You are on page 1of 7

European Journal of Pharmacology 893 (2021) 173840

Contents lists available at ScienceDirect

European Journal of Pharmacology


journal homepage: www.elsevier.com/locate/ejphar

Full length article

Effects of atorvastatin in combination with celecoxib and tipifarnib on


proliferation and apoptosis in pancreatic cancer sphere-forming cells
Xue-Tao Xu a, b, Jie Chen a, Xiang Ren a, Yu-Ran Ma a, Xiao Wang a, Yan-Yan Ma a, b,
Den-Gao Zhao a, b, Ren-Ping Zhou c, Kun Zhang a, b, Susan Goodin d, Dong-Li Li a, b, Xi Zheng c, d, *
a
School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
b
International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529020, PR China
c
Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
d
Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Cancer stem cell (CSC) plays an important role in pancreatic cancer pathogenesis and treatment failure. CSCs are
Pancreatic cancer characterized by their ability to form tumor spheres in serum-free medium and expression of CSC related
Atorvastatin markers. In the present study, we investigated the effect atorvastatin, celecoxib and tipifarnib in combination on
Celecoxib
proliferation and apoptosis in Panc-1 sphere-forming cells. The sphere-forming cells were isolated from Panc-1
Tipifarnib
Stem cells
cells by sphere-forming method. These sphere-forming cells showed CSC properties. The levels of CD44, CD133
Chemoresistant and ALDH1A1 in the sphere-forming cells were increased. Moreover, Panc-1 sphere-forming cells were resistant
Apoptosis to chemotherapeutic drug gemcitabine. Combined atorvastatin with celecoxib and tipifarnib synergistically
decreased the sphere forming ability of Panc-1 cells and the drug combination also strongly inhibited cell pro­
liferation and promoted apoptosis in the sphere-forming cells. The effects of the drug combination on the Panc-1
sphere-forming cells were associated with decreases in the levels of CD44, CD133 and ALDH1A1, and sup­
pression of Akt and NF-κB activation. Results of the present study indicate that the combination of atorvastatin,
celecoxib and tipifarnib may represent an effective approach for inhibiting pancreatic CSCs.

1. Introduction The discovery of stem cells for multiple malignant tumors has great
implications for study of tumor progression and therapy development.
Pancreatic cancer, one of the leading causes of cancer-related death The subpopulation of cancer cells commonly known as tumor-initiating
in Western countries, has very poor prognosis (Ferlay et al., 2015; Siegel cells, or cancer stem cells (CSCs), plays a critical role in tumorigenesis
et al., 2019). This disease is usually diagnosed at advanced stages when and is present in various types of cancer (Bjerkvig et al., 2005). CSCs are
little effective therapies are available (Stathis and Moore, 2010). About characterized by extensive abilities in self-renewal and differentiation as
60% of patients exhibit distant metastasis within 24 months after sur­ unidirectional cellular hierarchies and may contribute to tumor pro­
gery and the 5-year survival rate of pancreatic cancer patients is still low gression, recurrence and metastasis (Ajani et al., 2015; Vinogradov and
(O’Reilly and Abou-Alfa, 2007; Stathis and Moore, 2010; Yan et al., Wei, 2012; Reya et al., 2001). These characteristics suggest that CSCs
2019). Chemotherapy with gemcitabine remains the standard treatment could be a critical target for cancer therapy. Development of novel
for patients with advanced pancreatic cancer. Gemcitabine has a tumor therapies that inhibit CSCs would hold great promises in improving the
response rate of approximately 15% and treatment of patients with clinical management of pancreatic cancer.
gemcitabine extends survival by a mere 5 weeks on average (Wang et al., In an earlier study, our laboratory found that a combination of
2011; Ina et al., 2010). The development of resistance to gemcitabine atorvastatin (Fig. 1), celecoxib (Fig. 1) and tipifarnib (Fig. 1) strongly
and severe side effects associated with gemcitabine-based chemotherapy inhibited the growth and induced apoptosis in human pancreatic cancer
limits its clinical success. Clearly, it holds high clinical significance to cells (Ding et al., 2014). Atorvastatin is a statin drug that inhibits the
develop novel approach for effective treatment of pancreatic cancer. enzyme HMG-CoA reductase and thus decreased the cholesterol

* Corresponding author. Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersy, Piscataway, NJ, 08854,
USA
E-mail address: xizheng@pharmacy.rutgers.edu (X. Zheng).

https://doi.org/10.1016/j.ejphar.2020.173840
Received 2 June 2020; Received in revised form 21 December 2020; Accepted 22 December 2020
Available online 24 December 2020
0014-2999/© 2020 Elsevier B.V. All rights reserved.
X.-T. Xu et al. European Journal of Pharmacology 893 (2021) 173840

and formation of apoptotic bodies.

2.4. Caspase-3 activity assay

Caspase-3 activation was measured using an EnzoLyte AMC Caspase-


3 Assay Fluorimetric kit (AnaSpec, Fremont, CA) following the manu­
facturer instructions. Briefly, 1 × 105 cells were plated in triplicate in a
Fig. 1. Structures of atorvastatin, celecoxib and tipifarnib. flat-bottomed 96-well plate. After drug treatment, caspase-3 substrate
was added to each well. Plates were incubated for 30 min at room
temperature. Fluorescence intensity was measured in a Tecan Infinite
biosynthesis (Farnier and Davignon, 1998). Celecoxib is a selective
M200 plate reader (Tecan US Inc., Durham NC).
cyclooxygenase-2 (COX-2) inhibitor and has been shown to inhibit
pancreatic cancer (Hawkey, 1999; Zuo et al., 2018). Tipifarnib is a se­
2.5. Panc-1 sphere-forming cells
lective non-peptidomimetic inhibitor of farnesyltransferase (Karp et al.,
2009). Preclinical studies demonstrated that tipifarnib competitively
Isolation of Panc-1 sphere-forming cells was carried out using the
inhibits farnesylation of K-ras peptides at nanomolar concentrations.
sphere forming assay as described elsewhere. Briefly, Panc-1 cells (1000
Tipifarnib also inhibits proliferation of pancreatic cancer cells cultured
cells/mL) were seeded in a 24-well ultralow attachment surface culture
in vitro and grown as xenograft tumors in immunodeficient mice (Siegel
plate (Corning Co, NY, USA) and cultured in DMEM/F12 serum free
et al., 2013).
medium supplemented with basic fibroblast growth factor, epidermal
The present study was aimed to explore the effects and mechanisms
growth factor, insulin, selenium, transferrin, and bovine serum albumin
of the combination of atorvastatin, celecoxib and tipifarnib on pancre­
for 12 days. For the comparison of sphere-forming efficiency of Panc-1
atic cancer sphere-forming cells. The Panc-1 sphere-forming cells were
cells and the sphere-forming cells, Panc-1 cells and sphere-forming cells
isolated by the suspension sphere forming method. Our study demon­
(collected from the 12-day sphere culture as described above) were
strated for the first time that a combination of atorvastatin, celecoxib
seeded in ultralow attachment plate and cultured in DMEM/F12 serum
and tipifarnib strongly inhibited the formation of Panc-1 spheres,
free medium with atorvastatin, celecoxib and tipifarnib alone or in
inhibited proliferation and promoted apoptosis in sphere-forming cells.
combination for 12 days, and the number of spheres were counted in the
The effects of the triple combination on Panc-1 sphere-forming cells
end of the experiment.
were associated with suppression of Akt and NF-κB activation.

2.6. Immunofluorescence staining


2. Materials and methods

The cells were fixed in 4% paraformaldehyde for 30 min and then


2.1. Cells culture
treated with 0.1% Triton X-100 for 10 min at room temperature. After
blocking with phosphate-buffered saline (PBS) containing 5% bovine
Human pancreatic cancer cell line Panc-1 was purchased from the
serum albumin for 2 h, the cells were incubated with antibodies against
American Type Culture Collection (ATCC, Rockville, MD, USA). Panc-1
CD44 (#3570, Cell Signaling Co., Beverly, MA, USA) and caspase-3
cells sustained exponential growth by culturing in DMEM medium
(active form, AF835, R&D System, Minneapolis, MN, USA) overnight
supplemented with 10% fetal bovine serum (Gibco, Grand Island, NY,
at 4 ◦ C. After washing with PBS, the cells were incubated with FITC-
USA), penicillin (100 units/ml), streptomycin (100 mg/ml), and L-
conjugated and Rhodamine-conjugated secondary antibody for 3 h at
glutamine (300 μg/ml). The cells were grown at 37 ◦ C in a humidified
room temperature. The cells were counterstained with DAPI and
atmosphere of 5% CO2. Atorvastatin and celecoxib were obtained from
examined using a fluorescence cell imager (Nikon Eclipse TE200, Tokyo,
Sigma-Aldrich (St. Luis, MO, USA). Tipifarnib was kindly provided by
Japan).
J&J Co. Ltd. Atorvastatin, celecoxib and tipifarnib were dissolved in
DMSO, and the final concentration of DMSO in all experiments was
2.7. Western blotting
0.1%.

After treatment, the cell lysates were prepared (Ponta et al., 2003).
2.2. Determination of the number of viable cells Proteins were subjected to sodium dodecyl sulfatepolyacrylamide gel
electrophoresis (SDS-PAGE) and transferred to nitrocellulose mem­
The number of viable cells after each treatment was determined by branes. After blocking nonspecific binding sites with blocking buffer, the
using the trypan blue exclusion assay. After each experiment, single cell membranes were incubated overnight at 4 ◦ C with primary antibodies
suspension was made by trypsinized the cells. The trypan blue exclusion (#3570 for CD44, #5860 for CD133, #12035 for ALDH1A1, #3031 for
assay was performed by mixing 80 μl of cell suspension and 20 μl of 0.4% p-p65, #6956 for p65, #9271 for p-Akt and #4691 for Akt, Cell
trypan blue solution for 2 min. Blue cells were marked dead and the cells Signaling Co., Beverly, MA, USA). β-actin (sc-47778, Santa Cruz
that did not absorb dye were marked alive. We used a hemacytometerto Biotechnology Inc., Dallas, TX, USA) was used as a loading control.
count the number of viable cells under a Nikon light microscope Following the removal of the primary antibodies, the membranes were
(Optiphot, Nikon, Tokyo, Japan). then washed three times with TBS (PBS containing 0.05% Tween 20)
buffer at room temperature and later incubated with
2.3. Assessment of apoptotic cells fluorochrome-conjugated secondary antibody (925–32,211, Li-Cor
Biotechnology, Lincoln, NE, USA). The membrane was then washed
Apoptosis was determined by morphological assessment in cells with TBS three times. Final detection was done with an Odyssey infrared
stained with propidium iodide. After each experiment, cells were tryp­ imaging system (Li-Cor Biotechnology).
sinized and cytospin slides were prepared. The cells were fixed with
acetone/methanol (1:1) for 10 min at room temperature, followed by 10 2.8. Statistical analyses
min with propidium iodide staining (1 μg/ml in PBS), and finally
analyzed using a Nikon fluorescence microscope (Eclipse TE200, Nikon, Statistical analyses were done by using the software InStat (Graph­
Tokyo, Japan). Apoptotic cells were identified by their classical Pad Software, Inc., La Jolla, CA, USA). Comparisons of treatment
morphological features such as nuclear condensation, cell shrinkage, outcome were analyzed for statistical difference by ANOVA. Statistical

2
X.-T. Xu et al. European Journal of Pharmacology 893 (2021) 173840

significance was assumed at a value of P < 0.05. Analysis of synergy was 3.2. Inhibitory effect of atorvastatin, celecoxib and tipifarnib on sphere
performed using CompuSyn software. The combination index-fraction formation
affected (CI-Fa) curve demonstrates the relationship between the CI
value and the effective level of a certain biological indicator. If the CI is To explore whether atorvastatin, celecoxib and tipifarnib could exert
< 1, then the compounds are considered to have synergistic effects. If the suppressive effect on formation of spheres, Panc-1 cells were cultured in
CI is > 1 or = 1, then the compounds are thought to have an antagonistic ultralow attachment plate and treated with these drugs alone or in
or additive manner. combination at different ratios. As shown in Fig. 3A, atorvastatin, cel­
ecoxib or tipifarnib alone dose-dependently suppressed the formation of
3. Results spheres in Panc-1 cells. Combinations of atorvastatin, celecoxib and
tipifarnib had stronger effect on suppressing the formation of spheres
3.1. Sphere formation of Panc-1 cells than each drug used alone (Fig. 3A). Analysis of synergy based on iso­
bologram method showed that the triple combination synergistically
Cancer stem cells (CSCs) are characterized by their ability to form decreased the number of spheres in Panc-1 cells (Fig. 3B and C). The
three dimensional tumorspheres in vitro, and the sphere formation combination index (CI) was 0.49 for the combination of atorvastatin (5
method is widely used in the isolation and enrichment of CSCs in vitro. In μM), celecoxib (5 μM) and tipifarnib (0.2 μM). The CI was 0.61 for the
our studies, Panc-1 cells cultured in DMEM medium supplemented with combination of 2 μM atorvastatin, 2 μM celecoxib and 0.1 μM tipifarnib,
10% FBS showed adherent cell culture morphology (Fig. 2A). The Panc- and the CI was 0.89 for the combination of 10 μM atorvastatin, 10 μM
1 cells cultured in ultralow attachment plate with serum free medium celecoxib and 0.5 μM tipifarnib (Fig. 3C). Since the combination with
formed spheres (Fig. 2B). the ratio of 5: 5: 0.2 (μM) had the lowest CI, we used this dose ratio for
We next investigated the expression of CSC related molecular the combination in subsequent experiments. We further determined the
markers in Panc-1 sphere forming cells. The levels of CD133, CD44 and effect of atorvastatin, celecoxib or tipifarnib alone or in combination on
ALDH1A1 in parental Panc-1 cells and Panc-1 sphere-forming cells were the sphere formation in Panc-1 sphere-forming cells. As shown in
determined by Western blotting using corresponding antibodies. Results Fig. 3D, the drug combination strongly inhibited the sphere formation in
of the experiments showed that the abundances of CD133, CD44 and the sphere-forming cells. The inhibitory effect of the combination sphere
ALDH1A1in the sphere-forming cells were higher than the parental
Panc-1 cells (Fig. 2C). Since CSCs are resistant to chemotherapeutic
drug, the Panc-1 sphere-forming cells were also tested for their sensi­
tivity to gemcitabine which is a commonly used chemotherapeutic drug
for pancreatic cancer treatment. As shown in Fig. 2D, the effect of
gemcitabine on decreasing the cell viability was significantly stronger in
the parental Panc-1 cells than the sphere-forming cells (P < 0.001)
indicating that the sphere-forming cells were partially resistant to
gemcitabine-induced growth inhibition. Taken together, the results
described above demonstrated that the sphere-forming cells derived
from Panc-1 cells through sphere-forming method exhibited both the
classical cellular and molecular properties of pancreatic CSCs.

Fig. 3. Inhibition of sphere formation by atorvastatin, celecoxib and tipifarnib.


Panc-1 cells were cultured in ultralow attachment plate with serum free me­
dium and treated with atorvastatin (LIP), celecoxib (CEL) and tipifarnib (ZAN)
alone or in combination for 12 days. The number of spheres was counted under
microscope. (A) Sphere formation in Panc-1 cells treated with LIP, CEL and ZAN
Fig. 2. Panc-1 cells and the Panc-1 sphere-forming cells. (A) Representative alone or in combination. (B) Effect of LIP, CEL and ZAN alone or in combina­
micrograph of Panc-1 cells cultured in DMEM medium with 10% FBS. (B) tionon sphere formation plotted as fraction affected (Fa). (C) Combination
Representative micrograph of Panc-1 spheres cultured in ultralow attachment index (CI) for different ratios of the three drugs in combination. (D) Effects of
plate with serum free medium. (C) Protein levels of CSC related markers CD44, LIP, CEL and ZAN alone or in combinationon sphere formation as compared
CD133, ALDH1A1 in PCs and the SFCs. (D) Effects of gemcitabine (GEM; 5 μM) with negative control (NC) in Panc-1 cells (PCs) and the Panc-1 sphere forming
on the viability of PCs and SFCs. Superscript a indicates significant difference cells (SFCs). Each value represents mean ± S.D from three separate
(P < 0.001) as compared to the parental Panc-1 cells. experiments.

3
X.-T. Xu et al. European Journal of Pharmacology 893 (2021) 173840

formation was similar in parental Panc-1 cells and Panc-1 sphere- of viable cells was observed in the Panc-1 sphere-forming cells after
forming cells. Panc-1 cell population is heterogenous with a small treatment with atorvastatin, celecoxib and tipifarnib in combination
fraction of cancer stem cells while sphere-forming cells are enriched while each drug alone only had small effect on decreasing the number of
with cancer stem cells. Our result of similar inhibitory effects of the viable cells. The levels of proliferating cell nuclear antigen (PCNA) and
combination on sphere formation in Panc-1 and the sphere-forming cells Cyclin D1 were also decreased in the sphere-forming cells treated with
indicate that the combination consistently target the cancer stem cells. the triple combination (Fig. 4B). Effect of the combination treatment on
apoptosis was determined by propidium iodide staining assay and
3.3. Decrease in cell proliferation and increase in apoptosis in Panc-1 caspase-3 assay. As shown in Fig. 4C, treatment of the sphere-forming
sphere-forming cells treated with atorvastatin, celecoxib and tipifarnib cells with the combination resulted in a strong increase in the number
of apoptotic cells. Result of the caspase-3 assay also showed that the
To investigate the potential inhibitory effect of atorvastatin, cele­ combination strongly increased apoptosis in the sphere-forming cells
coxib and tipifarnib on pancreatic CSCs, proliferation and apoptosis in (Fig. 4D). Fig. 4E shows representative micrographs of immunofluo­
sphere-forming cells treated with these drugs alone or in combination rescence staining in Panc-1 sphere-forming cells. Cells in the control
were determined. As shown in Fig. 4A, a strong decrease in the number group showed positive staining with CSC marker CD44 and negative

Fig. 4. Effects of atorvastatin (LIP), celecoxib (CEL)


and tipifarnib (ZAN) alone or in combination (COM)
on proliferation and apoptosis in Panc-1 sphere-
forming cells. Cell viability was measured by the
trypan blue assay. PCNA and Cyclin D1 were deter­
mined by Western blotting. Apoptosis was deter­
mined by propidium iodide staining and caspase-3
assay. (A) Percent viable cells. (B) Expression levels of
PCNA and Cyclin D1. (C) Percent apoptotic cells. (D)
Caspase-3 activity. (E) Immunofluoresence staining of
CD44 and caspase-3 in the control and the
combination-treated cells. (F) Percent viable cells
Each value represents mean ± S.D from three sepa­
rate experiments. Superscript a indicates significant
differences (P < 0.001) as compared to cells treated
with LIP, CEL or ZAN alone. Superscriptb indicates
significant difference (P < 0.01) as compared to cells
treated with the triple combination (COM).

4
X.-T. Xu et al. European Journal of Pharmacology 893 (2021) 173840

staining of caspase-3. The cells treated with the combination had posi­ forming cells. We found that atorvastatin, celecoxib or tipifarnib alone
tive staining for both CD44 and caspase-3 indicating apoptosis in CSCs. had small effect on decreasing the levels of phosphorylated p65 while
The results described above indicated that the triple combination the triple combination strongly blocked the phosphorylation of p65
inhibited proliferation and induced apoptosis in Panc-1 CSCs cells. Since (Fig. 5). The levels of phosphorylated Akt in the sphere-forming cells
the sphere-forming cells were partially resistance to gemcitabine, we treated with atorvastatin, celecoxib and tipifarnib alone or in combi­
further determined if the triple combination could enhance the growth nation was also determined. As shown in Fig. 5, atorvastatin alone had
inhibitory effect of gemcitabine. As shown in Fig. 4F, atorvastatin, cel­ little effect, and celecoxib or tipifarnib alone had moderate effect on the
ecoxib or tipifarnib alone did not enhance the effect of gemcitabine on level of phosphorylated Akt. However, the triple combination potently
cell viability. However, the drug combination significantly enhance the reduced the level of phosphorylated Akt. These results demonstrated
growth inhibitory effect of gemcitabine (P < 0.01; Fig. 4F). that combined atorvastatin with celecoxib and tipifarnib strongly
inhibited the activation of NF-κB and Akt.
3.4. Down regulation of CD133, CD44 and ALDH1A1 in Panc-1 sphere-
forming cells treated with the triple combination 4. Discussion

To evaluate the effect of atorvastatin, celecoxib and tipifarnib on the Accumulating experimental evidence indicates that CSCs drive
stemness of the sphere-forming cells, we examined the levels of CSC tumor initiation, invasion and metastasis, and contributes to chemo­
related markers CD133, CD44 and ALDH1A1 in these cells. In these resistance, consequently promoting unrestricted tumor progression
experiments, the sphere-forming cells were treated with the combina­ (Elshamy and Duhe, 2013; Koury et al., 2017; Yoshida and Saya, 2016).
tion of atorvastatin, celecoxib and tipifarnib for 24 h. The CSC related The present study was carried out to test our hypothesis that combined
markers were examined by the Western blot analysis. Results of the atorvastatin with celecoxib and tipifarnib will suppress pancreatic CSCs.
experiment showed small to moderate decreases in the levels of CD44, We showed in the present study that the combination of atorvastatin,
CD133 and ALDH1A1 in the cells treated with atorvastatin, celecoxib or celecoxib and tipifarnib strongly inhibited Panc-1 CSCs as evidenced by
tipifarnib alone (Fig. 5). As expected, the expression levels of CD44, the decreases in tumorsphere formation and reduced levels of CSC
CD133 and ALDH1A1 were strongly reduced in the sphere-forming cells related markers in the sphere forming cells. Results of our study suggest
treated with the combination of atorvastatin, celecoxib and tipifarnib that the combination strategy using atorvastatin, celecoxib and tipi­
(Fig. 5). farnib may represent an effective approach for inhibiting pancreatic
CSCs.
3.5. Suppression of NF-κB and Akt in Panc-1 sphere-forming cells by the An important step in CSC-based research is the effective isolation and
triple combination reliable identification of CSCs from a large heterogenious cancer cell
population. Pancreatic CSCs can be isolated and enriched by their
We further determined the effects of atorvastatin, celecoxib and sphere-forming ability (Zeng et al., 2018). In the present study, Panc-1
tipifarnib alone or in combination on the activity of NF-κB in the sphere- cells were cultured in ultralow attachment culture plate with serum-free
medium. Panc-1 cells form spheres with this culture condition. The
sphere-forming cells were examined for expression levels of CSC related
markers. Based on existing studies, CD44, CD133 and ALDH1 are
considered pancreatic cancer CSC related markers (Raj et al., 2015; Zeng
et al., 2018; Zhou et al., 2019). Thus, these markers were chosen to
identify pancreatic cancer CSCs. We found that the levels of these
markers were much higher in the sphere-forming cells than in parental
Panc-1 cells. This result indicates that the pancreatic cancer CSCs can be
isolated and enriched by the sphere-forming method.
There is increasing interest in using combinations of different agents
for inhibiting pancreatic cancer cells. Suitable combinations may pro­
duce synergistic effect on suppressing pancreatic CSCs. In the present
study, we observed that combined atorvastatin with celecoxib and
tipifarnib synergistically suppressed Panc-1 sphere formation, while
each agent alone only had small inhibitory effect on Panc-1 sphere
formation. Moreover, the combination of atorvastatin, celecoxib and
tipifarnib effectively diminished Panc-1 CSC activity as evidenced by
decreasing proliferation and increasing apoptosis in the sphere-forming
cells. Combined atorvastatin with celecoxib and tipifarnib also strongly
downregulated CSC related markers CD44, CD133 and ALDH1.
CD44 is a transmembrane glycoprotein which possesses a particular
cell adhesion function and consequently assists the matrix adhesion and
migration of CSCs (Ponta et al., 2003). The expression of CD44 variants
was significantly correlated with poor prognosis in pancreatic cancer
(Garcea et al., 2005). In pancreatic cancer, CD44+ cells were more
tumorigenic than CD44-cells (Li et al., 2007). CD44 is one of the
important CSC markers in pancreatic cancer (Li et al., 2007). Gemcita­
bine resistant pancreatic cancer cells showed characteristics of CSC and
RNA interference of CD44 inhibited clonogenic growth of the cells
Fig. 5. Effects of atorvastatin (LIP), celecoxib (CEL) and tipifarnib (ZAN) alone
or in combination (COM) on expression levels of CD133, CD44, ALDH1A1, Akt (Hong et al., 2009). CD133 (also called Prominin-1 or AC133) was the
and NF-κB p65. Panc-1 sphere-forming cells were treated with LIP, CEL and first member verified in the prominin family of pentaspan trans­
ZAN alone or in combination for 24 h. The protein levels of CD133, CD44, membrane glycoproteins, and is now widely used as CSC related markers
ALDH1, Akt, p-Akt, p65 and p-p65 were determined by the Western in various types of cancer including pancreatic cancer (Gupta et al.,
blot analysis. 2018; Hermann et al., 2007). As few as 500 patient-derived CD133+

5
X.-T. Xu et al. European Journal of Pharmacology 893 (2021) 173840

pancreatic cancer cells were capable of forming orthotopic tumors in Appendix A. Supplementary data
immunodeficient mice. In contrast, as many as 106 CD133-pancreatic
cancer cells did not result in tumor formation (Hermann et al., 2007). Supplementary data to this article can be found online at https://doi.
In the present study, we found that the sphere forming cells expressed org/10.1016/j.ejphar.2020.173840.
high levels of CD44 and CD133, and treatment of the cells with the triple
combination decreased the levels of CD44 and CD133. This result in­ References
dicates that CD44 and CD133 are useful markers for the determination
of the effect of drug combination on the stemness of pancreatic cancer Ajani, J.A., Song, S., Hochster, H.S., Steinberg, I.B., 2015. Cancer stem cells: the promise
and the potential. Semin. Oncol. 42 (Suppl. 1), S3–S17. https://doi: 10.1053/j.sem
cells. inoncol.2015.01.001.
ALDH1A1 is a cytosolic enzyme belongs to the ALDH family of en­ Altomare, D.A., Testa, J.R., 2005. Perturbations of the AKT signaling pathway in human
zymes that catalyze the intracellular oxidation of different aldehydes cancer. Oncogene 24, 7455–7464. https://doi: 10.1038/sj.onc.1209085.
Arima, K., Ohmuraya, M., Miyake, K., Koiwa, M., Uchihara, T., Izumi, D., Gao, F.,
(Tomita et al., 2016). It plays an important role in retinoic acid signaling Yonemura, A., Bu, L., Okabe, H., Imai, K., Hashimoto, D., Baba, Y., Chikamoto, A.,
and used as a marker to identify various types of CSCs (Tomita et al., Yamashita, Y.I., Furukawa, T., Araki, K., Baba, H., Ishimoto, T., 2019. Inhibition of
2016; Duong et al., 2017; Jiang et al., 2009; Meng et al., 2014). A recent 15-PGDH causes Kras-driven tumor expansion through prostaglandin E2-ALDH1
signaling in the pancreas. Oncogene 38, 1211–1224. https://doi: 10.1038/s41388-0
study showed that accumulation of prostaglandin E2 (PGE2) resulted in 18-0510-y.
high levels of ALDH1A1 and enhanced KRAS driven pancreatic cancer Bjerkvig, R., Tysnes, B.B., Aboody, K.S., Najbauer, J., Terzis, A.J., 2005. Opinion: the
progression indicating a role of PGE2-ALDH1A1 pathway in pancreatic origin of the cancer stem cell: current controversies and new insights. Nat. Rev.
Canc. 5, 899–904. https://doi: 10.1038/nrc1740.
cancer pathogenesis (Arima et al., 2019). PGE2 is regulated by COX-2
Deeb, D., Gao, X., Liu, Y.B., Pindolia, K., Gautam, S.C., 2014. Pristimerin, a
and the COX-2 inhibitor celecoxib may decrease the level of ALDH1A1 quinonemethide triterpenoid, induces apoptosis in pancreatic cancer cells through
through the PGE2-ALDH1A1 pathway. ALDH1A1 reportedly is a target the inhibition of pro-survival Akt/NF-kappaB/mTOR signaling proteins and anti-
gene of the NF-κB pathway (Moreno et al., 2004; Yan et al., 2008) and apoptotic Bcl-2. Int. J. Oncol. 44, 1707–1715. https://doi: 10.3892/ijo.2014.2325.
Ding, N., Cui, X.X., Gao, Z., Huang, H., Wei, X., Du, Z., Lin, Y., Shih, W.J., Rabson, A.B.,
NF-κB may act as an upstream positive regulator of ALDH1A1 (Yang Conney, A.H., Hu, C., Zheng, X., 2014. A triple combination of atorvastatin,
et al., 2015). In the present study, we found that combined atorvastatin celecoxib and tipifarnib strongly inhibits pancreatic cancer cells and xenograft
with celecoxib and tipifarnib strongly inhibited NF-κB. Our findings pancreatic tumors. Int. J. Oncol. 44, 2139–2145. https://doi: 10.3892/ijo.2014.
2350.
raise the possibility that the strong decrease in ALDH1A1 by the com­ Duong, H.Q., You, K.S., Oh, S., Kwak, S.J., Seong, Y.S., 2017. Silencing of NRF2 reduces
bination of atorvastatin, celecoxib and tipifarnib in Panc-1 sphere the expression of ALDH1A1 and ALDH3A1 and sensitizes to 5-FU in pancreatic
forming cells was mediated by concurrent inhibition of COX-2 and cancer cells. Antioxidants 6. https://doi: 10.3390/antiox6030052.
Elshamy, W.M., Duhe, R.J., 2013. Overview: cellular plasticity, cancer stem cells and
NF-κB. metastasis. Canc. Lett. 341, 2–8. https://doi: 10.1016/j.canlet.2013.06.020.
Results of our study also indicate that activation of Akt was strongly Farnier, M., Davignon, J., 1998. Current and future treatment of hyperlipidemia: the role
inhibited by the combination of atorvastatin, celecoxib and tipifarnib. of statins. Am. J. Cardiol. 82, 3J–10J. https://doi: 10.1016/s0002-9149(98)00423-8.
Ferlay, J., Soerjomataram, I., Dikshit, R., Eser, S., Mathers, C., Rebelo, M., Parkin, D.M.,
The PI3K/Akt signaling pathway plays an important role in cell prolif­ Forman, D., Bray, F., 2015. Cancer incidence and mortality worldwide: sources,
eration, survival and malignant transformation, and is frequently acti­ methods and major patterns in GLOBOCAN 2012. Int. J. Canc. 136, E359–E386.
vated in human cancer (Tokunaga et al., 2008; Altomare and Testa, https://doi: 10.1002/ijc.29210.
Garcea, G., Neal, C.P., Pattenden, C.J., Steward, W.P., Berry, D.P., 2005. Molecular
2005). NF-κB and mTOR are down-stream targets of activated Akt (Deeb
prognostic markers in pancreatic cancer: a systematic review. Eur. J. Canc. 41,
et al., 2014; Huang et al., 2020), and mTOR plays critical roles in 2213–2236. https://doi: 10.1016/j.ejca.2005.04.044.
pancreatic CSCs through specific and stemness-related functions (Mat­ Gupta, V.K., Sharma, N.S., Kesh, K., Dauer, P., Nomura, A., Giri, B., Dudeja, V.,
subara et al., 2013). A strong inhibition of Akt activation in Banerjee, S., Bhattacharya, S., Saluja, A., Banerjee, S., 2018. Metastasis and
chemoresistance in CD133 expressing pancreatic cancer cells are dependent on their
sphere-forming cells by the triple combination was found in the present lipid raft integrity. Canc. Lett. 439, 101–112. https://doi: 10.1016/j.canlet.2018.0
study. Simultaneous suppression of Akt and NF-κB by suitable phar­ 9.028.
macological inhibitors may be an effective strategy for inhibition of Hawkey, C.J., 1999. COX-2 inhibitors. Lancet 353, 1440. https://doi:
10.1016/s0140-6736(05)75962-6.
pancreatic CSCs. Hermann, P.C., Huber, S.L., Herrler, T., Aicher, A., Ellwart, J.W., Guba, M., Bruns, C.J.,
In conclusion, results of this study demonstrate that combined Heeschen, C., 2007. Distinct populations of cancer stem cells determine tumor
atorvastatin with celecoxib and tipifarnib synergistically inhibited the growth and metastatic activity in human pancreatic cancer. Cell. Stem. Cell. 1,
313–323. https://doi: 10.1016/j.stem.2007.06.002.
formation of tumor sphere in Panc-1 cells. The combination also strongly Hong, S.P., Wen, J., Bang, S., Park, S., Song, S.Y., 2009. CD44-positive cells are
inhibited proliferation and promoted apoptosis in Panc-1 sphere-form­ responsible for gemcitabine resistance in pancreatic cancer cells. Int. J. Canc. 125,
ing cells. The levels of CSC related markers CD44, CD133 and ALDH1A1 2323–2331. https://doi: 10.1002/ijc.24573.
Huang, X., Fei, G.Q., Liu, W.J., Ding, J., Wang, Y., Wang, H., Ji, J.L., Wang, X., 2020.
were decreased in sphere-forming cells treated with this drug combi­ Adipose-derived mesenchymal stem cells protect against CMS-induced depression-
nation. The strong effects of the combination on sphere-forming cells like behaviors in mice via regulating the Nrf2/HO-1 and TLR4/NF-kappaB signaling
were associated with inhibition of Akt and NF-κB activation. Combined pathways. Acta Pharmacol. Sin. 41, 612–619. https://doi: 10.1038/s41401-019-03
17-6.
atorvastatin with celecoxib and tipifarnib may be an effective approach
Ina, S., Hirono, S., Noda, T., Yamaue, H., 2010. Identifying molecular markers for
to inhibit pancreatic CSCs. chemosensitivity to gemcitabine in pancreatic cancer: increased expression of
interferon-stimulated gene 15 kDa is associated with intrinsic chemoresistance.
Pancreas 39, 473–485. https://doi:10.1097/MPA.0b013e3181c0decc.
Declaration of competing interest
Jiang, F., Qiu, Q., Khanna, A., Todd, N.W., Deepak, J., Xing, L., Wang, H., Liu, Z., Su, Y.,
Stass, S.A., Katz, R.L., 2009. Aldehyde dehydrogenase 1 is a tumor stem cell-
The authors declare no conflict of interest. associated marker in lung cancer. Mol. Canc. Res. 7, 330–338. https://doi: 10.115
8/1541-7786.MCR-08-0393.
Karp, J.E., Flatten, K., Feldman, E.J., Greer, J.M., Loegering, D.A., Ricklis, R.M.,
Acknowledgments Morris, L.E., Ritchie, E., Smith, B.D., Ironside, V., Talbott, T., Roboz, G., Le, S.B.,
Meng, X.W., Schneider, P.A., Dai, N.T., Adjei, A.A., Gore, S.D., Levis, M.J., Wright, J.
The present study was supported by grants from the Department of J., Garrett-Mayer, E., Kaufmann, S.H., 2009. Active oral regimen for elderly adults
with newly diagnosed acute myelogenous leukemia: a preclinical and phase 1 trial of
Education of Guangdong Province (2016KCXTD005, 2017KZDXM084, the farnesyltransferase inhibitor tipifarnib (R115777, Zarnestra) combined with
2017KSYS010 and 2019KZDZX2003), the Department of Science and etoposide. Blood 113, 4841–4852. https://doi: 10.1182/blood-2008-08-172726.
Technology of Jiangmen (No: 2016350100170008351, Koury, J., Zhong, L., Hao, J., 2017. Targeting signaling pathways in cancer stem cells for
cancer treatment. Stem Cell. Int. 2017, 2925869. https://doi: 10.1155/201
2018110100330005446), Jiangmen Program for Innovative Research 7/2925869.
Team (No: 2017TD02), and the Youth Team Fund of Wuyi University Li, C., Heidt, D.G., Dalerba, P., Burant, C.F., Zhang, L., Adsay, V., Wicha, M., Clarke, M.
(2016td01). Dr. Xi Zheng is the Unilever Chair in Nutrition and Disease F., Simeone, D.M., 2007. Identification of pancreatic cancer stem cells. Canc. Res. 67,
1030–1037. https://doi: 10.1158/0008-5472.CAN-06-2030.
Prevention Research at Rutgers University.

6
X.-T. Xu et al. European Journal of Pharmacology 893 (2021) 173840

Matsubara, S., Ding, Q., Miyazaki, Y., Kuwahata, T., Tsukasa, K., Takao, S., 2013. mTOR Tomita, H., Tanaka, K., Tanaka, T., Hara, A., 2016. Aldehyde dehydrogenase 1A1 in stem
plays critical roles in pancreatic cancer stem cells through specific and stemness- cells and cancer. Oncotarget 7, 11018–11032. https://doi.org/10.18632/
related functions. Sci. Rep. 3, 3230. https://doi: 10.1038/srep03230. oncotarget.6920.
Meng, E., Mitra, A., Tripathi, K., Finan, M.A., Scalici, J., McClellan, S., Madeira da Vinogradov, S., Wei, X., 2012. Cancer stem cells and drug resistance: the potential of
Silva, L., Reed, E., Shevde, L.A., Palle, K., Rocconi, R.P., 2014. ALDH1A1 maintains nanomedicine. Nanomedicine 7, 597–615. https://doi: 10.2217/nnm.12.22.
ovarian cancer stem cell-like properties by altered regulation of cell cycle checkpoint Wang, Z., Li, Y., Ahmad, A., Banerjee, S., Azmi, A.S., Kong, D., Sarkar, F.H., 2011.
and DNA repair network signaling. PloS One 9, e107142. https://doi: 10.1371/journ Pancreatic cancer: understanding and overcoming chemoresistance. Nat. Rev.
al.pone.0107142. Gastroenterol. Hepatol. 8, 27–33. https://doi: 10.1038/nrgastro.2010.188.
Moreno, C.S., Ramachandran, S., Ashby, D.G., Laycock, N., Plattner, C.A., Chen, W., Yan, B., Chen, G., Saiga, l K., Yang, X., Jensen, S.T., Van, W.C., Stoeckert, C.J., Chen, Z.,
Hahn, W.C., Pallaset, D.C., 2004. Signaling and transcriptional changes critical for 2008. Systems biology-defined NF-kappaB regulons, interacting signal pathways and
transformation of human cells by simian virus 40 small tumor antigen or protein networks are implicated in the malignant phenotype of head and neck cancer cell
phosphatase2A B56gamma knockdown. Canc. Res. 64, 6978–6988. https://doi: 10 lines differing in p53 status. Genome Biol. 9, R53. https://doi.org/10.1186/gb-2008-
.1158/0008-5472.CAN-04-1150. 9-3-r53.
O’Reilly, E.M., Abou-Alfa, G.K., 2007. Cytotoxic therapy for advanced pancreatic Yan, H., Qiu, W., Koehne de Gonzalez, A.K., Wei, J.S., Tu, M., Xi, C.H., Yang, Y.R.,
adenocarcinoma. Semin. Oncol. 34, 347–353. https://doi: 10.1053/j.seminoncol. Peng, Y.P., Tsai, W.Y., Remotti, H.E., Miao, Y., Su, G.H., 2019. HHLA2 is a novel
2007.05.009. immune checkpoint protein in pancreatic ductal adenocarcinoma and predicts post-
Ponta, H., Sherman, L., Herrlich, P.A., 2003. CD44: from adhesion molecules to surgical survival. Canc. Lett. 442, 333–340. https://doi: 10.1016/j.canlet.20
signalling regulators. Nat. Rev. Mol. Cell Biol. 4, 33–45. https://doi: 10.103 18.11.007.
8/nrm1004. Yang, M.C., Wang, H.C., Hou, Y.C., Tung, H.L., Chiu, T.J., Shan, Y.S., 2015. Blockade of
Raj, D., Aicher, A., Heeschen, C., 2015. Concise review: stem cells in pancreatic cancer: autophagy reduces pancreatic cancer stem cell activity and potentiates the
from concept to translation. Stem Cell. 33, 2893–2902. https://doi: 10.1002/ tumoricidal effect of gemcitabine. Mol. Canc. 14, 179.
stem.2114. Yoshida, G.J., Saya, H., 2016. Therapeutic strategies targeting cancer stem cells. Canc.
Reya, T., Morrison, S.J., Clarke, M.F., Weissman, I.L., 2001. Stem cells, cancer, and Sci. 107, 5–11. https://doi: 10.1186/s12943-015-0449-3.
cancer stem cells. Nature 414, 105–111. https://doi: 10.1038/35102167. Zeng, D., Ma, J., Li, R., Yang, J., Yin, X., 2018. The inhibitory effect of 5,7-DMF on
Siegel, R., Naishadham, D., Jemal, A., 2013. Cancer statistics, 2013. CA. Cancer. J. Clin. pancreatic sphere-forming cell function mediated by FoxM1 gene expression. J. Cell.
63, 11–30. https://doi:10.3322/caac.21166. Biochem. 119, 1855–1865. https://doi: 10.1002/jcb.26346.
Siegel, R.L., Miller, K.D., Jemal, A., 2019. Cancer statistics, 2019. CA. Cancer. J. Clin. 69, Zhou, C., Qian, W., Ma, J., Cheng, L., Jiang, Z., Yan, B., Li, J., Duan, W., Sun, L., Cao, J.,
7–34. https://doi: 10.3322/caac.21551. Wang, F., Wu, E., Wu, Z., Ma, Q., Li, X., 2019. Resveratrol enhances the
Stathis, A., Moore, M.J., 2010. Advanced pancreatic carcinoma: current treatment and chemotherapeutic response and reverses the stemness induced by gemcitabine in
future challenges. Nat. Rev. Clin. Oncol. 7, 163–172. https://doi: 10.1038/nrclinonc pancreatic cancer cells via targeting SREBP1. Cell Prolif 52, e12514. https://doi:
.2009.236. 10.1111/cpr.12514.
Tokunaga, E., Oki, E., Egashira, A., Sadanaga, N., Morita, M., Kakeji, Y., Maehara, Y., Zuo, C., Hong, Y., Qiu, X., Yang, D., Liu, N., Sheng, X., Zhou, K., Tang, B., Xiong, S.,
2008. Deregulation of the Akt pathway in human cancer. Curr. Cancer Drug Targets Ma, M., Liu, Z., 2018. Celecoxib suppresses proliferation and metastasis of pancreatic
8, 27–36. https://doi.org/10.2174/156800908783497140. cancer cells by down-regulating STAT3/NF-kB and L1CAM activities. Pancreatology
18, 328–333. https://doi: 10.1016/j.pan.2018.02.006.

You might also like