ArticlePDF Available

Insulin-Like Growth Factor-1 Receptor Inhibition Induces a Resistance Mechanism via the Epidermal Growth Factor Receptor/HER3/AKT Signaling Pathway: Rational Basis for Cotargeting Insulin-Like Growth Factor-1 Receptor and Epidermal Growth Factor Receptor in Hepatocellular Carcinoma

Authors:

Abstract and Figures

The insulin-like growth factor (IGF) signaling axis is frequently dysregulated in hepatocellular carcinoma (HCC). Therefore, we investigated whether the specific targeting of the IGF type 1 receptor (IGF-1R) might represent a new therapeutic approach for this tumor. Total and phosphorylated levels of IGF-1R were measured in 21 paired samples of human HCCs and adjacent nontumoral livers using ELISA. The antineoplastic potency of a novel anti-IGF-1R antibody, AVE1642, was examined in five human hepatoma cell lines. Overexpression of IGF-1R was detected in 33% of HCCs and increased activation of IGF-1R was observed in 52% of tumors. AVE1642 alone had moderate inhibitory effects on cell viability. However, its combination with gefitinib, an epidermal growth factor receptor (EGFR) inhibitor, induced supra-additive effects in all cell lines that were associated with cell cycle blockage and inhibition of AKT phosphorylation. The combination of AVE1642 with rapamycin also induced a synergistic reduction of viability and of AKT phosphorylation. Of marked interest, AVE1642 alone up-regulated the phosphorylated and total levels of HER3, the main partner of EGFR, and AVE1642-induced phosphorylation of HER3 was prevented by gefitinib. Moreover, the down-regulation of HER3 expression with siRNA reduced AKT phosphorylation and increased cell sensitivity to AVE1642. These findings indicate that hepatoma cells overcome IGF-1R inhibition through HER3 activation in an EGFR-dependent mechanism, and that HER3 represents a critical mediator in acquired resistance to anti-IGF-1R therapy. These results provide a strong rational for targeting simultaneously EGFR and IGF-1R in clinical trials for HCC].
Content may be subject to copyright.
Cancer Therapy: Preclinical
Insulin-Like Growth Factor-1 Receptor Inhibition Induces a
Resistance Mechanism via the Epidermal Growth Factor
Receptor/HER3/AKT Signaling Pathway: Rational Basis for
Cotargeting Insulin-Like Growth Factor-1 Receptor and
Epidermal Growth Factor Receptor in Hepatocellular Carcinoma
Christèle Desbois-Mouthon,
1,2
Aurore Baron,
1,2
Marie-José Blivet-Van Eggelpoël,
1,2
Laetitia Fartoux,
1,2,3
Corinne Venot,
4
Friedhelm Bladt,
4
Chantal Housset,
1,2
and Olivier Rosmorduc
1,2,3
Abstract Purpose: The insulin-like growth factor (IGF) signaling axis is frequently dysregulated in
hepatocellular carcinoma (HCC). Therefore, we investigated whether the specific target-
ing of the IGF type 1 receptor (IGF-1R) might represent a new therapeutic approach for
this tumor.
Experimental Design: Total and phosphorylated levels of IGF-1R were measured in 21
paired samples of human HCCs and adjacent nontumoral livers using ELISA. The anti-
neoplastic potency of a novel antiIGF-1R antibody, AVE1642, was examined in five hu-
man hepatoma cell lines.
Results: Overexpression of IGF-1R was detected in 33% of HCCs and increased activation
of IGF-1R was observed in 52% of tumors. AVE1642 alone had moderate inhibitory ef-
fects on cell viability. However, its combination with gefitinib, an epidermal growth factor
receptor (EGFR) inhibitor, induced supra-additive effects in all cell lines that were asso-
ciated with cell cycle blockage and inhibition of AKT phosphorylation. The combination
of AVE1642 with rapamycin also induced a synergistic reduction of viability and of AKT
phosphorylation. Of marked interest, AVE1642 alone up-regulated the phosphorylated
and total levels of HER3, the main partner of EGFR, and AVE1642-induced phosphoryla-
tion of HER3 was prevented by gefitinib. Moreover, the down-regulation of HER3 expres-
sion with siRNA reduced AKT phosphorylation and increased cell sensitivity to AVE1642.
Conclusions: These findings indicate that hepatoma cells overcome IGF-1R inhibition
through HER3 activation in an EGFR-dependent mechanism, and that HER3 represents
a critical mediator in acquired resistance to anti-IGF-1R therapy. These results provide a
strong rational for targeting simultaneously EGFR and IGF-1R in clinical trials for HCC].
(Clin Cancer Res 2009;15(17):544556)
Hepatocellular carcinoma (HCC) is the fifth most common
malignancy and the third most common cause of mortality
worldwide. Its incidence is increasing in Western countries,
mainly due to hepatitis C virus and excessive alcohol consump-
tion. At the time of diagnosis, most of the patients suffer ad-
vanced tumor disease and a curative treatment is possible
only in a minority of patients and overall survival is poor (1).
In this context, the recent development of molecular therapies
targeting specific receptors and/or signaling proteins has
opened promising perspectives for the treatment of advanced
HCC (2, 3). However, until now, the targeting of vascular en-
dothelial growth factor or epidermal growth factor receptor
(EGFR) has shown disappointing results in HCC treatment
(4) leading to the concept of multitarget therapies. This concept
has been recently validated by the approval of sorafenib, a multi-
kinase inhibitor, as a reference treatment of advanced HCC (5).
Authors' Affiliations:
1
UPMC Univ Paris 06;
2
Institut National de la Santé et de
la Recherche Médicale, UMR_S 938; and
3
AP-HP,Hôpital Saint-Antoine, Service
d'Hépatologie, Paris, France; and
4
Sanofi-aventis, Vitry-sur-Seine, France
Received 11/18/08; revised 6/19/09; accepted 6/22/09; published OnlineFirst
8/25/09.
Grant support: Association pour la Recherche sur le Cancer (O. Rosmorduc),
sanofi-aventis (O. Rosmorduc and C.D. Mouthon), and Académie Nationale
de Médecine (A. Baron).
The costs of publication of this article were defrayed in part by the payment of
page charges. This article must therefore be hereby marked advertisement
in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Note: Supplementary data for this article are available at Clinical Cancer
Research Online (http://clincancerres.aacrjournals.org/).
Requests for reprints: Christèle Desbois-Mouthon, Institut National de la Santé
et de la Recherche Médicale UMR_S 938, Faculté de Médecine Pierre et Marie
Curie, Site Saint-Antoine, 27 rue Chaligny, 75012, Paris, France. Phone: 33-1-40-
01-13-56; Fax: 33-1-40-01-14-26; E-mail: christele.desbois-mouthon@inserm.fr.
F2009 American Association for Cancer Research.
doi:10.1158/1078-0432.CCR-08-2980
5445 Clin Cancer Res 2009;15(17) September 1, 2009www.aacrjournals.org
Evidence has accumulated showing that the insulin-like
growth factor (IGF) signaling axis is activated in HCC (6, 7):
IGF-II may be overexpressed as a result of loss of promoter-
specific imprinting and reactivation of fetal promoters; increased
amounts of bioactive IGF-II are also the result of a reduced ex-
pression of IGF binding protein and/or inactivation of the
type 2 IGF receptor, which mediates IGF-II degradation. IGF-II
promotes proliferation, survival, and migration in hepatoma
cells through binding to its receptor IGF type 1 receptor (IGF-1R;
refs. 810). This receptor as well as its main substrates IRS-1 and
IRS-2 may be also overexpressed in HCC and in experimental
models of liver carcinogenesis (1114).
The potential interest of targeting IGF-1Rmediated signaling
pathways in HCC has been suggested both in vitro and in vivo:
first, the blockade of IGF-II overexpression impairs HCC devel-
opment in murine models (15, 16), and second, inhibition of
IGF-1R with a monoclonal antibody (mAb; αIR-3;ref.17)or
with tyrosine kinase inhibitors (TKI; AG1024, NVP-AEW541) re-
duces HCC cell proliferation and/or increases apoptosis (9, 18).
However, we and others recently reported that the combination
of AG1024 with an EGFR TKI (erlotinib, gefitinib) resulted in
synergistic antineoplastic effects in hepatoma cell lines (9, 19),
suggesting that the blockage of either EGFR or IGF-1R may allow
the other receptor to compensate for.
Having shown that IGF-1R was overexpressed or hyperacti-
vated in human HCC tumors, the study was then designed to
examine the antineoplastic effects of cotargeting IGF-1R and
EGFR in HCC cells by using a novel mAb against IGF-1R
(AVE1642), currently in phase I/II clinical trials (20), andgefitinib.
Although AVE1642 alone had moderate inhibitory effects on
cell viability, we observed that its combination with gefitinib ex-
erted supra-additive effects in the five cell lines tested. We char-
acterized the molecular mechanisms of this synergy by showing
that (a) both the total and the phosphorylated levels of HER3
(erbB-3), the main partner of EGFR, were up-regulated in the
presence of AVE1642; (b) gefitinib reduced the phosphoryla-
tion of HER3 induced by AVE1642, which was paralleled by a
marked inhibition of AKT phosphorylation; and (c)thedown-
regulation of HER3 expression using a siRNA strategy also reduced
AKT phosphorylation and significantly increased hepatoma
cell sensitivity to AVE1642. These data highlight an original
compensatory mechanism between IGF-1R and EGFR signaling
pathways in hepatoma cells and provide a strong rational ba-
sis for targeting simultaneously EGFR and IGF-1R in advanced
HCC to prevent the early development of EGFR-mediated
resistance.
Materials and Methods
Reagents. AVE1642, a humanized version of the murine mAb EM164
(21), was provided by sanofi-aventis/Immunogen; cetuximab (Erbitux) was
purchased from Merck KGaA; and gefitinib was provided by AstraZeneca.
Bovine insulin and human recombinant IGF-I and IGF-II were purchased
from Sigma-Aldrich.
Cell culture. HepG2 (HBs
neg
,Rb
wt
,p53
wt
,β-catenin
mut
), Hep3B
(HBs
pos
, undetected Rb, deleted p53, β-catenin
wt
), and HuH7 (HBs
neg
,
Rb
wt
,p53
mut
,β-catenin
wt
) cells were from the American Type Culture Col-
lection.HuH6 (HBs
neg
,p53
wt
,β-catenin
mut
), and PLC/P RF5 (HBs
pos
,Rb
wt
,
p53
mut
, axin
mut
) were provided by Dr Christine Perret (Institut Cochin,
Paris, France). Normal human hepatocytes were isolated from liver tissue
obtained from patients undergoing partial liver resection for liver metasta-
sis, and primary cultures were established as previously described (22).
Pools of HCC cells resistant to gefitinib were generated as previously re-
ported in nonsmall cell lung carcinoma cell lines (23). Briefly, HuH6
and HepG2 cell lines were continuously exposed to gefitinib (10 μmol/L)
in routine culture medium that was replaced every 4 d. Initially, cell
number was dramatically reduced, and for the next 2 mo, the surviving
cells were passaged approximatively every 15 d. Cell proliferation slowly
increased the next 2 mo and a stable growth rate was reached after a
total of 5 mo.
Western blotting and ELISA. The following antibodies were used to
detect electrotransferred proteins: phospho-Tyr
204
extracellular signal-
regulated kinase (ERK)1/2 (E-4), ERK1 (C-16), polyADP-ribose poly-
merase (H-250), IRS-1 (C-20), insulin receptor (IR, β-subunit, C-19;
all from Santa Cruz Biotechnology, Inc.), phospho-EGFR (Tyr
992
), total
EGFR, phospho-AKT (Ser
473
), total AKT, phospho-IGF-1R (Tyr
1131
)/
IR (Tyr
1146
), total IGF-1R (β-subunit), phospho-HER3 (Tyr1289;
21D3), PTEN (138G6), phospho-tyrosine mouse mAb (P-Tyr-100; all
from Cell Signaling Technology, Inc., IRS-2, total HER3 (clone 2F12;
all from Upstate), and β-actin (clone AC-15; Sigma-Aldrich). Immune
complexes were visualized by enhanced chemiluminescence (Pierce
Biotechnology, Inc.). Total amounts of HER3 were quantified by ELISA
according to manufacturer's instructions (R&D systems, Inc.). For HER3
immunoprecipitation, cell extracts (1,000-1,500 μg) were incubated
overnight at 4°C with 4 μg of anti-HER3 antibody (clone 2F12) together
with 30 μL of protein A/G PLUS-agarose (Santa Cruz Biotechnology,
Inc.). Immunoprecipitates were washed thrice with lysis buffer, re-
suspended in gel loading buffer, and analyzed by Western blotting.
Cell surface protein biotinylation. Biotinylation of surface proteins
was done for 30 min at 4°C using the cell surface protein isolation kit
(Pierce Biotechnology) and following instructions from the manufacturer.
Liver tissue specimens. Tumoral and nontumoral liver tissue speci-
mens were obtained from 21 patients with HCC who underwent partial
hepatectomy. Histologic analyses of nontumoral livers showed cirrhosis
in all cases. Cirrhosis was related to HCV infection (14 cases), to HBV
infection (3 cases), and to alcohol abuse (4 cases). Liver tissue samples
were flash frozen in liquid nitrogen and stored at -80°C until analysis.
This study was done with informed consent of patients in accordance
with the French legislation.
Translational Relevance
Based on a recent phase III study (Llovet 2008),
sorafenib, a multikinase inhibitor, has been approved
in Europe and the United States as the reference treat-
ment for patients with advanced hepatocellular carci-
noma (HCC). However, despite the therapeutic
advance allowed with sorafenib, medical need for
new compounds is still important in this indication.
Activation of insulin-like growth factor (IGF) signaling
pathways is strongly implicated in the pathogenesis
of HCC and, based on preclinical data, clinical trials
using IGF type 1 receptor (IGF-1R) inhibitors are cur-
rently ongoing. Here, we show that the specific inhibi-
tion of IGF-1R using a monoclonal antibody induces a
resistance mechanism to this drug in HCC cells via the
activation of the epidermal growth factor receptor
(EGFR)/HER3/AKT pathway, which could be pre-
vented by a combination of EGFR and IGF-1R inhibi-
tors. Therefore, these results together with our
previous data showing cross-talks between EGFR
and IGF-1R in HCC (Desbois-Mouthon 2006) provide
a strong rationale for IGF-1R and EGFR cotargeting
in future clinical trials.
5446Clin Cancer Res 2009;15(17) September 1, 2009 www.aacrjournals.org
Cancer Therapy: Preclinical
Measurements of IGF-1R expression and phosphorylation in human liver
tissue specimens. Liver tissue specimens were homogenized in cell ex-
traction buffer (Biosource, Invitrogen) and centrifuged at 14,000× gfor
15 min at 4°C. The amounts of phosphorylated (Y1135/Y1136) and total
IGF-1R were quantified in supernatants using ELISA following manufac-
turer's instructions (Biosource, Invitrogen). The anti-phospho(Y1135/
Y1136) IGF-1R antibody did not cross-react with phosphorylated IR.
Cell viability. Cells were seeded in 24-well plates (3 × 10
5
cells per
well) and allowed to proliferate for 24 h. Then cells were incubated for
a further 72 h in serum-free or in complete medium [10% fetal bovine
serum (FBS)] containing or not AVE1642, gefitinib, rapamycin, and/or
cetuximab. At the end of the treatment period, cell viability was measured
using the MTT assay.
Flow cytometry analysis of cell cycle and apoptosis. Cells plated in
60-mm dishes were exposed to complete medium containing AVE1642
and/or gefitinib for 48 and 72 h. Both adherent and floating cells were
collected, washed, fixed in ice-cold 70% ethanol at -20°C, and stained
with 20 μg/mL propidium iodide in the presence of 100 μg/mL RNase
A for 30 min at 37°C in the dark. DNA content was analyzed by flow
cytometry (FACS Calibur, Becton Dickinson). Apoptotic cells with hypo-
diploid DNA staining were found in the sub-G
1
peak.
Reverse transcription-PCR analysis. Total RNA was extracted using
RNA Plus lysis solution (MP Biomedicals) and reverse-transcribed as pre-
viously described (24). Transcripts coding for IGF-II and β-actin were
analyzed by semiquantitative PCR as previously described (14). HER3
mRNA transcripts were quantified by real-time PCR using previously
published primers (25). Each sample was normalized on the basis of
its 18S mRNA content as reported (24). PCR was done on a LightCycler
instrument (Roche Applied Science).
Immunofluorescence microscopy. Cells seeded on glass coverslips
were incubated with a 1:50 dilution of a mouse monoclonal anti-
HER3 antibody directed against the extracellular domain (Ab-4, clone
H3.90.6, Thermo Fisher Scientific) or of a control IgG (Jackson Immu-
noResearch Laboratories, Inc.) in HBSS for 1 h at 4°C. Cells were then
rinsed and fixed with 4% paraformaldehyde. Cells were blocked and in-
cubated with a 1:200 dilution of FITC-conjugated goat anti-mouse anti-
body [Alexa Fluor 488 F(ab)
2
; Invitrogen] in 1% bovine serum albumin
in PBS for 1.5 h at room temperature. The slides were counterstained with
4,6-diamidino-2-phenylindole (Sigma-Aldrich) for nuclei detection.
Fluorescence was visualized using a Leica differentially methylated re-
gion immunofluorescence microscope with DFC300 FX digital camera.
Down-regulation of HER3 expression using a siRNA strategy. HER3
expression was down-regulated by using a mixture of four siRNAs tar-
geting human HER3 (ON-TARGETplus SMARTpool; Dharmacon). Con-
trol siRNA was a pool from Dharmacon (siGENOME nontargeting
siRNA pool). Cells plated in 35-mm dishes (35 × 10
4
cells/dish) or in
24-well plates (15 × 10
4
cells per well) were cultured for 24 h to 40%
confluency and then transfected with 100 nmol/L siRNA using Lipo-
fectamine 2000 (InVitrogen). Twenty-four h later, cells were stimulated
or not with AVE1642 for further 48 to 72 h before being harvested for
protein extraction or analyzed for cell viability in a MTT assay.
Statistical analysis. Results are given as means ± SEM. Statistical
comparison of mean values was done using the Student's ttest.
Results
The IGF-II/IGF-1R signaling axis is dysregulated in human
HCC cell lines and tissue. Five human hepatoma cell lines
(HepG2, Hep3B, HuH7, HuH6, and PLC/PRF5) were used in
the present study. All these cell lines overexpressed IGF-1R
and its substrates IRS-1 and IRS-2 compared with normal human
hepatocytes (Fig. 1A). The heterogeneity of IGF-1R bands be-
tween cell lines has been reported by others (26) and may reflect
differential phosphorylations and glycosylations. IGF-II was
more expressed in Hep3B, HepG2, and HuH7 cells compared
with HuH6 and PLC/PRF5 cells (Fig. 1B). The overexpression
of both IGF-II and IGF-1R suggested the existence of an autocrine
stimulating loop in these cells.
IGF-1R status was examined in frozen tumors. The amounts
of total and phosphorylated IGF-1R were quantified by ELISA
in paired tumoral and nontumoral liver specimens from 21 pa-
tients with HCC. The ratio of phospho-IGF-1R to total IGF-1R
was calculated to evaluate the activation level of IGF-1R. We ob-
served increased expression of IGF-1R in 7 tumors (33%; Fig. 1C,
left) and increased activation of IGF-1R in 11 tumors (52%;
Fig. 1C, right). Of note, phospho-IGF-1R levels did not correlate
well with overall IGF-1R levels. These observations fully justified
the need for subsequent investigations examining the effects of
the specific targeting of IGF-1R in HCC cells.
The antiIGF-1R antibody AVE1642 specifically inhibits IGF
but not insulin signaling in hepatoma cells. As a first step, experi-
ments were conducted in HepG2, HuH7, and Hep3B cells to en-
sure the specificity of the antiIGF-1R antibody AVE1642 against
IGF signaling. IGF-1R/IR phosphorylation was examined by
Western blotting using a dual phospho-IGF-1R (Y1131)/IR
(Y1146) antibody. Similar results were obtained in the three cell
lines but, for the clarity of the presentation, only data obtained in
Hep3B cells are shown in Fig. 2A. In control cells, IGF-I, IGF-II,
and insulin (5 × 10
-9
mol/L; 15 min) increased the phosphoryla-
tion of both IGF-1R/IR and Akt. In the presence of AVE1642
(0.5 μg/mL), the stimulatory effects of IGF-I and IGF-II were
markedly inhibited, whereas those of insulin were fully main-
tained. In addition, we observed that long-term treatment (48
and 72 h) with AVE1642 decreased IGF-1R but not IR expression
in HepG2 and Hep3B (Fig. 2B). Therefore, besides its neutralizing
effect on IGF-1R, AVE1642 can also promote IGF-1R down-
regulation and degradation in hepatoma cells as previously re-
ported in other cancer cell types (21, 27). Altogether, these results
show that AVE1642 specifically inhibits IGF but not insulin in-
duction of receptor and AKT phosphorylation.
AVE1642 exerted a moderate antitumoral effect in hepatoma
cells. The blockade of IGF-1R by AVE1642 may impact both
autocrine and serum-induced cell growth. To examine this
point, cell viability was examined in cells cultured for 72 h in
serum-free medium or in complete medium (10% FBS) con-
taining or not AVE1642 (0.5 μg/mL). In serum-free conditions
(Fig. 2C, top), AVE1642 decreased by 41% the viability in
HepG2 cells, whereas its effects were less potent in Hep3B,
PLC/PRF5, and HuH7 cells (24.5%, 17.9%, and 12% decrease,
respectively). AVE1642 was without effect in HuH6 cells. In
complete medium (Fig. 2C, bottom), similar results were ob-
tained, HepG2 cells being the most sensitive cell line (45% in-
hibition). Of note, even higher concentrations of AVE1642 (up
to 2 μg/mL) did not further inhibit hepatoma cell viability (data
not shown). AVE1642 was without any significant effect on via-
bility in normal hepatocytes (Fig. 2D). These findings show that
AVE1642 by itself has an inhibitory effect, even if moderate, on
both autocrine and serum-induced cell growth in almost all hep-
atoma cell lines.
Combining AVE1642 to gefitinib induces a synergistic inhibi-
tion of proliferation in hepatoma cell lines. Our previous study
showing cross-talks between EGFR and IGF-1R (9) led us to ex-
amine whether the antineoplastic effect of AVE1642 might be
influenced by EGFR in hepatoma cells. To this aim, cell lines
were cultured in complete medium and treated with AVE1642
(0.5 μg/mL) in presence or absence of gefitinib. A 2-μmol/L con-
centration of gefitinib was used for these experiments because in
5447 Clin Cancer Res 2009;15(17) September 1, 2009www.aacrjournals.org
IGF-1R Inhibition Restores Gefitinib Sensitivity in HCC
preliminary studies, this concentration was found to be submax-
imal on cell viability (data not shown). In addition, this concen-
tration approaches the serum concentrations reported in patients
orally treated with gefitinib (400-600 mg/day). We observed
that the combination of AVE1642 with gefitinib induced a
supra-additive inhibition of cell growth in all cell lines, even in
HuH6 cells in which AVE1642 exhibited no effect by its own
(Fig. 3A). Similar synergistic effects were obtained when
AVE1642 was combined with a submaximal dose of cetuximab,
an EGFR-neutralizing antibody (Fig. 3B).
To discriminate whether the synergistic reduction in cell via-
bility induced by AVE1642 plus gefitinib resulted from reduced
proliferation and/or to increased cell death, we did flow cyto-
metric analyses. Forty-eight-hour treatment with the AVE1642/
gefitinib combination decreased the percentage of HepG2 cells
in the S phase and arrested cells in the G
0
-G
1
phase of the cell
cycle (Fig. 3C). No increased rate of apoptosis (% of sub-G
1
cells
after 48 and 72 hours of incubation) was evidenced in these ex-
perimental conditions (data not shown; Fig. 3C). In addition,
cell incubation with a nontoxic dose (10 μmol/L) of the pan-
caspase inhibitor ZVAD-fmk did not alter the synergistic effect
of AVE1642 combined to gefitinib on cell viability (data not
shown). Altogether, these findings indicate that the combination
of AVE1642 and gefitinib synergistically inhibits hepatoma
cell viability through cell cycle inhibition without apoptosis
induction.
The synergistic effect of AVE1642 combined with gefitinib routes
through the down-regulation of AKT signaling. As a next step,
we analyzed the molecular mechanisms whereby AVE1642 and
gefitinib had a synergistic inhibitory effect on hepatoma cell
proliferation. Cells were treated for 48 hours in the presence
of AVE1642 combined or not with gefitinib and then stimulated
for 20 minutes with 10% FBS. Similar results were obtained in
the five cell lines. When used alone, AVE1642 slightly decreased
serum-induced AKT phosphorylation (by about 1.5- to 2-fold),
whereas gefitinib had no effect (Fig. 4A). When AVE1642 was
combined with gefinitib, serum-induced AKT phosphorylation
was markedly reduced (by about 3- to 5-fold) compared with
the effects of each drug used alone. In contrast, the combination
of drugs had no effect on serum-induced ERK phosphorylation
(Fig. 4A). This strongly suggested that the inhibition of AKT sig-
naling rather than the inhibition of ERK signaling contributed
to the growth inhibitory effect of AVE1642 combined with
gefitinib. Down-regulation of AKT phosphorylation upon treat-
ment with the AVE1642/gefitinib combinationdid not result from
the modulation of PTEN expression (Fig. 4A). The combination
Fig. 1. Expression of components of
the IGF-II/IGF-1R signaling axis in
human HCC cell lines and tissue
specimens. A, whole-cell extracts
obtained from normal human
hepatocytes and HCC cell lines were
analyzed by Western blotting for
IGF-1R, IRS-1, and IRS-2 expressions.
The membrane was reblotted with
an AKT antibody to ensure equivalent
loading. B, total RNA (2 μg) extracted
from the five hepatoma cell lines were
analyzed by semiquantitative reverse
transcription-PCR using human IGF-II
primers. β-Actin PCR products were
run in parallel to ensure that equivalent
amounts of cDNA were amplified.
C, the levels of phosphorylated
(Y1135/Y1136) and total IGF-1R were
examined in 21 paired tumoral
(tumor) and nontumoral (adjacent)
liver specimens using ELISA.
5448Clin Cancer Res 2009;15(17) September 1, 2009 www.aacrjournals.org
Cancer Therapy: Preclinical
of AVE1642 with rapamycin (1 nmol/L), a specific inhibitor of
mammalian target of rapamycin (a downstream target of AKT),
also induced a synergistic inhibition of cell viability in hepatoma
cells (Fig. 4B). Previous studies conducted in diverse cancer cell
types (2830) have shown that rapamycin triggers a rapid feed-
back mechanism resulting in AKT activation and which may
be dependent on IGF-1R signaling. We examined whether such
a mechanism also occurred in HCC cells and, if so, whether
AVE1642 had the potential to block it. Rapamycin alone
(1 and 10 nmol/L) markedly increased the phospho-AKT content
in HCC cell lines and this effect was abolished by AVE1642
(Fig. 4C). These findings indicate that rapamycin up-regulates
the AKT pathway in HCC cells through an IGF-1Rdependent
pathway.
AVE1642 increases HER3 phosphorylation and expression in
hepatoma cell lines. AKT activation by EGFR is due to the ability
of this receptor to dimerize with and to transphosphorylate the
kinase-inactive receptor HER3 (ErbB-3), leading to the recruitment
Fig. 2. Effects of the antiIGF-1R
antibody AVE1642 on IGF and insulin
signaling in hepatoma cells. A, Hep3B
cells were treated for 1 h with or
without AVE1642 (0.5 μg/mL) and then
stimulated for 15 min with IGF-I, IGF-II,
or insulin (10
-9
mol/L). Whole-cell
extracts were analyzed by Western
blotting for levels of phosphorylated
and total IGF-1R/IR and AKT. B, HepG2
and Hep3B cells were treated with
AVE1642 (0.5 μg/mL) for 48 and
72 h and total expression levels of
IGF-1R and IR were assessed by
Western blotting. Blots are
representative of two independent
experiments. C, HCC cell lines were
treated for 72 h with or without AVE1642
(0.5 μg/mL) in serum-free (top)orin
complete (bottom) medium and then
cell viability was measured. D, normal
hepatocytes were treated for 72 h with
increasing concentrations of AVE1642
(0-2 μg/mL) and cell viability was
measured. Columns, means of three to
four independent experiments done in
quadruplet; bars, SEM. *, P< 0.05; **,
P< 0.002; ***, P< 0.001 compared with
untreated cells.
5449 Clin Cancer Res 2009;15(17) September 1, 2009www.aacrjournals.org
IGF-1R Inhibition Restores Gefitinib Sensitivity in HCC
of the p85 regulatory subunit of phosphoinositide 3-kinase
(PI3K) to HER3 and subsequent activation of PI3K (31). Because
AKT phosphorylation was almost completely abrogated in the
presence of AVE1642 and gefitinib but not in the presence of
AVE1642 alone, we examined by Western blot analyses whether
the EGFR/HER3/AKT pathway may be modulated by AVE1642.
All hepatoma cell lines expressed EGFR and HER3, whereas HER-2
(ErbB-2) expression was barely detected (data not shown). HepG2
Fig. 3. Antitumoral effects of AVE1642
alone or in combination with gefitinib in
hepatoma cell lines. A, cell lines were
treated for 72 h with or without
AVE1642 (AVE; 0.5 μg/mL) and/or
gefitinib (gef;2μmol/L) in complete
medium, and then cell viability was
measured. B, the same experiments
were done with cetuximab (cetux;
1-2 μg/mL) instead of gefitinib.
Columns, means of three independent
experiments done in quadruplet; bars,
SEM. Black lines, the additivity
threshold. C, HepG2 cells were treated
for 48 h with or without AVE1642
(0.5 μg/mL) and/or gefitinib (2 μmol/L)
and analyzed for cell cycle repartition
by flow cytometry. A representative
experiment out of two is shown.
5450Clin Cancer Res 2009;15(17) September 1, 2009 www.aacrjournals.org
Cancer Therapy: Preclinical
and HuH7 cell stimulation with 10% FBS induced the tyrosine
phosphorylation of both EGFR and HER3 and these effects were
inhibited by gefitinib (Fig. 5A). In the presence of AVE1642 alone,
the phosphorylation of EGFR and more especially that of HER3
were further increased. The stimulatory effects of AVE1642
on both EGFR and HER3 were strongly inhibited by gefitinib
(Fig. 5A). AVE1642 increased HER3 protein expression, although
it did not modify the amounts of EGFR (Fig. 5A). These find-
ings were also observed in Hep3B and PLC/PRF5 cells. Increased
tyrosine phosphorylation of HER3 parallel to increased expres-
sion of HER3 was also seen in HER3 immunoprecipitates ob-
tained from HCC cells treated with AVE1642 (Supplementary
Fig. S1). An accurate measurement of HER3 protein amounts
done by ELISA showed that AVE1642 increased HER3 by 1.5-,
2.7-, and 4.6-fold in HepG2, HuH7, and PLC/PRF5 cells, respec-
tively (Fig. 5B). Immunofluorescence microscopy (Fig. 5C) and
surface protein biotinylation (Fig. 5D) showed that AVE1642 in-
creased the amount of membrane-associated HER3. No increase
of HER3 expression was observed in normal hepatocytes treated
with increasing doses of AVE1642 (Supplementary Fig. S2). Alto-
gether, these findings show that, in all tested hepatoma cells, the
specific targeting of IGF-1R stimulates a cross-talk toward the
EGFR signaling pathway via the up-regulation of the total
HER3 cellular content. Interestingly, a reciprocal up-regulation
of IGF-1R signaling consecutive to EGFR inhibition was not ob-
served in HCC cell lines after short-term and long-term treat-
ments with gefitinib. Thus, a 48-hour treatment with gefitinib
(5 and 10 μmol/L) modified neither the phosphorylated nor
Fig. 4. Effects of AVE1642 combined
with gefitinib on AKT and ERK signaling
in hepatoma cells. A, serum-starved
HepG2 and HuH7 cells were treated
for 48 h with or without AVE1642
(0.5 μg/mL) and/or gefitinib (2 μmol/L)
and then stimulated with 10% FBS for
20 min. Whole-cell extracts were
analyzed by Western blotting for
phosphorylated and/or total AKT, ERKs,
and PTEN. B, HepG2, Hep3B, and HuH7
were treated for 72 h with or without
AVE1642 (0.5 μg/mL) and/or rapamycin
(rapa; 1 nmol/L) in complete medium,
and cell viability was measured.
Columns, mean of three independent
experiments done in quadruplet; bars,
SEM. Black lines, additivity threshold.
C, Hep3B and HuH7 cells were
pretreated for 2 h with or without
AVE1642 (0.5 μg/mL) and then
incubated or not with rapamycin (1 and
10 nmol/L; 3 h). Whole-cell extracts
were analyzed by Western blotting for
phosphorylated and/or total AKT levels.
Blots are representative of two
independent experiments.
5451 Clin Cancer Res 2009;15(17) September 1, 2009www.aacrjournals.org
IGF-1R Inhibition Restores Gefitinib Sensitivity in HCC
the total levels of IGF-1R (Supplementary Fig. S3A). Moreover,
HepG2 and HuH6 cells continuously exposed to 10 μmol/L
gefinitib for 5 months (resistant cells) showed no increased levels
of either phosphorylated or total IGF-1R (Supplementary
Fig. S3B) nor elevated sensitivity to growth inhibition by
AVE1642 (Supplementary Fig. S3C) compared with the untreat-
ed counterparts (parental cells). Rather, a decreased expression of
IGF-1R together with a decreased IGF-1R phosphorylation was
observed in HepG2 cells chronically incubated with gefitinib
(Supplementary Fig. S3B).
To further characterize the mechanisms whereby AVE1642
increased HER3 expression in HCC cells, we compared HER3
mRNA expression in control and AVE1642-treated cells by quan-
titative real-time PCR. AVE1642 increased the levels of HER3
transcripts by 1.6-, 1.9-, and 3.8-fold in HepG2, HuH7, and
PLC/PRF5 cells, respectively (Fig. 6A). Given that rapamycin
synergized with AVE1642 to reduce cell viability (Fig. 4B), we ex-
amined whether rapamycin could affect AVE1642 induction of
HER3 expression and phosphorylation. In HepG2 (Supplemen-
tary Fig. S4), HuH7, and Hep3B cells (data not shown), the stim-
ulatory effect of AVE1642 on HER3 remained unchanged in the
presence of rapamycin suggesting that AVE1642 increases HER3
expression and phosphorylation through a mammalian target of
rapamycin (mTOR)-independent pathway.
HER3 limits the antitumoral action of AVE1642 in hepatoma
cells. We examined whether the up-regulation of HER3 ex-
pression interfered with the antitumoral effect of AVE1642. To
address this issue, HepG2 and PLC/PRF5 cells were transiently
Fig. 5. Effects of AVE1642 on EGFR/HER3 signaling in hepatoma cells. A, serum-starved HepG2 and HuH7 cells were treated for 48 h with or without
AVE1642 (0.5 μg/mL) and/or gefitinib (2 μmol/L) and then stimulated with 10% FBS for 20 min. Whole-cell extracts were analyzed by Western blotting for
phosphorylated and total expression levels of EGFR and HER3. B, serum-starved HepG2, HuH7, and PLC/PRF5 cells were treated for 48 h with or without
AVE1642 (0.5 μg/mL) and the levels of total HER3 were quantified for by ELISA. Columns, mean of three independent experiments; bars, SEM. C, HepG2 and
HuH7 cells treated or not with AVE1642 for 24 h were incubated with an anti-HER3 antibody at 4°C to minimize internalization and to enhance cell surface
labeling of receptors. Cells were then fixed and receptor-antibody complexes were visualized with a secondary antibody conjugated with FITC. Parallel
experiments conducted with a control IgG showed no labeling (data not shown). D, cells treated or not with AVE1642 for 24 h were processed for cell surface
protein biotinylation. Biotinylated proteins were purified and analyzed for HER3 expression by Western blot analysis. E-cadherin detection was used as a
loading control.
5452Clin Cancer Res 2009;15(17) September 1, 2009 www.aacrjournals.org
Cancer Therapy: Preclinical
transfected with HER3-specific siRNAs or with control siRNAs.
As shown in Fig. 6B, HER3-specific siRNAs induced a partial
knock-down of HER3 expression (approximately 50% and
80% reduction in HepG2 and PLC/PRF5 cells, respectively) both
in unstimulated and in AVE1642-treated cells compared with
cells transfected with the control siRNAs. HER3 down-regulation
was accompanied by a subsequent decrease in the phosphoryla-
tion state of HER3. AKT phosphorylation was also reduced but
only in cells treated with AVE1642. When transfected cells were
analyzed in an MTT assay, we observed that the antitumoral
effect of AVE1642 was significantly increased in cells in which
HER3 was down-regulated compared with control cells (Fig. 6C).
This result confirms the major role of HER3 in the regulation of
hepatoma cell sensitivity to the antiIGF-1R antibody. Taken as a
whole, our findings show that hepatoma cell lines may adapt
to IGF-1R inhibition by inducing early compensatory inputs
leading to the activation of an EGFR/HER3/AKT-dependent
pathway (Fig. 6D).
Discussion
The dysregulation of the IGF signaling axis, notably via an
increased IGF-II availability, has been well documented in hu-
man HCC (6). However, data concerning the IGF-1R status in
HCC tumors are limited and conflictory. A study analyzing 15
paired samples of HCC and adjacent nontumoral liver tissue
showed no significant increase of IGF-1R mRNA expression in
tumors (32). A recent study showed by Western blot analysis
Fig. 6. Contribution of HER3 to hepatoma cell sensitivity to AVE1642. A, serum-starved HepG2, HuH7, and PLC/PRF5 cells were treated for 48 h with or
without AVE1642 (0.5 μg/mL), and HER3 mRNA transcripts were quantified by real-time PCR. Band C, HepG2 and PLC/PRF5 cells were transiently transfected
with HER3 siRNA or with control (ctl) siRNA and analyzed for HER3 and AKT expression by Western blotting after 48 h of treatment with AVE1642 (B), and for
cell viability after 72 h of treatment with AVE1642 (C). Representative blots of three independent experiments are shown. Columns, means of three
independent experiments; bars, SEM. *, P< 0.001 compared with cells transfected with the control siRNA and treated with AVE1642. D, proposed
mechanism for the synergistic antitumoral effect of AVE1642 combined with gefitinib. Left, IGF-1R is a strong activator of the AKT pathway in hepatoma cells,
which may contribute to the proliferative effect of IGF-II. In contrast, EGFR is a poor activator of AKT in these cells, maybe due to a nonefficient coupling
between EGFR and HER3. As a result, hepatoma cells are poorly sensitive to gefitinib. Right, in the presence of AVE1642, IGF-1R is blocked and an escape
mechanism is induced, resulting in the up-regulation of HER3 expression. EGFR, which is also activated consecutively to IGF-1R inhibition probably by
ligand-dependent mechanism, homodimerizes with and trans-phosphorylated HER3, thus restoring an efficient coupling to the PI3K/AKT pathway. Under
this condition, hepatoma cell sensitivity to gefitinib is increased.
5453 Clin Cancer Res 2009;15(17) September 1, 2009www.aacrjournals.org
IGF-1R Inhibition Restores Gefitinib Sensitivity in HCC
that IGF-1R was frequently overexpressed in a subgroup of
tumors associated with a low copy number of HBV (7 of 9 tu-
mors; ref. 12). In addition, some HCC tumors associated with
hepatitis C virus and increased proliferation presented frequent-
ly with IGF-1R phosphorylation by immunohistochemistry
analysis (11 of 23 tumors; ref. 33). In our panel of HCC, ELISA
measurements revealed that the levels of IGF-1R expression and
activation were increased in approximately 30% and 50% of tu-
mors compared with matched cirrhotic liver tissues, respectively.
As we did not find a clear correlation between total and phos-
phorylated levels of IGF-1R, our data suggest that both phos-
phorylated and total levels of IGF-1R might be predictive of
IGF-1R signaling dependency and of potential sensitivity to
IGF-1R targeting in HCC cells.
Two classes of IGF-1R inhibitors are currently under clinical
development for cancer therapy: (a) mAbs such as AVE1642
that specifically bind to the extracellular domain of IGF-1R,
block ligand/receptor interaction, and promote IGF-1R down-
regulation and degradation and (b) small-molecule TKIs.
However, due to the high sequence homology between IGF-1R
and IR (84%), these latter may be not fully specific for IGF-1R.
AVE1642 is the humanized version of the murine mAb EM164,
which has been shown to potently and specifically inhibit IGF-
1stimulated proliferation in different cancer cell lines including
breast, lung, colon, and myeloma cells (21, 27). Accordingly, we
observed that, in hepatoma cells, AVE1642 inhibited signaling in
response to exogenously added IGF-I, IGF-II, but not to insulin.
In MTT assays, the antitumoral potency of AVE1642 was compa-
rable in the presence or absence of serum, especially in cell lines
with strong IGF-II production (i.e, HepG2, HuH7, and Hep3B),
suggesting that autocrine IGF-II is an important driver for
hepatoma cell growth. The strongest effect of AVE1642 on cell
viability was observed in HepG2 cells, whereas HuH6 was the
most resistant cell line. There was no correlation between cell
sensitivity to AVE1642 and IGF-II mRNA expression in serum-
free conditions because AVE1642 induced the same inhibition
of cell viability in PLC/PRF5 (low IGF-II mRNA expression levels)
and HuH7 (high IGF-II mRNA expression levels) cells. Other-
wise, it has been shown that the antiproliferative effect of
AVE1642 was not correlated to IGF-1R expression in a panel
of cancer cells.
5
We observed that the combination of AVE1642 with gefitinib
or with cetuximab induced a supra-additive inhibition of viability
in all cell lines compared with drugs alone. These findings suggest
that ligand-dependent activation of EGFR limits the antineoplasic
action of AVE1642 in hepatoma cells, irrespective of their pheno-
type (i.e, wild-type or mutated p53, β-catenin, and/or Rb), their
viral status (HBs
neg
or HBs
pos
antigen), or their own sensitivity
to AVE1642. We show that the synergy between AVE1642 and
anti-EGFR therapy is a general phenomenon and not a peculiar-
ity of a few cell lines. The synergy between AVE1642 and gefitinib
led mainly to a cytostatic effect with growth arrest in G
0
-G
1
without apoptosis. The analysis of the underlying mechanisms
revealed that AVE1642 increased the amounts of total and phos-
phorylated HER3. Therefore, our findings indicate that, follow-
ing blockade of IGF-1R signaling, hepatoma cells are able to
switch from IGF-1R to EGFR dependency to maintain cell growth
and AKT phosphorylation. During the preparation of this article,
an adaptive up-regulation of the EGFR pathway in response to
IGF-1R inhibition with small TKIs has been reported in ovarian
and colon cancer cells (34, 35). A reciprocal regulation seems not
to occur in hepatoma cells because we did not observe any stim-
ulating effect of gefitinib on IGF-1R signaling after short-term
and even long-term exposures to this drug. Such a regulation
had been reported in breast, prostate, and nonsmall cell
lung cancer cells in which EGFR TKIs induced a significant
activation of IGF-1R signaling through different mechanisms:
up-regulation of IGF-1R activity (36), increased availability of
IRS-1 (37), formation of EGFR/IGF-1R heterodimers (38), or loss
of IGF binding protein expression (39).
HER3 is a peculiar member of the EGFR family because it
lacks a functional intrinsic tyrosine kinase activity and is able
to directly bind PI3K and activate the donwstream AKT pathway
(31). HER3 functions through heterodimerization with EGFR
or HER-2. In EGFR/HER3 and HER-2/HER3 heterodimers, the
transphosphorylation of HER3 allows the recruitment of the
p85 regulatory subunit of PI3K. A growing body of literature
has recently emerged showing that HER3 is a crucial sensor of
sensitivity of cancer cells to EGFR TKIs. First, there is a strong cor-
relation between the amounts of phosphorylated and total HER3
and response to gefitinib or to erlotinib in lung, pancreas, and
liver cancer cells (4043). Second, it has been shown that cancer
cells that rely mainly on the EGFR/HER3 pathway for AKT acti-
vation displayed exquisite sensitivity to gefitinib and erlotinib
(44, 45). In this context, the increased expression of phosphory-
lated HER3 observed in hepatoma cells exposed to AVE1642
might be the main mechanism accounting for the synergy be-
tween AVE1642 and gefitinib in these cells. Arguments for this
hypothesis are the following: first, all hepatoma cell lines that
express wild-type EGFR (9) were poorly sensitive to gefitinib
alone in terms of inhibition of cell viability (IC
50
, approximately
8-10 μmol/L in all cell lines; data not shown) and of AKT phos-
phorylation; second, gefitinib, by preventing AVE1642-induced
phosphorylation of HER3, led to the down-regulation of AKT phos-
phorylation; and third, counter-acting the effect of AVE1642 on
HER3 expression using siRNAs sensitized hepatoma cells to
AVE1642. Of note, there was no correlation between basal levels
of HER3 and cell response to the bitherapy AVE1642/gefitinib.
Moreover, HER3 expression did not correlate with IGF-II ex-
pression (data not shown), suggesting that baselines of HER3 and
IGF-II are not predictive markers of HCC response to the bitherapy.
AVE1642 synergized not only with gefitinib but also with
rapamycin to reduce cell viability. In both combinations, a
marked down-regulation of AKT phosphorylation was observed
but the underlying mechanisms seemed to be different. Indeed,
contrasting with what was observed in the presence of gefitinib,
rapamycin did not prevent the induction of HER3 phosphory-
lation by AVE1642. Nevertheless, AVE1642 had the potential to
impede the induction of AKT phosphorylation by rapamycin.
These latter findings corroborate with those obtained in other
cancer cell types showing that in HCC cells rapamycin is able to
up-regulate the AKT pathway through an IGF-1R-dependent
pathway (2830). The inhibitory effect of AVE1642 on this pro-
cess may contribute to the antitumoral effect of the rapamycin/
AVE1642 combination in HCC cells.
It is highly probable that the up-regulation of HER3 expres-
sion consecutive to IGF-1R inhibition may be responsible for
the increase of HER3 phosphorylation. Indeed, this could favor
the formation of EGFR/HER3 heterodimers and the subsequent
5
Unpublished results.
5454Clin Cancer Res 2009;15(17) September 1, 2009 www.aacrjournals.org
Cancer Therapy: Preclinical
trans-phosphorylation of HER3 by EGFR. This could also be en-
hanced by the fact that AVE1642 promoted a slight but repro-
ducible increase of EGFR phosphorylation. Otherwise,
AVE1642 did not seem to modulate the availability of specific
ligands for HER3 because HER3 neutralization with a specific
antibody was without effect on cell response to AVE1642 (data
not shown).
In summary, we present evidence for the interest of targeting
IGF-1R in HCC because this receptor may be overexpressed
and/or activated in tumors. However, we also show that the
antitumoral efficiency of a specific antibody against IGF-1R is
counteracted in HCC cells due to its ability to stimulate the
EGFR/HER3/AKT signaling pathway. These results provide
therefore a strong rational for IGF-1R and EGFR cotargeting
in human HCC to maximize antitumoral effect and to prevent
the early development of resistance. Clinical studies are current-
ly ongoing to validate this combinatory approach.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Acknowledgments
We thank the Tumorothèque Cancerest, hôpital Saint-Antoine, Paris
(Pr Jean-François Fléjou, Pr Dominique Wendum), for providing us with
human biopsies, Dr Christine Perret for providing us with cell lines and to
Roland Delelo for human hepatocytes, and cytometry platform (IFR65) for
fluorescence-activated cell sorting analysis.
References
1. Bruix J, Hessheimer AJ, Forner A, Boix L, Vilana
R, Llovet JM. New aspects of diagnosis and ther-
apy of hepatocellular carcinoma. Oncogene 2006;
25:384856.
2. Avila MA, Berasain C, Sangro B, Prieto J. New
therapies for hepatocellular carcinoma. Onco-
gene 2006;25:386684.
3. Hopfner M, Schuppan D, Scherubl H. Growth
factor receptors and related signalling pathways
as targets for novel treatment strategies of hepa-
tocellular cancer. World J Gastroenterol 2008;14:
114.
4. Llovet JM, Bruix J. Molecular targeted therapies
in hepatocellular carcinoma. Hepatology 2008;
48:131227.
5. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib
in advanced hepatocellular carcinoma. N Engl J
Med 2008;359:37890.
6. Breuhahn K, Longerich T, Schirmacher P. Dys-
regulation of growth factor signaling in human
hepatocellular carcinoma. Oncogene 2006;25:
3787800.
7. Scharf JG, Braulke T. The role of the IGF axis in
hepatocarcinogenesis. Horm Metab Res 2003;
35:68593.
8. Lund P, Schubert D, Niketeghad F, Schirmacher
P. Autocrine inhibition of chemotherapy response
in human liver tumor cells by insulin-like growth
factor-II. Cancer Lett 2004;206:8596.
9. Desbois-Mouthon C, Cacheux W, Blivet-Van
Eggelpoel MJ, et al. Impact of IGF-1R/EGFR
cross-talks on hepatomacell sensitivityto gefitinib.
Int J Cancer 2006;119:255766.
10. Nussbaum T, Samarin J, Ehemann V, et al.
Autocrine insulin-like growth factor-II stimula-
tion of tumor cell migration is a progression step
in human hepatocarcinogenesis. Hepatology
2008;48:14656.
11. Kim SO, Park JG, Lee YI. Increased expression
of the insulin-like growth factor I (IGF-I) receptor
gene in hepatocellular carcinoma cell lines: im-
plications of IGF-I receptor gene activation by
hepatitis B virus X gene product. Cancer Res
1996;56:38316.
12. Boyault S, Rickman DS, de Reynies A, et al.
Transcriptome classification of HCC is related to
gene alterations and to new therapeutic targets.
Hepatology 2007;45:4252.
13. Tanaka S, Wands JR. Insulin receptor sub-
strate 1 overexpression in human hepatocellular
carcinoma cells prevents transforming growth
factor β1-induced apoptosis. Cancer Res 1996;
56:33914.
14. Boissan M, Beurel E, Wendum D, et al. Over-
expression of insulin receptor substrate-2 in hu-
man and murine hepatocellular carcinoma.
Am J Pathol 2005;167:86977.
15. Haddad R, Held WA. Genomic imprinting and
Igf2 influence liver tumorigenesis and loss of het-
erozygosity in SV40 T antigen transgenic mice.
Cancer Res 1997;57:461523.
16. Yao X, Hu JF, Daniels M, et al. A methylated
oligonucleotide inhibits IGF2 expression and en-
hances survival in a model of hepatocellular car-
cinoma. J Clin Invest 2003;111:26573.
17. Zhang YC, Wang XP, Zhang LY, Song AL, Kou
ZM, Li XS. Effect of blocking IGF-I receptor on
growth of human hepatocellular carcinoma
cells. World J Gastroenterol 2006;12:397782.
18. Hopfner M, Huether A, Sutter AP, Baradari V,
Schuppan D, Scherubl H. Blockade of IGF-1 re-
ceptor tyrosine kinase has antineoplastic effects
in hepatocellular carcinoma cells. Biochem Phar-
macol 2006;71:143548.
19. Huether A, Hopfner M, Sutter AP, Baradari V,
Schuppan D, Scherubl H. Signaling pathways in-
volved in the inhibition of epidermal growth fac-
tor receptor by erlotinib in hepatocellular cancer.
World J Gastroenterol 2006;12:51607.
20. Moreau P, Hulin C, Facon T, et al. Phase I study
of AVE1642 anri IGF-1R monoclonal antibody in
patients with advanced multiple myeloma. Blood
2007;110: abstract 1166.
21. MaloneyEK, McLaughlin JL,Dagdigian NE, et al.
An anti-insulin-like growth factor I receptor anti-
body that is a potent inhibitor of cancer cell prolif-
eration. Ca ncer Res 200 3;63:5073 83.
22. Corpechot C, Barbu V, Wendum D, et al.
Hypoxia-induced VEGF and collagen I expres-
sions are associated with angiogenesis and
fibrogenesis in experimental cirrhosis.Hepatology
2002;35:101021.
23. Morgillo F, Woo JK, Kim ES, Hong WK, Lee HY.
Heterodimerization of insulin-like growth factor re-
ceptor/epidermal growth factor receptor and induc-
tion of survivin expression counteract the antitumor
action of erlotinib. Cancer Res 2006;66:1010011.
24. ChignardN, Mergey M, Barbu V,et al. VPAC1 ex-
pression is regulated by FXR agonists in the hu-
man gallbladder epithelium. Hepatology 2005;42:
54957.
25. Memon AA, Sorensen BS, Melgard P, Fokdal L,
Thykjaer T, Nexo E. Expression of HER3, HER4
and their ligand heregulin-4 is associated with
better survival in bladder cancer patients. Br J
Cancer 2004;91:203441.
26. Shimizu M, Shirakami Y, Sakai H, et al. EGCG
inhibits activation of the insulin-like growth factor
(IGF)/IGF-1 receptor axis in human hepatocellular
carcinoma cells. Cancer Lett 2007;262:108.
27. Descamps G, Wuilleme-Toumi S, Trichet V,
et al. CD45neg but not CD45pos human myelo-
ma cells are sensitive to the inhibition of IGF-1
signaling by a murine anti-IGF-1R monoclonal
antibody, mAVE1642. J Immunol 2006;177:
421823.
28. O'Reilly KE, Rojo F, She QB, et al. mTOR inhi-
bition induces upstream receptor tyrosine kinase
signaling and activates Akt. Cancer Res 2006;66:
15008.
29. Shi Y, Yan H, Frost P, Gera J, Lichtenstein A.
Mammalian target of rapamycin inhibitors acti-
vate the AKT kinase in multiple myeloma cel ls
by up-regulating the insulin-like growth factor
receptor/insulin receptor substrate-1/phosphati-
dylinositol 3-kinase cascade. Mol Cancer Ther
2005;4:153340.
30. Wan X, Harkavy B, Shen N, Grohar P, Helman
LJ. Rapamycin induces feedback activation of Akt
signaling through an IGF-1R-dependent mecha-
nism. Oncogene 2007;26:193240.
31. Sithanandam G, Anderson LM. The ERBB3 re-
ceptor in cancer and cancer gene therapy. Cancer
Gene Ther 2008;15:41348.
32. Cantarini MC, de la Monte SM, Pang M, et al.
Aspartyl-asparagyl βhydroxylase over-expres-
sion in human hepatoma is linked to activation
of insulin-like growth factor and notch signaling
mechanisms. Hepatology 2006;44:44657.
33. Chiang DY, Villanueva A, Hoshida Y, et al. Fo-
cal gains of VEGFA and molecular classification
of hepatocellular carcinoma. Cancer Res 2008;
68:677988.
34. Buck E, Eyzaguirre A, Rosenfeld-Franklin M,
et al. Feedback mechanisms promote coopera-
tivity for small molecule inhibitors of epidermal
and insulin-like growth factor receptors. Cancer
Res 2008;68:832232.
35. Haluska P, Carboni JM, TenEyck C, et al. HER
receptorsignalingconfers resistanceto the insulin-
like growth factor-I receptor inhibitor, BMS-
536924. Mol Cancer Ther 2008;7:258998.
36. Jones HE, Goddard L, Gee JM, et al. Insulin-
like growth factor-I receptor signalling and ac-
quired resistance to gefitinib (ZD1839; Iressa) in
human breast and prostate cancer cells. Endocr
Relat Cancer 2004;11:793814.
37. Knowlden JM, Jones HE, Barrow D, Gee JM,
Nicholson RI, Hutcheson IR. Insulin receptor sub-
strate-1 involvement in epidermal growth factor
receptor and insulin-like growth factor receptor
signalling: implication for Gefitinib ('Iressa') re-
sponse and resistance. Breast Cancer Res Treat
2007.
38. Morgillo F, Kim WY, Kim ES, Ciardiello F,
Hong WK, Lee HY. Implication of the insulin-
like growth factor-IR pathway in the resistance
of non-small cell lung cancer cells to treat-
ment with gefitinib. Clin Cancer Res 2007;13:
2795803.
39. Guix M, Faber AC, Wang SE, et al. Acquired
5455 Clin Cancer Res 2009;15(17) September 1, 2009www.aacrjournals.org
IGF-1R Inhibition Restores Gefitinib Sensitivity in HCC
resistance to EGFR tyrosine kinase inhibitors in
cancer cells is mediated by loss of IGF-binding
proteins. J Clin Invest 2008;118:260919.
40. Thomson S, Buck E, Petti F, et al. Epithelial
to mesenchymal transition is a determinant of
sensitivity of non-small-cell lung carcinoma cell
lines and xenografts to epidermal growth fac-
tor receptor inhibition. Cancer Res 2005;65:
945562.
41. Fujimoto N, Wislez M, Zhang J, et al. High
expression of ErbB family members and their
ligands in lung adenocarcinomas that are sensi-
tive to inhibition of epidermal growth factor
receptor. Cancer Res 2005;65:1147885.
42. Amann J, Kalyankrishna S, Massion PP, et al.
Aberrant epidermal growth factor receptor
signaling and enhanced sensitivity to EGFR in-
hibitors in lung cancer. Cancer Res 2005;65:
22635.
43. Fuchs BC, FujiiT, Dorfman JD, et al. Epithelial-to-
mesenchymal transitionand integrin-linkedkinase
mediate sensitivity to epidermal growth factor
receptor inhibition in human hepatoma cells.
Cancer Res 2008;68:23919.
44. Engelman JA, Janne PA, Mermel C, et al.
ErbB-3 mediates phosphoinositide 3-kinase ac-
tivity in gefitinib-sensitive non-small cell lung
cancer cell lines. Proc Natl Acad Sci U S A
2005;102:378893.
45. Buck E, Eyzaguirre A, Haley JD, Gibson NW,
Cagnoni P, Iwata KK. Inactivation of Akt by the
epidermal growth factor receptor inhibitor erlo-
tinib is mediated by HER-3 in pancreatic and
colorectal tumor cell lines and contributes to
erlotinib sensitivity. Mol Cancer Ther 2006;5:
20519.
5456Clin Cancer Res 2009;15(17) September 1, 2009 www.aacrjournals.org
Cancer Therapy: Preclinical
... Notably, breast cancer is more dependent on the dysfunctional PI3K/Akt/mTOR pathway than the MAPK/ERK pathway (30). In addition, HER3 allows the recruitment of the p85 regulatory subunit of PI3K to activate PI3K/Akt/mTOR signaling (31,32). These results suggest that a subset of TNBC cells preferentially employ the HER3/Akt/mTOR pathway and not the HER3/ERK pathway. ...
Article
Full-text available
Triple‑negative breast cancer (TNBC), a highly metastatic subtype of breast cancer, and it has the worst prognosis among all subtypes of breast cancer. However, no effective systematic therapy is currently available for TNBC metastasis. Therefore, novel therapies targeting the key molecular mechanisms involved in TNBC metastasis are required. The present study examined whether the expression levels of human epidermal growth factor receptor 3 (HER3) were associated with the metastatic phenotype of TNBC, and evaluated the potential of HER3 as a therapeutic target in vitro and in vivo. A new highly metastatic 4T1 TNBC cell line, termed 4T1‑L8, was established. The protein expression levels in 4T1‑L8 cells were measured using luminex magnetic bead assays and western blot analysis. The HER3 expression levels and distant metastasis‑free survival (DMFS) in TNBC were analyzed using Kaplan‑Meier Plotter. Transwell migration and invasion assays were performed to detect migration and invasion. The anti‑metastatic effects were determined in an experimental mouse model of metastasis. The results revealed that the increased expression of the HER3/Akt/mTOR pathway was associated with a greater level of cell migration, invasion and metastasis of TNBC cells. In addition, it was found that high expression levels of HER3 were associated with a poor DMFS. The inhibition of the HER3/Akt/mammalian target of rapamycin (mTOR) pathway decreased the migration, invasion and metastasis of TNBC cells by decreasing the expression of C‑X‑C chemokine receptor type 4 (CXCR4). Furthermore, treatment of metastatic TNBC cells with everolimus inhibited their migration, invasion and metastasis by decreasing CXCR4 expression. Thus, targeting the HER3/Akt/mTOR pathway opens up a new avenue for the development of therapeutics against TNBC metastasis; in addition, everolimus may prove to be an effective therapeutic agent for the suppression of TNBC metastasis.
... Similarly, based on their highly consistent co-expression profile, the approach of dual inhibition of IGF1R and EGFR/HER2 was adopted and have shown significant success in multiple preclinical models of different cancer -viz. (i) a bi-specific (anti-IGF1R and -EGFR) antibody XGFR in pancreatic cancer (Schanzer et al. 2016), (ii) IGF1R inhibitor linsitinib with EGFR inhibitors lapatinib or gefitinib in esophageal squamous cell carcinoma (Kang et al. 2022), (iii) a ligandbased enediyne-energized bi-specific fusion protein (anti-IGF1R and -EGFR) in esophageal ) and non-small-cell lung cancer , (iv) AVE1642 (anti-IGF1R mAb) and gefitinib in HCC (Desbois-Mouthon et al. 2009), (v) ganitumab and panitumumab (anti-EGFR mAb) in advanced cancers (non-small-cell lung cancer and sarcoma) (Vlahovic et al. 2018) and (vi) ganitumab and panitumumab in metastatic colorectal cancer (Van Cutsem et al. 2014), to name a few. Interestingly, the antidiabetic 'wonder drug' metformin is reported to affect a downregulation of IGF1R and thereby enhance the efficacy of figitumumab (mAb against IGF1R) in small-cell lung cancer (Cao et al. 2015) and non-small-cell lung cancer (Cao et al. 2016). ...
Article
Despite landmark advances in cancer treatments over the last 20 years, cancer remains the second highest cause of death worldwide, much ascribed to intrinsic and acquired resistance to the available therapeutic options. In this review, we address this impending issue, by focusing the spotlight on the rapidly emerging role of growth hormone action mediated by two intimately related tumoral growth factors - growth hormone (GH) and insulin-like growth factor 1 (IGF1). Here, we not only catalog the scientific evidences relating specifically to cancer therapy resistance inflicted by GH and IGF1, but also discuss the pitfalls, merits, outstanding questions and the future need of exploiting GH-IGF1 inhibition to tackle cancer treatment successfully.
... Thus, increased expression and activation of HER3 accompanied by expression of NRG have been reported in HER2 + breast cancer cells resistant to the antibody-drug conjugate (ADC) trastuzumab-emtansine (T-DM1) [68]. This increase in the expression/signaling of HER3 has also been associated with resistance to the insulin-like growth factor 1 receptor (IGF1R) inhibitors in hepatocarcinoma [69]. In this line, high expression of NRG has been reported to be a possible mechanism of resistance to cetuximab in colorectal cancer [70]. ...
Article
Full-text available
The HER3 protein, that belongs to the ErbB/HER receptor tyrosine kinase (RTK) family, is expressed in several types of tumors. That fact, together with the role of HER3 in promoting cell proliferation, implicate that targeting HER3 may have therapeutic relevance. Furthermore, expression and activation of HER3 has been linked to resistance to drugs that target other HER receptors such as agents that act on EGFR or HER2. In addition, HER3 has been associated to resistance to some chemotherapeutic drugs. Because of those circumstances, efforts to develop and test agents targeting HER3 have been carried out. Two types of agents targeting HER3 have been developed. The most abundant are antibodies or engineered antibody derivatives that specifically recognize the extracellular region of HER3. In addition, the use of aptamers specifically interacting with HER3, vaccines or HER3-targeting siRNAs have also been developed. Here we discuss the state of the art of the preclinical and clinical development of drugs aimed at targeting HER3 with therapeutic purposes.
... By forming an asymmetric kinase dimer, HER3 stabilizes its dimer partner in the active conformation Jura et al., 2009). ErbB3/HER3 overexpression is associated with lung, colon, gastric, and other cancers (Amin et al., 2010) and has been also found to be responsible for resistance to HER2, IGF1R, and EGFR inhibitors in the treatment of several types of cancers (Erjala et al., 2006;Sergina et al., 2007;Desbois-Mouthon et al., 2009;Miller et al., 2009). ...
Article
Full-text available
Kinases still remain the most favorable members of the druggable genome, and there are an increasing number of kinase inhibitors approved by the FDA to treat a variety of cancers. Here, we summarize recent developments in targeting kinases and pseudokinases with some examples. Targeting the cell cycle machinery garnered significant clinical success, however, a large section of the kinome remains understudied. We also review recent developments in the understanding of pseudokinases and discuss approaches on how to effectively target in cancer.
... To account this process in the model, the EGFR activation is proportional to the ERK phosphorylated concentration. Crosstalks between EGFR and the insulin family receptors, as well as between IGF1-R and IR, have been reported in the literature [DMBea09,RDea14]. ...
Thesis
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer. It appears mainly as a complication of cirrhosis of the liver in patients exposed to toxic agents (alcohol, hepatic viruses). The study of therapeutic targeting of oncogenic pathways brings new perspectives on cancer treatments that are more effective and better tolerated by patients. An oncogenic pathway is found to be activated in almost all types of tumors. This is the RAS-RAF-MEK-ERK path. The understanding of its regulation is still poorly known today. Sorafenib is prescribed as an inhibitor of RAF kinases in the advanced stages of hepatocellular carcinoma. Its effectiveness is relative since it prolongs the survival of certain patients by a few months, and it does not bring the same clinical benefit to all patients. No biomarker predictive of its effectiveness has been highlighted to date, which makes the customization of its prescription impossible. This thesis provides a study of the modalities of regulation of the RAS-RAF-MEK-ERK pathway in CHC through mathematical modelling. The aim is to build mathematical models to better understand the action of cancer therapy on RAS-RAF-MEK-ERK signal transduction networks, and to bring new perspectives on targeting therapeutic
... Importantly, molecular analysis conducted in these patients indicated that downregulation of IGF1R levels were associated with a concomitant activation of the PI3K/Akt and MAPK pathways due to compensatory upregulation of the epidermal growth factor receptor (EGFR) [111]. Similarly, compensatory activation of the IR-A and IR-B by insulin and IGF2 [86,113], activation of HER3/AKT pathway, or the induction of a mesenchymal-epithelial transition factor (c-MET) all contribute in conferring resistance to anti-IGF1R mAbs [114,115]. ...
Article
Full-text available
Aberrant bioactivity of the insulin-like growth factor (IGF) system results in the development and progression of several pathologic conditions including cancer. Preclinical studies have shown promising anti-cancer therapeutic potentials for anti-IGF targeted therapies. However, a clear but limited clinical benefit was observed only in a minority of patients with sarcomas. The molecular complexity of the IGF system, which comprises multiple regulators and interactions with other cancer-related pathways, poses a major limitation in the use of anti-IGF agents and supports the need of combinatorial therapeutic strategies to better tackle this axis. In this review, we will initially highlight multiple mechanisms underlying IGF dysregulation in cancer and then focus on the impact of the IGF system and its complexity in sarcoma development and progression as well as response to anti-IGF therapies. We will also discuss the role of Ephrin receptors, Hippo pathway, BET proteins and CXCR4 signaling, as mediators of sarcoma malignancy and relevant interactors with the IGF system in tumor cells. A deeper understanding of these molecular interactions might provide the rationale for novel and more effective therapeutic combinations to treat sarcomas.
... The computational model is constructed with the aim to capture the signaling dynamics of key components of the signaling pathway including receptor homo-and heterodimerization. It is not intended to be a complete compendium of all the known molecular interactions (35,38,39). Size and complexity of the computational model were chosen to reflect the available experimental data, and to facilitate efficient computation. ...
Preprint
Targeted therapies have shown significant patient benefit in about 5-10% of solid tumors that are addicted to a single oncogene. Here, we explore the idea of ligand addiction as a driver of tumor growth. High ligand levels in tumors have been shown to be associated with impaired patient survival, but targeted therapies have not yet shown great benefit in unselected patient populations. Using a novel approach of applying Bagged Decision Trees (BDT) to high-dimensional signaling features derived from a computational model, we can predict ligand dependent proliferation across a set of 58 cell lines. This mechanistic, multi-pathway model that features receptor heterodimerization, was trained on seven cancer cell lines and can predict signaling across two independent cell lines by adjusting only the receptor expression levels for each cell line. Interestingly, for patient samples the predicted tumor growth response correlates with high growth factor expression in the tumor microenvironment, which argues for a co-evolution of both factors in vivo . Summary Prediction of ligand-induced growth of cancer cell lines, which correlates with ligand-blocking antibody efficacy, could be significantly improved by learning from features of a mechanistic signaling model, and was applied to reveal a correlation between growth factor expression and predicted response in patient samples.
... Inhibition of one member of RTKs usually triggers the compensation by other members. While active IGF-IR can compensate EGFR inhibition, activation of the EGFR pathway also contributes to anti-IGF-IR drugs resistance in cancer [174,175]. Of note, the adaptor protein IRS1 not only binds to IGF-IR, but also interacts with EGFR/ErbB3. ...
Article
Full-text available
Insulin-like growth factors (IGFs) play important roles in mammalian growth, development, aging, and diseases. Aberrant IGFs signaling may lead to malignant transformation and tumor progression, thus providing the rationale for targeting IGF axis in cancer. However, clinical trials of the type I IGF receptor (IGF-IR)-targeted agents have been largely disappointing. Accumulating evidence demonstrates that the IGF axis not only promotes tumorigenesis, but also confers resistance to standard treatments. Furthermore, there are diverse pathways leading to the resistance to IGF-IR-targeted therapy. Recent studies characterizing the complex IGFs signaling in cancer have raised hope to refine the strategies for targeting the IGF axis. This review highlights the biological activities of IGF-IR signaling in cancer and the contribution of IGF-IR to cytotoxic, endocrine, and molecular targeted therapies resistance. Moreover, we update the diverse mechanisms underlying resistance to IGF-IR-targeted agents and discuss the strategies for future development of the IGF axis-targeted agents.
... In HER2-overexpressing breast cancers and in EGFRmutant-driven lung cancer and head and neck squamous cell carcinoma (SCCHN), HER3 promotes resistance to HER2 and EGFR inhibitors, respectively [37][38][39] . HER3 is also associated with resistance to anti-oestrogen therapies in oestrogen receptor-positive breast cancer and with resistance to insulin-like growth factor 1 receptor (IGF1R) inhibitors in hepatocellular carcinomas 40,41 . Drug-induced elevation of HER3 expression and phosphorylation are the primary mechanisms that contribute to this resistance (Fig. 2a). ...
Article
Full-text available
Pseudokinases are members of the protein kinase superfamily but signal primarily through noncatalytic mechanisms. Many pseudokinases contribute to the pathologies of human diseases, yet they remain largely unexplored as drug targets owing to challenges associated with modulation of their biological functions. Our understanding of the structure and physiological roles of pseudokinases has improved substantially over the past decade, revealing intriguing similarities between pseudokinases and their catalytically active counterparts. Pseudokinases often adopt conformations that are analogous to those seen in catalytically active kinases and, in some cases, can also bind metal cations and/or nucleotides. Several clinically approved kinase inhibitors have been shown to influence the noncatalytic functions of active kinases, providing hope that similar properties in pseudokinases could be pharmacologically regulated. In this Review, we discuss known roles of pseudokinases in disease, their unique structural features and the progress that has been made towards developing pseudokinase-directed therapeutics.
Article
Full-text available
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and is one of the leading causes of cancer-related deaths worldwide. The multi-target inhibitor sorafenib is a first-line treatment for patients with advanced unresectable HCC. Recent clinical studies have evidenced that patients treated with sorafenib together with the anti-diabetic drug metformin have a survival disadvantage compared to patients receiving sorafenib only. Here, we examined whether a clinically relevant dose of metformin (50 mg/kg/d) could influence the antitumoral effects of sorafenib (15 mg/kg/d) in a subcutaneous xenograft model of human HCC growth using two different sequences of administration, i.e concomitant versus sequential dosing regimens. We observed that the administration of metformin six hours prior to sorafenib was significantly less effective in inhibiting tumor growth (15.4% tumor growth inhibition) than concomitant administration of the two drugs (59.5% tumor growth inhibition). In vitro experiments confirmed that pretreatment of different human HCC cell lines with metformin reduced the effects of sorafenib on cell viability, proliferation and signaling. Transcriptomic analysis confirmed significant differences between xenografted tumors obtained under the concomitant and the sequential dosing regimens. Taken together, these observations call into question the benefit of parallel use of metformin and sorafenib in patients with advanced HCC and diabetes, as the interaction between the two drugs could ultimately compromise patient survival. Significance Statement When drugs are administrated sequentially, metformin alters the anti-tumor effect of sorafenib, the reference treatment for advanced hepatocellular carcinoma, in a preclinical murine xenograft model of liver cancer progression as well as in hepatic cancer cell lines. Defective activation of the AMPK pathway as well as major transcriptomic changes are associated with the loss of the anti-tumor effect. These results echo recent clinical work reporting a poorer prognosis for patients with liver cancer who were co-treated with metformin and sorafenib.
Article
Full-text available
No effective systemic therapy exists for patients with advanced hepatocellular carcinoma. A preliminary study suggested that sorafenib, an oral multikinase inhibitor of the vascular endothelial growth factor receptor, the platelet-derived growth factor receptor, and Raf may be effective in hepatocellular carcinoma. In this multicenter, phase 3, double-blind, placebo-controlled trial, we randomly assigned 602 patients with advanced hepatocellular carcinoma who had not received previous systemic treatment to receive either sorafenib (at a dose of 400 mg twice daily) or placebo. Primary outcomes were overall survival and the time to symptomatic progression. Secondary outcomes included the time to radiologic progression and safety. At the second planned interim analysis, 321 deaths had occurred, and the study was stopped. Median overall survival was 10.7 months in the sorafenib group and 7.9 months in the placebo group (hazard ratio in the sorafenib group, 0.69; 95% confidence interval, 0.55 to 0.87; P<0.001). There was no significant difference between the two groups in the median time to symptomatic progression (4.1 months vs. 4.9 months, respectively, P=0.77). The median time to radiologic progression was 5.5 months in the sorafenib group and 2.8 months in the placebo group (P<0.001). Seven patients in the sorafenib group (2%) and two patients in the placebo group (1%) had a partial response; no patients had a complete response. Diarrhea, weight loss, hand-foot skin reaction, and hypophosphatemia were more frequent in the sorafenib group. In patients with advanced hepatocellular carcinoma, median survival and the time to radiologic progression were nearly 3 months longer for patients treated with sorafenib than for those given placebo. (ClinicalTrials.gov number, NCT00105443.)
Article
Full-text available
Epidermal growth factor receptor (EGFR) and insulin-like growth factor-I receptor (IGF-IR) can cooperate to regulate tumor growth and survival, and synergistic growth inhibition has been reported for combined blockade of EGFR and IGF-IR. However, in preclinical models, only a subset of tumors exhibit high sensitivity to this combination, highlighting the potential need for patient selection to optimize clinical efficacy. Herein, we have characterized the molecular basis for cooperative growth inhibition upon dual EGFR and IGF-IR blockade and provide biomarkers that seem to differentiate response. We find for epithelial, but not for mesenchymal-like, tumor cells that Akt is controlled cooperatively by EGFR and IGF-IR. This correlates with synergistic apoptosis and growth inhibition in vitro and growth regression in vivo upon combined blockade of both receptors. We identified two molecular aspects contributing to synergy: (a) inhibition of EGFR or IGF-IR individually promotes activation of the reciprocal receptor; (b) inhibition of EGFR-directed mitogen-activated protein kinase (MAPK) shifts regulation of Akt from EGFR toward IGF-IR. Targeting the MAPK pathway through downstream MAPK/extracellular signal-regulated kinase kinase (MEK) antagonism similarly promoted IGF-driven pAkt and synergism with IGF-IR inhibition. Mechanistically, we find that inhibition of the MAPK pathway circumvents a negative feedback loop imposed on the IGF-IR- insulin receptor substrate 1 (IRS-1) signaling complex, a molecular scenario that parallels the negative feedback loop between mTOR-p70S6K and IRS-1 that mediates rapamycin-directed IGF-IR signaling. Collectively, these data show that resistance to inhibition of MEK, mTOR, and EGFR is associated with enhanced IGF-IR-directed Akt signaling, where all affect feedback loops converging at the level of IRS-1.
Article
Full-text available
Hepatocellular carcinomas represent the third leading cause of cancer-related deaths worldwide. The vast majority of cases arise in the context of chronic liver injury due to hepatitis B virus or hepatitis C virus infection. To identify genetic mechanisms of hepatocarcinogenesis, we characterized copy number alterations and gene expression profiles from the same set of tumors associated with hepatitis C virus. Most tumors harbored 1q gain, 8q gain, or 8p loss, with occasional alterations in 13 additional chromosome arms. In addition to amplifications at 11q13 in 6 of 103 tumors, 4 tumors harbored focal gains at 6p21 incorporating vascular endothelial growth factor A (VEGFA). Fluorescence in situ hybridization on an independent validation set of 210 tumors found 6p21 high-level gains in 14 tumors, as well as 2 tumors with 6p21 amplifications. Strikingly, this locus overlapped with copy gains in 4 of 371 lung adenocarcinomas. Overexpression of VEGFA via 6p21 gain in hepatocellular carcinomas suggested a novel, non-cell-autonomous mechanism of oncogene activation. Hierarchical clustering of gene expression among 91 of these tumors identified five classes, including "CTNNB1", "proliferation", "IFN-related", a novel class defined by polysomy of chromosome 7, and an unannotated class. These class labels were further supported by molecular data; mutations in CTNNB1 were enriched in the "CTNNB1" class, whereas insulin-like growth factor I receptor and RPS6 phosphorylation were enriched in the "proliferation" class. The enrichment of signaling pathway alterations in gene expression classes provides insights on hepatocellular carcinoma pathogenesis. Furthermore, the prevalence of VEGFA high-level gains in multiple tumor types suggests indications for clinical trials of antiangiogenic therapies.
Article
Rationale: CD221 (IGF-1R) is aberrantly expressed in multiple myeloma (MM) and is associated with disease severity (Bataille et al, Haematologica2005;90:706). IGF-1R is thus an attractive therapeutic target in patients with advanced disease. Patients and methods: We have conducted an open-label dose escalation phase I study of AVE1642, anti IGF-1R monoclonal antibody, in patients with advanced MM. The primary objective was to determine the selected dose of AVE1642 administered every 3 weeks (q3w) based on pharmacokinetic (PK), pharmacodynamic (PD) parameters and dose limiting toxicities. The secondary objectives were to assess the safety profile, the biological activity (saturation of receptors) on peripheral granulocytes, the potential immunogenicity and preliminary clinical activity of AVE1642. Results: 14 patients have been treated with AVE1642 as IV infusion administered q3w (day 1 = day 22) at 3 different dose levels: 3 (n = 4), 6 (n = 6) and 12 mg/kg (n = 4). A median number of 2 infusions (1–8) were administered. AVE1642 was well tolerated, except reversible grade 3 hyperglycemia observed in 2 diabetic patients. No hypersensitivity during infusion was reported. No human antibody anti AVE1642 was detected. One patient with Bence-Jones MM experienced a decrease in proteinuria and relief of bone pain. Based on PK/PD results, the dose of 12 mg/kg of AVE1642 has been selected for further clinical evaluation in MM patients. Based on the in vitro synergistic activity of AVE1642 + bortezomib (Descamps et al, ASH2006, 845a), we have started a combination trial of AVE1642, 12 mg/kg q3w + bortezomib (1.3 mg/m2 at d1, d4, d8 and d11 q3w) in patients with advanced MM.
Article
AIM: To study the expression level and localization of insulin-like growth factor -I receptor (IGF-IR) in HepG2 cells and Chang liver cells, and to observe the effect of anti-IGF-IR monoclonal antibody (αIR3) on the growth of HepG2 cells. METHODS: The expression of IGF-IR in HepG2 cells and Chang liver cells was detected by immunohistochemistry. The influences of αIR3 on proliferation and apoptosis were examined by the 3- (4, 5-dimethylthiazol-2-yl)-2, 5- diphenyltetrazolium bromide (MTT) assay and electron microscopy, respectively. Flow cytometry (FCM) was applied for the analysis of cell cycle and apoptosis was observed under electron microscope. RESULTS: IGF-IR was located in the membranes of both HepG2 and Chang liver cell lines, and the expression level of IGF-IR was higher in HepG2 cells than in Chang liver cells. Treated with 0.1 μg/mL αIR3 for 48 h in vitro, the cell growth index (GI) of HepG2 cells was significantly higher than that of control (103.41% vs 100%, P < 0.01). However, the αIR3 for 24 h at final concentration of 4.0 μg/mL made the GI of HepG2 cells lower than that of control (93.37% vs 100%, P < 0.01). Compared with control, treated with αIR3 for 48 h at final concentrations ranging from 1.0 μg/mL to 4.0 μg/mL markedly reduced the GIs of HepG2 cells (97.63%, 97.16%, 95.13%, 92.53% vs 100%, P < 0.05 or P < 0.01), treated with αIR3 for 72 h at final concentrations ranging from 0.2 μg/mL to 4.0 μg/mL decreased the GIs of HepG2 cells obviously (95%, 91.63%, 90.77%, 89.84%, 88.51% vs 100%, P < 0.01), and treated with αIR3 for 96 h at final concentrations ranging from 0.5 μg/mL to 4.0 μg/mL made GIs of HepG2 cells lower significantly (88.86%, 83.97%, 79.81%, 77.24%, 70.51% vs 100%, P < 0.05 or P < 0.01). Moreover, treated with αIR3 from 24 h to 96 h at final concentrations ranging from 0.2 μg/mL to 4.0 μg/mL reduced the GI of HepG2 cells from 97.63% to 70.51% in a dose- and time-dependent manner. Also, αIR3 treatment for 72 h at final concentration from 0.5 μg/mL to 2.0 μg/mL increased the proportion of G0/G1 phase cells(61.73%, 67.1%, 83.7%,76.87% vs 44.47%, P < 0.01) and significantly decreased that of S phase cells(28.63%, 25.13%, 15.63%, 23.13% vs 53.17%, P < 0.01), in contrast to the proportion of G2/M phase cells. The apoptotic rates of HepG2 cells were increased more than that of control (7.83%, 16.13%, 21.1%, 37.73% vs 4.13%, P < 0.01). CONCLUSION: The malignant cell phenotype of human hepatocarcinoma cell is related to overexpression of IGF-IR. The blockage of IGF-IR with αIR3 may contribute to the inhibition of proliferation and induction of apoptosis in HepG2 cells.
Article
No effective systemic therapy exists for patients with advanced hepatocellular carcinoma. A preliminary study suggested that sorafenib, an oral multikinase inhibitor of the vascular endothelial growth factor receptor, the platelet-derived growth factor receptor, and Raf may be effective in hepatocellular carcinoma. Methods In this multicenter, phase 3, double-blind, placebo-controlled trial, we randomly assigned 602 patients with advanced hepatocellular carcinoma who had not received previous systemic treatment to receive either sorafenib (at a dose of 400 mg twice daily) or placebo. Primary outcomes were overall survival and the time to symptomatic progression. Secondary outcomes included the time to radiologic progression and safety. Results At the second planned interim analysis, 321 deaths had occurred, and the study was stopped. Median overall survival was 10.7 months in the sorafenib group and 7.9 months in the placebo group (hazard ratio in the sorafenib group, 0.69; 95% confidence interval, 0.55 to 0.87; P
Article
Hepatocellular carcinoma (HCC) is a complex and heterogeneous tumor with several genomic alterations. There is evidence of aberrant activation of several signaling cascades such as epidermal growth factor receptor (EGFR), Ras/extracellular signal-regulated kinase, phosphoinositol 3-kinase/mammalian target of rapamycin (mTOR), hepatocyte growth factor/mesenchymal-epithelial transition factor, Wnt, Hedgehog, and apoptotic signaling. Recently a multikinase inhibitor, sorafenib, has shown survival benefits in patients with advanced HCC. This advancement represents a breakthrough in the treatment of this complex disease and proves that molecular therapies can be effective in HCC. It is becoming apparent, however, that to overcome the complexity of genomic aberrations in HCC, combination therapies will be critical. Phase II studies have tested drugs blocking EGFR, vascular endothelial growth factor/platelet-derived growth factor receptor, and mTOR signaling. No relevant data has been produced so far in combination therapies. Future research is expected to identify new compounds to block important undruggable pathways, such as Wnt signaling, and to identify new oncogenes as targets for therapies through novel high-throughput technologies. Recent guidelines have established a new frame for the design of clinical trials in HCC. Randomized phase II trials with a time-to-progression endpoint are proposed as pivotal for capturing benefits from novel drugs. Survival remains the main endpoint to measure effectiveness in phase III studies. Patients assigned to the control arm should receive standard-of-care therapy, that is, chemoembolization for patients with intermediate-stage disease and sorafenib for patients with advanced-stage disease. Biomarkers and molecular imaging should be part of the trials, in order to optimize the enrichment of study populations and identify drug responders. Ultimately, a molecular classification of HCC based on genome-wide investigations and identification of patient subclasses according to drug responsiveness will lead to a more personalized medicine.
Article
We have reported previously the activity of the insulin-like growth factor-I (IGF-IR)/insulin receptor (InsR) inhibitor, BMS-554417, in breast and ovarian cancer cell lines. Further studies indicated treatment of OV202 ovarian cancer cells with BMS-554417 increased phosphorylation of HER-2. In addition, treatment with the pan-HER inhibitor, BMS-599626, resulted in increased phosphorylation of IGF-IR, suggesting a reciprocal cross-talk mechanism. In a panel of five ovarian cancer cell lines, simultaneous treatment with the IGF-IR/InsR inhibitor, BMS-536924 and BMS-599626, resulted in a synergistic antiproliferative effect. Furthermore, combination therapy decreased AKT and extracellular signal-regulated kinase activation and increased biochemical and nuclear morphologic changes consistent with apoptosis compared with either agent alone. In response to treatment with BMS-536924, increased expression and activation of various members of the HER family of receptors were seen in all five ovarian cancer cell lines, suggesting that inhibition of IGF-IR/InsR results in adaptive up-regulation of the HER pathway. Using MCF-7 breast cancer cell variants that overexpressed HER-1 or HER-2, we then tested the hypothesis that HER receptor expression is sufficient to confer resistance to IGF-IR-targeted therapy. In the presence of activating ligands epidermal growth factor or heregulin, respectively, MCF-7 cells expressing HER-1 or HER-2 were resistant to BMS-536924 as determined in a proliferation and clonogenic assay. These data suggested that simultaneous treatment with inhibitors of the IGF-I and HER family of receptors may be an effective strategy for clinical investigations of IGF-IR inhibitors in breast and ovarian cancer and that targeting HER-1 and HER-2 may overcome clinical resistance to IGF-IR inhibitors.
Article
Hepatitis B virus infection is associated with acute and chronic liver disease and the development of hepatocellular carcinoma (hcc). Several lines of evidence have suggested that hepatitis B virus X protein (HBx), which is a transcriptional trans-activator, plays a role in the process of liver carcinogenesis. We have investigated the expression of insulin-like growth factor I (IGF-I) receptor in human hepatocellular carcinoma cell lines using SNU368 cells containing HBx and SNU387 cells, which lack HBx gene transcript (J-G. Park et al., Int. J. Cancer, 62: 276-282, 1995), in an attempt to understand its possible relationship to the HBx-induced hcc. The binding of 125I-labeled IGF-I to the SNU368 cells was 5-fold higher than that of SNU387 cells. The Scatchard analysis of the binding data revealed a single class binding site for IGF-I with Kd of 7.6 and 8.8 nM and maximum binding capacities of 169 and 33 fmol/10(5) cells, respectively. Therefore, the difference observed in 125I-labeled IGF-I binding between SNU368 and SNU387 cells was due to an increase in the number of IGF-I binding sites with no change in affinity for the IGF-I receptor. An enhanced level of IGF-I receptors in SNU368 cells was also observed by fluorescence-activated cell sorting analysis using a monoclonal antibody against human IGF-I receptor, alpha IR3. The level of IGF-I receptor RNA and the basal IGF-I receptor gene promoter activity in SNU368 cells were 5 and 10 times higher than those observed in SNU387 cells, respectively. To substantiate further that HBx could transactivate the expression of the endogenous IGF-I receptor gene, Hep G2 cells were transiently transfected with a HBx expression vector. The transfection of Hep G2 cells with an HBx expression vector resulted in increased levels of IGF-I receptor RNA, promoter activity, and 125I-labeled IGF-I binding by 2.6-, 2.8-, and 2-fold, respectively. As a result of higher levels of IGF-I receptor, the mitogenic effect of IGFs (IGF-I and IGF-II) on SNU368 cells was 6 times higher than that of SNU387 cells. These results suggest that HBx may play a role in the process of hcc by activating IGF-I receptor gene expression.