You are on page 1of 9

Tumor Biol.

DOI 10.1007/s13277-015-3232-6

RESEARCH ARTICLE

Celecoxib sensitizes gastric cancer to rapamycin via inhibition


of the Cbl-b-regulated PI3K/Akt pathway
Yubo Cao & Jinglei Qu & Ce Li & Dan Yang & Kezuo Hou &
Huachuan Zheng & Yunpeng Liu & Xiujuan Qu

Received: 22 September 2014 / Accepted: 5 February 2015


# International Society of Oncology and BioMarkers (ISOBM) 2015

Abstract Mammalian target of rapamycin (mTOR) has celecoxib and rapamycin. Our results suggest that celecoxib-
emerged as a new potential therapeutic target for gastric can- mediated upregulation of Cbl-b is responsible, at least in part,
cer. However, a phase III clinical trial found that monotherapy for the additive antitumor effect of celecoxib and rapamycin
with the mTOR inhibitor everolimus did not significantly im- via inhibition of rapamycin-induced Akt activation.
prove the overall survival of patients with advanced gastric
cancer. This has led to the exploration of more effective com-
Keywords Cbl-b . PI3K/Akt pathway . Gastric carcinoma .
binatorial regimens to enhance the effectiveness of mTOR
Celecoxib . COX-2 . mTOR inhibitor
inhibitors. Here, we demonstrate that Akt phosphorylation is
increased in the rapamycin-resistant gastric cancer cell lines
MGC803 and SGC7901. We further show that combined
treatment with celecoxib and rapamycin results in an additive
inhibitory effect on the growth of gastric cancer cells through Introduction
suppression of rapamycin-induced Akt activation. Moreover,
celecoxib upregulated the expression of the ubiquitin ligase Gastric cancer remains a leading cause of cancer-related
casitas B-lineage lymphoma-b (Cbl-b). Knockdown of Cbl-b deaths worldwide [1]. Most patients are diagnosed at an
significantly attenuated celecoxib-mediated inhibition of Akt advanced stage and the 5-year survival rate is less than
phosphorylation and impaired the additive anticancer effect of 10 % [2, 3]. At present, there is no globally accepted
standard chemotherapeutic regimen for the treatment of
patients with advanced gastric cancer [4]. However, recent
Electronic supplementary material The online version of this article
(doi:10.1007/s13277-015-3232-6) contains supplementary material, studies indicate that targeted therapy has great potential
which is available to authorized users. therapeutic value for the management of advanced gastric
Y. Cao : J. Qu : C. Li : K. Hou : Y. Liu (*) : X. Qu (*)
cancer. Trastuzumab has recently been recommended for
Department of Medical Oncology, The First Hospital of China the treatment of human epidermal growth factor receptor
Medical University, NO.155, North Nanjing Street, Heping District, 2-positive advanced gastric cancer [5]. Additionally, agents
Shenyang 110001, People’s Republic of China targeting the mammalian target of rapamycin (mTOR) are
e-mail: cmuliuyunpeng@hotmail.com
e-mail: qu_xiujuan@hotmail.com
currently undergoing clinical trials.
Given its central role in the regulation of cell growth, pro-
Y. Cao liferation, and survival, the phosphatidylinositol 3 kinase
Department of Medical Oncology, The Fourth Hospital of China (PI3K)-related serine/threonine kinase mTOR represents a po-
Medical University, Shenyang 110032, China
tential molecular target for anticancer therapy [6]. mTOR ex-
D. Yang ists as two functionally distinct complexes, mTORC1 and
Department of Pharmacology, Shenyang Medical College, mTORC2. Rapamycin-sensitive mTORC1 regulates messen-
Shenyang 110034, China ger ribonucleic acid (mRNA) translation by activating ribo-
somal p70S6 kinase (p70S6K) and inhibiting the translational
H. Zheng
Department of Biochemistry and Molecular Biology, College of repressor eIF4E-binding protein 1 [7]. Rapamycin-insensitive
Basic Medicine, China Medical University, Shenyang 110001, China mTORC2 phosphorylates the serine-473 residue of Akt to
Tumor Biol.

promote Akt activation [8, 9]. mTOR inhibitors such as the potential utility of celecoxib for overcoming rapamycin
rapamycin and its derivative everolimus have been shown to resistance in gastric cancer cells. We reveal that celecoxib
exhibit potent preclinical anticancer activities in gastric cancer sensitizes human gastric cancer cells to rapamycin by
both in vitro and in animal models [10, 11]. Phase I/II clinical inhibiting rapamycin-induced Akt activation. Additionally,
trials have shown that everolimus is well tolerated and exhibits celecoxib-mediated upregulation of Cbl-b is responsible, at
promising efficacy in the treatment of patients with advanced least in part, for this inhibition of the PI3K/Akt pathway in
gastric cancer [12, 13]. However, a recently published phase gastric cancer cells.
III clinical trial reported that everolimus monotherapy did not
significantly improve the overall survival of patients with ad-
vanced gastric cancer who had been previously treated with Materials and methods
one or two lines of systemic chemotherapy when compared
with best supportive care [14]. Therefore, there is a need to Reagents and antibodies
understand the mechanisms underlying drug resistance and
also to develop alternative treatment strategies for gastric 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-
cancer. mide (MTT) and dimethylsulphoxide (DMSO), LY294002
Activation of Akt under conditions of mTOR inhibition is were from Sigma-Aldrich (St. Louis, MO, USA). Celecoxib
tightly associated with development of cell resistance to was obtained from Sigma-Aldrich (St. Louis, MO, USA).
rapamycin [15]. He et al. have reported that resveratrol can Rapamycin were purchased from Sigma-Aldrich (St. Louis,
enhance the antitumor activity of rapamycin in breast cancer MO, USA). Antibodies against Cbl-b and β-actin were pur-
cell lines by suppressing rapamycin-induced Akt activation chased from Santa Cruz Biotechnology (Santa Cruz, CA,
[16]. Furthermore, Ji et al. have reported that the Akt inhibitor USA). Antibodies against mTOR and phospho-mTOR,
MK-2206 could overcome everolimus resistance in PTEN- p70S6K and phospho-p70S6K, Akt and phospho-Akt, and
mutant gastric cancer cells [17]. However, these agents have COX-2 were obtained from Cell Signaling Inc. (Beverly,
not been widely subjected to clinical trials in cancer therapy. MA, USA).
Therefore, we investigated whether combining rapamycin
with conventional and low-toxicity Akt-suppressing drugs
could improve therapeutic efficacy. Celecoxib is a new- Cells and cell culture
generation non-steroidal anti-inflammatory drug (NSAID)
that selectively inhibits COX-2 activity without inhibiting Gastric cancer MGC803 and SGC7901 cell lines were ob-
COX-1, and therefore, does not induce the side-effects associ- tained from the Type Culture Collection of the Chinese
ated with traditional NSAIDs [18]. Previous studies have dem- Academy of Sciences (Shanghai, China). The cells were
onstrated that celecoxib possesses anticancer effects attributable cultured in Roswell Park Memorial Institute (RPMI) 1640
to inhibition of Akt signaling [19, 20]. Recently, Chen et al. medium (GIBCO, Gaithersburg, MD, USA) containing
reported that celecoxib could enhance the sensitivity of lung 10 % foetal bovine serum (FBS), penicillin (100 U/mL),
cancer cells to the EGFR-tyrosine kinase inhibitor ZD1839 by and streptomycin (100 mg/mL) at 37 °C in an atmosphere
suppressing Akt signaling [21]. Furthermore, celecoxib en- of 95 % air and 5 % CO2.
hanced the anti-proliferative response of estrogen-dependent
breast cancer cells to nimotuzumab through downregulation Cell viability assay
of p-Akt expression [22]. However, whether celecoxib can
sensitize gastric cancer cells to rapamycin by suppressing The effects of celecoxib and rapamycin on cell proliferation
Akt signaling remains unclear. were measured using the 3-(4,5-dimethyl thiazol-2-yl)-2,5-
Regulation of the Akt signaling pathway is affected by diphenyl tetrazolium bromide (MTT) assay. Cells were seeded
many factors, among which ubiquitin ligase casitas B- at 5,000–1,0000/well in 96-well plates and incubated over-
lineage lymphoma-b (Cbl-b) is an important negative regula- night; various concentrations of test agents were then added
tor [23, 24]. Cbl-b is an E3 ubiquitin ligase that ubiquitinates and incubation continued for 48 or 72 h. Thereafter, 20 μL of
tyrosine kinase receptors and other signaling proteins, MTT solution (5 mg/mL) was added to each well and the
resulting in their downregulation [25]. Previous studies have cells incubated for a further 4 h at 37 °C. After removal
identified that Cbl-b proteins interact with the p85-regulatory of the culture medium, cells were lysed in 200 μL of
subunit of PI3K, resulting in PI3K ubiquitination and degra- DMSO and the optical density (OD) was measured at
dation [26]. However, a role for Cbl-b in celecoxib-induced 570 nm using a microplate reader (Bio-Rad, Hercules,
antitumor activity has not yet been identified. CA, USA). Change in percentage viability was calculated
We have investigated the mechanisms underlying the resis- using the formula: cell viability=(OD of the experimental
tance of human gastric cancer cells to mTOR inhibition and sample/OD of the control group)×100 %.
Tumor Biol.

Western blotting hairpin RNA (shRNA) targeting Cbl-b using lipofectamine


2000 reagent (Invitrogen, USA), according to the manufac-
The cells were washed twice with ice-cold phosphate-buffered turer’s instructions. After 48 h, stably transfected cell lines
saline (PBS), lysed in lysis buffer (1 % Triton X-100, 50 mM were selected by 0.4 mg/mL active G418 (Invitrogen, USA).
Tris–Cl, pH 7.4, 150 mM NaCl, 10 mM EDTA, 100 mM NaF, The expression of Cbl-b was verified by the Western blot with
1 mM Na3VO4, 1 mM PMSF, and 2 μg/mL aprotinin) and anti-Cbl-b antibody.
quantified with Lowry method. Cell lysate proteins were sepa-
rated to SDS-PAGE electrophoresis then transferred to a nitro- Small interfering RNA transfections
cellulose membrane (Immoblin-P, Millipore, Bedford, MA,
USA). After the membranes were blocked with 5 % skim milk Cbl-b small interfering RNA (siRNA) was obtained from
in TBST buffer (10 mM Tris–HCl pH 7.4, 150 mM NaCl, 0.1 % Shanghai GeneChem Co. Ltd (China). Cbl-b siRNA was syn-
Tween 20) at room temperature for 2 h, the blots were incubated thesized: 5′-GGAUGUGUUUGGGACUAAUTT-3′ (sense).
with indicated antibodies overnight at 4 °C. After washing three The control sequence was: AATTCTCCGAACGTGTCACG
times with TBST buffer, the membrane was incubated with T. The cells were transiently transfected with Cbl-b siRNA
secondary antibodies for 30 min at room temperature. Finally, using Lipofectamine 2000 reagent (Invitrogen, USA), accord-
proteins were detected with enhanced chemiluminescence ing to the manufacturer’s instructions.
reagent (SuperSignal Western Pico Chemiluminescent
Substrate; Pierce, Rockford, IL, USA) and visualized with Statistical analysis
the Electrophoresis Gel Imaging Analysis System (DNR
Bio-Imaging Systems, Jerusalem, Israel). Data were confirmed in three independent experiments and
were expressed as the mean ± standard deviation (S.D.).
Clonogenic assay Differences between groups were evaluated by Student’s t test.
SPSS 16.0 computer software (SPSS Inc., Chicago, IL, USA)
Cells were seeded at 200 cells/well in 12-well plates and treat- was used for statistical analysis and P<0.05 was considered
ed with either 100 nM rapamycin or 10 mM LY294002 or statistically significant.
30 μM celecoxib, or co-treatment with rapamycin and
LY294002 or celecoxib after plating for 24 h in 10 % FBS
medium. Cells were allowed to grow for additional 14 days. Results
Followed by staining with Wright Giemsa, the number of
colonies was counted. Rapamycin-induced Akt activation was responsible
for rapamycin resistance in MGC803 and SGC7901 gastric
Reverse-transcription-polymerase chain reaction (RT-PCR) cancer cells

MGC803 and SGC7901 cells either treated or untreated with We assessed the effect of rapamycin on cell survival in the
celecoxib were cultured and harvested at the indicated time. human gastric cancer cell lines MGC803 and SGC7901 by
Cell pellets were washed twice with ice-cold PBS and total performing an MTT assay 72 h after rapamycin treatment.
RNA extracted with the RNeasy mini kit (Qiagen, Carlsbad, Over 70 % of cells remained viable following exposure to
CA, USA) as described by the manufacturer. RT-PCR was up to 1,000 nM rapamycin (Fig. 1a). This indicated that both
performed with primer pairs for Cbl-b: forward (5′-CCGG cell lines were resistant to rapamycin.
TTAAGTTGCACTCGAT-3′) and reverse (5′-CAAAGGGG To investigate the mechanism of resistance to rapamycin in
TCCACGATTATG-3′) and for actin as a control: forward MGC803 and SGC7901 cells, we measured protein expres-
(5′-GTGGGGCGCCCCAGGCACCA-3′) and reverse sion levels of p-mTOR, p-p70S6K, and p-Akt following
(5′-CTCCTTAATGTCACGCACGATTTC-3′). PCR con- rapamycin treatment. In both MGC803 and SGC7901 cell
ditions were 95 °C for 5 min; 30 cycles of 95 °C for lines, p-mTOR and p-p70S6K levels decreased within 8 h of
30 s, 59 °C for 30 s, and 72 °C for 30 s; and one cycle exposure to rapamycin, and this decrease was sustained for up
of 72 °C for 7 min. The amplifed products were then to 24 h. Concurrently, p-Akt levels increased within 8 h of
separated on 1.5 % agarose gels, stained with ethidium exposure to rapamycin, and this increase was apparent up to
bromide and visualized under UV illumination. 24 h after treatment (Fig. 1b). We also examined the effects of
rapamycin on the protein expression levels of p70S6K, Akt,
RNA interference stable infection and mTOR and found that rapamycin did not alter their
expression.
The method of plasmid construction is discussed in our pre- To further explore whether the activation of Akt is respon-
vious study [23]. MGC803 cells were transfected with short sible for rapamycin resistance in MGC803 and SGC7901
Tumor Biol.

Fig. 1 Akt activation was associated with development of cell resistance of 100 nM of rapamycin for 24 h. The phosphorylation status of mTOR
to rapamycin in MGC803 and SGC7901 gastric cancer cells. a MGC803 and Akt was analyzed by Western blotting. d MGC803 or SGC7901 cells
or SGC7901 cells were treated with various concentrations of rapamycin were treated with or without LY294002 (25 mM) for 1 h before the
(1–1,000 nM) for 72 h. Cell viability was determined by MTT assay. b addition of 100 nM of rapamycin for 72 h. Cell survival was determined
MGC803 or SGC7901 cells were treated with 100 nM rapamycin for 8, by MTT assay. Results from three independent experiments are shown.
16, or 24 h; the phosphorylation status of mTOR and Akt was analyzed Student’s t test, **P<0.01 for the comparisons between the groups as
by Western blotting. c MGC803 or SGC7901 cells were treated with or indicated
without the PI3K inhibitor LY294002 (25 mM) for 1 h before the addition

gastric cancer cells, we blocked PI3K/Akt signaling Celecoxib sensitized gastric cancer cells to rapamycin
with a PI3K-specific inhibitor (LY294002) and then ex- by inhibiting rapamycin-induced Akt phosphorylation
posed cells to rapamycin. Treatment with LY294002
prior to rapamycin exposure diminished rapamycin- We next examined the effect of celecoxib alone and in com-
induced Akt phosphorylation in both MGC803 and bination with rapamycin on the survival of MGC803 and
SGC7901 cells (Fig. 1c). Rapamycin (100 nM) inhibited SGC7901 cells. An MTT assay conducted 72 h after exposure
cell growth by 15.70 ± 2.97 % and 20.22 ± 2.84 % in to celecoxib revealed a dose-dependent inhibitory effect upon
MGC803 and SGC7901 cells, respectively (Fig. 1d). cell survival in both cell lines. The 50 % inhibitory concen-
However, a combination of rapamycin and LY294002 tration of celecoxib was 47.25±6.43 μM in COX-2-negative
significantly increased the level of growth inhibition MGC803 cells and 43.53 ± 5.12 μM in COX-2-positive
(41.17 ± 3.00 % and 49.57 ± 2.92 % in MGC803 and SGC7901 cells (Fig. 2a, b). To assess whether celecoxib sen-
SGC7901 cells, respectively; P < 0.01). The effect of sitized gastric cancer cells to rapamycin, we incubated
the drugs in combination was greater than the inhibitory MGC803 and SGC7901 cells with rapamycin (100 nM),
effects caused by each agent alone, indicating an addi- celecoxib (40 μM), or both for 72 h. Co-treatment with
tive effect. Collectively, these results indicate that celecoxib enhanced the anti-proliferative response to
rapamycin-induced Akt activation was associated with rapamycin in both MGC803 and SGC7901 cells (Fig. 2c).
rapamycin resistance in both MGC803 and SGC7901 Colony formation assays confirmed that both celecoxib and
gastric cancer cells. LY294002 enhanced the sensitivity of gastric cancer cells to
Tumor Biol.

Fig. 2 Celecoxib enhanced the anti-tumor activity of rapamycin in for 72 h. Cell viability was determined by MTT assay. Results from three
gastric cancer cell lines by suppressing rapamycin-induced Akt independent experiments are shown. Student’s t test, **P<0.01 for the
signaling. a The protein expression levels of COX-2 in MGC803 or comparisons between the groups as indicated. d Clonogenic assay to
SGC7901 cells were analyzed by Western blotting. b MGC803 or determine the long-term effects of a combination of 100 nM rapamycin
SGC7901 cells were treated with various concentrations of celecoxib and 10 mM LY294002 or 30 μM celecoxib on the growth of MGC803 or
(10–60 μM) for 72 h. Cell viability was determined by MTT assay. SGC7901 cells after 14 days of treatment. e MGC803 or SGC7901 cells
Results from three independent experiments are shown. Student’s t test, were treated with rapamycin (100 nM) and/or celecoxib (40 μM) for 24 h;
*P<0.05, **P<0.01, and ***P<0.001 for the comparisons between the phosphorylation status of mTOR and Akt was analyzed by Western
celecoxib-treated and vehicle-treated group. c MGC803 or SGC7901 blotting
cells were treated with rapamycin (100 nM) and/or celecoxib (40 μM)

rapamycin (Fig. 2d). Collectively, these results reveal that Treatment with celecoxib alone significantly increased Cbl-b
treatment with a combination of rapamycin and celecoxib protein levels within 8 h of exposure to celecoxib, and this
can inhibit growth of gastric cancer cells. increase was evident up to 24 h after treatment (Fig. 3a).
To elucidate whether celecoxib sensitizes gastric cancer Combined treatment with celecoxib and rapamycin also up-
cells to rapamycin by suppressing rapamycin-induced Akt regulated Cbl-b to a similar level (Fig. 3b). Furthermore, RT-
phosphorylation, we treated MGC803 and SGC7901 cells PCR analysis showed that Cbl-b mRNA expression was in-
with rapamycin (100 nM), celecoxib (40 μM), or both for creased following treatment with celecoxib (Fig. 3c). These
24 h, and measured p-Akt levels by Western blotting. Levels results suggest that celecoxib-mediated Akt deactivation
of p-Akt protein were significantly reduced in celecoxib- might be at least in part triggered by upregulation of Cbl-b
treated cells exposed to rapamycin compared with control protein.
cells or those treated with rapamycin alone (Fig. 2e). These
results indicate that celecoxib sensitized MGC803 and Celecoxib-mediated Cbl-b upregulation enhanced sensitivity
SGC7901 cells to rapamycin by suppressing rapamycin- of MCG803 and SGC7901 cells to rapamycin
induced Akt phosphorylation..
Next, Cbl-b gene expression in MGC803 and SGC7901 cells
Celecoxib upregulated Cbl-b expression in MGC803 was inhibited by treatment with shRNA and siRNA to deter-
and SGC7901 cells mine whether Cbl-b was responsible for the impaired Akt
phosphorylation following celecoxib treatment. Repression
We next measured levels of Cbl-b to determine whether it is of Cbl-b resulted in increased levels of Akt phosphorylation,
involved with celecoxib-mediated Akt deactivation. whereas control had no effect on p-Akt levels (Fig. 4a). These
Tumor Biol.

Fig. 3 Celecoxib increased Cbl-b


protein levels. a MGC803 or
SGC7901 cells were treated with
40 μM Celecoxib for 8, 16, or
24 h; the protein expression level
of Cbl-b was analyzed by Western
blotting. b MGC803 or SGC7901
cells were treated with rapamycin
(100 nM) and/or celecoxib
(40 μM) for 24 h; the protein
expression level of Cbl-b was
analyzed by Western blotting. c
MGC803 or SGC7901 cells were
treated with 40 μM celecoxib for
8, 16, or 24 h; the mRNA
expression level of Cbl-b was
analyzed by RT-PCR. Actin was
used as an internal control. M
indicated marker

results suggest that Cbl-b downregulated the levels of p-Akt. inhibition of Akt signaling increased cell sensitivity to
Furthermore, combination treatment with celecoxib and rapamycin, consistent with previous findings from Ji et al.
rapamycin following knockdown of Cbl-b expression demon- [17]. These results suggest that Akt activation is critical for
strated that knockdown of Cbl-b attenuated celecoxib- the development of rapamycin resistance in gastric cancer
mediated Akt deactivation (Fig. 4b). Compared with controls, cells.
combined treatment with celecoxib and rapamycin signifi- Combinatorial cancer therapies can provide a more syner-
cantly attenuated growth inhibition by 26.01±3.81 % and gistic anticancer effect and less systemic toxicity than the use
36.69±2.82 % in MGC803 and SGC7901 cells, respectively of a lone therapeutic agent [29, 30]. Recent studies have dem-
(P<0.05; Fig. 4c). These results suggest that upregulation of onstrated that celecoxib can enhance the antitumor activity of
Cbl-b protein, at least in part, contributed to the additive effect rapamycin in malignant melanoma and angiosarcoma cell
of celecoxib and rapamycin on cell growth inhibition. lines [31, 32]. However, the mechanisms underlying this ef-
fect were not elucidated. Suppression of COX-2 activity has
been identified as the mechanism responsible for the chemo-
Discussion preventive effects of NSAIDs [30, 33]. However, more recent
reports have suggested that celecoxib may exert antitumor
The mTOR inhibitors temsirolimus and everolimus have been activities through COX-2-independent mechanisms [30, 34].
approved for the treatment of patients with metastatic renal To address this, we treated COX-2 over-expressing SGC7901
cell carcinoma [27, 28]. However, monotherapy with mTOR and COX-2-deficient MGC803 cell lines with celecoxib and
inhibitors often yields only modest therapeutic activity in ad- measured effects on cell survival. Celecoxib inhibited prolif-
vanced gastric cancer [14], and the emergence of drug resis- eration in a dose-dependent manner in both cell lines, consis-
tance may ultimately limit the utility of mTOR inhibitors [15]. tent with the findings of Cho et al. who performed similar
Our present data and those from others [17] indicate that gas- experiments using the gastric cancer cell lines AGS and
tric cancer cells are resistant to rapamycin. We detected in- MKN-45 [35]. Furthermore, the growth inhibitory effects of
creased levels of p-Akt in rapamycin-resistant MGC803 and combined celecoxib and rapamycin were similar in both cell
SGC7901 cells following rapamycin exposure. Moreover, lines. This finding suggests that the additive anticancer effect
Tumor Biol.

Fig. 4 Celecoxib-mediated Cbl-b upregulation enhanced the antitumor treated with the combination of 100 nM rapamycin and 40 μM celecoxib
activity of rapamycin in MGC803 and SGC7901 cells. a MGC803 cells for 48 h. Cell survival was determined by MTT assay. Results from three
were stably transfected with control shRNA or Cbl-b shRNA. The control independent experiments are shown. Student’s t test, *P<0.05 for the
shRNA and Cbl-b.shRNA MGC803 cells were untreated or treated with comparisons between two arms as indicated. c, d After the transfection,
100 nM rapamycin and/or 40 μM celecoxib for 48 h. Cell survival was MGC803 or SGC7901 cells were untreated or treated with 100 nM
determined by MTT assay. Results from three independent experiments rapamycin and/or 40 μM celecoxib for 24 h. The protein expression
are shown. Student’s t test, *P<0.05 for the comparisons between two levels of Cbl-b and the phosphorylation status of mTOR and Akt were
arms as indicated. b SGC7901 cells were transiently transfected with analyzed by Western blotting
control siRNA or Cbl-b siRNA. At 48 h after transfection, the cells were

of combined celecoxib and rapamycin therapy is at least cells lacking the PDK1 gene [37]. This indicates that other
partially mediated by a COX-2-independent pathway. factors may be involved in the celecoxib-mediated suppres-
Given that recent studies have identified the Akt signaling sion of Akt phosphorylation. Recent studies by us and others
pathway as a major COX-2-independent target of have shown that Cbl-b is a negative regulator of the PI3K/Akt
celecoxib [19, 20], we further investigated the role of this pathway [23, 26]. Additionally, we have demonstrated that
pathway in mediating the anticancer effect of celecoxib Cbl proteins can regulate the sensitivity of gastric and breast
and rapamycin. We found that celecoxib significantly re- cancer cells to treatment with a combination of tumor necrosis
pressed rapamycin-induced Akt phosphorylation in both cell factor-related apoptosis-inducing ligand and interferon-α or
lines. Together, these results demonstrate that celecoxib can bufalin [38, 39]. Therefore, we investigated whether Cbl-b is
sensitize gastric cancer cells to rapamycin by suppressing Akt involved in the suppression of Akt phosphorylation in gastric
signaling, at least in part, independently of its COX-2 inhibi- cancer cells exposed to celecoxib. To the best of our knowl-
tory function. edge, this is the first report of Cbl-b upregulation by celecoxib.
The mechanisms by which celecoxib suppresses Akt phos- Moreover, knockdown of Cbl-b attenuated celecoxib-
phorylation in the proposed COX-2-independent pathway are mediated inhibition of Akt signaling, which partially
not fully understood. Previous studies identified abolished the additive anticancer effect of celecoxib and
phosphoinositide-dependent kinase (PDK)-1 as a direct target rapamycin. Together, these results suggest that Cbl-b upregu-
of celecoxib [36]. However, more recent studies have found lation contributes to the additive anticancer effect of celecoxib
that celecoxib can induce apoptosis in mouse embryonic stem and rapamycin in gastric cancer cells.
Tumor Biol.

We demonstrate that celecoxib can sensitize human gastric 13. Yoon DH, Ryu MH, Park YS, Lee HJ, Lee C, Ryoo BY, et al. Phase II
study of everolimus with biomarker exploration in patients with ad-
cancer cells to rapamycin by inhibiting rapamycin-induced
vanced gastric cancer refractory to chemotherapy including
Akt signaling. Furthermore, celecoxib upregulates Cbl-b pro- fluoropyrimidine and platinum. Br J Cancer. 2012;106:1039–44.
tein expression, which likely plays a role in this celecoxib- 14. Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, Pan HM, et al.
mediated Akt deactivation and sensitization to rapamycin. Everolimus for previously treated advanced gastric cancer: results of
These results have important implications for the design of the randomized, double-blind, phase III GRANITE-1 study. J Clin
Oncol. 2013;31:3935–43.
future combination therapies for gastric cancer treatment in- 15. Carew JS, Kelly KR, Nawrocki ST. Mechanisms of mTOR inhibitor
volving rapamycin. resistance in cancer therapy. Targeted Oncol. 2011;6:17–27.
16. He X, Wang Y, Zhu J, Orloff M, Eng C. Resveratrol enhances the
Acknowledgments This research was supported by the following anti-tumor activity of the mTOR inhibitor rapamycin in multiple
grants: National Natural Science Foundation of China (nos. 81372546, breast cancer cell lines mainly by suppressing rapamycin-induced
81372547, and 81372485), Science and Technology Project of Liaoning Akt signaling. Cancer Lett. 2011;301:168–76.
Province (no. 2012225001), and Science and Technology Plan Project of 17. Ji D, Zhang Z, Cheng L, Chang J, Wang S, Zheng B, et al. The
Liaoning Province (no. 2011404013-1). combination of RAD001 and MK-2206 exerts synergistic cytotoxic
effects against PTEN mutant gastric cancer cells: involvement of
Conflicts of interest None MAPK-dependent autophagic, but not apoptotic cell death pathway.
PLoS One. 2014;9:e85116.
18. Leng J, Han C, Demetris AJ, Michalopoulos GK, Wu T.
Cyclooxygenase-2 promotes hepatocellular carcinoma cell growth
References through Akt activation: evidence for Akt inhibition in celecoxib-
induced apoptosis. Hepatology. 2003;38:756–68.
19. Kim N, Kim CH, Ahn DW, Lee KS, Cho SJ, Park JH, et al. Anti-
1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global
gastric cancer effects of celecoxib, a selective cox-2 inhibitor, through
cancer statistics. CA Cancer J Clin. 2011;61:69–90.
inhibition of Akt signaling. J Gastroenterol Hepatol. 2009;24:480–7.
2. Wagner AD, Grothe W, Haerting J, Kleber G, Grothey A, Fleig WE.
20. Pang RP, Zhou JG, Zeng ZR, Li XY, Chen W, Chen MH, et al.
Chemotherapy in advanced gastric cancer: a systematic review and
Celecoxib induces apoptosis in COX-2 deficient human gastric can-
meta-analysis based on aggregate data. J Clin Oncol. 2006;24:
cer cells through Akt/GSK3beta/NAG-1 pathway. Cancer Lett.
2903–09.
2007;251:268–77.
3. Power DG, Kelsen DP, Shah MA. Advanced gastric cancer–slow but
21. Chen L, He Y, Huang H, Liao H, Wei W. Selective COX-2 inhibitor
steady progress. Cancer Treat Rev. 2010;36:384–92.
celecoxib combined with EGFR-TKI ZD1839 on non-small cell lung
4. Menges M, Hoehler T. Current strategies in systemic treatment of
cancer cell lines: in vitro toxicity and mechanism study. Med Oncol.
gastric cancer and cancer of the gastroesophageal junction. J
2008;25:161–71.
Cancer Res Clin Oncol. 2009;135:29–38.
5. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki 22. Wang YX, Gao JX, Wang XY, Zhang L, Liu CM. Antiproliferative
A, et al. ToGA Trial Investigators. Trastuzumab in combination with effects of selective cyclooxygenase-2 inhibitor modulated by
chemotherapy versus chemotherapy alone for treatment of HER2- nimotuzumab in estrogen-dependent breast cancer cells. Tumour
positive advanced gastric or gastro-oesophageal junction cancer Biol. 2012;33:957–66.
(ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 23. Qu X, Zhang Y, Li Y, Hu X, Xu Y, Xu L, et al. Ubiquitin ligase Cbl-b
2010;376:687–97. sensitizes leukemia and gastric cancer cells to anthracyclines by ac-
6. Al-Batran SE, Ducreux M, Ohtsu A. mTOR as a therapeutic target in tivating the mitochondrial pathway and modulating Akt and ERK
patients with gastric cancer. Int J Cancer. 2012;130:491–6. survival signals. FEBS Lett. 2009;583:2255–62.
7. Fingar DC, Richardson CJ, Tee AR, Cheatham L, Tsou C, Blenis J. 24. Zhang Y, Qu X, Hu X, Yang X, Hou K, Teng Y, et al. Reversal of P-
mTOR controls cell cycle progression through its cell growth effec- glycoprotein-mediated multi-drug resistance by the E3 ubiquitin li-
tors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. gase Cbl-b in human gastric adenocarcinoma cells. J Pathol.
Mol Cell Biol. 2004;24:200–16. 2009;218:248–55.
8. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley AF, 25. Rao N, Dodge I, Band H. The Cbl family of ubiquitin ligases: critical
et al. Prolonged rapamycin treatment inhibits mTORC2 assembly negative regulators of tyrosine kinase signaling in the immune sys-
and Akt/PKB. Mol Cell. 2006;22:159–68. tem. J Leukoc Biol. 2002;71:753–63.
9. Gupta M, Ansell SM, Novak AJ, Kumar S, Kaufmann SH, Witzig 26. Brennan T, Adapala NS, Barbe MF, Yingling V, Sanjay A.
TE. Inhibition of histone deacetylase overcomes rapamycin-mediated Abrogation of Cbl-PI3K interaction increases bone formation and
resistance in diffuse large B-cell lymphoma by inhibiting Akt signal- osteoblast proliferation. Calcif Tissue Int. 2011;89:396–410.
ing through mTORC2. Blood. 2009;114:2926–35. 27. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A,
10. Fuereder T, Jaeger-Lansky A, Hoeflmayer D, Preusser M, Strommer et al. Temsirolimus, interferon alfa, or both for advanced renal-cell
S, Cejka D, et al. mTOR inhibition by everolimus counteracts VEGF carcinoma. N Engl J Med. 2007;356:2271–81.
induction by sunitinib and improves anti-tumor activity against gas- 28. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S,
tric cancer in vivo. Cancer Lett. 2010;296:249–56. et al. Efficacy of everolimus in advanced renal cell carcinoma: a
11. Shigematsu H, Yoshida K, Sanada Y, Osada S, Takahashi T, Wada Y, double-blind, randomised, placebo-controlled phase III trial. Lancet.
et al. Rapamycin enhances chemotherapy-induced cytotoxicity by 2008;372:449–56.
inhibiting the expressions of TS and ERK in gastric cancer cells. Int 29. Mustafa A, Kruger WD. Suppression of tumor formation by a
J Cancer. 2010;126:2716–25. cyclooxygenase-2 inhibitor and a peroxisome proliferator-activated
12. Lim T, Lee J, Lee DJ, Lee HY, Han B, Baek KK, et al. Phase I trial of receptor gamma agonist in an in vivo mouse model of spontaneous
capecitabine plus everolimus (RAD001) in patients with previously breast cancer. Clin Cancer Res. 2008;14:4935–42.
treated metastatic gastric cancer. Cancer Chemother Pharmacol. 30. Liu B, Wen JK, Li BH, Fang XM, Wang JJ, Zhang YP, et al.
2011;68:255–62. Celecoxib and acetylbritannilactone interact synergistically to
Tumor Biol.

suppress breast cancer cell growth via COX-2-dependent and - 36. Arico S, Pattingre S, Bauvy C, Gane P, Barbat A, Codogno P, et al.
independent mechanisms. Cell Death Dis. 2011;2:e185. Celecoxib induces apoptosis by inhibiting 3-phosphoinositide-
31. Bundscherer A, Hafner C, Maisch T, Becker B, Landthaler M, Vogt T. dependent protein kinase-1 activity in the human colon cancer HT-
Antiproliferative and proapoptotic effects of rapamycin and celecoxib 29 cell line. J Biol Chem. 2002;277:27613–21.
in malignant melanoma cell lines. Oncol Rep. 2008;19:547–53. 37. Kardosh A, Wang W, Uddin J, Petasis NA, Hofman FM, Chen TC,
32. Bundscherer A, Vogt T, Kohl G, Landthaler M, Hafner C. et al. Dimethyl-celecoxib (DMC), a derivative of celecoxib that lacks
Antiproliferative effects of rapamycin and celecoxib in angiosarcoma cyclooxygenase-2-inhibitory function, potently mimics the anti-
cell lines. Anticancer Res. 2010;30:4017–23. tumor effects of celecoxib on burkitt’s lymphoma in vitro and
33. Liu M, Li CM, Chen ZF, Ji R, Guo QH, Li Q, et al. Celecoxib regulates in vivo. Cancer Biol Ther. 2005;4:571–82.
apoptosis and autophagy via the PI3K/Akt signaling pathway in SGC- 38. Qu J, Zhao M, Teng Y, Zhang Y, Hou K, Jiang Y, et al. Interferon-
7901 gastric cancer cells. Int J Mol Med. 2014;33:1451–8. alpha sensitizes human gastric cancer cells to TRAIL-induced apo-
34. Gallouet AS, Travert M, Bresson-Bepoldin L, Guilloton F, Pangault ptosis via activation of the c-CBL-dependent MAPK/ERK pathway.
C, Caulet-Maugendre S, et al. COX-2-independent effects of Cancer Biol Ther. 2011;12:494–502.
celecoxib sensitize lymphoma B cells to TRAIL-mediated apoptosis. 39. Yan S, Qu X, Xu C, Zhu Z, Zhang L, Xu L, et al. Down-regulation of
Clin Cancer Res. 2014;20:2663–73. Cbl-b by bufalin results in up-regulation of DR4/DR5 and sensitiza-
35. Cho SJ, Kim N, Kim JS, Jung HC, Song IS. The anti-cancer effect of tion of TRAIL-induced apoptosis in breast cancer cells. J Cancer Res
COX-2 inhibitors on gastric cancer cells. Dig Dis Sci. 2007;52:1713–21. Clin Oncol. 2012;138:1279–89.

You might also like