Additive antitumor effects of celecoxib and simvastatin on head and neck squamous cell carcinoma in vitro

  • Authors:
    • Thomas Gehrke
    • Agmal Scherzad
    • Stephan Hackenberg
    • Pascal Ickrath
    • Philipp Schendzielorz
    • Rudolf Hagen
    • Norbert Kleinsasser
  • View Affiliations

  • Published online on: July 10, 2017     https://doi.org/10.3892/ijo.2017.4071
  • Pages: 931-938
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lipid-lowering statins as well as non-steroidal anti-inflammatory drugs (NSAIDs) have been reported to possess cancer-protective effects in many epidemiologic cohort studies. However, the underlying mechanisms of these findings are mostly unknown. To evaluate possible additive antitumor effects of statins and NSAIDs in vitro, PJ-41 and HLaC78 head and neck squamous cell carcinoma cells (HNSCC) were treated with 40 µM celecoxib, 50 µM simvastatin or a combination of both. Analysis of tumor viability, proliferation, apoptosis, cell cycle changes and secretion of interleukin-6 (IL-6) and interleukin-8 (IL-8) was conducted via MTT assay, Annexin V-propidium iodide test, cell cycle analysis, colony assay and enzyme-linked immunosorbent assay (ELISA). Celecoxib and simvastatin alone as well as a combined treatment showed a significant reduction in tumor cell viability, proliferation and secretion of IL-6 and IL-8 compared to the control group. The combined treatment even proved to have significantly greater effects. We postulate that simvastatin and celecoxib have additive antitumor effects on HNSCC in vitro, which warrants further investigation.

Introduction

Squamous cell carcinoma of the head and neck (HNSCC) is one of the most commonly occurring malignancies, and is a major cause of cancer morbidity and mortality worldwide with an estimated incidence of 500,000 per year in the USA (1). The overall 5-year survival rate for pharyngeal and oral squamous cell carcinoma is approximately 60% in the USA, and has not changed significantly during the past 40 years (2). Due to the increasing spread of the human papillomavirus within the oropharyngeal tract, which represents a new pathogenetic factor, the incidence of HNSCC is estimated to rise even more (3). Thus, there is a tremendous need for new treatment options for HNSCC patients.

Statins are widely used as cholesterol-lowering drugs, being small-molecule inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (4). In addition to their common use in the treatment of lipid disorders, statins have also demonstrated anticarcinogenic properties in various preclinical in vitro studies (5,6). This has been attributed mainly to the inhibition of isoprenoid and cholesterol synthesis, which are both important processes in the intracellular signaling pathways (6,7). Several observational human studies have reported a potential beneficial effect of statin use against the overall risk of cancer (810). Other studies, however, reported no such protective effects (11,12). For several specific cancers, especially for colorectal cancer (13), lung cancer (14,15) and renal cell carcinoma (16), protective effects of statin use have been published.

NSAIDs have also been demonstrated to have a potential chemoprotective effect. This has been explained by an induction of cell cycle arrest in G1-Phase via inhibition of Akt (17), inhibition of Ca2+ ATPase activity (18) or activating p53 and p21 (19). Several studies have reported on a protective effect of Aspirin on colorectal adenoms as well as colorectal carcinomas, even in randomized, double-blinded and placebo-controlled study designs (2023). For selective cyclooxygenase (COX)-2 inhibitors such as celecoxib or rofecoxib, protective effects could also be demonstrated, again mainly for colorectal carcinoma (2426). However, since there have been serious cardiovascular complications regarding long-term therapy with COX-2-inhibitors (2729), the relatively high doses needed for the observed cancer-protective effect are being questioned. Therefore, a combination with other drugs with synergistic effects to reduce the dosage of NSAIDs is warranted.

A combined therapy of statins and NSAIDs has already been demonstrated to have a synergistic effect on the induction of apoptosis in prostate and colorectal cancer cells in vitro (30,31). In vivo, a low-dose combination of atorvastatin and celecoxib was reported to have synergistic antitumor effects on colorectal cancer in a xenograft animal model (32). For colorectal cancer, population-based studies have also shown synergistic cancer-protective effects of a combination therapy with statins and NSAIDs compared to the respective monotherapies (33,34). Yet for other entities, for example squamous cell carcinoma, few studies have been conducted to date.

The objective of the present study is to analyze the possible additive effects of the combined use of simvastatin and celecoxib on human HNSCC cells in vitro in terms of viability, cell growth, apoptosis and cell cycle changes. Additionally, secretion of selected interleukins, namely IL-6 and IL-8, was analyzed. IL-6 plays a key role in cell proliferation, apoptosis and differentiation. IL-6 induces activation of Janus kinase 1/2 (JAK1/2), resulting in phosphorylation of STAT3 at tyrosine-705 (Y705) (35). Activation of IL-6/STAT3 signaling has a significant role in self-renewal and acquisition of malignant features of cancer stem cells (CSC) (36). Furthermore, IL-6 levels are highly elevated in metastatic diseases and increased levels of serum IL-6 are associated with poor disease outcome and prognosis in human cancers (37,38). IL-8 is reported to be related to different malignancies due to the involvement of thrombophilia and angiogenesis (39). Increased secretion of IL-8 has been shown to increase the metastatic ability of different cancer entities (40,41). Therefore, analysis of IL-6 and IL-8 was included into the present study.

Materials and methods

Cell culture

The head and neck squamous carcinoma cell lines PE/CA-PJ 41 and HLaC78 were obtained from ECACC (European Collection of Cell Cultures, Salisbury, Wiltshire, UK). Cells were grown in RPMI-expansion medium (RPMI-EM) consisting of RPMI-1640 medium (Biochrom AG, Berlin, Germany) with 10% FCS, 100 U/ml penicillin, 100 µg/ml streptomycin, 1% sodium pyruvate (100 mM, Biochrom AG), and 1% non-essential amino acids (100-fold concentration, Biochrom AG). Cells were cultured at 37°C with 5% CO2 in culture flasks. Medium was replaced every other day and passaging was performed after reaching 70–80% confluence by trypsinization, with subsequent washing and seeding in new flasks or treatment wells. Experiments were performed using cells in the exponential growth phase.

Exposure to celecoxib and simvastatin

The half-maximal inhibitory concentrations (IC50) of celecoxib (Pfizer Pharma PFE, Berlin, Germany) and simvastatin (MIP Pharma, Blieskastel, Germany) on PE/CA-PJ 41 were evaluated with the MTT assay (Fig. 1). To this end, PE/CA-PJ-41 cells were treated with 40 µM/ml celecoxib, 50 µM/ml simvastatin or the combination of both. Analytical assays were performed after 24 h of incubation.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

After 3 days of co-culture, the MTT (Sigma-Aldrich, Taufkirchen, Germany) colorimetric staining method was performed according to Mosmann (42) to determine cell viability. Cells were seeded at 10,000 cells per well in a 12-well plate. All wells were incubated with 1 ml MTT (1 mg/ml) for 5 h at 37°C and 5% CO2. MTT was then removed and 1 ml isopropanol was added, followed by another incubation period of 1 h at 37°C and 5% CO2. Measurement of the color conversion of the blue formazan dye was performed using a multi-plate reader (Titertek Multiskan PLUS MK II; Thermo Labsystems, Thermo Fisher Scientific, Inc., Darmstadt, Germany) at a wavelength of 570 nm.

Colony assay

PE/CA-PJ 41 were seeded into 6-well plates at a concentration of 2.5×103 cells/well in triplicate. Celecoxib (40 µM), 50 µM simvastatin or the combination of both were added to defined well plates. PE/CA-PJ 41 cultivated in RPMI-EM served as the control. Cells were incubated for 14 days. After 2 weeks the well plates were stained with crystal violet, and colonies were counted manually.

Annexin V-propidium iodide test

The BD Pharmingen Annexin V-APC kit (BD Biosciences, Heidelberg, Germany) was used to evaluate apoptosis on HLaC78 and PE/CA-PJ 41. After 3 days of co-culture, cells in suspension and adherent cells were harvested, then washed twice with PBS and resuspended in 1:10 binding buffer [0.1 M HEPES (Sigma-Aldrich) (pH 7.4), 1.4 M NaCl, 25 mM CaCl2] at a concentration of 1×106 cells/ml. Aliquots of this cell suspension (100 µl; 1×105 cells) were then transferred to a 5 ml culture tube. Propidium iodide (5 µl) and Annexin V-APC (5 µl) were added to each aliquot. After 15 min of incubation at room temperature in the dark, the cells were resuspended with 400 µl 1:10 binding buffer. A FACSCanto flow cytometer was used to analyze the samples with BD FACSDiva version 5.0.3 software (BD Biosciences). Only cells with damaged membranes were stained by propidium iodide.

Cell cycle analysis

To analyze the effect of celecoxib and salinomycin on the cell cycle of PE/CA-PJ-41 and HLaC78, 1×105 cells were cultivated in 12-well plates in triplicate. Following a 48 h period, PE/CA-PJ-41 cells were trypsinized and washed twice with cold PBS. Cells were then fixed in 1ml of 70% cold ethanol in test tubes and incubated for 2 h at 4°C in the dark. After incubation, cells were centrifuged at 500 × g for 5 min at 4°C and resuspended in 500 µl propidium iodide (BD Bioscience). After another incubation at 4°C in the dark for 15 min, cells were analyzed with flow cytometry within 1 h. PE/CA-PJ-41 cultivated in RPMI-EM served as the control.

IL-6/IL-8 ELISA

For measurement of the secretion of IL-6 and IL-8, the super natants were collected (centrifugation, 150 × g for 5 min at 37°C) after 3 days of co-culture and stored at −20°C in sterile tubes until further use. RPMI-EM served as the control. Human IL-6 and IL-8 kits (catalog nos. 950.030.192 and 950.050.192, respectively; Diaclone SAS, Besançon, France) were used and the experiments were performed in duplicate. The ELISA plate was read at 450 nm (Titertek Multiskan PLUS MK II). The concentrations of IL-6 and IL-8 were determined by constructing a standard curve using recombinant IL-6 and IL-8.

Statistical analysis

The data collected was transferred to standard spreadsheets and statistically analyzed using GraphPad Prism software (version 4.0; GraphPad Software, Inc., San Diego, CA, USA). Data are presented as the mean ± standard deviation of three experiments, unless otherwise stated. The Gaussian distribution was tested via first column analysis. One-way analysis of variance followed by Tukey's multiple comparison test was used. Additionally, multiplicity adjusted p-values were determined. p<0.05 was used to indicate a statistically significant difference. The combination index (CI) was applied to evaluate the interaction between celecoxib and simvastatin for PE/CA-PJ 41. CI analysis provides qualitative information on the nature of drug interaction, and the CI index, a numerical value calculated as described below, also provides a quantitative measure of the extent of drug interaction (43).

CI=CA,X:ICX,A+CB,X:ICX,B CA,X and CB,X are the concentrations of drug A and drug B used in combination to achieve x% drug effect (IC75, IC50). ICX,A and ICX,B are the concentrations required for single agents to achieve the same effect. A CI of <0.85 was deemed to indicate synergy, a CI of >1.15 was deemed to indicate antagonism. Additive effects were assumed at an CI between 0.85 and 1.15.

Results

MTT assay

Viability of PE/CA-PJ-41 cells was analyzed using the MTT assay (Fig. 2). It revealed a significantly lower viability after addition of celecoxib, simvastatin and the combination of both compared to the control group (p<0.05 for all three). The combination of treatment with celecoxib and simvastatin also proved to decrease tumor cell viability significantly more compared to celecoxib and simvastatin alone (p<0.05 for both). Between treatment with celecoxib and simvastatin, no significant difference was found (p>0.05). The CI was calculated as 1.13, indicating a moderate additive effect (Fig. 3).

Colony assay

Tumor cell proliferation was analyzed using a colony assay (Figs. 4 and 5). Treatment with celecoxib, simvastatin and the combination of both showed significantly reduced cell colonies compared to the control group (p<0.05 for all three). Colony forming was higher when incubated with celecoxib alone versus simvastatin alone (p<0.05). The combination of both drugs showed no significant difference compared to celecoxib (p>0.05) or simvastatin (p>0.05) alone.

Annexin V-propidium iodide test

Annexin V-propidium iodide analysis of PE/CA-PJ 41 (Fig. 6A) and HLaC78 (Fig. 6B) revealed enhanced apoptosis and necrosis after simvastatin treatment alone and after the combination of both compared to the control group (p<0.05 for both). The combination of both drugs induced higher rates of apoptosis and necrosis compared to simvastatin and celecoxib alone (p<0.05). Addition of celecoxib alone had no significant difference on apoptosis or necrosis compared to the control (p>0.05).

Cell cycle analysis

For both cell lines used, cell cycle analysis showed a significant increase in cells in G0/G1-phase when treated with celecoxib and simvastatin in combination compared to the control group (p<0.05), while celecoxib (p>0.05) and simvastatin (p>0.05) alone had no significant effect. The effect of the combination therapy was also significant compared to celecoxib (p<0.05) and simvastatin (p<0.05) alone (Fig. 7A and B).

Quantitative analysis of IL-6

Treatment with celecoxib, simvastatin and the combination of both all showed a lower secretion of IL-6 compared to the control group (p<0.05 for all three). Addition of simvastatin proved to decrease IL-6 secretion significantly more than celecoxib (p<0.05). The combined treatment of both drugs in turn showed less IL-6 than celecoxib (p<0.05) or simvastatin alone (p<0.05; Fig. 8).

Quantitative analysis of IL-8

Treatment with celecoxib, simvastatin and the combination of both all revealed a lower secretion of IL-8 compared to the control group (p<0.05 for all three). Incubation with celecoxib showed significantly decreased IL-8 secretion than with simvastatin (p<0.05). The combined treatment of both drugs proved to reduce IL-8-production significantly more than celecoxib (p<0.05) or simvastatin alone (p<0.05; Fig. 9).

Discussion

Despite many advances in the therapy of HNSCC, survival rates remain low (44). Anticancer drug treatment for HNSCC today is mostly reserved for palliative chemotherapy regimens, which include cytostatic agents such as cisplatin, 5-FU or docetaxel as well as monoclonal antibodies such as cetuximab. However, these drugs offer small benefit with respect to progression-free survival, while in turn inducing severe side effects further limiting the use in cancer patients (45). Therefore, research for identifying new treatment options with reduced toxicities is warranted.

Simvastatin is an inhibitor of HNG-CoA reductase, an enzyme of the mevalonate synthesis pathway, which in turn inhibits formation of downstream lipid isoprenoids such as farnesyl pyrophosphate (FFP) and geranylgeranyl pyrophosphate (GGPP) (46). This in turn results in the side effect of decreasing cell proliferation via inhibition of Ras oncogenes (47). Statins have also been shown to induce apoptosis, reduce serum-stimulated Ras activity and increase messenger RNA (mRNA) and protein expression of the proapoptotic proteins Bax and Bad in esophageal carcinoma cell lines (48).

NSAIDs in general (2123), as well as selective COX-2-inhibitors (2426), have already proven to be potent tumor-protective substances in vitro and in vivo. However, relatively high doses of NSAIDs or selective COX-2-inhibitors are needed to achieve the desired effects, which causes problems for long-term therapy due to the cardiovascular risks of these drugs (2729). Thus, a combination of NSAIDs with other possibly synergistic drugs, for example statins, could be a solution for reducing the required doses for each.

The combination of statins and NSAIDs has already been demonstrated to have synergistic effects on colorectal cancer cells in vitro (30,31) and in an animal model in vivo (32). Yet, as of now few studies have evaluated the effects of this combination therapy on HNSCC cells. Thus, the present study focused on the synergistic effects of celecoxib and simvastatin on HNSCC cells in vitro.

The analyses showed significant reduction in PE/CA-PJ-41 tumor cell proliferation and viability after addition of celecoxib or simvastatin alone, with the effect increasing even more using a combination of both substances. This confirms results of colorectal cancer cells and prostate cancer cells treated with celecoxib and simvastatin in combination (3032,46). The underlying mechanisms of these anticarcinogenic effects are not completely understood, however.

In the present study, these antitumor effects were mainly caused by apoptosis and, to a much lesser extent, by necrosis. By inhibition of HMG-CoA reductase, statins inhibit the synthesis of isoprenoids essential for membrane localization and subsequent activation of signaling proteins such as Ras, Rho and Rac, leading to increased apoptosis (4). Moreover, the reduction of cholesterol synthesis via statins and their inhibition of the Akt pathway has been shown to promote apoptosis in cancer cells (7). NSAIDs, on the other hand, also have a variety of possible mechanisms that determine their anticarcinogenic properties. Besides inhibition of Ca2+ ATPase activity (18), increase in ceramid levels (49), and inhibition of transcription activity of NFκB (50), NSAIDs have also shown the potential for inhibition of the Akt pathway, as does simvastatin (17). Even in concentrations that do not induce direct inhibition of Akt by celecoxib itself, it could be demonstrated that celecoxib significantly synergized atorvastatin to inhibit Akt-phosphorylation, indicating a pivotal synergistic effect of both substances regarding Akt-pathway-induced apoptosis (46).

In addition, significant cell cycle arrest in G0/G1-phase could be demonstrated for the combination therapy in the present study. Celecoxib has already been shown to induce cell cycle arrest at G1-phase via increased expression of cyclin-dependent kinase (CDK) inhibitors for various tumor cell types (51,52). The combination of atorvastatin and celecoxib has also been demonstrated to cause cell cycle arrest at G0/G1-phase at a significantly higher level than both substances alone (46). Thus, the induction of cell cycle arrest at G0/G1-phase could also be a potential synergistic effect. Since G0/G1-arrest inhibits the proliferation of tumor cells, it is a vital target for anticancer therapeutics. However, whether this G0/G1-arrest is irreversible, as has been described for Terfenadine (53), or perhaps even reversible, remains unclear since the present study only measured one time point after treatment. Preliminary analysis hint at an increase of cyclin-dependent kinase inhibitors p21Cip1/Waf1 and p27Kip1 as a possible mechanism behind the G0/G1-arrest. However, in the present study no complete analysis of cell cycle protein expression has been conducted, limiting the information in this regard. It will be part of a future investigation at our institution.

The present study also revealed a significantly lower secretion of IL-6 and IL-8 by the tumor cells after addition of simvastatin and celecoxib combined rather than alone. IL-6 is a cytokine which, among other functions, induces STAT3 phosphorylation via IL-6 receptors and Janus family kinases (JAK), and is thereby involved in cell proliferation, angiogenesis and apoptosis (54,55). For hepatocellular carcinoma, celecoxib has already been demonstrated to inhibit IL-6/IL-6-receptor-induced JAK2/STAT3 phosphorylation (56). In a study focusing on arthritis, celecoxib significantly reduced secretion of IL-6 and IL-8 in synovial fluid (57). Similarly, simvastatin was also shown to inhibit IL-6 and IL-8 production in rheumatoid arthritis (58). However, the effect of a combination of celecoxib and simvastatin on IL-6-secretion has not been investigated thus far, and in particular not for HNSCC.

Interleukin-8 (IL-8), one of the ELR+ CXC family of chemokines, is a potent proangiogenic factor and its expression is associated with angiogenesis, tumor progression and survival in patients with cancer (59,60). NSAIDs have been shown to have inhibitory effects on angiogenesis for pancreatic tumors in a mouse model, even by COX-independent mechanisms (61), and have proven to inhibit other proangiogenetic factors such as matrixmetalloprotease (MMP)-2 and -9, as well as early growth response factor EGR-1 (6264). Simvastatin could be demonstrated to inhibit the production of IL-6 and IL-8 as well as cell proliferation in patients with rheumatoid arthritis (58). To our knowledge, the present study is the first to evaluate the effect of a combination of celecoxib and simvastatin on IL-8 production, and therefore the angiogenesis of tumor cells, which may be of great value in the treatment of metastatic cancer considering the critical role of angiogenesis.

Still, there is much controversy about the concentrations of simvastatin and celecoxib used in in vitro experiments, regarding the expected in vivo dose necessary to achieve similar effects in humans (65). Most in vitro studies regarding cancer therapy have come to use concentrations of 40–60 µM of simvastatin (66). Although it is true that the in vitro concentrations generally used in oncologic studies are higher than the expected dose in vivo, there is also the problem of accumulation in the target organ, the liver, which at least partially makes up for the difference in concentrations (67). The same is valid for celecoxib, where the concentrations generally used in vitro are also slightly higher than the expected in vivo doses achievable in humans (68). Therefore, to further reduce possible side effects when examining the co-medication in vivo, lower concentrations should also be tested for possible further synergistic effects in the future.

Since especially simvastatin intervenes in the synthesis of Cholesterol, a key in cellular integrity, there could be possible damage to regular human cells as well. Gauthaman et al, on the other hand, already showed that simvastatin decreased viability and proliferation of cancer cells and cancer stem cells, but had no effect on normal human stem cells (69). This, coupled with the years of clinical practice and experience with possible side effects of both substances, may still make it worthwhile to further examine their possible use as anticancer drugs.

In conclusion, it could be demonstrated that a combination of celecoxib and simvastatin has significant synergistic effects on reducing tumor cell proliferation and viability in HNSCC cells in vitro. These antitumor effects are based on apoptosis and cell cycle arrest in G0/G1-phase. Furthermore, a reduction in the secretion of IL-6 and IL-8 could be shown, indicating additional ways this synergism works to inhibit tumor growth, such as via antiangiogenesis. At present, medical tumor therapy for HNSCC is still limited. However, since both substances have a good risk-benefit ratio based on their long-term clinical use as lipid-lowering and anti-inflammational drugs, at least in concentrations used in the present study, their combined use for cancer therapy clearly warrants further investigation. Future studies will need to elucidate the intracellular mechanisms behind these effects. Especially analysis of mitogenic and other signaling pathways are relevant targets for further investigations.

Acknowledgments

This study was supported by the Rudolf Bartling Foundation.

References

1 

Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R and Jemal A: Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 66:271–289. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Herrero R, Castellsagué X, Pawlita M, Lissowska J, Kee F, Balaram P, Rajkumar T, Sridhar H, Rose B, Pintos J, et al IARC Multicenter Oral Cancer Study Group: Human papillomavirus and oral cancer: The International Agency for Research on Cancer multicenter study. J Natl Cancer Inst. 95:1772–1783. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Xiao H and Yang CS: Combination regimen with statins and NSAIDs: A promising strategy for cancer chemoprevention. Int J Cancer. 123:983–990. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Prasanna P, Thibault A, Liu L and Samid D: Lipid metabolism as a target for brain cancer therapy: Synergistic activity of lovastatin and sodium phenylacetate against human glioma cells. J Neurochem. 66:710–716. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Xia Z, Tan MM, Wong WW, Dimitroulakos J, Minden MD and Penn LZ: Blocking protein geranylgeranylation is essential for lovastatin-induced apoptosis of human acute myeloid leukemia cells. Leukemia. 15:1398–1407. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Zhuang L, Kim J, Adam RM, Solomon KR and Freeman MR: Cholesterol targeting alters lipid raft composition and cell survival in prostate cancer cells and xenografts. J Clin Invest. 115:959–968. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Blais L, Desgagné A and LeLorier J: 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors and the risk of cancer: A nested case-control study. Arch Intern Med. 160:2363–2368. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Graaf MR, Beiderbeck AB, Egberts AC, Richel DJ and Guchelaar HJ: The risk of cancer in users of statins. J Clin Oncol. 22:2388–2394. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Friis S, Poulsen AH, Johnsen SP, McLaughlin JK, Fryzek JP, Dalton SO, Sørensen HT and Olsen JH: Cancer risk among statin users: A population-based cohort study. Int J Cancer. 114:643–647. 2005. View Article : Google Scholar

11 

Olsen JH, Johansen C, Sørensen HT, McLaughlin JK, Mellemkjaer L, Steffensen FH and Fraumeni JF Jr: Lipid-lowering medication and risk of cancer. J Clin Epidemiol. 52:167–169. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Kaye JA and Jick H: Statin use and cancer risk in the General Practice research Database. Br J Cancer. 90:635–637. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Poynter JN, Gruber SB, Higgins PD, Almog R, Bonner JD, Rennert HS, Low M, Greenson JK and Rennert G: Statins and the risk of colorectal cancer. N Engl J Med. 352:2184–2192. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Khurana V, Bejjanki HR, Caldito G and Owens MW: Statins reduce the risk of lung cancer in humans: A large case-control study of US veterans. Chest. 131:1282–1288. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Farwell WR, Scranton RE, Lawler EV, Lew RA, Brophy MT, Fiore LD and Gaziano JM: The association between statins and cancer incidence in a veterans population. J Natl Cancer Inst. 100:134–139. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Khurana V, Caldito G and Ankem M: Statins might reduce risk of renal cell carcinoma in humans: Case-control study of 500,000 veterans. Urology. 71:118–122. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Kulp SK, Yang YT, Hung CC, Chen KF, Lai JP, Tseng PH, Fowble JW, Ward PJ and Chen CS: 3-phosphoinositide-dependent protein kinase-1/Akt signaling represents a major cyclooxygenase-2-independent target for celecoxib in prostate cancer cells. Cancer Res. 64:1444–1451. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Johnson AJ, Hsu AL, Lin HP, Song X and Chen CS: The cyclooxygenase-2 inhibitor celecoxib perturbs intracellular calcium by inhibiting endoplasmic reticulum Ca2+-ATPases: A plausible link with its anti-tumour effect and cardiovascular risks. Biochem J. 366:831–837. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Luciani MG, Campregher C and Gasche C: Aspirin blocks proliferation in colon cells by inducing a G1 arrest and apoptosis through activation of the checkpoint kinase ATM. Carcinogenesis. 28:2207–2217. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Thun MJ, Henley SJ and Patrono C: Nonsteroidal anti-inflammatory drugs as anticancer agents: Mechanistic, pharmacologic, and clinical issues. J Natl Cancer Inst. 94:252–266. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Giardiello FM, Hamilton SR, Krush AJ, Piantadosi S, Hylind LM, Celano P, Booker SV, Robinson CR and Offerhaus GJ: Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. N Engl J Med. 328:1313–1316. 1993. View Article : Google Scholar : PubMed/NCBI

22 

Labayle D, Fischer D, Vielh P, Drouhin F, Pariente A, Bories C, Duhamel O, Trousset M and Attali P: Sulindac causes regression of rectal polyps in familial adenomatous polyposis. Gastroenterology. 101:635–639. 1991. View Article : Google Scholar : PubMed/NCBI

23 

Nugent KP, Farmer KC, Spigelman AD, Williams CB and Phillips RK: Randomized controlled trial of the effect of sulindac on duodenal and rectal polyposis and cell proliferation in patients with familial adenomatous polyposis. Br J Surg. 80:1618–1619. 1993. View Article : Google Scholar : PubMed/NCBI

24 

Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB, Wakabayashi N, Saunders B, Shen Y, Fujimura T, et al: The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med. 342:1946–1952. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Bertagnolli MM, Eagle CJ, Zauber AG, Redston M, Solomon SD, Kim K, Tang J, Rosenstein RB, Wittes J, Corle D, et al APC Study Investigators: Celecoxib for the prevention of sporadic colorectal adenomas. N Engl J Med. 355:873–884. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Arber N, Eagle CJ, Spicak J, Rácz I, Dite P, Hajer J, Zavoral M, Lechuga MJ, Gerletti P, Tang J, et al PreSAP Trial Investigators: Celecoxib for the prevention of colorectal adenomatous polyps. N Engl J Med. 355:885–895. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Bresalier RS, Sandler RS, Quan H, Bolognese JA, Oxenius B, Horgan K, Lines C, Riddell R, Morton D, Lanas A, et al Adenomatous Polyp Prevention on Vioxx (APPROVe) Trial Investigators: Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med. 352:1092–1102. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Kerr DJ, Dunn JA, Langman MJ, Smith JL, Midgley RS, Stanley A, Stokes JC, Julier P, Iveson C, Duvvuri R, et al VICTOR Trial Group: Rofecoxib and cardiovascular adverse events in adjuvant treatment of colorectal cancer. N Engl J Med. 357:360–369. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Solomon SD, McMurray JJ, Pfeffer MA, Wittes J, Fowler R, Finn P, Anderson WF, Zauber A, Hawk E and Bertagnolli M; Adenoma Prevention with Celecoxib (APC) Study Investigators: Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med. 352:1071–1080. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Swamy MV, Cooma I, Reddy BS and Rao CV: Lamin B, caspase-3 activity, and apoptosis induction by a combination of HMG-CoA reductase inhibitor and COX-2 inhibitors: A novel approach in developing effective chemopreventive regimens. Int J Oncol. 20:753–759. 2002.PubMed/NCBI

31 

Agarwal B, Rao CV, Bhendwal S, Ramey WR, Shirin H, Reddy BS and Holt PR: Lovastatin augments sulindac-induced apoptosis in colon cancer cells and potentiates chemopreventive effects of sulindac. Gastroenterology. 117:838–847. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Zheng X, Cui XX, Avila GE, Huang MT, Liu Y, Patel J, Kong AN, Paulino R, Shih WJ, Lin Y, et al: Atorvastatin and celecoxib inhibit prostate PC-3 tumors in immunodeficient mice. Clin Cancer Res. 13:5480–5487. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Hoffmeister M, Chang-Claude J and Brenner H: Individual and joint use of statins and low-dose aspirin and risk of colorectal cancer: A population-based case-control study. Int J Cancer. 121:1325–1330. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Flick ED, Habel LA, Chan KA, Van Den Eeden SK, Quinn VP, Haque R, Orav EJ, Seeger JD, Sadler MC, Quesenberry CP Jr, et al: Statin use and risk of prostate cancer in the California Men's Health Study cohort. Cancer Epidemiol Biomarkers Prev. 16:2218–2225. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Taga T, Hibi M, Hirata Y, Yamasaki K, Yasukawa K, Matsuda T, Hirano T and Kishimoto T: Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell. 58:573–581. 1989. View Article : Google Scholar : PubMed/NCBI

36 

He G, Dhar D, Nakagawa H, Font-Burgada J, Ogata H, Jiang Y, Shalapour S, Seki E, Yost SE, Jepsen K, et al: Identification of liver cancer progenitors whose malignant progression depends on autocrine IL-6 signaling. Cell. 155:384–396. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Trikha M, Corringham R, Klein B and Rossi JF: Targeted anti-interleukin-6 monoclonal antibody therapy for cancer: a review of the rationale and clinical evidence. Clin Cancer Res. 9:4653–4665. 2003.PubMed/NCBI

38 

Zhang GJ and Adachi I: Serum interleukin-6 levels correlate to tumor progression and prognosis in metastatic breast carcinoma. Anticancer res. 19:1427–1432. 1999.PubMed/NCBI

39 

Zabaleta J, Su LJ, Lin HY, Sierra RA, Hall MC, Sartor AO, Clark PE, Hu JJ and Ochoa AC: Cytokine genetic polymorphisms and prostate cancer aggressiveness. Carcinogenesis. 30:1358–1362. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Li A, Varney ML and Singh RK: Expression of interleukin 8 and its receptors in human colon carcinoma cells with different metastatic potentials. Clin Cancer res. 7:3298–3304. 2001.PubMed/NCBI

41 

Ren Y, Poon RT, Tsui HT, Chen WH, Li Z, Lau C, Yu WC and Fan ST: Interleukin-8 serum levels in patients with hepatocellular carcinoma: correlations with clinicopathological features and prognosis. Clin Cancer res. 9:5996–6001. 2003.PubMed/NCBI

42 

Mosmann T: Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J Immunol Methods. 65:55–63. 1983. View Article : Google Scholar : PubMed/NCBI

43 

Chou TC and Talalay P: Quantitative analysis of dose-effect relationships: The combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 22:27–55. 1984. View Article : Google Scholar : PubMed/NCBI

44 

Chan GG, Tai BC, Liang S, Lim DT and Soo KC: Squamous cell carcinoma of the head and neck (HNSCC) - multi-modality treatment and impact on survival. Asian J Surg. 25:35–40. 2002.

45 

Patil V, Joshi A, Noronha V, Deodhar J, Bhattacharjee A, Dhumal S, Chandrakanth MV, Karpe A, Talreja V, ChandRasekharan A, et al: Expectations and preferences for palliative chemotherapy in head and neck cancers patients. Oral Oncol. 63:10–15. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Xiao H, Zhang Q, Lin Y, Reddy BS and Yang CS: Combination of atorvastatin and celecoxib synergistically induces cell cycle arrest and apoptosis in colon cancer cells. Int J Cancer. 122:2115–2124. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Goldstein JL and Brown MS: Regulation of the mevalonate pathway. Nature. 343:425–430. 1990. View Article : Google Scholar : PubMed/NCBI

48 

Ogunwobi OO and Beales IL: Statins inhibit proliferation and induce apoptosis in Barrett's esophageal adenocarcinoma cells. Am J Gastroenterol. 103:825–837. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Kundu N, Smyth MJ, Samsel L and Fulton AM: Cyclooxygenase inhibitors block cell growth, increase ceramide and inhibit cell cycle. Breast Cancer Res Treat. 76:57–64. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Kim SH, Song SH, Kim SG, Chun KS, Lim SY, Na HK, Kim JW, Surh YJ, Bang YJ and Song YS: Celecoxib induces apoptosis in cervical cancer cells independent of cyclooxygenase using NF-kappab as a possible target. J Cancer res Clin Oncol. 130:551–560. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Grösch S, Tegeder I, Niederberger E, Bräutigam L and Geisslinger G: COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib. FASEB J. 15:2742–2744. 2001.PubMed/NCBI

52 

Kardosh A, Blumenthal M, Wang WJ, Chen TC and Schönthal AH: Differential effects of selective COX-2 inhibitors on cell cycle regulation and proliferation of glioblastoma cell lines. Cancer Biol Ther. 3:55–62. 2004. View Article : Google Scholar : PubMed/NCBI

53 

Liu JD, Wang YJ, Chen CH, Yu CF, Chen LC, Lin JK, Liang YC, Lin SY and Ho YS: Molecular mechanisms of G0/G1 cell-cycle arrest and apoptosis induced by terfenadine in human cancer cells. Mol Carcinog. 37:39–50. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Berishaj M, Gao SP, Ahmed S, Leslie K, Al-Ahmadie H, Gerald WL, Bornmann W and Bromberg JF: Stat3 is tyrosine-phosphorylated through the interleukin-6/glycoprotein 130/Janus kinase pathway in breast cancer. Breast Cancer Res. 9:R322007. View Article : Google Scholar : PubMed/NCBI

55 

Bollrath J, Phesse TJ, von Burstin VA, Putoczki T, Bennecke M, Bateman T, Nebelsiek T, Lundgren-May T, Canli O, Schwitalla S, et al: gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis. Cancer Cell. 15:91–102. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Liu Y, Liu A, Li H, Li C and Lin J: Celecoxib inhibits interleukin-6/interleukin-6 receptor-induced JAK2/STAT3 phosphorylation in human hepatocellular carcinoma cells. Cancer Prev Res (Phila). 4:1296–1305. 2011. View Article : Google Scholar

57 

Bianchi M, Broggini M, Balzarini P, Franchi S and Sacerdote P: Effects of nimesulide on pain and on synovial fluid concentrations of substance P, interleukin-6 and interleukin-8 in patients with knee osteoarthritis: Comparison with celecoxib. Int J Clin Pract. 61:1270–1277. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Yokota K, Miyazaki T, Hirano M, Akiyama Y and Mimura T: Simvastatin inhibits production of interleukin 6 (IL-6) and IL-8 and cell proliferation induced by tumor necrosis factor-alpha in fibroblast-like synoviocytes from patients with rheumatoid arthritis. J Rheumatol. 33:463–471. 2006.PubMed/NCBI

59 

Yuan A, Yang PC, Yu CJ, Chen WJ, Lin FY, Kuo SH and Luh KT: Interleukin-8 messenger ribonucleic acid expression correlates with tumor progression, tumor angiogenesis, patient survival, and timing of relapse in non-small-cell lung cancer. Am J respir Crit Care Med. 162:1957–1963. 2000. View Article : Google Scholar : PubMed/NCBI

60 

Masuya D, Huang C, Liu D, Kameyama K, Hayashi E, Yamauchi A, Kobayashi S, Haba R and Yokomise H: The intratumoral expression of vascular endothelial growth factor and interleukin-8 associated with angiogenesis in nonsmall cell lung carcinoma patients. Cancer. 92:2628–2638. 2001. View Article : Google Scholar : PubMed/NCBI

61 

Wei D, Wang L, He Y, Xiong HQ, Abbruzzese JL and Xie K: Celecoxib inhibits vascular endothelial growth factor expression in and reduces angiogenesis and metastasis of human pancreatic cancer via suppression of Sp1 transcription factor activity. Cancer res. 64:2030–2038. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Lee HC, Park IC, Park MJ, An S, Woo SH, Jin HO, Chung HY, Lee SJ, Gwak HS, Hong YJ, et al: Sulindac and its metabolites inhibit invasion of glioblastoma cells via down-regulation of Akt/PKB and MMP-2. J Cell Biochem. 94:597–610. 2005. View Article : Google Scholar

63 

Ostrowski J, Wocial T, Skurzak H and Bartnik W: Do altering in ornithine decarboxylase activity and gene expression contribute to antiproliferative properties of COX inhibitors? Br J Cancer. 88:1143–1151. 2003. View Article : Google Scholar : PubMed/NCBI

64 

Peluffo GD, Stillitani I, Rodríguez VA, Diament MJ and Klein SM: Reduction of tumor progression and paraneoplastic syndrome development in murine lung adenocarcinoma by nonsteroidal antiinflammatory drugs. Int J Cancer. 110:825–830. 2004. View Article : Google Scholar : PubMed/NCBI

65 

Björkhem-Bergman L, Lindh JD and Bergman P: What is a relevant statin concentration in cell experiments claiming pleiotropic effects? Br J Clin Pharmacol. 72:164–165. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Gauthaman K, Fong CY and Bongso A: Statins, stem cells, and cancer. J Cell Biochem. 106:975–983. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Thelen KM, Rentsch KM, Gutteck U, Heverin M, Olin M, Andersson U, von Eckardstein A, Björkhem I and Lütjohann D: Brain cholesterol synthesis in mice is affected by high dose of simvastatin but not of pravastatin. J Pharmacol Exp Ther. 316:1146–1152. 2006. View Article : Google Scholar

68 

Lai GH, Zhang Z and Sirica AE: Celecoxib acts in a cyclooxygenase-2-independent manner and in synergy with emodin to suppress rat cholangiocarcinoma growth in vitro through a mechanism involving enhanced Akt inactivation and increased activation of caspases-9 and -3. Mol Cancer Ther. 2:265–271. 2003.PubMed/NCBI

69 

Gauthaman K, Manasi N and Bongso A: Statins inhibit the growth of variant human embryonic stem cells and cancer cells in vitro but not normal human embryonic stem cells. Br J Pharmacol. 157:962–973. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 51 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gehrke T, Scherzad A, Hackenberg S, Ickrath P, Schendzielorz P, Hagen R and Kleinsasser N: Additive antitumor effects of celecoxib and simvastatin on head and neck squamous cell carcinoma in vitro. Int J Oncol 51: 931-938, 2017
APA
Gehrke, T., Scherzad, A., Hackenberg, S., Ickrath, P., Schendzielorz, P., Hagen, R., & Kleinsasser, N. (2017). Additive antitumor effects of celecoxib and simvastatin on head and neck squamous cell carcinoma in vitro. International Journal of Oncology, 51, 931-938. https://doi.org/10.3892/ijo.2017.4071
MLA
Gehrke, T., Scherzad, A., Hackenberg, S., Ickrath, P., Schendzielorz, P., Hagen, R., Kleinsasser, N."Additive antitumor effects of celecoxib and simvastatin on head and neck squamous cell carcinoma in vitro". International Journal of Oncology 51.3 (2017): 931-938.
Chicago
Gehrke, T., Scherzad, A., Hackenberg, S., Ickrath, P., Schendzielorz, P., Hagen, R., Kleinsasser, N."Additive antitumor effects of celecoxib and simvastatin on head and neck squamous cell carcinoma in vitro". International Journal of Oncology 51, no. 3 (2017): 931-938. https://doi.org/10.3892/ijo.2017.4071