Skip to main content
Journal of Cancer Prevention logoLink to Journal of Cancer Prevention
. 2013 Jun;18(2):123–134. doi: 10.15430/JCP.2013.18.2.123

Cyclooxygenase-1 and -2: Molecular Targets for Cervical Neoplasia

Hee Seung Kim 1, Taehun Kim 1, Mi-Kyung Kim 1, Dong Hoon Suh 2, Hyun Hoon Chung 1, Yong Sang Song 1,3,4,
PMCID: PMC4189449  PMID: 25337538

Abstract

Cyclooxygenase (COX) is a key enzyme responsible for inflammation, converting arachidonic acid to prostaglandin and thromboxane. COX has at least two isoforms, COX-1 and COX-2. While COX-1 is constitutively expressed in most tissues for maintaining physiologic homeostasis, COX-2 is induced by inflammatory stimuli including cytokines and growth factors. Many studies have shown that COX-2 contributes to cancer development and progression in various types of malignancy including cervical cancer. Human papillomavirus, a necessary cause of cervical cancer, induces COX-2 expression via E5, E6 and E7 oncoproteins, which leads to prostaglandin E2 increase and the loss of E-cadherin, promotes cell proliferation and production of vascular endothelial growth factor. It is strongly suggested that COX-2 is associated with cancer development and progression such as lymph node metastasis. Many studies have suggested that non-selective COX-2 inhibitors such as non-steroidal anti-inflammatory drugs (NSAIDs), and selective COX-2 inhibitors might show anti-cancer activity in COX-2 -dependent and -independent manners. Two phase II trials for patients with locally advanced cervical cancer showed that celecoxib increased toxicities associated with radiotherapy. Contrary to these discouraging results, two phase II clinical trials, using rofecoxib and celecoxib, demonstrated the promising chemopreventive effect for patients with cervical intraepithelial neoplasia 2 or 3. However, these agents cause a rare, but serious, cardiovascular complication in spite of gastrointestinal protection in comparison with NSAIDs. Recent pharmacogenomic studies have showed that the new strategy for overcoming the limitation in clinical application of COX-2 inhibitors shed light on the use of them as a chemopreventive method.

Keywords: Cyclooxygenase, Cyclooxygenase-2 inhibitor, Cervical cancer

INTRODUCTION

Cyclooxygenase (COX) pathway is known to be one of major routes for producing bioactive prostanoids such as prostaglandin (PG) E2, D2, F2α, I2 (prostacyclin) and thromboxane (TX) A2. COX exists as at least two different enzymes in mammalian cells: COX-1 and COX-2, which are located on human chromosomes 9 and 1 respectively.1,2 COX-1 is constitutively expressed in many normal cells, and PGs produced by COX-1 are important for maintaining the integrity of gastric mucosa and allowing normal platelet aggregation and renal function. On the other hand, COX-2 is induced by oncogene, growth factors and cytokines, and COX-2-derived PGs can stimulate cell proliferation, promote angiogenesis, increase invasiveness and adhesion to the extracellular matrix and inhibit immune surveillance and apoptosis.35 Furthermore, COX-2-derived PGs have been shown to contribute to cancer development, progression and metastasis.6 Therefore, the inhibition of COX-2 has been anticipated to prevent the development and progression of cancer and to promote the response to cytotoxic agents as well as ionizing radiation.7

Although non-steroidal anti-inflammatory drugs (NSAIDs), which non-specifically inhibit both COX-1 and COX-2, induce adverse effects on gastrointestinal (GI) tract, selective COX-2 inhibitors such as rofecoxib and celecoxib reduce the adverse effects of NSAIDs on GI tract with relief of chronic pain.8,9 However, selective COX-2 inhibitors are known to be associated with increased cardiovascular adverse effects.10 Since many preclinical and clinical studies have shown that COX-2-derived PGs are associated with cervical neoplasia and COX-2 inhibitors have anti-cancer effect, we will show the role of COX-2 and the efficacy of COX-2 inhibitors in cervical neoplasia, and will suggest the new strategy for overcoming the limitation in clinical application of COX-2 inhibitors through this review.

COX-2, INFLAMMATION AND CARCINOGENESIS

Chronic inflammation mediated by COX-2 is associated with carcinogenesis and cancer progression. It is caused by various factors including bacterial infections and chemical irritants. The longer the inflammation persists, the higher is the risk of associated carcinogenesis. Moreover, neoplasia could be caused by inflammatory mediators inducing preneoplastic mutation, stimulation of angiogenesis and resistance to apoptosis, and these inflammatory mediators may activate signaling molecules involved in inflammation and carcinogenesis such as COX-2 and nuclear factor-kappa B (NF-kB).11

Carcinogenesis by COX-2 has been explored in terms of the inhibition of apoptosis, promotion of angiogenesis, invasiveness and immunosuppression in various types of malignancy.7 Especially, PG E2, an end product of COX-2, may increase the activity of mitogen-activated protein kinase (MAPK),12 affect ras-controlled signal transduction pathways,13 and suppress the activity of caspase-3, a key enzyme in apoptotic process.14 Besides, COX-2-derived PGs may increase the production of vascular endothelial growth factor (VEGF) and promote neovascularization in cancer.15,16

COX-2 overexpression may lead to the invasiveness of cancer to basement membrane, stroma, penetration to blood vessels and metastasis, which are mediated by matrix metalloproteinases (MMPs) such as MMP-1, -2 and -9.6,17 Additionally, carcinogenesis is related with immunosuppression because colony-stimulating factors secreted by cancer cells activate monocytes and macrophages resulting in the synthesis of PG E2 by COX-2. PG E2 shows the immunosuppressive effect by inhibiting the production of lymphokines and tumor necrosis factors, proliferation of T- and B-cells and cytotoxic activity of natural killer cells.18,19

INDUCTION OF COX-2 GENE BY HUMAN PAPILLOMAVIRUS ITSELF

Human papillomavirus (HPV) is the most prevalent sexually infectious agent and causes cervical cancer. Especially, HPV 16 E6 and E7 oncoproteins stimulate to produce amphiregulin, which induces the transcription of COX-2 gene by activating MAPK cascade (Fig. 1A).5 HPV 16 E5 oncoprotein also induces the transcription of COX-2 gene in a ligand-dependent and -independent activation of epidermal growth factor receptor (EGFR) and MAPK cascade,2022 and causes the increased expression of VEGF by activating MEK/ERK 1/2 and PI3K/Akt, which are associated with cervical carcinogenesis (Fig. 1B).20,23,24 Moreover, chronic infection of HPV in cervical epithelium increases PG E2 by COX-2, which leads to the loss of E-cadherin, increased cell proliferation and production of VEGF.2527

Fig. 1.

Fig. 1.

Schematic of pathway where human papillomavirus (HPV)16 E5, E6 and E7 oncoproteins regulate cyclooxygenase-2 (COX-2) expression associated with the cervical carcinogenesis. (A) HPV16 E6 and E7 oncoproteins stimulate production of amphiregulin and thereby activate EGFR → Ras → MAPK signaling. This results, in turn, in the phosphorylation of c-Jun, leading to transduction β-like protein 1-related protein (TBLR1)-dependent degradation of the nuclear receptor corepressor (NCoR)/histone deacetylase 3 (HDAC3) complex and recruitment of the coactivator cyclic AMP-responsive element binding protein-binding protein (CBP)/p300 and phosphorylated c-Jun/c-Fos heterodimer to the COX-2 promoter. This corepressor/coactivator exchange triggered by HPV onco-proteins leads to enhanced COX-2 transcription5; (B) HPV 16 E5 oncoprotein also causes the increase of phosphorylated EGFR, and thereby increases the transcription of COX-2 gene and secretion of VEGF, which enhances cervical carcinogenesis.20

COX-2 EXPRESSION IN CERVICAL CARCINOGENESIS

COX-2 is highly expressed in various types of cervical neoplasm such as cervical intraepithelial neoplasia (CIN) (7.4%), adenocarcinoma (13%) and squamous cell carcinoma (28.8%) of cervix, suggesting that COX-2 expression can be associated clinically with cervical cancer development and progression.2830 Besides, COX-2 gene has been shown to be involved in early cervical carcinogenesis and accelerate tumor progression by increasing VEGF.25

COX-2 has been also shown to be expressed in dysplastic epithelium (7.4%) but not in stromal cells of CIN (0%).31 This fact is contrary to previous studies of COX-2 overexpression in colon cancer where the increased COX-2 expression in stromal cells was related with carcinogenesis, suggesting that PGs derived from COX-2 in stromal cells would be secreted and bind to receptors on adjacent epithelial cells, then might promote carcinogenesis with the “landscaping effect”.32 Unlike colon cancer, the landscaping effect of stromal cells seems to have no role in cervical carcinogenesis because it may be influenced by HPV itself.

Interestingly, COX-2 overexpression may be also associated with old age and menopause in CIN.31 Although the reason is unclear, the lack of progesterone for menopausal women could explain this fact because progesterone has been shown to suppress COX-2 expression in some cells.33

COX-2 CONTRIBUTING TO PROGRESSION IN CERVICAL NEOPLASIA

COX-2 overexpression is associated with lymph node metastasis in cervical cancer.34,35 Although COX-2 over-expression was not an independent prognostic factor for survival,36,37 it may enhance metastatic potentials of tumors by inducing genes which promote lymphangiogenesis and increase metastatic properties of cervical cancer.38

Moreover, COX-2 overexpression is related with NF-kB activation, which is localized to the cytoplasm in resting cells and binds to the DNA recognition sites in the regulatory regions of target genes after it migrates into the nucleus on various stimuli.34,35,39 Many studies have been focused on NF-kB as a molecular target for chemoprevention, which plays a crucial role in the regulation of inflammatory and immunes responses and in carcinogenesis. Stimuli regulated by NF-kB during inflammation can be redirected as tumor growth signals. NF-kB has been found constitutively activated in many human cancer samples, supporting an important role of NF-kB in cancer development.40 Moreover, COX-2 is inducible via the activation of NF-kB by many factors such as cytokines and growth factors.41

EFFICACY OF COX-2 INHIBITORS AGAINST CERVICAL NEOPLASIA IN PRECLINICAL STUDIES

NSAIDs and selective COX-2 inhibitors such as celecoxib have been commonly used as analgesics, anti-inflammatory drugs. After several studies reported their apoptotic effect in various types of cancer cells,4244 the efficacy of COX-2 inhibitors has been evaluated for the prevention or treatment of cervical neoplasia. In detail, anti-cancer activity of COX-2 inhibitors is mediated in part through the inhibition of the COX-2 activity.4547 However, anti-cancer activity exerted by COX-2 inhibitors is independent of their COX-2 inhibitory properties because the growth of hematopoietic and epithelial tumor cells without COX-2 expression has been reported to be suppressed by COX-2 inhibitors.48,49 Besides, in cervical cancer cells, celecoxib induces apoptosis independent of COX-2 inhibition through two major pathways: death receptor pathway followed by the activation of caspase-8, which then activates the downstream effector caspases such as caspase-3, -6 and -7, triggering cell death; mitochondrial pathway by the activation of caspase-9, which leads to the loss of mitochondrial membrane potential.42,50

Celecoxib-induced apoptosis is mediated by a Fas/Fas- associated protein with death domain (FADD)-dependent mechanism in Fasligand (FasL)-independent manner, and involved in the activation of NF-kB.42 Growth arrest and DNA damage inducible gene (GADD153), a transcription factor involved in apoptosis, also plays a key role in celecoxib-induced apoptosis in cervical cancer cells by regulating the expression of proapoptotic proteins such as Bak.51

NSAIDs seem to have comparable efficacy to celecoxib. In a study on the association among COX-1, COX-2 and VEGF expression in cervical cancer, VEGF expression was strongly correlated with COX-1 expression, and COX-2 expression was associated with lymph node metastasis,28 suggesting that NSAIDs may be efficient to treat cervical cancer.52,53 Furthermore, NSAIDs including aspirin, sulindac and indomethacin have been reported to decrease cell proliferation and colony formation in a time and dose- dependent manner in cervical cancer cells, and increase apoptosis and radiotherapeutic efficacy by pretreatment of cervical cancer cells through bcl-2 repression and caspase-3 induction.54

On the other hand, COX expression in cervical cancer may be associated with the effect of radiotherapy.55,56 Especially, COX-1 expression decreases significantly radiosensitivity in cervical cancer cell lines in spite of no association between COX-2 expression and radio-resistance. These data suggest that COX-1 might imply more importance than COX-2 regarding the innate radiosensitivity of cervical cancer, and that NSAIDs, non-selective COX-2 inhibitors, might increase the radiotherapeutic effectiveness if cervical tumor cells have not yet lost their ability to express COX-1.56

CLINICAL APPLICATION OF COX-2 INHIBITORS IN CERVICAL CANCER

1. COX-2 inhibitors for the prevention of cervical cancer

The efficacy of COX-2 inhibitors has a definite advantage to treat CIN because cervical conization may be avoided, reducing obstetrical complications including preterm delivery, and preterm premature rupture of membrane. In a prospective, randomized, placebo-controlled, double-blind study with rofecoxib 25 mg daily for 6 months for the treatment of 16 patients with CIN 2 and CIN 3, regression rate was higher in patients treated with rofecoxib than those treated with placebo (25% vs. 12.5%) without no severe side effects although the results were statistically not significant due to early withdrawal of refecoxib from the market by increased cardiovascular adverse effect.57 Also, clinical response rate and complete pathologic response were higher for patients treated with celecoxib than in those treated with placebo (75% vs. 31%; 33% vs. 15%, respectively) in a randomized, double-blind, placebo-controlled phase II trial of celecoxib 200 mg twice a day or placebo for the treatment of 25 patients with CIN 2 or CIN 3.58

2. COX-2 inhibitors for the treatment of cervical cancer

The efficacy of selective COX-2 inhibitors has been mainly studied for patients with locally advanced cervical cancer receiving radiotherapy. However, the results were disappointing because COX-2 inhibitors showed no clinical benefit and higher toxicity by the addition to chemoradiation. In a phase I–II trial of celecoxib 400 mg twice per day for 2 weeks before and during chemoradiation using cisplatin, 31 patients with locally advanced cervical cancer were enrolled. Higher incidence of grade 3 or 4 acute toxicity (35.5%) was seen with no difference in 81% of response rate, compared with previous studies about the chemoradiation alone. Besides, there was an increase in late complication such as fistula (9.7%). Thus, celecoxib in combination with chemoradiation was associated with acceptable acute toxicity, but higher late complication.59

Furthermore, the Radiation Therapy Oncology Group (RTOG) 0128 trial was performed as a phase II study to evaluate the efficacy and toxicity of celecoxib and chemoradiation for patients with locally advanced cervical cancer. In this study, 83 patients were treated with chemoradiation using cisplatin and 5-fluorouracil with the addition of celecoxib at the dose of 400 mg twice daily for 1 year. However, grade 3 or 4 toxicities were developed in 47% and late toxicities such as GI and genitourinary side effects were observed in 13% of all patients, which were higher than expected rates of complication. These data suggest that the toxicities associated with celecoxib may limit the use of this drug.60

On the other hand, a randomized clinical trial showed that the treatment of oxyphenbutazone, a non-selective COX-2 inhibitor, at the dose of 300 mg daily improved 5- and 10-year survival rates, compared to placebo in patients undergoing radiotherapy only for cervical cancer (5-year survival rate, 70 vs. 55%; 10-year survival rate, 62 vs. 44%). Taken together, there are two possible explanations for these discrepant results. First, the improvement of survival rates might be due to slowing of tumor spread and improvement of cell repair after radiotherapy by the inhibition of PGs. Second, the inhibition of both COX-1 and -2 might be important to treat cervical cancer.52

Thus, many clinical trials are required to evaluate the role of COX-2 inhibitors in the management of cervical cancer. Table 1 depicts clinical studies about the efficacy of COX-2 inhibitors in cervical neoplasia. The clinical trials of selective COX-2 inhibitors, especially celecoxib, are being on the progress for the treatment of cervical neoplasia combined with chemotherapy or radiotherapy or alone.

Table 1.

Clinical trials of cyclooxygenase-2 (COX-2) inhibitors for the treatment of cervical neoplasia

Authors or protocol ID Sample size Interventions Targeted disease Response rate
Weppelmann and Monkemeier52 76 vs. 84 (control) Oxyphenbutazone Cervical cancer 5-year survival rate : 70% vs. 55%
10-year survival rate : 62% vs. 44%
Hefler et al.57 8 vs. 8 (control) Rofecoxib CIN* 2–3 25% vs.12.5%
Farley et al.58 12 vs. 13 (control) Celecoxib CIN* 2–3 75% vs. 31%
Herrera et al.59 31 Celecoxib Cervical cancer 81%
Gaffney et al.60 84 Celecoxib Cervical cancer Toxicity: 48%
NCT00081263 (GOG-0207) 100 Celecoxib CIN* 2–3 -
NCT00152828 45 Celecoxib Cervical cancer -
NCT00072540 (SWOG-S0212) 100 Celecoxib CIN* 2–3 -
*

Cervical intraepithelial neoplasia;

Active clinical trials (available at http://clinicaltrials.gov).

ADVERSE EFFECTS OF COX-2 INHIBITORS

After selective COX-2 inhibitors were introduced as alternative analgesics to NSAIDs due to fewer GI side effects, the approval of rofecoxib (Vioxx®) and celecoxib (Celebrex®) by the Food and Drug Administration in the United States came in 1999 with their market release. Moreover, selective COX-2 inhibitors had been investigated for chemoprevention because some studies have demonstrated that inhibiting COX-2 could prevent the formation of premalignant colorectal adenomas.6165 However, rofecoxib was withdrawn from the market on September 2004 because of the serious adverse event found in Adenomatous Polyp Prevention on Vioxx® (APPROVe) trial, demonstrating that the group assigned to rofecoxib had a fourfold increased risk of serious thromboembolic events including acute myocardial infarction and cerebrovascular accident compared with the placebo group.66 Furthermore, rofecoxib has been shown to increase cardiovascular adverse effects by meta-analysis when compared to placebo or NSAIDs (Table 2).

Table 2.

Cardiovascular adverse effect of selective cyclooxygenase-2 (COX-2) inhibitors by meta-analysis

Adverse effects Meta-analysis Comparison
Relative risk* with 95% CI
Control Intervention
Serious cardiovascular events Kearney et al.97 Placebo Selective COX-2 inhibitors 1.42 (1.13–1.78)
Naproxen Selective COX-2 inhibitors 1.57 (1.21–2.03)
Non-naproxen Selective COX-2 inhibitors 0.88 (0.69–1.12)
Mukherjee et al.98 Naproxen Rofecoxib 1.89 (1.03–3.45)
Jüni et al.99 Control§ Rofecoxib 1.55 (1.05–2.29)
Garner et al.100 Non-naproxen
- Diclofenac Rofecoxib 0.70 (0.25–1.93)
- Nabumetone Rofecoxib 2.90 (0.12–71.01)
- Arthrotec Rofecoxib 1.39 (0.63–3.08)
Cardiovascular mortality Kearney et al.97 Placebo Selective COX-2 inhibitors 1.49 (0.97–2.29)
Naproxen Selective COX-2 inhibitors 1.47 (0.90–2.40)
Jüni et al.99 Control§ Rofecoxib 0.79 (0.29–2.19)
Myocardial infarction Kearney et al.97 Naproxen Selective COX-2 inhibitors 2.04 (1.41–2.96)
Non-naproxen Selective COX-2 inhibitors 1.20 (0.85–1.68)
Jüni et al.99 Placebo Rofecoxib 1.04 (0.34–3.12)
Naproxen Rofecoxib 2.93 (1.36–6.33)
Non-naproxen Rofecoxib 1.55 (0.55–4.36)
Garner et al.100 Placebo Rofecoxib 1.48 (0.06–36.06)
Naproxen Rofecoxib 4.98 (0.58–42.57)
Non-naproxen
- Diclofenac Rofecoxib 0.52 (0.05–5.72)
Stroke** Kearney et al.97 Placebo Selective COX-2 inhibitors 1.02 (0.71–1.47)
Naproxen Selective COX-2 inhibitors 1.10 (0.73–1.65)
Non-naproxen Selective COX-2 inhibitors 0.62 (0.41–0.95)
Jüni et al.99 Control§ Rofecoxib 1.02 (0.54–1.93)
Garner et al.100 Naproxen Rofecoxib 0.08 (0.00–1.36)
*

A ratio of the probability of the event occurring in the interventiongroup versus the control group;

non-fatal myocardial infarction, non-fatal stroke or cardiovascular death;

including rofecoxib, celecoxib, etoricoxib, lumiracoxib and valdecoxib;

§

placebo and NSAIDs;

Death due to cardiovascular events;

fatal or non-fatal myocardial infarction;

**

fatal or non-fatal thrombotic or hemorrhagic stroke.

Benefit and risk by COX-2 inhibitors are summarized in Fig. 2. In spite of markedly less GI damage than NSAIDs, selective COX-2 inhibitors are doomed to increase cardiovascular adverse effects because selective COX-2 inhibition may reduce the production of prostacyclin, which normally inhibits platelet aggregation and vasodilation, while still allowing COX-1 mediated synthesis of TX A2 to induce platelet aggregation and vasoconstriction.67 After withdrawal of rofecoxib, the safety of celecoxib has also been investigated for cardiovascular adverse effects. Celecoxib has been shown to be safer than rofecoxib in most studies. The first reason is that the degree of COX-2 selectivity of celecoxib is a fifth of that of rofecoxib. Actually, the degree of COX-2 selectivity is known to correlate with cardiovascular and renal risks.68,69 The second reason is that a reactive metabolite of rofecoxib, a maleic anhydride derivative which contributes to atherothrombosis, cannot be derived from other COX-2 inhibitors including celecoxib, valdecoxib and lumaricoxib.70 Furthermore, the Celecoxib Long-term Arthritis Safety Study (CLASS) demonstrated no significant difference in cardiovascular event between celecoxib 800 mg/day and NSAIDs, suggesting the safety of celecoxib.71

Fig. 2.

Fig. 2.

Role of cyclooxygenase (COX) in human gastrointestinal, cardiovascular and renal functions. COX-1-derived thromboxane A2 decreases gastric acid secretion in gastrointestinal tract and renal vascular resistance in kidney, whereas it increases mucus production in gastrointestinal tract, vasoconstriction, platelet aggregation and smooth muscle proliferation in blood vessel, and vasodilation in kidney. Moreover, COX-2-derived prostaglandins E2 and I2 decrease platelet aggregation and smooth muscle proliferation in blood vessel while they increase vasodilation in gastrointestinal tract and blood vessel, and diuresis and natriuresis in kidney. On the other hand, selective COX-2 inhibitors increase thromboembolic risk, and decrease gastrointestinal side effects and renal function.

Nonetheless, the Adenoma Prevention with Celecoxib (APC) and Prevention of Spontaneous Adenomatous Polyps (PreSAP) trials comparing celecoxib with placebo for the reduction in recurrent colorectal polyps were stopped early because of significantly higher numbers of cardiovascular adverse effects in celecoxib-treated group.72,73 Thus, the safety of celecoxib is still on debate, and further trials designed to assess the incidence of cardiovascular adverse effects by celecoxib are needed.

NEW STRATEGY FOR OVERCOMING THE LIMITATION FOR USING COX-2 INHIBITORS

1. Natural products for the chemoprevention of cervical neoplasia

Many natural products are being investigated to inhibit COX-2 overexpression and NF-kB activation as molecular targets for chemoprevention of cervical neoplasia. First, curcumin is a yellow pigment of turmeric, a natural product with diverse biological activities. It has been shown to possess anti-inflammatory, anti-oxidant and anti-tumor properties. Much of its beneficial effect is found to be due to its inhibition of NF-kB and subsequent inhibition of proinflammatory pathways.74 Besides, curcumin synergistically augments the growth inhibitory effect of celecoxib by down-regulating COX-2 mRNA expression and inhibition of the catalytic activity of 5-lipoxygenase producing leukotrienes associated with carcinogenic process.75 Phase I trials on curcumin showed that it is safe to human up to 12,000 mg/day when taken orally and caused histological improvement of precancerous lesions including CIN.7678 Moreover, curcumin has been shown to confer the radiosensitizing effect in cervical cancer cells.79

Second, indole-3-carbinol (I3C) is derived from cruciferous vegetables such as broccoli and cabbage. I3C and its metabolite, 3,3′-diindoylmethane (DIM) target multiple aspects of cancer cell-cycle regulation and survival including NF-kB signaling, caspases activation and cyclin-dependent kinase activity.80 I3C and its metabolite have been shown to prevent cervical cancer and have the efficacy in the treatment of cervical dysplasia in the mouse model.81 A small randomized controlled clinical trial in patients with CIN 2 or 3 indicated the efficacy of I3C for the regression of CIN.82 In addition, some studies on HPV persistence or cervical neoplasia showed a possible protective effect of fruits, vegetables, vitamins C and E, α- and β-carotenes, lycopene, luterin/zeaxanthin and cryotoxanthin.83

2. New methods using COX-2 inhibitors

Since the safety of selective COX-2 inhibitors is controversial, patients treated with selective COX-2 inhibitors should be monitored regularly in terms of blood pressure, edema and cardiac status because regular interruptions of treatment can contribute a great deal to the safe use of selective COX-2 inhibitors.68 In addition, new methods are being investigated for overcoming the limitation of selective COX-2 inhibitors as follows.

The first is the combination of COX-2 inhibitors with other drugs. The prescription of a combined therapy of NSAIDs and proton pump inhibitors (PPIs) has been shown to have comparable ulcerous bleeding to COX-2 inhibitors (6.4% vs. 4.9%).84 However, it should be considered that PPIs may be associated with adverse effects independent of concomitant NSAID use, including pneumonia, bacterial diarrhea and hip fracture.8587 Moreover, it can be considered that selective COX-2 inhibitors are combined with low-dose aspirin for cardioprotection. However, the CLASS trial demonstrated that a fourfold increase in the incidence of GI bleeding occurred in a subgroup of patients taking celecoxib in combination with aspirin, suggesting that the combination should not be used in patients with high-risk GI bleeding.71 Furthermore, curcumin can be combined with selective COX-2 inhibitors because it induces cardioprotective effect by scavenging oxygen-free radical.88 However, large and well-controlled clinical trials are required to determine the role of selective COX-2 inhibitors and curcumin to prevent and treat cancer.

The second method is the structural modification of NSAIDs. Nitric oxide (NO)-donating NSAIDs have been claimed to exert a broader range of anti-inflammatory action while reducing markedly GI and cardiovascular toxicity.8991 However, these claims are poorly substantiated by clinical studies to date.

The third method is the modification of schedule for the use of selective COX-2 inhibitors. In some meta-analyses, celecoxib showed dose-dependent cardiovascular effect although rofecoxib was associated with cardiovascular adverse effect at all doses (at doses of 25 mg or less, or greater than 25 mg once daily), suggesting that celecoxib doses of up to 200 mg once daily was not related with increased cardiovascular adverse effect in spite of the need of clinical trials for evaluating dose-dependent toxicity of celecoxib.92,93 Since the combination of chemoradiation with celecoxib increased late toxicities compare to chemoradiation alone in patients with locally advanced cervical cancer,59,60 various schedules for the administration of celecoxib are being investigated in clinical trials for gynecologic cancers. For example, in a phase II study of weekly paclitaxel and celecoxib for the treatment of recurrent or persistent platinum-resistant epithelial ovarian or primary peritoneal cancer, patients receive paclitaxel on days 1, 8, and 15 and celecoxib twice daily on days 2–6, 9–13 and 16–27 with the repeat of courses every 28 days in the absence of disease progression or unacceptable toxicity.94

CONCLUSION

After withdrawal of rofecoxib from market, other selective COX-2 inhibitors including celecoxib have been focused on many clinical trials to prevent and treat various types of malignancy including cervical cancer. Since the safety of other selective COX-2 inhibitors remains controversial, it is important to select patients with low cardiovascular risk from selective COX-2 inhibitors, and to follow up them regularly for the prevention and early detection of GI, renal and cardiovascular adverse effects. For example, selective COX-2 inhibitors seem to be useful for the treatment of CIN which mainly develops in young women with HPV infection because most of them have relatively lower cardiovascular risk than old women.95 Besides, selective COX-2 inhibitors have the advantage that these agents can lessen the risk of preterm delivery by cervical conization for the treatment of CIN with lesser GI toxicity compared to non-selective COX-2 inhibitors.96

On the other hand, the role of COX-1 should be reevaluated for the prevention and treatment of cervical neoplasia because some preclinical and clinical studies have shown that the inhibition of COX-1 might increase the radiotherapeutic efficacy in cervical cancer.52,55,56 Furthermore, new strategies using natural products or COX-2 inhibitors should be proven through preclinical and clinical studies for overcoming the limitation of COX-2 inhibitors.

Acknowledgments

This research was supported by grant (No. 04-2012- 0890; 03-2012-0170) from the Seoul National University Hospital research fund, the World Class University program through the Korea Science and Engineering Foundation (No. R31-2008-000-10056-0), and the Priority Research Centers Program through the National Research Foundation of Korea (No. 2009-0093820) funded by the Ministry of Education, Science and Technology.

REFERENCES

  • 1.Yokoyama C, Tanabe T. Cloning of human gene encoding prostaglandin endoperoxide synthase and primary structure of the enzyme. Biochem Biophys Res Commun. 1989;165:888–94. doi: 10.1016/s0006-291x(89)80049-x. [DOI] [PubMed] [Google Scholar]
  • 2.Tay A, Squire JA, Goldberg H, Skorecki K. Assignment of the human prostaglandin-endoperoxide synthase 2 (PTGS2) gene to 1q25 by fluorescence in situ hybridization. Genomics. 1994;23:718–9. doi: 10.1006/geno.1994.1569. [DOI] [PubMed] [Google Scholar]
  • 3.Sheng H, Shao J, Washington MK, DuBois RN. Prostaglandin E2 increases growth and motility of colorectal carcinoma cells. J Biol Chem. 2001;276:18075–81. doi: 10.1074/jbc.M009689200. [DOI] [PubMed] [Google Scholar]
  • 4.Dohadwala M, Luo J, Zhu L, Lin Y, Dougherty GJ, Sharma S, et al. Non-small cell lung cancer cyclooxygenase-2-dependent invasion is mediated by CD44. J Biol Chem. 2001;276:20809–12. doi: 10.1074/jbc.C100140200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Subbaramaiah K, Dannenberg AJ. Cyclooxygenase-2 transcription is regulated by human papillomavirus 16 E6 and E7 oncoproteins: evidence of a corepressor/coactivator exchange. Cancer Res. 2007;67:3976–85. doi: 10.1158/0008-5472.CAN-06-4273. [DOI] [PubMed] [Google Scholar]
  • 6.Tsujii M, Kawano S, DuBois RN. Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci U S A. 1997;94:3336–40. doi: 10.1073/pnas.94.7.3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dempke W, Rie C, Grothey A, Schmoll HJ. Cyclooxygenase-2: a novel target for cancer chemotherapy? J Cancer Res Clin Oncol. 2001;127:411–7. doi: 10.1007/s004320000225. [DOI] [PubMed] [Google Scholar]
  • 8.Schmassmann A, Peskar BM, Stettler C, Netzer P, Stroff T, Flogerzi B, et al. Effects of inhibition of prostaglandin endoperoxide synthase-2 in chronic gastro-intestinal ulcer models in rats. Br J Pharmacol. 1998;123:795–804. doi: 10.1038/sj.bjp.0701672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Vane JR, Bakhle YS, Botting RM. Cyclooxygenases 1 and 2. Annu Rev Pharmacol Toxicol. 1998;38:97–120. doi: 10.1146/annurev.pharmtox.38.1.97. [DOI] [PubMed] [Google Scholar]
  • 10.Strand V. Are COX-2 inhibitors preferable to non-selective non-steroidal anti-inflammatory drugs in patients with risk of cardiovascular events taking low-dose aspirin? Lancet. 2007;370:2138–51. doi: 10.1016/S0140-6736(07)61909-6. [DOI] [PubMed] [Google Scholar]
  • 11.Shacter E, Weitzman SA. Chronic inflammation and cancer. Oncology (Williston Park) 2002;16:217–26. [PubMed] [Google Scholar]
  • 12.Sheng H, Shao J, Morrow JD, Beauchamp RD, DuBois RN. Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res. 1998;58:362–6. [PubMed] [Google Scholar]
  • 13.Gilhooly EM, Rose DP. The association between a mutated ras gene and cyclooxygenase-2 expression in human breast cancer cell lines. Int J Oncol. 1999;15:267–70. [PubMed] [Google Scholar]
  • 14.Villa P, Kaufmann SH, Earnshaw WC. Caspases and caspase inhibitors. Trends Biochem Sci. 1997;22:388–93. doi: 10.1016/s0968-0004(97)01107-9. [DOI] [PubMed] [Google Scholar]
  • 15.Marmé D. Tumor angiogenesis: the pivotal role of vascular endothelial growth factor. World J Urol. 1996;14:166–74. doi: 10.1007/BF00186896. [DOI] [PubMed] [Google Scholar]
  • 16.Chiarugi V, Magnelli L, Gallo O. Cox-2, iNOS and p53 as play-makers of tumor angiogenesis (review) Int J Mol Med. 1998;2:715–9. doi: 10.3892/ijmm.2.6.715. [DOI] [PubMed] [Google Scholar]
  • 17.Attiga FA, Fernandez PM, Weeraratna AT, Manyak MJ, Patierno SR, Patierno SR. Inhibitors of prostaglandin synthesis inhibit human prostate tumor cell invasiveness and reduce the release of matrix metalloproteinases. Cancer Res. 2000;60:4629–37. [PubMed] [Google Scholar]
  • 18.Balch CM, Dougherty PA, Cloud GA, Tilden AB. Prostaglandin E2-mediated suppression of cellular immunity in colon cancer patients. Surgery. 1984;95:71–7. [PubMed] [Google Scholar]
  • 19.Kambayashi T, Alexander HR, Fong M, Strassmann G. Potential involvement of IL-10 in suppressing tumor-associated macrophages. Colon-26-derived prostaglandin E2 inhibits TNF-alpha release via a mechanism involving IL-10. J Immunol. 1995;154:3383–90. [PubMed] [Google Scholar]
  • 20.Kim SH, Juhnn YS, Kang S, Park SW, Sung MW, Bang YJ, et al. Human papillomavirus 16 E5 up-regulates the expression of vascular endothelial growth factor through the activation of epidermal growth factor receptor, MEK/ERK1,2 and PI3K/Akt. Cell Mol Life Sci. 2006;63:930–8. doi: 10.1007/s00018-005-5561-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fehrmann F, Laimins LA. Human papillomaviruses: targeting differentiating epithelial cells for malignant transformation. Oncogene. 2003;22:5201–7. doi: 10.1038/sj.onc.1206554. [DOI] [PubMed] [Google Scholar]
  • 22.Straight SW, Hinkle PM, Jewers RJ, McCance DJ. The E5 oncoprotein of human papillomavirus type 16 transforms fibroblasts and effects the downregulation of the epidermal growth factor receptor in keratinocytes. J Virol. 1993;67:4521–32. doi: 10.1128/jvi.67.8.4521-4532.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kodama J, Seki N, Tokumo K, Hongo A, Miyagi Y, Yoshinouchi M, et al. Vascular endothelial growth factor is implicated in early invasion in cervical cancer. Eur J Cancer. 1999;35:485–9. doi: 10.1016/s0959-8049(98)00410-9. [DOI] [PubMed] [Google Scholar]
  • 24.Cheng WF, Chen CA, Lee CN, Wei LH, Hsieh FJ, Hsieh CY. Vascular endothelial growth factor and prognosis of cervical carcinoma. Obstet Gynecol. 2000;96:721–6. doi: 10.1016/s0029-7844(00)01025-5. [DOI] [PubMed] [Google Scholar]
  • 25.Dai Y, Zhang X, Peng Y, Wang Z. The expression of cyclooxygenase-2, VEGF and PGs in CIN and cervical carcinoma. Gynecol Oncol. 2005;97:96–103. doi: 10.1016/j.ygyno.2004.12.020. [DOI] [PubMed] [Google Scholar]
  • 26.Young JL, Jazaeri AA, Darus CJ, Modesitt SC. Cyclooxygenase-2 in cervical neoplasia: a review. Gynecol Oncol. 2008;109:140–5. doi: 10.1016/j.ygyno.2008.01.008. [DOI] [PubMed] [Google Scholar]
  • 27.Kim YM, Park JY, Lee KM, Kong TW, Yoo SC, Kim WY, et al. Does pretreatment HPV viral load correlate with prognosis in patients with early stage cervical carcinoma? J Gynecol Oncol. 2008;19:113–6. doi: 10.3802/jgo.2008.19.2.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kim MH, Seo SS, Song YS, Kang DH, Park IA, Kang SB, et al. Expression of cyclooxygenase-1 and -2 associated with expression of VEGF in primary cervical cancer and at metastatic lymph nodes. Gynecol Oncol. 2003;90:83–90. doi: 10.1016/s0090-8258(03)00224-5. [DOI] [PubMed] [Google Scholar]
  • 29.Kang S, Kim MH, Park IA, Kim JW, Park NH, Kang D, et al. Elevation of cyclooxygenase-2 is related to lymph node metastasis in adenocarcinoma of uterine cervix. Cancer Lett. 2006;237:305–11. doi: 10.1016/j.canlet.2005.06.027. [DOI] [PubMed] [Google Scholar]
  • 30.Kulkarni S, Rader JS, Zhang F, Liapis H, Koki AT, Masferrer JL, et al. Cyclooxygenase-2 is overexpressed in human cervical cancer. Clin Cancer Res. 2001;7:429–34. [PubMed] [Google Scholar]
  • 31.Kim K, Jeon YT, Park IA, Kim JW, Park NH, Kang SB, et al. Cyclooxygenase-2 expression in cervical intraepithelial neoplasia. Ann N Y Acad Sci. 2009;1171:111–5. doi: 10.1111/j.1749-6632.2009.04724.x. [DOI] [PubMed] [Google Scholar]
  • 32.Prescott SM. Is cyclooxygenase-2 the alpha and the omega in cancer? J Clin Invest. 2000;105:1511–3. doi: 10.1172/JCI10241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hardy DB, Janowski BA, Corey DR, Mendelson CR. Progesterone receptor plays a major antiinflammatory role in human myometrial cells by antagonism of nuclear factor-kappaB activation of cyclooxygenase 2 expression. Mol Endocrinol. 2006;20:2724–33. doi: 10.1210/me.2006-0112. [DOI] [PubMed] [Google Scholar]
  • 34.Yamamoto K, Arakawa T, Taketani Y, Takahashi Y, Hayashi Y, Ueda N, et al. TNF alpha-dependent induction of cyclooxygenase-2 mediated by NF kappa B and NF-IL6. Adv Exp Med Biol. 1997;407:185–9. [PubMed] [Google Scholar]
  • 35.Surh YJ, Chun KS, Cha HH, Han SS, Keum YS, Park KK, et al. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat Res. 2001;480–481:243–68. doi: 10.1016/s0027-5107(01)00183-x. [DOI] [PubMed] [Google Scholar]
  • 36.Kyzas PA, Stefanou D, Agnantis NJ. COX-2 expression correlates with VEGF-C and lymph node metastases in patients with head and neck squamous cell carcinoma. Mod Pathol. 2005;18:153–60. doi: 10.1038/modpathol.3800244. [DOI] [PubMed] [Google Scholar]
  • 37.Su JL, Shih JY, Yen ML, Jeng YM, Chang CC, Hsieh CY, et al. Cyclooxygenase-2 induces EP1- and HER-2/Neu-dependent vascular endothelial growth factor-C up-regulation: a novel mechanism of lymphangiogenesis in lung adenocarcinoma. Cancer Res. 2004;64:554–64. doi: 10.1158/0008-5472.can-03-1301. [DOI] [PubMed] [Google Scholar]
  • 38.Yao M, Lam EC, Kelly CR, Zhou W, Wolfe MM. Cyclooxygenase-2 selective inhibition with NS-398 suppresses proliferation and invasiveness and delays liver metastasis in colorectal cancer. Br J Cancer. 2004;90:712–9. doi: 10.1038/sj.bjc.6601489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lim JW, Kim H, Kim KH. Nuclear factor-kappaB regulates cyclooxygenase-2 expression and cell proliferation in human gastric cancer cells. Lab Invest. 2001;81:349–60. doi: 10.1038/labinvest.3780243. [DOI] [PubMed] [Google Scholar]
  • 40.Barnes OJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med. 1997;336:1066–71. doi: 10.1056/NEJM199704103361506. [DOI] [PubMed] [Google Scholar]
  • 41.Ohshima H, Tazawa H, Sylla BS, Sawa T. Prevention of human cancer by modulation of chronic inflammatory processes. Mutat Res. 2005;591:110–22. doi: 10.1016/j.mrfmmm.2005.03.030. [DOI] [PubMed] [Google Scholar]
  • 42.Kim SH, Song SH, Kim SG, Chun KS, Lim SY, Na HK, et al. Celecoxib induces apoptosis in cervical cancer cells independent of cyclooxygenase using NF-kappaB as a possible target. J Cancer Res Clin Oncol. 2004;130:551–60. doi: 10.1007/s00432-004-0567-6. [DOI] [PubMed] [Google Scholar]
  • 43.Grösch S, Tegeder I, Niederberger E, Bräutigam L, 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. 2001;15:2742–4. doi: 10.1096/fj.01-0299fje. [DOI] [PubMed] [Google Scholar]
  • 44.Hsu AL, Ching TT, Wang DS, Song X, Rangnekar VM, Chen CS. The cyclooxygenase-2 inhibitor celecoxib induces apoptosis by blocking Akt activation in human prostate cancer cells independently of Bcl-2. J Biol Chem. 2000;275:11397–403. doi: 10.1074/jbc.275.15.11397. [DOI] [PubMed] [Google Scholar]
  • 45.Smalley WE, DuBois RN. Colorectal cancer and nonsteroidal anti-inflammatory drugs. Adv Pharmacol. 1997;39:1–20. doi: 10.1016/s1054-3589(08)60067-8. [DOI] [PubMed] [Google Scholar]
  • 46.Elder DJ, Paraskeva C. COX-2 inhibitors for colorectal cancer. Nat Med. 1998;4:392–3. doi: 10.1038/nm0498-392. [DOI] [PubMed] [Google Scholar]
  • 47.Williams CS, Mann M, DuBois RN. The role of cyclooxygenases in inflammation, cancer, and development. Oncogene. 1999;18:7908–16. doi: 10.1038/sj.onc.1203286. [DOI] [PubMed] [Google Scholar]
  • 48.Waskewich C, Blumenthal RD, Li H, Stein R, Goldenberg DM, Burton J. Celecoxib exhibits the greatest potency amongst cyclooxygenase (COX) inhibitors for growth inhibition of COX-2-negative hematopoietic and epithelial cell lines. Cancer Res. 2002;62:2029–33. [PubMed] [Google Scholar]
  • 49.Totzke G, Schulze-Osthoff K, Jänicke RU. Cyclooxygenase-2 (COX-2) inhibitors sensitize tumor cells specifically to death receptor-induced apoptosis independently of COX-2 inhibition. Oncogene. 2003;22:8021–30. doi: 10.1038/sj.onc.1206837. [DOI] [PubMed] [Google Scholar]
  • 50.Hengartner MO. The biochemistry of apoptosis. Nature. 2000;407:770–6. doi: 10.1038/35037710. [DOI] [PubMed] [Google Scholar]
  • 51.Kim SH, Hwang CI, Juhnn YS, Lee JH, Park WY, Song YS. GADD153 mediates celecoxib-induced apoptosis in cervical cancer cells. Carcinogenesis. 2007;28:223–31. doi: 10.1093/carcin/bgl227. [DOI] [PubMed] [Google Scholar]
  • 52.Weppelmann B, Monkemeier D. The influence of prostaglandin antagonists on radiation therapy of carcinoma of the cervix. Gynecol Oncol. 1984;17:196–9. doi: 10.1016/0090-8258(84)90077-5. [DOI] [PubMed] [Google Scholar]
  • 53.Tsuji S, Tsujii M, Kawano S, Hori M. Cyclooxygenase-2 upregulation as a perigenetic change in carcinogenesis. J Exp Clin Cancer Res. 2001;20:117–29. [PubMed] [Google Scholar]
  • 54.Kim KY, Seol JY, Jeon GA, Nam MJ. The combined treatment of aspirin and radiation induces apoptosis by the regulation of bcl-2 and caspase-3 in human cervical cancer cell. Cancer Lett. 2003;189:157–66. doi: 10.1016/s0304-3835(02)00519-0. [DOI] [PubMed] [Google Scholar]
  • 55.Jeon YT, Seo SS, Kim JW, Park NH, Kang SB, Lee HP, et al. Cyclooxygenase expressions and response to radiation therapy in uterine cervix cancer. J Gynecol Oncol. 2006;17:105–11. [Google Scholar]
  • 56.Jeon YT, Song YC, Kim SH, Wu HG, Kim IH, Park IA, et al. Influences of cyclooxygenase-1 and -2 expression on the radiosensitivities of human cervical cancer cell lines. Cancer Lett. 2007;256:33–8. doi: 10.1016/j.canlet.2007.05.008. [DOI] [PubMed] [Google Scholar]
  • 57.Hefler LA, Grimm C, Speiser P, Sliutz G, Reinthaller A. The cyclooxygenase-2 inhibitor rofecoxib (Vioxx) in the treatment of cervical dysplasia grade II–III A phase II trial. Eur J Obstet Gynecol Reprod Biol. 2006;125:251–4. doi: 10.1016/j.ejogrb.2005.08.019. [DOI] [PubMed] [Google Scholar]
  • 58.Farley JH, Truong V, Goo E, Uyehara C, Belnap C, Larsen WI. A randomized double-blind placebo-controlled phase II trial of the cyclooxygenase-2 inhibitor Celecoxib in the treatment of cervical dysplasia. Gynecol Oncol. 2006;103:425–30. doi: 10.1016/j.ygyno.2006.03.036. [DOI] [PubMed] [Google Scholar]
  • 59.Herrera FG, Chan P, Doll C, Milosevic M, Oza A, Syed A, et al. A prospective phase I–II trial of the cyclooxygenase-2 inhibitor celecoxib in patients with carcinoma of the cervix with biomarker assessment of the tumor microenvironment. Int J Radiat Oncol Biol Phys. 2007;67:97–103. doi: 10.1016/j.ijrobp.2006.08.024. [DOI] [PubMed] [Google Scholar]
  • 60.Gaffney DK, Winter K, Dicker AP, Miller B, Eifel PJ, Ryu J, et al. A Phase II study of acute toxicity for Celebrex (celecoxib) and chemoradiation in patients with locally advanced cervical cancer: primary endpoint analysis of RTOG 0128. Int J Radiat Oncol Biol Phys. 2007;67:104–9. doi: 10.1016/j.ijrobp.2006.08.002. [DOI] [PubMed] [Google Scholar]
  • 61.Marnett LJ, Kalgutkar AS. Cyclooxygenase 2 inhibitors: discovery, selectivity and the future. Trends Pharmacol Sci. 1999;20:465–9. doi: 10.1016/s0165-6147(99)01385-1. [DOI] [PubMed] [Google Scholar]
  • 62.Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB, et al. The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med. 2000;342:1946–52. doi: 10.1056/NEJM200006293422603. [DOI] [PubMed] [Google Scholar]
  • 63.Giardiello FM, Yang VW, Hylind LM, Krush AJ, Petersen GM, Trimbath JD, et al. Primary chemoprevention of familial adenomatous polyposis with sulindac. N Engl J Med. 2002;346:1054–9. doi: 10.1056/NEJMoa012015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Baron JA. Epidemiology of non-steroidal anti-inflammatory drugs and cancer. Prog Exp Tumor Res. 2003;37:1–24. doi: 10.1159/000071364. [DOI] [PubMed] [Google Scholar]
  • 65.Hawk ET, Viner J, Richmond E, Umar A. Non-steroidal anti-inflammatory drugs (NSAIDs) for colorectal cancer prevention. Cancer Chemother Biol Response Modif. 2003;21:759–89. doi: 10.1016/s0921-4410(03)21036-x. [DOI] [PubMed] [Google Scholar]
  • 66.Bresalier RS, Sandler RS, Quan H, Bolognese JA, Oxenius B, Horgan K, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med. 2005;352:1092–102. doi: 10.1056/NEJMoa050493. [DOI] [PubMed] [Google Scholar]
  • 67.Clark DW, Layton D, Shakir SA. Do some inhibitors of COX-2 increase the risk of thromboembolic events?: Linking pharmacology with pharmacoepidemiology. Drug Saf. 2004;27:427–56. doi: 10.2165/00002018-200427070-00002. [DOI] [PubMed] [Google Scholar]
  • 68.Jaksch W, Dejaco C, Schirmer M. 4 years after withdrawal of rofecoxib: where do we stand today? Rheumatol Int. 2008;28:1187–95. doi: 10.1007/s00296-008-0650-4. [DOI] [PubMed] [Google Scholar]
  • 69.Grosser T. The pharmacology of selective inhibition of COX-2. Thromb Haemost. 2006;96:393–400. [PubMed] [Google Scholar]
  • 70.Mason RP, Walter MF, McNulty HP, Lockwood SF, Byun J, Day CA, et al. Rofecoxib increases susceptibility of human LDL and membrane lipids to oxidative damage: a mechanism of cardiotoxicity. J Cardiovasc Pharmacol. 2006;47 (Suppl 1):S7–14. doi: 10.1097/00005344-200605001-00003. [DOI] [PubMed] [Google Scholar]
  • 71.Juni P, Rutjes AW, Dieppe PA. Are selective COX 2 inhibitors superior to traditional non steroidal anti-inflammatory drugs? BMJ. 2002;324:1287–8. doi: 10.1136/bmj.324.7349.1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Solomon LA, Munkarah AR, Schimp VL, Arabi MH, Morris RT, Nassar H, et al. Maspin expression and localization impact on angiogenesis and prognosis in ovarian cancer. Gynecol Oncol. 2006;101:385–9. doi: 10.1016/j.ygyno.2005.11.049. [DOI] [PubMed] [Google Scholar]
  • 73.Armstrong DJ. Celecoxib and CVS risk-lessons from the APC and PreSAP studies. Rheumatology (Oxford) 2007;46:561–2. doi: 10.1093/rheumatology/kel398. [DOI] [PubMed] [Google Scholar]
  • 74.Thangapazham RL, Sharma A, Maheshwari RK. Maheshwari, Multiple molecular targets in cancer chemoprevention by curcumin. Aaps J. 2006;8:E443–9. doi: 10.1208/aapsj080352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Shishodia S, Chaturvedi MM, Aggarwal BB. Role of curcumin in cancer therapy. Curr Probl Cancer. 2007;31:243–305. doi: 10.1016/j.currproblcancer.2007.04.001. [DOI] [PubMed] [Google Scholar]
  • 76.Cheng AL, Hsu CH, Lin JK, Hsu MM, Ho YF, Shen TS, et al. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. 2001;21:2895–900. [PubMed] [Google Scholar]
  • 77.Lao CD, Ruffin MT, 4th, Normolle D, Heath DD, Murray SI, Bailey JM, et al. Dose escalation of a curcuminoid formulation, BMC Complement. Altern Med. 2006;6:10. doi: 10.1186/1472-6882-6-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sharma RA, McLelland HR, Hill KA, Ireson CR, Euden SA, Manson MM, et al. Pharmacodynamic and pharmacokinetic study of oral Curcuma extract in patients with colorectal cancer. Clin Cancer Res. 2001;7:1894–900. [PubMed] [Google Scholar]
  • 79.Javvadi P, Segan AT, Tuttle SW, Koumenis C. The chemopreventive agent curcumin is a potent radiosensitizer of human cervical tumor cells via increased reactive oxygen species production and overactivation of the mitogen-activated protein kinase pathway. Mol Pharmacol. 2008;73:1491–501. doi: 10.1124/mol.107.043554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Weng JR, Tsai CH, Kulp SK, Chen CS. Indole-3-carbinol as a chemopreventive and anti-cancer agent. Cancer Lett. 2008;262:153–63. doi: 10.1016/j.canlet.2008.01.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Jin L, Qi M, Chen DZ, Anderson A, Yang GY, Arbeit JM, Auborn KJ. Indole-3-carbinol prevents cervical cancer in human papilloma virus type 16 (HPV16) transgenic mice. Cancer Res. 1999;59:3991–7. [PubMed] [Google Scholar]
  • 82.Bell MC, Crowley-Nowick P, Bradlow HL, Sepkovic DW, Schmidt-Grimminger D, Howell P, et al. Placebo-controlled trial of indole-3-carbinol in the treatment of CIN. Gynecol Oncol. 2000;78:123–9. doi: 10.1006/gyno.2000.5847. [DOI] [PubMed] [Google Scholar]
  • 83.García-Closas R, Castellsagué X, Bosch X, González CA. The role of diet and nutrition in cervical carcinogenesis: a review of recent evidence. Int J Cancer. 2005;117:629–37. doi: 10.1002/ijc.21193. [DOI] [PubMed] [Google Scholar]
  • 84.Chan FK, Hung LC, Suen BY, Wu JC, Lee KC, Leung VK, et al. Celecoxib versus diclofenac and omeprazole in reducing the risk of recurrent ulcer bleeding in patients with arthritis. N Engl J Med. 2002;347:2104–10. doi: 10.1056/NEJMoa021907. [DOI] [PubMed] [Google Scholar]
  • 85.Laheij RJ, Sturkenboom MC, Hassing RJ, Dieleman J, Stricker BH, Jansen JB. Risk of community-acquired pneumonia and use of gastric acid-suppressive drugs. JAMA. 2004;292:1955–60. doi: 10.1001/jama.292.16.1955. [DOI] [PubMed] [Google Scholar]
  • 86.Vakevainen S, Tillonen J, Salaspuro M, Jousimies-Somer H, Nuutinen H, Farkkila M. Hypochlorhydria induced by a proton pump inhibitor leads to intragastric microbial production of acetaldehyde from ethanol. Aliment Pharmacol Ther. 2000;14:1511–8. doi: 10.1046/j.1365-2036.2000.00858.x. [DOI] [PubMed] [Google Scholar]
  • 87.Sturkenboom MC, Burke TA, Tangelder MJ, Dieleman JP, Walton S, Goldstein JL. Adherence to proton pump inhibitors or H2-receptor antagonists during the use of non-steroidal anti-inflammatory drugs. Aliment Pharmacol Ther. 2003;18:1137–47. doi: 10.1046/j.1365-2036.2003.01795.x. [DOI] [PubMed] [Google Scholar]
  • 88.Miriyala S, Panchatcharam M, Rengarajulu P. Cardioprotective effects of curcumin. Adv Exp Med Biol. 2007;595:359–77. doi: 10.1007/978-0-387-46401-5_16. [DOI] [PubMed] [Google Scholar]
  • 89.Bannwarth B. Do selective cyclo-oxygenase-2 inhibitors have a future? Drug Saf. 2005;28:183–9. doi: 10.2165/00002018-200528030-00001. [DOI] [PubMed] [Google Scholar]
  • 90.Fiorucci S, Santucci L, Wallace JL, Sardina M, Romano M, del Soldato P, et al. Interaction of a selective cyclo-oxygenase-2 inhibitor with aspirin and NO-releasing aspirin in the human gastric mucosa. Proc Natl Acad Sci U S A. 2003;100:10937–41. doi: 10.1073/pnas.1933204100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wallace JL, Del Soldato P. The therapeutic potential of NO-NSAIDs. Fundam Clin Pharmacol. 2003;17:11–20. doi: 10.1046/j.1472-8206.2003.00125.x. [DOI] [PubMed] [Google Scholar]
  • 92.Hernandez-Diaz S, Varas-Lorenzo C, Garcia Rodriguez LA. Non-steroidal antiinflammatory drugs and the risk of acute myocardial infarction. Basic Clin Pharmacol Toxicol. 2006;98:266–74. doi: 10.1111/j.1742-7843.2006.pto_302.x. [DOI] [PubMed] [Google Scholar]
  • 93.McGettigan P, Henry D. Cardiovascular risk and inhibition of cyclooxygenase: a systematic review of the observational studies of selective and nonselective inhibitors of cyclooxygenase 2. JAMA. 2006;296:1633–44. doi: 10.1001/jama.296.13.jrv60011. [DOI] [PubMed] [Google Scholar]
  • 94.RTOG-C-0128. Radiation therapy plus celecoxib, fluorouracil, and cisplatin in patients with locally advanced cervical cancer. http://clinicaltrials.gov/ct2/show/NCT00023660?term=COX-2+inhibitor+and+cervical+cancer&rank=1. Accessed May, 2013.
  • 95.Peto J, Gilham C, Deacon J, Taylor C, Evans C, Binns W, et al. Cervical HPV infection and neoplasia in a large population-based prospective study: the Manchester cohort. Br J Cancer. 2004;91:942–53. doi: 10.1038/sj.bjc.6602049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sadler L, Saftlas A. Cervical surgery and preterm birth. J Perinat Med. 2007;35:5–9. doi: 10.1515/JPM.2007.001. [DOI] [PubMed] [Google Scholar]
  • 97.Kearney PM, Baigent C, Godwin J, Halls H, Emberson JR, Patrono C. Do selective cyclo-oxygenase-2 inhibitors and traditional non-steroidal anti-inflammatory drugs increase the risk of atherothrombosis? Meta-analysis of randomised trials. BMJ. 2006;332:1302–8. doi: 10.1136/bmj.332.7553.1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mukherjee D, Nissen SE, Topol EJ. Risk of cardiovascular events associated with selective COX-2 inhibitors. JAMA. 2001;286:954–9. doi: 10.1001/jama.286.8.954. [DOI] [PubMed] [Google Scholar]
  • 99.Jüni P, Nartey L, Reichenbach S, Sterchi R, Dieppe PA, Egger M. Risk of cardiovascular events and rofecoxib: cumulative meta-analysis. Lancet. 2004;364:2021–9. doi: 10.1016/S0140-6736(04)17514-4. [DOI] [PubMed] [Google Scholar]
  • 100.Garner SE, Fidan DD, Frankish R, Maxwell L. Rofecoxib for osteoarthritis. Cochrane Database Syst Rev. 2005:CD005115. doi: 10.1002/14651858.CD005115. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Cancer Prevention are provided here courtesy of Korean Society of Cancer Prevention

RESOURCES