ArticlePDF Available

Combination of cetuximab with met inhibitor in control of cetuximab-resistant oral squamous cell carcinoma

Authors:

Abstract and Figures

Objective: To investigate the underlying molecular mechanisms contributing to oral squamous cell carcinoma (OSCC) cell resistance to the epidermal growth factor receptor (EGFR) inhibitor. Materials and methods: OSCC cell lines HSC-2 and HSC-3 were assessed in vitro for drug treatment, cell viability, and gene expression and the online gene expression in OSCC tissues was analyzed for association with OSCC prognosis. Results: HSC-2 and HSC-3 cells expressed high EGFR levels, but hepatocyte growth factor (HGF) treatment induced cetuximab resistance, whereas the Met inhibitor PHA-665752 as well as Met siRNA was able to restore OSCC cell sensitivity to cetuximab. HGF treatment induced tumor cells to express p-Akt and p-ERK1/2. In contrast, the activity of Akt and ERK1/2 was suppressed by treatment with PHA-665752, Met siRNA, or their combination. Furthermore, Met was highly expressed in OSCC tissues and associated with a poor patient survival, while Met/HGF-activated Akt also was associated with a poor patient survival. Conclusions: This study demonstrates that Met/HGF expression results in OSCC resistance to cetuximab and tumor recurrence after cetuximab therapy; thus, inhibition of Met/HGF activity could restore OSCC sensitivity to cetuximab.
Content may be subject to copyright.
Am J Transl Res 2019;11(4):2370-2381
www.ajtr.org /ISSN:1943-8141/AJTR0088717
Original Article
Combination of cetuximab with met inhibitor in control
of cetuximab-resistant oral squamous cell carcinoma
Hua Yang1, Chuzi Mo1, Yang Xun1, Leyna G Liu2, Wenxing Li3,4, Jieying Guan1, Jing Liu1, Jianquan Wu1, Anping
Yang1, Songguo Zheng5, Dahai Liu1, Fang Liu1
1Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan
University, Foshan 528000, Guangdong, China; 2Portola High School, 1001 Cadence, Irvine, CA 92618, U.S.A;
3The Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese
Academy of Sciences, Kunming 650223, Yunnan, China; 4Kunming College of Life Science, University of Chinese
Academy of Sciences, Kunming 650204, Yunnan, China; 5Department of Medicine, Division of Rheumatology,
Milton S. Hershey Medical Center at Penn State University, Hershey, PA 17033, USA
Received November 20, 2018; Accepted January 29, 2019; Epub April 15, 2019; Published April 30, 2019
Abstract: Objective: To investigate the underlying molecular mechanisms contributing to oral squamous cell car-
cinoma (OSCC) cell resistance to the epidermal growth factor receptor (EGFR) inhibitor. Materials and methods:
OSCC cell lines HSC-2 and HSC-3 were assessed in vitro for drug treatment, cell viability, and gene expression and
the online gene expression in OSCC tissues was analyzed for association with OSCC prognosis. Results: HSC-2 and
HSC-3 cells expressed high EGFR levels, but hepatocyte growth factor (HGF) treatment induced cetuximab resis-
tance, whereas the Met inhibitor PHA-665752 as well as Met siRNA was able to restore OSCC cell sensitivity to
cetuximab. HGF treatment induced tumor cells to express p-Akt and p-ERK1/2. In contrast, the activity of Akt and
ERK1/2 was suppressed by treatment with PHA-665752, Met siRNA, or their combination. Furthermore, Met was
highly expressed in OSCC tissues and associated with a poor patient survival, while Met/HGF-activated Akt also was
associated with a poor patient survival. Conclusions: This study demonstrates that Met/HGF expression results in
OSCC resistance to cetuximab and tumor recurrence after cetuximab therapy; thus, inhibition of Met/HGF activity
could restore OSCC sensitivity to cetuximab.
Keywords: Oral squamous cell carcinoma, cetuximab resistance, HGF, Met, Akt, ERK1/2
Introduction
Head and neck cancer, which occurs in the oral
cavity, nose, throat, larynx, sinuses, salivary
glands, nasopharynx, or hypopharynx, signi-
cantly affects the quality of life of patients [1].
Oral squamous cell carcinoma (OSCC) is a com-
monly diagnosed head and neck squamous cell
carcinoma (HNSCC). The risk factors include
tobacco smoking and alcohol consumption [2]
as well as excessive consumption of processed
meats and red meat [3], human papillomavirus
infection [4], and Frequently chewing betel nuts
[5]. OSCC management is usually surgery,
chemoradiotherapy, targeted therapy, or photo-
dynamic therapy [6, 7]. Early-stage OSCC has a
favorable prognosis after treatment, but drug
resistance, disease recurrence, and metasta-
sis result in overall 5-year survival rates ≤ 50%
[8]. Targeted therapy is a hot topic for the treat-
ment of OSCC, and clinical drugs used frequent-
ly include epidermal growth factor receptor
(EGFR) inhibitors, such as cetuximab, bevaci-
zumab, and erlotinib, which have shown
improvement of OSCC patient survival [9, 10].
Indeed, EGFR is a transmembrane receptor
tyrosine kinase that is highly overexpressed in
head and neck, lung, and breast cancers [11,
12]. EGFR promotes cancer development by
increasing cell growth, migration, and survival
[13]. Cetuximab is a chimeric monoclonal anti-
body that can bind to EGFR and in turn inhibit
EGFR tyrosine kinase activity to suppress EGFR-
positive cancer progression [14]. In the treat-
ment of HNSCC, cetuximab has gained much
attention as a novel therapeutic strategy and
was approved by the European Medicines
Agency in 2004 and the US Food and Drug
Drug combination in treatment of oral cancer
2371 Am J Transl Res 2019;11(4):2370-2381
Administration (FDA) in 2006 [15]. Unfor-
tunately, long-term cetuximab treatment results
in HNSCC drug resistance, even in tumors with
high EGFR expression [16, 17]. Thus, a better
understanding of the underlying molecular
events of this drug resistance is critical to
restore the sensitivity of cancer cells to cetux-
imab [18-20]. To date, there is increasing evi-
dence implicating that receptor tyrosine kinas-
es play a pivotal role in regulating cetuximab
resistance in colon and lung cancers [21, 22],
and the combined treatment with receptor tyro-
sine kinase inhibitors may overcome cetuximab
resistance [22].
Hepatocyte growth factor (HGF) plays a crucial
role in cell motility, growth, and morphogenesis
through binding to hepatocyte growth factor
receptor (also known as Met) to activate the
receptor tyrosine kinase cascade, which is
related to cancer development [23-25]. HGF is
highly expressed in OSCC, and the HGF/Met
signaling pathway has been shown to induce
OSCC cell migration, invasion, and metastasis
through lamellipodia and lopodia formation
[26] and the destruction of E-cadherin [27]. The
cross-talk of Met with other signaling proteins,
like EGFR, vascular endothelial growth factor
receptor, and Wnt, also has been revealed in
various cell lines [28-30], indicating the role of
HGF/Met in cancer development. In addition, it
has been shown that Met activation during
cetuximab treatment of recurrent and meta-
static HNSCC is associated with poor outcomes
[31].
In this study, we investigated the underlying
molecular events contributing to OSCC cell
resistance to cetuximab using an in vitro OSCC
cell line model. Our data provide novel insight
into a therapeutic strategy to restore cetuximab
sensitivity of OSCC.
Materials and methods
Cell culture
The human OSCC cell lines HSC-2 and HSC-3
(Immuno-Biological Laboratories Co. Shanghai,
China) were maintained in Dulbecco’s modied
Eagle medium (DMEM; Gibco, Gaithersburg,
MD, USA) supplemented with 10% fetal bovine
serum (Gibco), 100 units/mL penicillin, and
100 µg/mL streptomycin (Gibco) in a humidi-
ed incubator with 5% CO2 at 37°C.
Cell viability assay
Cell viability was measured by using the 3-(4,
5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium
bromide (MTT) method. In brief, 2 × 103 tumor
cells/well were seeded into 96-well plates and
grown for 24 h and then treated with various
concentrations of cetuximab (Merck, Germany),
PHA-665752 (Selleck Chemicals, TX, USA),
HGF (R&D Systems, Minneapolis, MN, USA),
and a goat anti-human HGF neutralizing anti-
body, normal goat IgG (R&D Systems) for 72 h.
At the end of each experiment, 50 μL of MTT
solution (2 mg/mL; Sigma Chemicals, St. Louis,
MO, USA) was added to each well of the cell cul-
ture plate, and the cells were incubated for an
additional 2 h. The culture medium was
replaced with 100 μL of dimethyl sulfoxide. The
absorbance rate was measured with a micro-
plate reader (BioTek, Winooski, VT, USA) at a
wavelength of 490 nm. The percentage of cell
viability was calculated by comparison to
untreated controls.
Western blot
A western blot assay was performed, as
described previously [32]. Briey, cells after
treatment were lysed in cell lysis buffer, and the
protein concentration was measured using a
bicinchoninic acid protein assay kit (Beyotime
Biotechnology, Shanghai, China). Each total
protein sample (30 µg per lane) was separated
by sodium dodecyl sulfate-polyacrylamide gel
electrophoresis and transferred onto a polyvi-
nylidene uoride membrane (Millipore, Billerica,
MA, USA). After that, the membranes were
blocked by 5% skimmed dry milk solution in
phosphate-buffered saline (PBS) at room tem-
perature for 1 h and then incubated with a pri-
mary antibody at 4°C overnight (Table 1). On
the following day, the membranes were washed
briey three times with PBS-Tween 20 and then
incubated with the corresponding horseradish
peroxidase-conjugated secondary antibody at
room temperature for 1 h. The target protein
bands were visualized by ECL plus western blot-
ting detection reagent and exposed to X-ray
lms.
RNA interference (RNAi) assay
For the RNAi assay, Duplexed Stealth RNAi tar-
geting ErbB3 and Met or Stealth RNAi Negative
Control Low GC Duplex #3 were purchased
from Invitrogen (Carlsbad, CA, USA). OSCC cells
Drug combination in treatment of oral cancer
2372 Am J Transl Res 2019;11(4):2370-2381
were seeded into 24-well plates at a density of
2 × 104 per well in 400 μL of antibiotic-free
DMEM and grown overnight. On the next day,
the cells were transfected with siRNA (50 μmol)
using Lipofectamine 2000 (Invitrogen) for 24 h,
according to the manufacturer’s instructions,
then washed with ice-cold PBS and reseeded
into 96-well plates for treatment with cetux-
imab (1 μg/mL) and/or recombinant human
HGF (20 ng/µL) for 72 h, and nally subjected
to the MTT assay or another assay. The siRNA
sequences targeting ErbB3 and Met were
5’-GGCCAUGAAUGAAUUCUCUACUCUA-3’ and
5’-UCCAGAAGAUCAGUUUCCUAAUUCA-3’, res-
pectively.
Online gene expression database and survival
analysis
Transcriptome data of OSCC tissues were
retrieved from the NCBI-GEO database with the
series ID of GSE42743 (https://www.ncbi.nlm.
nih.gov/geo/query/acc.cgi?acc=GSE42743). R
statistical software v3.3.3 (https://www.rpro-
ject.org/) was used for data analysis. In brief,
we utilized the Robust Multichip Average algo-
rithm in the “oligo” package [33] to normalize
the raw data on gene expression and generat-
ed the normalized expression matrix. The gene
annotation and integration of the expression
matrix was conducted by using a custom-
designed Python code, according to a previous
study [34]. We then removed probes without
any gene annotations or those that matched
multiple gene symbols. After that, we calculat-
ed the average expression value for each gene
when there were multiple probe IDs that
matched one ofcial gene symbol and made
this value to represent the expression intensity
“survival” package in R. Kaplan-Meier survival
curves were used to show the prognostic differ-
ences between two groups.
Statistical analysis
All data were expressed as the mean ± stan-
dard deviation of triplicate experiments. The
two-tailed unpaired Student’s t test or the one-
way analysis of variance test was used to deter-
mine the p values; P < 0.05 was considered as
a signicant difference.
Results
HGF induction of OSCC cell resistance to ce-
tuximab by an increase in akt and ERK1/2
phosphorylation
First, we detected the levels of EGFR and c-Met
expression in the OSCC cell lines HSC-2 and
HSC-3 using western blot. We found that both
cell lines signicantly expressed EGFR and MET
(Figure 1A). We then assessed the effect of
cetuximab on OSCC viability using the MTT
assay and found that HSC-2 and HSC-3 cell
growth was moderately inhibited by cetuximab
in a dose-dependent manner (Figure 2B). To
obtain cetuximab resistance, we cotreated
HSC-2 and HSC-3 cells with cetuximab (5 µg/
mL) and HGF (20 µg/mL); however, we did not
nd a signicant change in cell viability (Figure
1B), indicating that both OSCC cell lines became
resistant to cetuximab after HGF treatment. At
the protein level, HGF treatment induced Met
activity (Figure 1C), which could be partially
blocked by cetuximab treatment. Specically,
cetuximab treatment downregulated the phos-
phorylation of Akt and ERK1/2, but it recovered
back to the previous levels after the addition of
Table 1. Antibodies used for Western blot
Antibody Cat # Source Dilution
Anti-EGFR R&D Systems, AF231 Rabbit 1 µg/mL
Anti-phospho-EGFR Abcam, ab32578 Rabbit 1:1000
Anti-Met CST, 3127 Mouse 1:1000
Anti-phospho-Met Abcam, ab47402 Rabbit 1:1000
Anti-Akt CST, 9272 Rabbit 1:1000
Anti-phospho-Akt CST, 4060 Rabbit 1:1000
Anti-ErbB3 CST, 4754 Rabbit 1:1000
Anti-phospho-ErbB3 CST, 4791 Rabbit 1:1000
Anti-ERK1/ERK2 R&D Systems, AF1576 Rabbit 0.2 μg/mL
Anti-phospho-ERK1/ERK2 R&D Systems, AF1018 Rabbit 0.1 μg/mL
CST, Cell Signaling Technology (Danvers, MA, USA); Abcam, Cambridge, MA, USA.
of the corresponding gene
symbol. Differentially expres-
sed genes were then obtained
by using the empirical Bayes
method in the “limma” pack-
age [35]. The upregulated
genes were considered to be
those with logarithmic trans-
formed fold-change (log2FC)
≥ 1, and the downregulated
genes were considered to be
log2FC ≤ -1. A false discovery
rate-adjusted P value ≤ 0.05
indicated statistical signi-
cance. All survival analyses
were conducted using the
Drug combination in treatment of oral cancer
2373 Am J Transl Res 2019;11(4):2370-2381
HGF (Figure 1C). Under these conditions,
p-ErbB3 was dramatically inhibited with cetux-
imab, even after the addition of HGF, indicating
that p-ErbB3 is the target of cetuximab but not
involved in in the HGF-induced cetuximab resis-
tance of OSCC cells; whereas the Met, PI3K/
Akt, and MAPK pathways may be involved in the
HGF-induced cetuximab resistance of OSCC
cells. For further conrmation, we pretreated
these cells with the HGF neutralizing antibody
to block HGF signaling and found that HSC-2
cells regained their sensitivity to cetuximab
treatment (Figure 1D).
Met inhibitor restoration of cetuximab sensitiv-
ity of HGF-treated OSCC cells by decreasing
akt and ERK1/2 phosphorylation
The biological effects of HGF are through bind-
ing to its receptor, Met; thus, we treated the
cells with the Met inhibitor PHA-665752 in the
same setting. Our MTT assay data showed that
PHA-665752 signicantly reduced the tumor
cell viability in a dose-dependent manner
(Figure 2A). Without the stimulus of HGF, PHA-
665752 and cetuximab together strongly
induced inhibition of cell proliferation, even at a
low dose of PHA-665752 (< 1 µM). After stimu-
lus with HGF, the OSCC cells showed cetuximab
resistance in low-dose PHA-665752-treated
cells, but the cell viability was dramatically
reduced after the PHA-665752 dose reached 1
µM, which was similar to that without the HGF
stimulus.
To evaluate the mechanism by which HGF res-
cues OSCC cancer cells from cetuximab-
induced inhibition of proliferation, we deter-
mined activation of the downstream signaling
molecules and found that cetuximab not only
Figure 1. HGF induction of OSCC resistance to cetuximab. A. Western blot. HSC-2 and HSC-3 cells were subjected
to protein extraction and showed high protein expression levels of EGFR and MET by western blot. B. The MTT as-
say. HSC-2 and HSC-3 cells were treated with different doses of cetuximab (0-5 µg/mL) and HGF (20 ng/mL) for
72 h, and the cell proliferation levels were shown by the MTT assay. C. Western blot. HSC-2 and HSC-3 cells were
pretreated with control, HGF (20 ng/mL), cetuximab (1 µg/mL), or their combination. D. The MTT assay. HSC-2 cells
treated with HGF (20 ng/mL) and the HGF neutralizing antibody or cetuximab (1 µg/mL) for 72 h showed different
proliferation abilities by the MTT assay. *P < 0.05.
Drug combination in treatment of oral cancer
2374 Am J Transl Res 2019;11(4):2370-2381
inhibited EGFR phosphorylation, but also AKT
and ERK1/2 activation in HSC-2 and HSC-3
cells. Treating the cells with a combination of
cetuximab with HGF led to MET phosphoryla-
tion and reactivation of AKT and ERK1/2.
However, when treating the cells with a combi-
nation of cetuximab with PHA-665752, inhibi-
tion of EGFR and MET phosphorylation led to
sustained inhibition of the AKT and MAPK path-
ways in the presence of HGF (Figure 2B).
Met siRNA restoration of cetuximab sensitivity
of HGF-treated OSCC cells
Next, we assessed whether Met siRNA pos-
sesses the same effects as PHA-665752 on
HSC-2 cells; it is because cetuximab had even
better inhibitory effects on the tumor cells and
ErbB3 siRNA was used as a negative control
since ErbB3 is not involved in this effect. The
western blot data showed that Met siRNA was
able to knock down Met expression in OSCC
cells, compared to scrambled and ErbB3 siRNA
transfection (Figure 3A). Moreover, Met siRNA
reduced the OSCC cell viability and reversed
the cetuximab resistance induced by HGF treat-
ment, while ErbB3 siRNA did not show these
effects (Figure 3B). At the protein level, p-Akt
and p-ERK1/2 expression was blocked by
cetuximab treatment and was restored partially
by HGF; however, the expression of these pro-
Figure 2. Treatment with the Met inhibitor restored OSCC cell sensitivity to cetuximab in HGF-induced cetuximab-
resistant OSCC cells and decreased Akt and ERK1/2 phosphorylation. (A) The MTT assay. HSC-2 and HSC-3 cells
were grown and treated with different doses of PHA-665752 or pretreated with or without HGF (20 ng/mL) and
cetuximab (1 µg/mL). (B) Western blot. The cells described in (A) were subjected to western blotting.
Drug combination in treatment of oral cancer
2375 Am J Transl Res 2019;11(4):2370-2381
teins was almost completely blocked by Met
siRNA transfection (Figure 3C).
Association of met and akt expression with
poor OSCC outcomes
To relate our current data clinically, we assess-
ed and retrieved data on the differentially
expressed genes in OSCC tissue samples from
the NCBI-GEO database and then identied the
expression of HGF, Met, ERK1 (also called
MAPK3 in Figure 4D), ERK2 (also called MAPK1
in Figure 4E), and Akt in OSCC tissues. The
Kaplan-Meier curves were plotted against the
expression of these genes and then statistical-
ly analyzed by using the log rank test. We found
that the expression of Met and Akt was associ-
ated with a poor survival of OSCC patients (P =
0.021 and 0.011, respectively; Figure 4A and
4F), whereas HGF expression was associated
with a better patient survival (P = 0.026; Figure
4B). Moreover, interactive analysis of HGF and
Met expression did not yield any statistical sig-
nicance for patient survival (Figure 4C). In
addition, the expression of ERK1 and ERK2
also did not show any signicant association
with patient survival (P = 0.20 and 0.51; Figure
4D and 4E).
Discussion
Cetuximab is a monoclonal antibody used to
treat various human cancers by blockage of
EGFR activation to inhibit the downstream
pathways in tumor cell growth and invasion
[36]. However, primary and acquired cetuximab
resistance occurs in most cancer cases, even if
the tumor cells express a high level of EGFR
[37]. The underlying molecular mechanisms of
drug resistance are complicated. For example,
in colon cancer, resistance to anti-EGFR anti-
bodies is mainly through the irregular activation
of the EGFR downstream proteins by other pro-
teins, like RAS [38], STAT [39], PTEN [40], or
Figure 3. Met siRNA restoration of OSCC cell sensitivity to cetuximab in HGF-induced cetuximab-resistant OSCC
cells. A. Western blot. HSC-2 cells were grown and transiently transfected with Met, negative control, or ErbB3 siRNA
for 48 h and then subjected to western blotting. B. The MTT assay. HSC-2 cells were grown and transiently trans-
fected with Met, negative control, or ErbB3 siRNA for 48 h and then subjected to the MTT assay. C. Western blot.
HSC-2 cells were grown and transiently transfected with Met siRNA for 48 h and then treated with cetuximab (1 µg/
mL) for 72 h before being subjected to western blotting.
Drug combination in treatment of oral cancer
2376 Am J Transl Res 2019;11(4):2370-2381
Met [41]. A single nucleotide polymorphism of
the EGFR gene has been reported to occur in
more than 40% of HNSCC patients, a change
that could affect the afnity of cetuximab to
bind to EGFR protein [42]. In another previous
study, Smad4 expression was found to be criti-
Figure 4. Association of gene expression with the survival of OSCC patients. The online database data were retrieved
and statistically analyzed. A. Association of Met expression with the overall survival (OS) of OSCC patients. High Met
expression predicted a poor OS (P = 0.021). B. Association of HGF expression with the OS of OSCC patients. A high
HGF level was associated with a better OS (P = 0.026). C. Association of Met/HGF expression with the OS of OSCC
patients. The combined expression of Met and HGF failed to predict the OS of OSCC patients. D and E. Association
of ERK1 and ERK2 expression with the OS of OSCC patients. ERK1 and ERK2 expression failed to predict the OS
of OSCC patients. F. Association of Akt expression with the OS of OSCC patients. A high Akt expression predicted a
poor OS of OSCC patients (P = 0.011).
Drug combination in treatment of oral cancer
2377 Am J Transl Res 2019;11(4):2370-2381
cal in mediating cetuximab sensitivity and
resistance [43]. In addition, in cetuximab-sen-
sitive HNSCC A431 cells, cetuximab downregu-
lated p-ERK and p-Akt expression, whereas Akt
phosphorylation was unchanged in cetuximab-
resistant cells [44]. In our current study, we
found that HGF-induced cetuximab resistance
occurred in both HSC-2 and HSC-3 cells with
high levels of p-Akt and p-ERK1/2 when they
were cotreated with cetuximab, indicating that
Akt and ERK1/2 were responsible for mediat-
ing cetuximab resistance in OSCC.
Met protein is frequently highly expressed in
various human cancers. It is activated by its
ligand HGF to trigger activation of the down-
stream signaling pathways and multiple cellular
events [45], like cancer development and
metastasis. However, it has been shown in lung
cancer and hepatocellular carcinoma that aber-
rant HGF-independent Met activation occurs
and then in turn triggers the ErbB3/PI3K/Akt
signaling pathway, leading to cancer progres-
sion [46, 47]. In HNSCC, a previous study has
revealed that ErbB3 is required to activate
EGFR and drug resistance [48]. However, in our
current study, we did not provide any evidence
showing the importance of ErbB3 in HGF-
induced cetuximab resistance in OSCC cells.
Furthermore, in non-small cell lung cancer, it
has been demonstrated that there is a syner-
gistic effect of HGF and EGF on cancer cell
growth and that the cross-talk between Met
and HGF, at the molecular level, is crucial for
tumor cell growth [49]. In colorectal cancer,
HGF induction of Met activation resulted in
cetuximab resistance [50]. Furthermore, Met
siRNA was able to reduce HNSCC cell viability
and migration in vitro and tumor xenograft
growth in vivo [51]. In addition, in human OSCC
tissues, Met expression was signicantly upreg-
ulated compared with that of the adjacent nor-
mal tissues [35]. Met inhibitors exhibited a
strong effect on inhibition of OSCC cell growth
and induction of OSCC cell apoptosis by sup-
pression of Akt and ERK1/2 activities [52]. In
our current study, HGF induced Akt and ERK1/2
phosphorylation, leading to cetuximab-resis-
tant OSCC cells. With the combination of the
Met inhibitor and Met siRNA, OSCC cell viability
was signicantly reduced and Akt and ERK1/2
phosphorylation was dramatically decreased.
Taken together, we speculated that Met and
EGFR cross-talk could synergistically promote
cancer progression and that anti-Met therapy
could be effective even in OSCC cells resistant
to anti-EGFR therapy. In this context, the combi-
nation of cetuximab and Met inhibitor could be
a better option for the treatment of OSCC
patients with primary or acquired cetuximab
resistance.
Indeed, different Met inhibitors are under clini-
cal trials, and crizotinib and cabozantinib were
approved by the US FDA to treat NSCLC and
medullary thyroid cancer, respectively [53].
Another Met inhibitor, PHA-665752, is a com-
pound used to inhibit Met phosphorylation and
the downstream signaling cascades [54]. In
ovarian cancer, PHA-665752 and Met siRNA
have been shown to inhibit tumor cell growth
and overcome cisplatin resistance [55]. While
in colorectal cancer, the combination of PHA-
665752 and cetuximab signicantly decreased
tumor cell proliferation compared with that of
either agent alone [56]. In our current study,
PHA-665752 was shown to resensitize OSCC
cells to cetuximab resistance, and its combina-
tion with cetuximab had a better antitumor ef-
cacy. At the molecular level, our current data
showed that both HGF and cetuximab were
able to regulate Akt and ERK1/2 activities.
Indeed, Yu et al. have demonstrated that the
PI3K/Akt/mTOR pathway is an ideal target for
controlling OSCC because the combination of
PI3K/Akt inhibitor with radiation improved the
radiation efcacy for the treatment of OSCC
[57]. Targeting of the PI3K/Akt/mTOR pathway
also improved the effects of doxorubicin on its
antitumor activity in OSCC [58]. Furthermore,
sulfasalazine (SSZ), an anti-inammatory drug,
has been demonstrated to have a potential
therapeutic ability in the treatment of OSCC by
promoting autophagy-induced tumor cell death
and inhibiting the PI3K/Akt and MAPK path-
ways [59]. Another previous study has revealed
that activation of the ERK, Akt, and p38 path-
ways is involved in HGF- and EGF-induced OSCC
cell migration [60]. Taken together, the PI3K/
Akt and MAPK pathways could be involved in
OSCC development and progression, and these
signaling pathways are activated by different
upstream factors, e.g. EGFR and Met. When
OSCC cells became sensitive to cetuximab
treatment, the Akt and ERK activities were sig-
nicantly suppressed in OSCC cells, leading to
a decrease in OSCC progression; whereas in
cetuximab-resistant cells, the cetuximab-EGFR-
Drug combination in treatment of oral cancer
2378 Am J Transl Res 2019;11(4):2370-2381
Akt/ERK axis was aberrant, but there was an
increase in HGF-induced Met activity to induce
Akt/ERK phosphorylation, leading to OSCC cell
proliferation. However, blockage of these two
signaling pathways using the combination of
cetuximab and Met inhibitor or Met siRNA inhib-
ited Akt and ERK phosphorylation, resulting in
cell growth inhibition. Thus, our current data
provide insightful information regarding the
effects of the combined treatment of cetux-
imab and Met inhibitor as a novel therapeutic
strategy in the control of OSCC.
In addition, our ex vivo data analysis showed
that the expression of the cell growth signaling
proteins Akt and Met was associated with a
poor survival of OSCC patients. However, due to
the small sample size, the expression of Met
with HGF did not show statistically signicant
data that predict OSCC patient survival. Future
studies with a large sample size should be per-
formed to conrm our current ndings. However,
our current study does have some limitations;
for instance, all the experiments were per-
formed in vitro, and in vivo experiments are
needed to assess and validate the effects of
the combined treatment of cetuximab and Met
inhibitor on the control of OSCC in the future.
Furthermore, our ex vivo data lacked a suf-
cient sample size as well as cetuximab or Met
inhibitor treatment data for OSCC patients.
Conclusions
Our current study revealed that HGF activated
Met, subsequently increasing the phosphoryla-
tion of the downstream PI3K/Akt and MAPK
pathways, which were responsible for the
cetuximab resistance of OSCC, and that inhibi-
tion of Met expression or activity restored OSCC
sensitivity to cetuximab treatment. Future stud-
ies are needed to conrm the effects of the
combination treatment of cetuximab with Met
inhibitor in the control of OSCC clinically.
Acknowledgements
This study was supported in part by grants from
the Foshan Science and Technology Innovation
Project (#2017AB002001 and #2015AG1-
0010), the National Natural Science Founda-
tion of China (#81570202, #81570376 and
#81870307), the University Special Innovative
Research Program of Department of Education
of Guangdong Province (#2017KTSCX189) and
a Project of DEGP (#2015KTSCX154).
Disclosure of conict of interest
None.
Abbreviations
Akt, Protein kinase B; c-Met, hepatocyte growth
factor receptor; EGFR, epidermal growth factor
receptor; ERK1/2, Extracellular signal-regulat--regulat-
ed kinase; HGF, Hepatocyte growth factor;
OSCC, Oral squamous cell carcinoma.
Address correspondence to: Dahai Liu and Fang Liu,
Department of Basic Medicine and Biomedical
Engineering, The School of Stomatology and Me-
dicine, Foshan University, 5 Hebin Road, Chancheng
District, Foshan 528000, Guangdong, China. E-mail:
seansean2014@126.com (DHL); 542998816@qq.
com (FL)
References
[1] Olshan AF, Weissler MC, Watson MA, Bell DA.
GSTM1, GSTT1, GSTP1, CYP1A1, and NAT1
polymorphisms, tobacco use, and the risk of
head and neck cancer. Cancer Epidemiol
Biomarkers Prev 2000; 9: 185-191.
[2] Heidel R. Reducing the health consequences
of smoking: 25 years of progress. a report of
the surgeon general. Us Department of Health
and Human Services. Public Health Service
Cdc 1989; 89: 8411.
[3] Levi F, Pasche C, La Vecchia C, Lucchini F,
Franceschi S, Monnier P. Food groups and risk
of oral and pharyngeal cancer. Int J Cancer
1998; 77: 705-709.
[4] D’Souza G, Kreimer AR, Viscidi R, Pawlita M,
Fakhry C, Koch WM, Westra WH, Gillison ML.
Case-control study of human papillomavirus
and oropharyngeal cancer. N Engl J Med 2007;
356: 1944-1956.
[5] Jeng JH, Chang MC, Hahn LJ. Role of areca nut
in betel quid-associated chemical carcinogen-
esis: current awareness and future perspec-
tives. Oral Oncol 2001; 37: 477-492.
[6] Maxwell JH, Thompson LD, Brandwein-Gensler
MS, Weiss BG, Canis M, Purgina B, Prabhu AV,
Lai C, Shuai Y, Carroll WR, Morlandt A, Duvvuri
U, Kim S, Johnson JT, Ferris RL, Seethala R,
Chiosea SI. Early oral tongue squamous cell
carcinoma: sampling of margins from tumor
bed and worse local control. JAMA Otolaryngol
Head Neck Surg 2015; 141: 1104-1110.
[7] Al-Sarraf M. Treatment of locally advanced
head and neck cancer: historical and critical
review. Cancer Control 2002; 9: 387-399.
[8] Wu CF, Lee CH, Hsi E, Chen CH, Tang JY. Interval
between intra-arterial infusion chemotherapy
and surgery for locally advanced oral squa-
Drug combination in treatment of oral cancer
2379 Am J Transl Res 2019;11(4):2370-2381
mous cell carcinoma: impacts on effectiveness
of chemotherapy and on overall survival.
ScienticWorldJournal 2014; 2014: 568145.
[9] Bonner JA, Harari PM, Giralt J, Azarnia N, Shin
DM, Cohen RB, Jones CU, Sur R, Raben D,
Jassem J, Ove R, Kies MS, Baselga J,
Youssouan H, Amellal N, Rowinsky EK, Ang
KK. Radiotherapy plus cetuximab for squa-
mous-cell carcinoma of the head and neck. N
Engl J Med 2006; 354: 567-578.
[10] Burtness B, Goldwasser MA, Flood W, Mattar
B, Forastiere AA. Phase III randomized trial of
cisplatin plus placebo compared with cisplatin
plus cetuximab in metastatic/recurrent head
and neck cancer: an eastern cooperative on-
cology group study. J Clin Oncol 2005; 23:
8646-8654.
[11] Lynch TJ, Bell DW, Sordella R, Gurubhagavatula
S, Okimoto RA, Brannigan BW, Harris PL,
Haserlat SM, Supko JG, Haluska FG, Louis DN,
Christiani DC, Settleman J, Haber DA. Activating
mutations in the epidermal growth factor re-
ceptor underlying responsiveness of non-
small-cell lung cancer to getinib. N Engl J Med
2004; 350: 2129-2139.
[12] Abbas A, Aster J, Kumar V. Robbins Basic
Pathology. 9th edition. 2012.
[13] Seshacharyulu P, Ponnusamy MP, Haridas D,
Jain M, Ganti AK, Batra SK. Targeting the EGFR
signaling pathway in cancer therapy. Expert
Opin Ther Targets 2012; 16: 15-31.
[14] Wong SF. Cetuximab: an epidermal growth fac-
tor receptor monoclonal antibody for the treat-
ment of colorectal cancer. Clin Ther 2005; 27:
684-694.
[15] Concu R, Cordeiro M. Cetuximab and the head
and neck squamous cell cancer. Curr Top Med
Chem 2018; 18: 192-198.
[16] Shaib W, Kono S, Saba N. Antiepidermal
growth factor receptor therapy in squamous
cell carcinoma of the head and neck. J Oncol
2012; 2012: 521215.
[17] Pao W, Miller VA, Politi KA, Riely GJ, Somwar R,
Zakowski MF, Kris MG, Varmus H. Acquired re-
sistance of lung adenocarcinomas to getinib
or erlotinib is associated with a second muta-
tion in the EGFR kinase domain. PLoS Med
2005; 2: e73.
[18] Bardelli A, Corso S, Bertotti A, Hobor S, Valtorta
E, Siravegna G, Sartore-Bianchi A, Scala E,
Cassingena A, Zecchin D, Apicella M, Migliardi
G, Galimi F, Lauricella C, Zanon C, Perera T,
Veronese S, Corti G, Amatu A, Gambacorta M,
Diaz LA Jr, Sausen M, Velculescu VE, Comoglio
P, Trusolino L, Di Nicolantonio F, Giordano S,
Siena S. Amplication of the MET receptor
drives resistance to anti-EGFR therapies in
colorectal cancer. Cancer Discov 2013; 3:
658-673.
[19] Diaz LA Jr, Williams RT, Wu J, Kinde I, Hecht JR,
Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA,
Kinzler KW, Oliner KS, Vogelstein B. The mo-
lecular evolution of acquired resistance to tar-
geted EGFR blockade in colorectal cancers.
Nature 2012; 486: 537-540.
[20] Brand TM, Iida M, Wheeler DL. Molecular
mechanisms of resistance to the EGFR mono-
clonal antibody cetuximab. Cancer Biol Ther
2011; 11: 777-792.
[21] Wheeler DL, Huang S, Kruser TJ, Nechrebecki
MM, Armstrong EA, Benavente S, Gondi V, Hsu
KT, Harari PM. Mechanisms of acquired resis-
tance to cetuximab: role of HER (ErbB) family
members. Oncogene 2008; 27: 3944-3956.
[22] Hu S, Dai H, Li T, Tang Y, Fu W, Yuan Q, Wang F,
Lv G, Lv Y, Fan X, Zhang S, Jin R, Shen Y, Lin F,
Ye X, Ding M, Yang Y, Lei C. Broad RTK-targeted
therapy overcomes molecular heterogeneity-
driven resistance to cetuximab via vectored
immunoprophylaxis in colorectal cancer.
Cancer Lett 2016; 382: 32-43.
[23] Bottaro DP, Rubin JS, Faletto DL, Chan AM,
Kmiecik TE, Vande Woude GF, Aaronson SA.
Identication of the hepatocyte growth factor
receptor as the c-met proto-oncogene product.
Science 1991; 251: 802-804.
[24] Johnson M, Koukoulis G, Matsumoto K,
Nakamura T, Iyer A. Hepatocyte growth factor
induces proliferation and morphogenesis in
nonparenchymal epithelial liver cells. He-
patology 1993; 17: 1052-1061.
[25] Blumenschein GR Jr, Mills GB, Gonzalez-
Angulo AM. Targeting the hepatocyte growth
factor-cMET axis in cancer therapy. J Clin Oncol
2012; 30: 3287-3296.
[26] Yasui H, Ohnishi Y, Nakajima M, Nozaki M.
Migration of oral squamous cell carcinoma
cells are induced by HGF/c-Met signalling via
lamellipodia and lopodia formation. Oncol
Rep 2017; 37: 3674-3680.
[27] Murai M, Shen X, Huang L, Carpenter WM,
Lin CS, Silverman S, Regezi J, Kramer RH.
Overexpression of c-met in oral SCC promotes
hepatocyte growth factor-induced disruption of
cadherin junctions and invasion. Int J Oncol
2004; 25: 831-840.
[28] Engelman JA, Zejnullahu K, Mitsudomi T, Song
Y, Hyland C, Park JO, Lindeman N, Gale CM,
Zhao X, Christensen J, Kosaka T, Holmes AJ,
Rogers AM, Cappuzzo F, Mok T, Lee C, Johnson
BE, Cantley LC, Janne PA. MET amplication
leads to getinib resistance in lung cancer by
activating ERBB3 signaling. Science 2007;
316: 1039-1043.
[29] Sulpice E, Ding S, Muscatelli-Groux B, Berge M,
Han ZC, Plouet J, Tobelem G, Merkulova-
Rainon T. Cross-talk between the VEGF-A and
Drug combination in treatment of oral cancer
2380 Am J Transl Res 2019;11(4):2370-2381
HGF signalling pathways in endothelial cells.
Biol Cell 2009; 101: 525-539.
[30] Vermeulen L, De Sousa EMF, van der Heijden
M, Cameron K, de Jong JH, Borovski T, Tuynman
JB, Todaro M, Merz C, Rodermond H, Sprick
MR, Kemper K, Richel DJ, Stassi G, Medema
JP. Wnt activity denes colon cancer stem cells
and is regulated by the microenvironment. Nat
Cell Biol 2010; 12: 468-476.
[31] Madoz-Gurpide J, Zazo S, Chamizo C, Casado
V, Carames C, Gavin E, Cristobal I, Garcia-
Foncillas J, Rojo F. Activation of MET pathway
predicts poor outcome to cetuximab in pa-
tients with recurrent or metastatic head and
neck cancer. J Transl Med 2015; 13: 282.
[32] Liu T, Sun Q, Li Q, Yang H, Zhang Y, Wang R, Lin
X, Xiao D, Yuan Y, Chen L. Dual PI3K/mTOR in-
hibitors, GSK2126458 and PKI-587, suppress
tumor progression and increase radiosensitiv-
ity in nasopharyngeal carcinoma. Mol Cancer
Ther 2015; 14: 429-439.
[33] Carvalho BS, Irizarry RA. A framework for
oligonucleotide microarray preprocessing.
Bioinformatics 2010; 26: 2363-2367.
[34] Li WX, Dai SX, Wang Q, Guo YC, Hong Y, Zheng
JJ, Liu JQ, Liu D, Li GH, Huang JF. Integrated
analysis of ischemic stroke datasets revealed
sex and age difference in anti-stroke targets.
PeerJ 2016; 4: e2470.
[35] Ritchie ME, Phipson B, Wu D, Hu Y, Law CW,
Shi W, Smyth GK. limma powers differential ex-
pression analyses for RNA-sequencing and mi-
croarray studies. Nucleic Acids Res 2015; 43:
e47.
[36] Wong SF. Cetuximab: an epidermal growth fac-
tor receptor monoclonal antibody for the treat-
ment of colorectal cancer. Clin Ther 2005; 27:
684-94.
[37] Iida M, Brand TM, Starr MM, Huppert EJ,
Luthar N, Bahrar H, Coan JP, Pearson HE,
Salgia R, Wheeler DL. Overcoming acquired re-
sistance to cetuximab by dual targeting HER
family receptors with antibody-based therapy.
Mol Cancer 2014; 13: 242.
[38] Misale S, Arena S, Lamba S, Siravegna G, Lallo
A, Hobor S, Russo M, Buscarino M, Lazzari L,
Sartore-Bianchi A, Bencardino K, Amatu A,
Lauricella C, Valtorta E, Siena S, Di Nicolantonio
F, Bardelli A. Blockade of EGFR and MEK inter-
cepts heterogeneous mechanisms of acquired
resistance to anti-EGFR therapies in colorectal
cancer. Sci Transl Med 2014; 6: 224ra226.
[39] Li Q, Zhang D, Chen X, He L, Li T, Xu X, Li M.
Nuclear PKM2 contributes to getinib resis-
tance via upregulation of STAT3 activation in
colorectal cancer. Sci Rep 2015; 5: 16082.
[40] Zhang YJ, Tian XQ, Sun DF, Zhao SL, Xiong H,
Fang JY. Combined inhibition of MEK and
mTOR signaling inhibits initiation and progres-
sion of colorectal cancer. Cancer Invest 2009;
27: 273-285.
[41] Gherardi E, Birchmeier W, Birchmeier C, Vande
Woude G. Targeting MET in cancer: rationale
and progress. Nat Rev Cancer 2012; 12: 89-
103.
[42] Braig F, Kriegs M, Voigtlaender M, Habel B,
Grob T, Biskup K, Blanchard V, Sack M,
Thalhammer A, Ben Batalla I, Braren I, Laban
S, Danielczyk A, Goletz S, Jakubowicz E, Markl
B, Trepel M, Knecht R, Riecken K, Fehse B,
Loges S, Bokemeyer C, Binder M. Cetuximab
resistance in head and neck cancer is mediat-
ed by EGFR-K521 polymorphism. Cancer Res
2017; 77: 1188-1199.
[43] Ozawa H, Ranaweera RS, Izumchenko E,
Makarev E, Zhavoronkov A, Fertig EJ, Howard
JD, Markovic A, Bedi A, Ravi R, Perez J, Le QT,
Kong CS, Jordan RC, Wang H, Kang H, Quon H,
Sidransky D, Chung CH. SMAD4 loss is associ-
ated with cetuximab resistance and induction
of MAPK/JNK activation in head and neck can-
cer cells. Clin Cancer Res 2017; 23: 5162-
5175.
[44] Rebucci M, Peixoto P, Dewitte A, Wattez N, De
Nuncques MA, Rezvoy N, Vautravers-Dewas C,
Buisine MP, Guerin E, Peyrat JP, Lartigau E,
Lansiaux A. Mechanisms underlying resis-
tance to cetuximab in the HNSCC cell line: role
of AKT inhibition in bypassing this resistance.
Int J Oncol 2011; 38: 189-200.
[45] Stella MC, Comoglio PM. HGF: a multifunction-
al growth factor controlling cell scattering. Int J
Biochem Cell Biol 1999; 31: 1357-1362.
[46] Engelman JA, Zejnullahu K, Mitsudomi T, Song
Y, Hyland C, Park JO, Lindeman N, Gale CM,
Zhao X, Christensen J. MET amplication leads
to getinib resistance in lung cancer by activat-
ing ERBB3 signaling. Science 2007; 316:
1039-1043.
[47] Steinway SN, Dang H, You H, Rountree CB,
Ding W. The EGFR/ErbB3 pathway acts as a
compensatory survival mechanism upon c-met
inhibition in human c-met+ hepatocellular car-
cinoma. PLoS One 2015; 10: e0128159.
[48] Redlich N, Robinson AM, Nickel KP, Stein AP,
Wheeler DL, Adkins DR, Uppaluri R, Kimple RJ.
Anti-Trop2 blockade enhances the therapeutic
efcacy of ErbB3 inhibition in head and neck
squamous cell carcinoma. Cell Death Dis
2018; 9: 5.
[49] Puri N, Salgia R. Synergism of EGFR and c-Met
pathways, cross-talk and inhibition, in non-
small cell lung cancer. J Carcinog 2008; 7: 9.
[50] Liska D, Chen CT, Bachleitner-Hofmann T,
Christensen JG, Weiser MR. HGF rescues
colorectal cancer cells from EGFR inhibition
via MET activation. Clin Cancer Res 2011; 17:
472-482.
Drug combination in treatment of oral cancer
2381 Am J Transl Res 2019;11(4):2370-2381
[51] Tao X, Hill KS, Gaziova I, Sastry SK, Qui S,
Szaniszlo P, Fennewald S, Resto VA, Elferink
LA. Silencing Met receptor tyrosine kinase sig-
naling decreased oral tumor growth and in-
creased survival of nude mice. Oral Oncol
2014; 50: 104-112.
[52] Sun Z, Liu Q, Ye D, Ye K, Yang Z, Li D. Role of
c-Met in the progression of human oral squa-
mous cell carcinoma and its potential as a
therapeutic target. Oncol Rep 2018; 39: 209-
216.
[53] Kazandjian D, Blumenthal GM, Chen HY, He K,
Patel M, Justice R, Keegan P, Pazdur R. FDA
approval summary: crizotinib for the treatment
of metastatic non-small cell lung cancer with
anaplastic lymphoma kinase rearrangements.
Oncologist 2014; 19: e5-11.
[54] Christensen JG, Schreck R, Burrows J,
Kuruganti P, Chan E, Le P, Chen J, Wang X,
Ruslim L, Blake R, Lipson KE, Ramphal J, Do S,
Cui JJ, Cherrington JM, Mendel DB. A selective
small molecule inhibitor of c-Met kinase inhib-
its c-met-dependent phenotypes in vitro and
exhibits cytoreductive antitumor activity in
vivo. Cancer Res 2003; 63: 7345-7355.
[55] Li E, Hu Z, Sun Y, Zhou Q, Yang B, Zhang Z, Cao
W. Small molecule inhibitor of c-Met
(PHA665752) suppresses the growth of ovari-
an cancer cells and reverses cisplatin resis-
tance. Tumour Biol 2016; 37: 7843-7852.
[56] Jia YT, Yang DH, Zhao Z, Bi XH, Han WH, Feng
B, Zhi J, Gu B, Duan Z, Wu JH, Ju YC, Wang MX,
Li ZX. Effects of PHA-665752 and cetuximab
combination treatment on in vitro and murine
xenograft growth of human colorectal cancer
cells with KRAS or BRAF mutations. Curr
Cancer Drug Targets 2018; 18: 278-286.
[57] Yu CC, Hung SK, Lin HY, Chiou WY, Lee MS,
Liao HF, Huang HB, Ho HC, Su YC. Targeting the
PI3K/AKT/mTOR signaling pathway as an ef-
fectively radiosensitizing strategy for treating
human oral squamous cell carcinoma in vitro
and in vivo. Oncotarget 2017; 8: 68641-
68653.
[58] Smolensky D, Rathore K, Bourn J, Cekanova
M. Inhibition of the PI3K/AKT pathway sensi-
tizes oral squamous cell carcinoma cells to
anthracycline-based chemotherapy in vitro. J
Cell Biochem 2017; 118: 2615-2624.
[59] Han HY, Kim H, Jeong SH, Lim DS, Ryu MH.
Sulfasalazine induces autophagic cell death in
oral cancer cells via akt and ERK pathways.
Asian Pac J Cancer Prev 2014; 15: 6939-
6944.
[60] Brusevold IJ, Aasrum M, Bryne M, Christoffersen
T. Migration induced by epidermal and hepato-
cyte growth factors in oral squamous carcino-
ma cells in vitro: role of MEK/ERK, p38 and
PI-3 kinase/Akt. J Oral Pathol Med 2012; 41:
547-558.
... Another resistance mechanism involves the abnormal activation of parallel signaling pathways to counteract the blockade of the EGFR signaling by CTX. Thus, cancer cell survival is ensured by increased expression/activation of alternative receptor tyrosine kinases (RTK) (50, 77,79,123,[135][136][137][138][139][140][141][142][143][144][145][146][147][148][149][150], ensuring the activation of several parallel pathways, including the VEGF pathway (72,(151)(152)(153). ...
... The binding ligand-extracellular domain induces receptor dimerization, activation of the intrinsic tyrosine-kinase activity of the RTK, and activation of downstream signaling cascades implicated in cell proliferation, differentiation, motility, survival, and cell-cell communication (155). Thus, activation of these RTKs is a mechanism of resistance to CTX during HNSCC treatment (50, 77,79,123,[135][136][137][138][139][140][141][142][143][144][145][146][147][148][149][150]. ...
... The BET inhibitor JQ1, which binds preferentially to the bromodomains of BRD4, abrogates the expression of the alternative RTK (HER3 and AXL), resulting in significantly delayed acquired resistance in two PDX models of HNSCC (148). Combined with CTX, MET inhibitor PHA-665752 is also shown to restore CTX sensitivity in vitro and in vivo, especially by decreasing akt and ERK1/2 phosphorylation (146,147). Inhibition of the AXL receptor is explored by using imatinib (which targets c-Abl) in CTXresistant HNSCC PDX (156). This led to complete tumor regression and a prolonged effect (no recurrence up to 3 months after cessation of treatment). ...
Article
Full-text available
Head and neck squamous cell carcinoma (HNSCC) is the sixth most incident cancer worldwide. More than half of HNSCC patients experience locoregional or distant relapse to treatment despite aggressive multimodal therapeutic approaches that include surgical resection, radiation therapy, and adjuvant chemotherapy. Before the arrival of immunotherapy, systemic chemotherapy was previously employed as the standard first-line protocol with an association of cisplatin or carboplatin plus 5-fluorouracil plus cetuximab (anti-EFGR antibody). Unfortunately, acquisition of therapy resistance is common in patients with HNSCC and often results in local and distant failure. Despite our better understanding of HNSCC biology, no other molecular-targeted agent has been approved for HNSCC. In this review, we outline the mechanisms of resistance to the therapeutic strategies currently used in HNSCC, discuss combination treatment strategies to overcome them, and summarize the therapeutic regimens that are presently being evaluated in early- and late-phase clinical trials.
... In OSCC, Yang et al reported that the inhibition of hepatocyte growth factor-MET activity sensitizes OSCC cells to cetuximab. 19 However, little is known about the molecular mechanism by which OSCC cells escape the antiproliferative effect of EGFR-targeted drugs. Therefore, it would be of particular interest to discover alternative signaling for the survival of OSCC under EGFR inhibition. ...
Article
Full-text available
Background: Epidermal growth factor receptor (EGFR) is frequently overexpressed in oral squamous cell carcinoma (OSCC), and EGFR-targeting therapeutics have been widely employed to treat patients with a variety of carcinomas including OSCC. Here, we aimed to investigate alternative signaling for OSCC survival under the disruption of EGFR signaling. Methods: OSCC cell lines, namely HSC-3 and SAS, were utilized to investigate how EGFR disruption affects cell proliferation. Gene set enrichment analysis was performed to examine how EGFR disruption affects oncogenic signaling in OSCC cells. Disruption of KDR gene was performed using CRISPR/Cas9 techniques. A VEGFR inhibitor, vatalanib was used to research the impact of VEGFR inhibition on OSCC survival. Results: EGFR disruption significantly decreased the proliferation and oncogenic signaling including Myc and PI3K-Akt, in OSCC cells. Chemical library screening assays revealed that VEGFR inhibitors continued to inhibit the proliferation of EGFR-deficient OSCC cells. In addition, CRISPR-mediated disruption of KDR/VEGFR2 retarded OSCC cell proliferation. Furthermore, combined erlotinib-vatalanib treatment exhibited a more potent anti-proliferative effect on OSCC cells, compared to either monotherapy. The combined therapy effectively suppressed the phosphorylation levels of Akt but not p44/42. Conclusion: VEGFR-mediated signaling would be an alternative signaling pathway for the survival of OSCC cells under the disruption of EGFR signaling. These results highlight the clinical application of VEGFR inhibitors in the development of multi-molecular-targeted therapeutics against OSCC.
... Therefore, early detection and prevention play a vital role in controlling the burden of oral cancer. Currently used therapies for oral cancer include surgery, radiation therapy and chemotherapy alone or their combination, such as anlotinib [6], cetuximab [7] and PD-1/PD-L1 [8]. Cisplatin and 5-fluorouracil are commonly used in combination, and cisplatin is effective when used alone as well [9], however, the survival rate remains poor. ...
Article
Full-text available
Oral cancer (OC) has been attracted research attention in recent years as result of its high morbidity and mortality. Costunolide (CTD) possesses potential anticancer and bioactive abilities that have been confirmed in several types of cancers. However, its effects on oral cancer remain unclear. This study investigated the potential anticancer ability and underlying mechanisms of CTD in OC in vivo and in vitro. Cell viability and anchorage-independent colony formation assays were performed to examine the antigrowth effects of CTD on OC cells; assessments for migration and invasion of OC cells were conducted by transwell; Cell cycle and apoptosis were investigated by flow cytometry and verified by immunoblotting. The results revealed that CTD suppressed the proliferation, migration and invasion of oral cancer cells effectively and induced cell cycle arrest and apoptosis; regarding the mechanism, CTD bound to AKT directly by binding assay and repressed AKT activities through kinase assay, which thereby downregulating the downstream of AKT. Furthermore, CTD remarkably promotes the generation of reactive oxygen species by flow cytometry assay, leading to cell apoptosis. Notably, CTD strongly suppresses cell-derived xenograft OC tumor growth in an in vivo mouse model. In conclusion, our results suggested that costunolide might prevent progression of OC and promise to be a novel AKT inhibitor.
... In recent years, treatment options, particularly for advanced (III and IV) malignancies, have been applied including surgery, radiation and/or chemotherapy as well as targeted therapy. [1,7]. Despite huge advances in diagnosis and treatment, the toxic side effects and drug resistance remain the main causes of high mortality and poor mortality in OSCC [1,3]. ...
Preprint
Full-text available
Natural autoantibodies serve as an important anti-tumorigenic system due to its immune surveillance function. The present study aims to investigate whether circulating natural IgG autoantibodies against cluster of differentiation 47 (CD47) could suppress the proliferation of oral squamous cell carcinoma (OSCC) cells. Three OSCC cell lines were used for this study. The expression of fourteen tumor related genes including CD47 were tested. OSCC cells were grown, respectively, with 20% human plasma positive and negative for anti-CD47 IgG. Cell proliferation, apoptosis and invasion/metastasis were examined. The results showed that CD47 presented highest expression among all 14 genes detected in OSCC cells. Plasma anti-CD47 IgG significantly inhibited the viability of all three OSCC cell lines. The proportions of apoptotic cells were remarkably higher in OSCC cells treated with anti-CD47 IgG-positive plasma than those treated with IgG-negative plasma. Furthermore, the cell invasion/metastasis was attenuated evidently with the attribution of plasma anti-CD47 IgG. In conclusion, natural autoantibodies against CD47 may be a potential target for OSCC immunotherapy.
... During epithelial to mesenchymal transition (EMT), CD44 expression increases conferring resistance to Cetuximab [110]. In another study, Cetuximab resistant cells had shown increased MET (HGF binding receptor), which activates AKT signalling and treating those cells with MET and Cetuximab inhibited cell growth by inactivating ERK and AKT [111]. This study provides a base for synergistic usage of MET inhibitor and EGFR inhibitor in Cetuximab-resistant OSCC. ...
Article
Full-text available
Oral cancer ranks sixth most prevalent type of cancer worldwide due to its alarming increase every year. Progression of oral cancer depends on various heterogeneity pathways, but their exact mechanism remains unclear. Genetic aberrations on oral cancer cells set back the effectiveness of existing therapies and make it more challenging by triggering drug resistance. To understand the intricate details of oral cancer pathogenesis and for advancing current therapies, genetic modifications are the most promising approach. In this review, we tabulated the information on genetic alterations, microbial associations, aberrant signalling pathways and their clinicopathological characters on the pathogenesis of oral cancer. We primarily discussed the pitfalls of the current treatment regimen and its associated drug resistance pattern, which will provide a clear insight into developing new drugs. We also highlighted the genetic-molecular targets with their current clinical status on drug development and its outcome.
Article
Objective: Integrin subunit beta 4 (ITGB4), a receptor for laminins, was an oncoprotein in several malignancies. However, its clinical role in oral squamous cell carcinoma (OSCC) remains unclear. Materials and methods: Firstly, 99 OSCC and 13 normal oral epithelium samples were employed for immunohistochemistry (IHC) for detecting the expression level of ITGB4 protein in OSCC. Subsequently, 971 OSCC and 281 non-cancerous specimens from RNA-seq and 18 microarrays were applied for investigating the expression of ITGB4 mRNA. Furthermore, to explore the potential mechanism of ITGB4 in OSCC, the co-expressed genes of ITGB4 were initially screened using all available datasets, and were further utilized for the gene enrichment analysis. Results: First, IHC showed a distinctively higher expression level of the ITGB4 protein in the OSCC group than that in the normal controls. Second, expression profile from RNA-seq and microarrays reflected that ITGB4 mRNA was dramatically overexpressed in OSCC tissues compared with non-tumor tissues. Third, standardized mean difference (SMD) with the area under the summary receiver operating characteristic (sROC) curve combining all incorporated data revealed that ITGB4 was consistently significantly upregulated in OSCC tissues, with the SMD value being 1.31 and the area under the sROC curve being 0.82. Lastly, 184 upregulated and 179 downregulated co-expressed genes of ITGB4 were utilized for enrichment analysis, which demonstrated that ITGB4 might influence the pathogenesis of OSCC through cell cycle, ECM-receptor interaction and focal adhesion pathways. Conclusions: ITGB4 might play a pivotal role in the tumorigenesis and progression of OSCC, making it a promising biomarker of OSCC.
Article
Epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein, which has been considered as one of the key targets for cancer therapy. However, currently approved therapeutic anti‐EGFR antibody may cause the hypersensitivity reaction induced by galactose‐α‐1,3‐galactose (α‐Gal) structure, which is inevitable in insect cell expression system. In this study, the Chinese hamster ovary cell line was used to produce a monoclonal antibody containing simplified glycosylation patterns (code: AB01). And cetuximab was used as a control. The two antibodies were highly similar in molecular weight, secondary structure, binding affinity and endocytosis behavior, whereas the glycotypes are extremely distinct. The flow cytometry assay suggested that AB01 induced cell cycle arrest in G1, thus inhibit cell proliferation. Moreover, both cetuximab and AB01 showed similar sensitivity for all tested cell lines in this research. In conclusion, AB01 could be a potential anti‐EGFR drug candidate for cancer therapy.
Article
Objectives Natural autoantibodies serve as an important anti-tumorigenic component in the body. This study was thus designed to investigate whether circulating natural IgG autoantibodies against cluster of differentiation 47 (CD47) could exert inhibitory effects on oral squamous cell carcinoma (OSCC). Subjects and Methods The expression levels of 13 tumor-targeted genes in three OSCC cell lines were analyzed by qPCR, and CD47 expression in OSCC tissues was also verified with IHC staining. An in-house ELISA was performed to analyze circulating anti-CD47 IgG levels in control subjects, oral benign tumor and OSCC patients, and to detect anti-CD47 IgG-abundant plasma. Three OSCC cell lines were treated with anti-CD47 IgG-abundant and -deficient plasma, respectively, followed by analysis of cell proliferation, apoptosis and invasion/metastasis. Results The CD47 gene showed the highest expression among 13 genes detected in three OSCC cell lines; its expression was significantly higher in OSCC tissues than adjacent tissues. Plasma anti-CD47 IgG levels showed different between control subjects, oral benign tumor and OSCC patients. Anti-CD47 IgG-abundant plasma could evidently reduce cell viability via suppressing p-AKT expression and inducing cell apoptosis, and inhibit invasion of all three OSCC cell lines. Conclusions Natural autoantibodies against CD47 may be a potential agent for OSCC immunotherapy.
Article
MET receptor has emerged as a druggable target across several human cancers. Agents targeting MET and its ligand hepatocyte growth factor (HGF) including small molecules such as crizotinib, tivantinib and cabozantinib or antibodies including rilotumumab and onartuzumab have proven their values in different tumors. Recently, capmatinib was approved for treatment of metastatic lung cancer with MET exon 14 skipping. In this review, we critically examine the current evidence on how HGF/MET combination therapies may take advantage of synergistic effects, overcome primary or acquired drug resistance, target tumor microenvironment, modulate drug metabolism or tackle pharmacokinetic issues. Preclinical and clinical studies on the combination of HGF/MET-targeted agents with conventional chemotherapeutics or molecularly targeted treatments (EGFR, VEGFR, HER2, RAF/MEK, and PI3K/Akt targeting agents) and also the value of biomarkers are examined. Our deeper understanding of molecular mechanisms underlying successful pharmacological combinations is crucial to find the best personalized treatment regimens for cancer patients.
Article
Full-text available
ErbB3 has been widely implicated in treatment resistance, but its role as a primary treatment target is less clear. Canonically ErbB3 requires EGFR or ErbB2 for activation, whereas these two established treatment targets are thought to signal independently of ErbB3. In this study, we show that ErbB3 is essential for tumor growth of treatment-naive HNSCC patient-derived xenografts. This ErbB3 dependency occurs via ErbB3-mediated control of EGFR activation and HIF1α stabilization, which require ErbB3 and its ligand neuregulin-1. Here, we show that ErbB3 antibody treatment selects for a population of ErbB3-persister cells that express high levels of the transmembrane protein Trop2 that we previously identified as an inhibitor of ErbB3. Co-treatment with anti-ErbB3 and anti-Trop2 antibodies is synergistic and produces a greater anti-tumor response than either antibody alone. Collectively, these data both compel a revision of ErbB-family signaling and delineate a strategy for its effective inhibition in HNSCC.
Article
Full-text available
Mesenchymal-epithelial transition factor (c-Met) is the only high affinity receptor for hepatocyte growth factor (HGF), and is frequently activated in many human cancers. However, little is known about the role of the HGF/c-Met signaling pathway in the progression of human oral squamous cell carcinoma (OSCC). This study evaluated the role of the HGF/c-Met signaling pathway in the progression of human OSCC. We found that the expression of c-Met was significantly increased in human OSCC tissues than in normal mucosa adjacent to the tumor (P<0.05), but was not correlated with clinicopathological parameters. Additionally, the selective c-Met inhibitor JNJ was found to inhibit cell viability and migration and promote apoptosis in OSCC cell lines, and also blocked the activation AKT, ERK1/2, and NF-κB p65; thus, suggesting that HGF/c-Met signaling may play an important role in the tumorigenic properties of OSCC cells via the AKT, ERK1/2, and NF-κB pathways. Collectively, these results indicated that HGF/c-Met signaling may serve essential roles in the progression of human OSCC, and may thus be a basis for the development of novel therapeutic approaches in the treatment of OSCC.
Article
Full-text available
Radiation therapy (RT) is the current standard adjuvant approach for oral squamous cell carcinoma (OSCC) patients. Radioresistance is a major contributor to radiotherapy failure. In this study, we used patient-derived cells and a radiation-resistant cell line in vitro and in vivo for two purposes: evaluate the anti-tumor effects and understand the mechanisms in the dual PI3K/mTOR signaling pathway regulation of radiosensitization. Our findings indicate that in OML1-R cells, the radioresistance phenotype is associated with activation of the PI3K/AKT/mTOR signaling pathway. Compared to a combination of PI3K or mTOR inhibitors and radiation, dual blockade of the PI3K and mTOR kinases significantly improved radiation efficacy in oral cancer and patient-derived OSCC cells. Dual PI3K/mTOR inhibition enhanced the effect of radiation by inhibiting AKT/mTOR signaling pathways and caused G1 phase arrest, which is associated with downregulation of cyclin D1/CDK4 activity, leading to growth inhibition. In nude mice xenografted with radioresistant OML1-R cells, the combined treatment was also more effective than RT alone in reducing tumor growth. This treatment was also demonstrated to be dependent on the inhibition of protein kinase-dependent S6 kinase pathway and eIF4E-mediated cap-dependent translation. These findings indicate that activation of the PI3K/AKT/mTOR signaling pathway has a role in radioresistance of OSCC. We determined that a PI3K/mTOR inhibitor combined with radiation exhibits synergistic inhibition of the AKT/mTOR axis and induces cell cycle arrest. Our results show the therapeutic potential of drugs targeting the PI3K/AKT/mTOR signaling pathway should be new candidate drugs for radiosensitization in radiotherapy.
Article
Full-text available
The activation of receptor tyrosine kinases (RTKs) results in cellular effects including cell proliferation, survival, migration and invasion; RTKs also play an important role in tumourigenesis. It has been reported that EGFR signalling controls the migration of oral squamous cell carcinoma (OSCC) SAS and HSC3 cells but not of HSC4 cells, although the proliferation of HSC4 cells is regulated by EGF/EGFR. In the present study, we investigated the roles of EGFR and the c-Met signalling pathway in cell migration via filopodia and lamellipodia formation, which may be prerequisites for migration. To explore the role of c-Met in cell migration, we inhibited c-Met RTK activity using the c-Met inhibitor SU11274 and activated c-Met using hepatocyte growth factor (HGF) in three OSCC cell lines HSC4, SAS and Ca9-22 and investigated migration potency using a wound healing assay. We showed that inhibition of c-Met significantly suppressed, and activation of c-Met significantly promoted, the migration of OSCC cells. Additionally, the migration of SAS and Ca9-22 cells was inhibited by the EGFR inhibitors AG1478 and cetuximab and promoted by EGF treatment. Moreover, migration potency was correlated with lamellipodia formation. Furthermore, western blot analyses demonstrated that SU11274 decreased and HGF increased lamellipodin protein levels as well as phosphorylated c-Met levels. Collectively, we demonstrated that c-Met signalling induced lamellipodia formation by upregulating lamellipodin, thereby promoting the migration of OSCC cells.
Article
The head and neck squamous cell cancer (HNSCC) is the most common type of head and neck cancer (more than 90%), and all over the world more than a half million people have been developing this cancer in the last years. This type of cancer is usually marked by a poor prognosis with a really significant morbidity and mortality. Cetuximab received early favor as an exciting and promising new therapy with relatively mild side effect, and due to this received authorization in the 2004 from the European Medicines Agency (EMA) and in the 2006 from the Food and Drug Association (FDA) for the treatment of patients with squamous cell cancer of the head and neck in combination with radiation therapy for locally advanced disease. In this work we will review the application and the efficacy of the Cetuximab in the treatment of the HNSCC.
Article
Purpose: We previously demonstrated an association between decreased SMAD4 expression and cetuximab resistance in head and neck squamous cell carcinoma (HNSCC). The purpose of this study was to further elucidate the clinical relevance of SMAD4 loss in HNSCC. Experimental Design: SMAD4 expression was assessed by IHC in 130 newly diagnosed and 43 patients with recurrent HNSCC. Correlative statistical analysis with clinicopathologic data was also performed. OncoFinder, a bioinformatics tool, was used to analyze molecular signaling in TCGA tumors with low or high SMAD4 mRNA levels. The role of SMAD4 was investigated by shRNA knockdown and gene reconstitution of HPV-negative HNSCC cell lines in vitro and in vivo. Results: Our analysis revealed that SMAD4 loss was associated with an aggressive, HPV-negative, cetuximab-resistant phenotype. We found a signature of prosurvival and antiapoptotic pathways that were commonly dysregulated in SMAD4-low cases derived from TCGA-HNSCC dataset and an independent oral cavity squamous cell carcinoma (OSCC) cohort obtained from GEO. We show that SMAD4 depletion in an HNSCC cell line induces cetuximab resistance and results in worse survival in an orthotopic mouse model in vivo. We implicate JNK and MAPK activation as mediators of cetuximab resistance and provide the foundation for the concomitant EGFR and JNK/MAPK inhibition as a potential strategy for overcoming cetuximab resistance in HNSCCs with SMAD4 loss. Conclusions: Our study demonstrates that loss of SMAD4 expression is a signature characterizing the cetuximab-resistant phenotype and suggests that SMAD4 expression may be a determinant of sensitivity/resistance to EGFR/MAPK or EGFR/JNK inhibition in HPV-negative HNSCC tumors. Clin Cancer Res; 23(17); 5162–75. ©2017 AACR.
Article
Background: It remains unknown whether blockade of c-Met signaling and epidermal growth factor receptor signaling is effective in suppressing the growth of human colorectal cancer (CRC) cells. In this study, we investigated the effects of the c-Met inhibitor PHA-665752 alone and in combination with cetuximab on the growth of human CRC cells in vitro and in mouse xenografts. Methods: Human CRC cell lines (Caco2, HCT-116, and HT-29) and mice bearing HCT-116 xenografts were treated with cetuximab in the absence or presence of PHA-665752. Cell viability and apoptosis were examined using the MTT and TUNEL assays, respectively. Vimentin was measured by immunohistochemistry as a marker for epithelial-to-mesenchymal transition. Western blotting was used to determine signaling protein expression levels. Results: The MTT assay showed that the growth of Caco2, HCT-116, and HT-29 cells was inhibited by PHA-665752 in a dose-dependent manner, but only Caco2 cell growth was suppressed by cetuximab. Combination treatment with PHA-665752 and cetuximab inhibited the proliferation of Caco2 cells and RAS mutant CRC cell lines. However, relative to the PHA-665752-alone treatment group, HT-29 cells with a BRAF mutation showed no noticeable effect. The mean tumor volume in mice treated with cetuximab in combination with PHA-665752 was significantly smaller than that in the mice treated with only cetuximab (P = 0.033) or PHA-665752 (P < 0.01). Similarly, the expression of vimentin in the mice treated with PHA-665752 in combination with cetuximab was significantly lower than that in the mice treated with cetuximab or PHA-665752 alone (P < 0.05 in each case). TUNEL assays revealed that treatment with PHA-665752 in combination with cetuximab markedly increased CRC cell apoptosis. Western blotting analysis of signaling protein expression showed that PHA-665752 inhibited Met phosphorylation (P < 0.05). In addition, treatment with cetuximab alone or in combination with PHA-665752 effectively inhibited EGFR phosphorylation (P < 0.05). Conclusion: Combination treatment with PHA-665752 and cetuximab suppressed in vitro and in vivo CRC cell growth more than treatment with either agent alone did.