolic-Vukosavljevic D,
Brankovic-Magic M et al. Expression of epidermal growth
factor receptor in breast cancer, from early stages to advanced
disease. J Exp Clin Cancer Res 1999;18:347-355.
20 Tsutsui S, Ohno S, Murakami S et al. Prognostic value of epidermal growth factor receptor (EGFR) and its relationship to
the estrogen receptor status in 1029 patients with breast cancer.
Breast Cancer Res Treat 2002;71:67-75.
21 Ohsaki Y, Tanno S, Fujita Y et al. Epidermal growth factor
receptor expression correlates with poor prognosis in nonsmall cell lung cancer patients with p53 overexpression. Oncol
Rep 2000;7:603-607.
22 Volm M, Rittgen W, Drings P. Prognostic value of ERBB-1,
VEGF, cyclin A, FOS, JUN and MYC in patients with squamous cell lung carcinomas. Br J Cancer 1998;77:663-669.
23 Knoop AS, Bentzen SM, Nielsen MM et al. Value of epidermal
growth factor receptor, HER2, p53, and steroid receptors in predicting the efficacy of tamoxifen in high-risk postmenopausal
breast cancer patients. J Clin Oncol 2001;19:3376-3384.
9 Baselga J. New technologies in epidermal growth factor
receptor-targeted cancer therapy. Signal 2000;1:12-21.
24 Kurokawa H, Arteaga CL. Inhibition of erbB receptor (HER)
tyrosine kinases as a strategy to abrogate antiestrogen resistance
in human breast cancer. Clin Cancer Res 2001;7(suppl
12):4436-4442.
10 Rowinsky EK. The pursuit of optimal outcomes in cancer
therapy in a new age of rationally designed target-based anticancer agents. Drugs 2000;60(suppl 1):1-14.
25 Wakeling AE, Nicholson RI, Gee JM. Prospects for combining hormonal and nonhormonal growth factor inhibition. Clin
Cancer Res 2001;7(suppl 12):4350-4355.
11 Vogel CL, Cobleigh MA, Tripathy D et al. Efficacy and
safety of trastuzumab as a single agent in first-line treatment
of HER2-overexpressing metastatic breast cancer. J Clin
Oncol 2002;20:719-726.
26 Nicholson RI, Hutcheson IR, Harper ME et al. Modulation of
epidermal growth factor receptor in endocrine-resistant, oestrogen receptor-positive breast cancer. Endocr Relat Cancer
2001;8:175-182.
12 Birner P, Oberhuber G, Stani J et al. Evaluation of the United
States Food and Drug Administration-approved scoring and
test system of HER-2 protein expression in breast cancer.
Clin Cancer Res 2001;7:1669-1675.
27 Harari PM, Huang SM. Radiation response modification following molecular inhibition of epidermal growth factor
receptor signaling. Semin Radiat Oncol 2001;11:281-289.
13 Albanell J, Rojo F, Averbuch S et al. Pharmacodynamic studies
of the epidermal growth factor receptor inhibitor ZD1839 in
skin from cancer patients: histopathologic and molecular consequences of receptor inhibition. J Clin Oncol 2002;20:110-124.
14 Mandic R, Eikelkamp N, Peldszus R et al. Variations of EGFR surface expression in squamous cell carcinomas of the head
and neck region. Anticancer Res 2001;21:3413-3418.
15 Kumar RR, Meenakshi A, Sivakumar N. Enzyme immunoassay of human epidermal growth factor receptor (hEGFR).
Hum Antibodies 2001;10:143-147.
16 Robertson KW, Reeves JR, Lannigan AK et al. Radioimmunohistochemistry of epidermal growth factor receptor in breast
cancer. Arch Pathol Lab Med 2002;126:177-181.
28 Corvo R, Antognoni P, Sanguineti G. Biological predictors of
response to radiotherapy in head and neck cancer: recent
advances and emerging perspectives. Tumori 2001;87:355-363.
29 Meye A, Fiedler U, Kunert K et al. Growth inhibitory effects
of ZD1839 (‘Iressa’) on human bladder cancer cell lines. Proc
Am Assoc Cancer Res 2001;42:805 (A4320).
30 Janmaat ML, Kruyt FAE, Rodriguez JA et al. Inhibition of
the epidermal growth factor receptor induces apoptosis in
A431 cells but not in non-small-cell lung cancer cell lines.
Proc Am Assoc Cancer Res 2002;43:A3901.
31 Budillon A, Guarrasi R, Di Gennaro E et al. ZD1839
(‘Iressa’), an EGFR tyrosine kinase inhibitor, potentiates nonMHC restricted cytotoxicity in human cancer cell lines. Proc
Am Assoc Cancer Res 2001;42:A4306.
Downloaded from www.TheOncologist.com by on January 16, 2010
5 Chen X, Yeung TK, Wang Z. Enhanced drug resistance in cells
coexpressing ErbB2 with EGF receptor or ErbB3. Biochem
Biophys Res Commun 2000;277:757-763.
17 Nicholson RI, Gee JM, Harper ME. EGFR and cancer prognosis.
Eur J Cancer 2001;37(suppl 4):S9-S15.
38
32 Sirotnak FM, Zakowski MF, Miller VA et al. Efficacy of cytotoxic agents against human tumor xenografts is markedly
enhanced by coadministration of ZD1839 (Iressa), an inhibitor
of EGFR tyrosine kinase. Clin Cancer Res 2000;6:4885-4892.
33 Ciardiello F, Caputo R, Bianco R et al. Antitumor effect and
potentiation of cytotoxic drugs activity in human cancer cells by
ZD-1839 (‘Iressa’), an epidermal growth factor receptor-selective tyrosine kinase inhibitor. Clin Cancer Res 2000;6:20532063.
34 Ranson M, Hammond LA, Ferry D et al. ZD1839, a selective
oral epidermal growth factor receptor-tyrosine kinase
inhibitor, is well tolerated and active in patients with solid,
malignant tumors: results of a phase I trial. J Clin Oncol
2002;20:2240-2250.
36 Perez-Soler R, Chachoua A, Huberman M et al. A Phase II trial
of the epidermal growth factor receptor (EGFR) tyrosine kinase
inhibitor OSI-774, following platinum-based chemotherapy, in
patients (pts) with advanced, EGFR-expressing, non-small cell
lung cancer (NSCLC). Proc Am Soc Clin Oncol 2001;20:310a
(A1235).
37 Baselga J, Pfister D, Cooper MR et al. Phase I studies of antiepidermal growth factor receptor chimeric antibody C225
alone and in combination with cisplatin. J Clin Oncol
2000;18:904-914.
38 Voldborg BR, Damstrup L, Spang-Thomsen M et al.
Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol
1997;8:1197-1206.
39 Gorgoulis V, Aninos D, Mikou P et al. Expression of EGF,
TGF-α and EGFR in squamous cell lung carcinomas.
Anticancer Res 1992;12:1183-1187.
40 Cohen DW, Simak R, Fair WR et al. Expression of transforming growth factor-alpha and the epidermal growth factor receptor in human prostate tissues. J Urol 1994;152:2120-2124.
41 Rusch V, Klimstra D, Venkatraman E et al. Overexpression
of the epidermal growth factor receptor and its ligand transforming growth factor alpha is frequent in resectable nonsmall cell lung cancer but does not predict tumor progression.
Clin Cancer Res 1997;3:515-522.
42 Hsieh ET, Shepherd FA, Tsao MS. Co-expression of epidermal
growth factor receptor and transforming growth factor-alpha is
independent of ras mutations in lung adenocarcinoma. Lung
Cancer 2000;29:151-157.
43 Umekita Y, Ohi Y, Sagara Y et al. Co-expression of epidermal growth factor receptor and transforming growth factoralpha predicts worse prognosis in breast-cancer patients. Int J
Cancer 2000;89:484-487.
44 Mizukami Y, Nonomura A, Noguchi M et al. Immunohistochemical study of oncogene product ras p21, c-myc and growth
factor EGF in breast carcinomas. Anticancer Res 1991;11:14851494.
45 Tateishi M, Ishida T, Mitsudomi T et al. Immunohistochemical evidence of autocrine growth factors in adenocarcinoma of the human lung. Cancer Res 1990;50:7077-7080.
46 Karunagaran D, Tzahar E, Beerli RR et al. ErbB-2 is a common
auxiliary subunit of NDF and EGF receptors: implications for
breast cancer. EMBO J 1996;15:254-264.
47 Worthylake R, Opresko LK, Wiley HS. ErbB-2 amplification
inhibits down-regulation and induces constitutive activation
of both ErbB-2 and epidermal growth factor receptors. J Biol
Chem 1999;274:8865-8874.
48 Tateishi M, Ishida T, Kohdono S et al. Prognostic influence
of the co-expression of epidermal growth factor receptor and
c-erbB-2 protein in human lung adenocarcinoma. Surg Oncol
1994;3:109-113.
49 Xia W, Lau YK, Zhang HZ et al. Combination of EGFR,
HER-2/neu, and HER-3 is a stronger predictor for the outcome of oral squamous cell carcinoma than any individual
family members. Clin Cancer Res 1999;5:4164-4174.
50 Nicholson RI, McClelland RA, Finlay P et al. Relationship
between EGF-R, c-erbB-2 protein expression and Ki67
immunostaining in breast cancer and hormone sensitivity.
Eur J Cancer 1993;29A:1018-1023.
51 Iwase H, Itoh Y, Kuzushima T et al. Simultaneous quantitative
analyses of c-erbB-2 protein, epidermal growth factor receptor,
cathepsin D, and hormone receptors in breast cancer. Cancer
Detect Prev 1997;21:29-35.
52 Brabender J, Danenberg KD, Metzger R et al. Epidermal
growth factor receptor and HER2-neu mRNA expression in
non-small cell lung cancer is correlated with survival. Clin
Cancer Res 2001;7:1850-1855.
53 Christensen JG, Schreck RE, Chan E et al. High levels of HER2 expression alter the ability of epidermal growth factor receptor (EGFR) family tyrosine kinase inhibitors to inhibit EGFR
phosphorylation in vivo. Clin Cancer Res 2001;7:4230-4238.
54 Moulder SL, Yakes FM, Muthuswamy SK et al. Epidermal
growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839
(Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer
cells in vitro and in vivo. Cancer Res 2001;61:8887-8895.
55 Anderson NG, Ahmad T, Chan K et al. ZD1839 (Iressa), a
novel epidermal growth factor receptor (EGFR) tyrosine
kinase inhibitor, potently inhibits the growth of EGFR-positive cancer cell lines with or without erbB2 overexpression.
Int J Cancer 2001;94:774-782.
56 Prenzel N, Zwick E, Leserer M et al. Tyrosine kinase signalling in breast cancer. Epidermal growth factor receptor:
convergence point for signal integration and diversification.
Breast Cancer Res 2000;2:184-190.
57 Carpenter G. Employment of the epidermal growth factor
receptor in growth factor-independent signaling pathways. J
Cell Biol 1999;146:697-702.
58 Gschwind A, Zwick E, Prenzel N et al. Cell communication
networks: epidermal growth factor receptor transactivation as
the paradigm for interreceptor signal transmission. Oncogene
2001;20:1594-1600.
59 Filardo EJ. Epidermal growth factor receptor (EGFR) transactivation by estrogen via the G-protein-coupled receptor, GPR30:
a novel signaling pathway with potential significance for breast
cancer. J Steroid Biochem Mol Biol 2002;80:231-238.
Downloaded from www.TheOncologist.com by on January 16, 2010
35 Moasser MM, Basso A, Averbuch SD et al. The tyrosine
kinase inhibitor ZD1839 (“Iressa”) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing
tumor cells. Cancer Res 2001;61:7184-7188.
EGFR Dependence in Cancer: More Than Just Expression?
Arteaga
60 Nishikawa R, Ji XD, Harmon RC et al. A mutant epidermal
growth factor receptor common in human glioma confers
enhanced tumorigenicity. Proc Natl Acad Sci USA 1994;91:77277731.
61 Moscatello DK, Holgado-Madruga M, Godwin AK et al.
Frequent expression of a mutant epidermal growth factor receptor in multiple human tumors. Cancer Res 1995;55:5536-5539.
62 Wikstrand CJ, Hale LP, Batra SK et al. Monoclonal antibodies against EGFRvIII are tumor specific and react with breast
and lung carcinomas and malignant gliomas. Cancer Res
1995;55:3140-3148.
63 Sauter G, Maeda T, Waldman FM et al. Patterns of epidermal
growth factor receptor amplification in malignant gliomas.
Am J Pathol 1996;148:1047-1053.
64 Learn CA, Archer GE, Heimberger AH et al. Oral administration of the specific epidermal growth factor receptor tyrosine
39
kinase inhibitor (EGFR-TKI) ZD1839 (Iressa) is efficacious
against EGFR-overexpressing intracranial tumors. Proc Am
Assoc Cancer Res 2002;43:A3890.
65 Fukuoka M, Yano S, Giaccone G et al. Final results from a Phase
II trial of ZD1839 (‘Iressa’) for patients with advanced nonsmall-cell lung cancer (IDEAL 1). Proc Am Soc Clin Oncol
2002;21:298a (A1188).
66 Kris MG, Natale RB, Herbst RS et al. A Phase II trial of ZD1839
(‘Iressa’) in advanced non-small-cell lung cancer (NSCLC)
patients who had failed platinum- and docetaxel-based regimens
(IDEAL 2). Proc Am Soc Clin Oncol 2002;21:292a (A1166).
67 Lorimer I, Parolin D, Robertson S et al. NCIC CTG IND.122:
immunohistochemical analysis of tumor biopsy specimens
from patients undergoing treatment with ZD1839. Clin
Cancer Res 2001;7(suppl):A277.
Downloaded from www.TheOncologist.com by on January 16, 2010
Epidermal Growth Factor Receptor Dependence in Human Tumors: More Than
Just Expression?
Carlos L. Arteaga
Oncologist 2002;7;31-39
DOI: 10.1634/theoncologist.7-suppl_4-31
This information is current as of January 16, 2010
Updated Information
& Services
including high-resolution figures, can be found at:
http://www.TheOncologist.com/cgi/content/full/7/suppl_4/31
Related Articles
A related article has been published:
http://www.TheOncologist.com/cgi/content/full/8/1/124-a
Downloaded from www.TheOncologist.com by on January 16, 2010