Open Access

Potency of non-steroidal anti-inflammatory drugs in chemotherapy (Review)

  • Authors:
    • Lucia Hiľovská
    • Rastislav Jendželovský
    • Peter Fedoročko
  • View Affiliations

  • Published online on: October 16, 2014     https://doi.org/10.3892/mco.2014.446
  • Pages: 3-12
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer cell resistance, particularly multidrug resistance (MDR), is the leading cause of chemotherapy failure. A number of mechanisms involved in the development of MDR have been described, including the overexpression of ATP-dependent membrane-bound transport proteins. The enhanced expression of these proteins, referred to as ATP‑binding cassette (ABC) transporters, results in an increased cellular efflux of the cytotoxic drug, thereby reducing its intracellular concentration to an ineffective level. Non-steroidal anti-inflammatory drugs (NSAIDs) are the most frequently consumed drugs worldwide. NSAIDs are mainly used to treat pain, fever and inflammation. Numerous studies suggest that NSAIDs also show promise as anticancer drugs. NSAIDs have been shown to reduce cancer cell proliferation, motility, angiogenesis and invasiveness. In addition to these effects, NSAIDs have been shown to induce apoptosis in a wide variety of cancer types. Moreover, several studies have indicated that NSAIDs may sensitise cancer cells to the antiproliferative effects of cytotoxic drugs by modulating ABC transporter activity. Therefore, combining specific NSAIDs with chemotherapeutic drugs may have clinical applications. Such treatments may allow for the use of a lower dose of cytotoxic drugs and may also enhance the effectiveness of therapy. The objective of this review was to discuss the possible role of NSAIDs in the modulation of antitumour drug cytotoxicity. We particularly emphasised on the use of COX-2 inhibitors in combination with chemotherapy and the molecular and cellular mechanisms underlying the alterations in outcome that occur in response to this combination therapy.

Introduction

Non-steroidal anti-inflammatory drugs (NSAIDs) are primarily used as analgesics, antipyretics and anti-inflammatory agents. NSAIDs mainly act by inhibiting prostaglandin (PG) production. A number of experimental, epidemiological and clinical studies have revealed the antitumour properties of NSAIDs, particularly of cyclooxygenase (COX)-2 inhibitors (1, 2). NSAIDs have been shown to inhibit malignant transformation in several cancer cell lines. Moreover, the frequent use of NSAIDs has been associated with a reduced risk of colorectal, gastrointestinal, breast, prostate and lung cancer (36). The mechanism underlying the antitumour activity of NSAIDs has not been fully elucidated; however, it may involve the inhibition of COXs or other non-COX enzymatic pathways.

NSAIDs

The use of herbal extracts containing salicylates dates back thousands of years. In 1874, Maclagan successfully used salicylic acid isolated from willow bark for the treatment of the inflammation associated with rheumatic fever (7). A more effective and tolerable synthetic acetylated form of salicylic acid was introduced by Felix Hoffman in 1897; this derivative was named aspirin (8, 9). Over time, several other drugs with the same antipyretic, analgesic and anti-inflammatory properties were introduced, including antipyrine, acetaminophen, phenylbutazone, naproxen and indomethacin. As these drugs share a similar mechanism of action and are clearly distinct from other groups of drugs used in the treatment of inflammation (glucocorticoids), they were collectively named non-steroidal anti-inflammatory drugs (NSAIDs) (10, 11).

The main mechanism through which NSAIDs exert their effects is the inhibition of PG biosynthesis. PGs have been implicated in a number of physiological and pathological disorders, such as inflammation, pain, pyrexia, cancer, osteoporosis, cardiovascular diseases and asthma (12, 13). Following exposure to physiological and pathological stimuli, polyunsaturated fatty acids, including arachidonic acid (AA), are released from membrane phospholipids through the action of phospholipase A2 enzymes. Free AA is subsequently converted via one of three enzymatic pathways (1416) (Fig. 1): In the COX pathway, AA is converted to PGs, prostacyclins (PCs) and thromboxanes (TXs); in the lipoxygenase (LOX) pathway, AA is converted to hydroxyeicosatetraenoic acids (HETEs), leukotrienes (LTs) and lipoxins (LXs); lastly, in the cytochrome P450 (CYP450) monooxygenase pathway, AA release leads to the production of HETEs and epoxyeicosatrienoic acids (EETs). Additionally, in a non-enzymatic pathway, AA release results in the synthesis of isoprostanes. The products of these metabolic pathways are referred to as eicosanoids. Eicosanoids represent important intercellular and intracellular signalling molecules that participate in a wide range of physiological processes, such as the regulation of smooth muscle tone, vascular permeability, platelet aggregation, transporter proteins and proliferation. In addition, eicosanoids are involved in inflammation, autoimmunity, angiogenesis, allergic diseases and cancer (1720). Extensive research has been focused on PGs and other COX-derived metabolites. However, a number of studies suggested that LOX-derived products also affect the development and progression of several malignancies (2125).

COXs and their inhibitors in cancer treatment

There are 3 COX isoforms, commonly referred to as COX-1, COX-2 and COX-3. COX-1, also referred to as PGH synthase, is the key enzyme responsible for the oxidation of AA to PGG2 and PGH2. COX-1 is constitutively expressed, with its levels remaining constant under most physiological and pathological conditions. By contrast, the expression of COX-2 is highly inducible in response to mitogenic and inflammatory stimuli, such as fibroblast growth factor (26), transforming growth factor β (27), epidermal growth factor (28), vascular endothelial growth factor, tumour necrosis factor α and interleukins 1α and 1β (29). The function of COX-3 remains unclear (3032). An aberrant constitutive expression of COX-2 has been demonstrated during the early stages of carcinogenesis (33, 34). There is compelling evidence supporting a role for COX-2 in tumour development. COX-2 expression has been shown to be elevated in several human tumours, including colorectal (35, 36), gastric (37) and pancreatic cancer (38), oesophageal adenocarcinoma (39), lung (40) and breast cancer (41). The tumour-promoting effect of COX-2 may be a consequence of the numerous effects that COX-2 exerts on cells. COX-2 may promote proliferation, angiogenesis and invasiveness, prevent apoptosis and enhance cell adhesion and motility (42). Treatment with COX-2-specific inhibitors results in a wide range of cellular effects, including induction of apoptosis, reduction of cell proliferation, inhibition of angiogenesis and enhanced anticancer drug-induced cytotoxicity (4346). These findings suggest that NSAIDs may exert their anticancer effects through COX-2 inhibition. Although the significance of COX-2 inhibitors is well established, the mechanism underlying their chemopreventive and chemotherapeutic actions is largely unknown. Indeed, there is evidence suggesting that the antitumour effect of NSAIDs may not only be mediated by the inhibition of COX-2 activity, but that other cellular targets may also play a role (46). This hypothesis is supported by the observation that NSAID treatment reduced cell survival in COX-2-overexpressing as well as COX-deficient cancer cell lines (4749).

LOXs and their inhibitors in cancer treatment

Information regarding the role of LOXs in the promotion of cancer growth is limited. The identification of LOX isoforms in cancer, stromal and immune cells has led to the hypothesis that these enzymes may contribute to tumour development and growth (50), with interest mainly focused on 5-LOX, 12-LOX and 15-LOX. Under physiological conditions, the expression of 5-LOX is limited to immune cells (51, 52). 5-LOX may directly control tumour cell function or indirectly affect the tumour microenvironment. Increased 5-LOX activity has been demonstrated to play a role in the early stages of colon cancer (53) and in carcinogenesis in human oral cavity tissues (54). It was also reported that 5-LOX expression may be involved in the development of BCR-ABL-induced chronic myeloid leukaemia (55). Moreover, the 5-LOX pathway may be involved in the metastatic process of pancreatic, intestinal and prostate cancers (56, 57). The inhibition of 5-LOX expression and activity promotes cell apoptosis and tumour growth arrest. Additionally, 5-LOX inhibition affects epithelial-to-mesenchymal transition in certain cancer cell lines and suppresses metastasis in pancreatic cancer. These effects are likely due to the upregulation of E-cadherin and paxillin (5861). The finding that 12-LOX is overexpressed in murine lung carcinoma and human prostate cancer cells suggests a possible role for this enzyme in cancer development (22, 62). The 12-LOX inhibitor baicalein induces apoptosis in cancer cells. This induction is mediated through the regulation of the B-cell lymphoma-2 (Bcl-2) protein (6365). Furthermore, 12-LOX controls G1/S-phase arrest by inhibiting Akt and mitogen-activated protein kinases and regulating the expression of nuclear factor (NF)-κB (66). A proangiogenic function for 12-LOX products has also been suggested. The downregulation of 15-LOX expression has been shown in breast and prostate cancer and colorectal adenocarcinomas (6770). The 15-LOX-2 isoform suppresses cell cycle progression and promotes cell senescence (7072). Taken together, these findings suggest that LOXs may be potential targets for anticancer therapy.

P450 monooxygenases and their inhibitors in cancer treatment

CYP450s are monooxygenases that catalyse a variety of reactions. These enzymes have variable substrates, including fatty acids, steroids and xenobiotics. CYP450 enzymes are localised to the mitochondria and the endoplasmic reticulum. Mitochondrial CYP450s metabolise endogenous substrates, whereas microsomal CYP450s are involved in the metabolic reactions of exo- and endogenous substrates. Significant attention has been focused on the roles of COX- and LOX-derived products in carcinogenesis; however, little is known regarding the role of CYP450-derived products in this process. CYP450 activity in cancer cells may lead to the deactivation of antitumour drugs, thereby limiting therapeutic efficacy. The CYP1, CYP2 and CYP3 families are important enzymes that metabolise a significant number of clinically important drugs (73). Aberrant CYP450 enzymatic activity has been detected in a variety of human cancer cell lines and has been shown to contribute to neoangiogenesis, cancer cell migration, tumour growth and metastasis (7478).

NSAIDs in cancer treatment

Drug resistance is considered to be a major hindrance to the success of chemotherapeutic treatment. Multidrug resistance (MDR) is a multifactorial phenomenon and is often associated with the overexpression of ATP-binding cassette (ABC) transporter proteins (79, 80). Accumulating evidence indicates that NSAIDs exert a chemosensitising effect; however, the exact mechanism underlying this action remains unknown, although several molecular mechanisms have been suggested.

Combination of NSAIDs with chemotherapeutic drugs in vitro

NSAIDs, particularly COX-2 inhibitors, may supress MDR by inhibiting ABC transporters and sensitise cancer cells to the antiproliferative effects of anticancer drugs. These effects of NSAIDs have been demonstrated in several different malignancies (8185). Permeability glycoprotein (P-gp), which acts on a broad substrate range, is one of the most extensively investigated and best characterised transporter proteins. NSAIDs have been shown to suppress the expression and function of this transporter in a variety of cancer cell types. Zatelli et al (85) demonstrated that treatment with the selective COX-2 inhibitor NS-398 resulted in significantly increased doxorubicin accumulation and sensitivity in chemoresistant MCF7 breast cancer cells. Those effects depended on the inhibition of P-gp expression and function. By contrast, it was suggested that NSAIDs are not involved in the regulation of P-gp activity and function and that their chemosensitising effect is mediated through different mechanisms (86). However, the majority of the studies contradict this hypothesis. Awara et al (87) reported an enhancement of doxorubicin antitumour activity with celecoxib-induced P-gp inhibition. This was demonstrated by a significant reduction in the efflux of the P-gp substrate Rhodamine 123. Similar findings were reported by other research groups (82, 85, 88, 89). Indomethacin and a COX-2 selective inhibitor, SC236, sensitised HepG2 human hepatocellular carcinoma cells to the cytotoxic effects of doxorubicin. This effect was the result of increased intracellular retention and accumulation of doxorubicin via the inhibition of P-gp and MDR associated protein 1 (MRP1) expression and activity (90). Kang et al (91) detected an inhibition of the MRP1 efflux pump and enhanced doxorubicin cytotoxicity with celecoxib treatment. Similar results were obtained by Ko et al (92), where celecoxib not only reverted MRP1-related drug resistance, but also inhibited the function of breast cancer resistance protein (BCRP). Due to its expression in malignant hematopoietic and lymphoid cells, BCRP potentially plays an important role in drug resistance, not only in breast cancer, but also in hematological malignancies. Furthermore, BCRP is expressed in leukaemic stem cells, contributing to the resistance of these cancers to chemotherapy or targeted therapy (93). The drugs used to treat these cancers are often BCRP substrates. Little is known regarding the effects of NSAIDs on antitumour drug cytotoxicity in hematological malignancies. Accumulating evidence indicates a positive effect of NSAIDs on chemotherapeutic drug action in BCRP-overexpressing solid tumours. Co-treatment with mitoxantrone and indomethacin sensitised resistant MCF-7/MX cells to mitoxantrone (94). Studies that combined NSAIDs with cisplatin-based chemotherapy have yielded opposing results. A recent study revealed that celecoxib and SC-236 antagonised the cytotoxicity of cisplatin in human gastric cells, whereas indomethacin and nimesulid exerted no effects (95). By contrast, the use of another COX-2 selective inhibitor, JTE-522, in combination with cisplatin, resulted in synergistic antitumour activity in a gastric cancer cell line (96). In other cancer cell lines, celecoxib potentiated the cytotoxicity of cisplatin (97, 98). The discrepancy regarding the effects of NSAIDs on cisplatin action may be partially explained by the different chemical structures of the utilised NSAIDs and by the different tumour cell types employed (95).

Apart from ABC transporter inhibition, other mechanisms have been suggested to explain the chemosensitising effect of NSAIDs, including the inhibition of several transcriptional factors, varying functions of COX-2 in cancer cells, ceramide production and DNA hypermethylation (Table I). NF-κB inhibition may play a role in NSAID-enhanced antitumour drug cytotoxicity (99). NF-κB has been shown to be involved in chemoresistance in different cancer types. The constitutive expression of this transcription factor in tumours protects against apoptotic stimuli. Moreover, the inhibition of NF-κB activity may affect intracellular drug accumulation and transport. The enhanced accumulation of doxorubicin in MDA-MB-231 human breast cancer cells upon celecoxib treatment was not mediated by changes in COX-2 enzyme activity or through P-gp, MRP1 or BCRP inhibition, but rather due to the inhibition of NF-κB. Xia et al also demonstrated that NSAIDs may sensitise cancer cells to antitumour drugs by inducing DNA hypermethylation (100). The ability of celecoxib to modulate DNA methylation has also been demonstrated (101). The expression of the MDR1 gene, which codes for the P-gp protein, is regulated through the methylation of CpG islands located within the MDR1 promoter (102104). Xia et al observed that treatment with celecoxib significantly enhanced CpG island methylation, which led to the suppression of P-gp expression (100). The ability of celecoxib to repress the activity of the transcription factor Sp1 was previously demonstrated (105). The MDR1 gene promoter contains a binding side for this factor. This binding site may be susceptible to celecoxib-induced hypermethylation, thereby limiting the ability of Sp1 to bind DNA. Celecoxib, in combination with the 5-LOX inhibitor MK-886, exerted a significant additive cytotoxic effect on Caco-2 and HT-29 cancer cells, which was, in part, mediated by ceramide-induced apoptosis (106). El-Awady et al (107) demonstrated the diverse effects of celecoxib on the anticancer activity of etoposide, cisplatin, 5-fluorouracil (5-FU) and doxorubicin in five cancer cell lines, namely the HeLa, HCT-116, HepG2, MCF7 and U251. In the MCF7 breast cancer cell line, the interaction of celecoxib with these four chemotherapeutics was antagonistic, indicating that celecoxib is of little value when used in combination with antitumour drugs in the treatment of breast cancer. By contrast, other data indicate that celecoxib enhances the cytotoxicity of anticancer drugs in breast cancer cells (99, 108). The interaction of celecoxib with etoposide, cisplatin and 5-FU was shown to be dependent on the cancer cell line employed, the drug type used and the incubation schedule. The combination of celecoxib and the same antitumour drug also exerted different effects on different cell lines. One plausible explanation for this finding may be that COX-2 has different roles in different cancer types (107). In cancers where COX-2 increases tumour growth and progression (109), COX-2 inhibitors may be of therapeutic benefit. However, in other malignancies, COX-2 has been reported to exert proapoptotic and tumour-suppressing effects (110112). In such cancer types, COX-2 inhibition may lead to enhanced tumour growth, inhibition of apoptosis and decreased efficacy of anticancer drugs (107). Several studies reported a direct association between COX-2 expression and the ABC transporters P-gp and MRP1. Patel et al (113) demonstrated that the overexpression of COX-2 led to increased P-gp expression and activity, whereas the COX-2 inhibitor NS398 was able to block this increase. In colon cancer, a resistance to cisplatin resulted from COX-2 overexpression, which induced MRP1 expression (114). A positive correlation between the expression of COX-2 and P-gp was also reported by studies on hepato­cellular carcinoma, breast and ovarian cancer (115117). COX-2 was found to be involved in the regulation of P-gp, MRP1 and BCRP transporter expression via the COX-2/PGE2/PGE receptor 4/phosphatidyl inositol 3-kinase pathway (116, 118).

Table I

Effects of NSAIDs on cytotoxic drug efficacy in different cancer cell lines.

Table I

Effects of NSAIDs on cytotoxic drug efficacy in different cancer cell lines.

NSAIDs/chemotherapeuticsCancer cell lineEffectMechanism of actionAuthor (Refs.)
Celecoxib/doxorubicinECCSynInhibition of P-gpAwara et al (87)
H460SynInhibition of MRP1Kang et al (91)
MDA-MB-231SynInhibition of NF-κBvan Wijngaarden et al (99)
MCF7AntDNA damage repairEl-Awady et al (107)
Celecoxib/vincristineKB/VCRSynInhibition of P-gpYan et al (88)
Celecoxib/imatinibK562SynInhibition of P-gpArunasree et al (82)
Celecoxib/cisplatinTMK1AntEnhanced efflux, reduced influxChen et al (95)
MCF7AntDNA damage repairEl-Awady et al (107)
Celecoxib/5-FUMCF7AntDNA damage repairEl-Awady et al (107)
JTE-522/cisplatinMKN-45SynInhibition of ABCSugiura et al (96)
Celecoxib/tamoxifenMCF7SynInhibition of P-pg due to DNA methylationXia et al (100)
NS-398/doxorubicinMCF7SynInhibition of P-gpZatelli et al (85)
Indomethacin, SC-236/doxorubicinHepG2SynInhibition of P-gp, MRP1Ye et al (90)
Indomethacin/mitoxantroneMCF7/MXSynInhibition of BCRPElahian et al (94)

[i] Syn, synergiSTIc effect of combinatory therapy; ant, antagonistic effect; ECC, Ehrlich carcinoma cell line; H460, non-small-cell lung carcinoma cell line; MDA-MB-231 AND MCF7, breast carcinoma cell lines; MCF7/MX, mitoxantrone-resistant breast carcinoma cell line; KB/VCR, oral cancer cell line; K562, erythromyeloblastoid leukaemia cell line; TMK1 AND MKN-45, human gastric adenocarcinoma cell lines; HepG2, hepatocellular carcinoma cell line; 5-FU, 5-fluorouracil; P-gp, permeability glycoprotein; MRP, multidrug resistance protein; NF, nuclear factor; ABC, ATP-binding cassette; BCRP, breast cancer resistance protein.

Synergistic effects of NSAIDs with hypericin (HY)-mediated photodynamic therapy (PDT) have also been reported (119122). The specific inhibition of COX, LOX and CYP450 activity increased the efficacy of HY-PDT in the HT-29 cancer cell line (121). An important role for the MRP1 and BCRP transporters in HY efflux was also demonstrated (119). Proadifen, a P450 monooxygenase inhibitor, was shown to inhibit these transport proteins, resulting in a significant increase in intracellular HY accumulation in HT-29 cells and MRP1 and BCRP-overexpressing cells.

Taken together, the abovementioned findings indicate that the mechanism through which NSAIDs affect the action and effectiveness of cytotoxic drugs varies. The exact mechanism may depend on the cancer cell line, the structures of the NSAIDs and chemotherapeutics, the specific interactions between the drugs and the incubation schedule. The mechanism underlying the NSAID-induced increase in antitumour drug cytotoxicity may be one of the abovementioned processes. However, more than one mechanisms are likely involved.

Combination of NSAIDs with chemotherapeutic drugs in vivo

A growing amount of evidence from various animal models suggests positive effects of NSAID use in combination with antitumour drugs (87, 123129) (Table II). However, the exact mechanism through which this combined treatment results in improved antitumour activity in in vivo models is not clearly understood. Given the complexity of animal models in comparison to in vitro systems, the effects of the tumour microenvironment, tumour angiogenesis, the immune system and pharmacokinetic processes must be taken into consideration (123, 126, 130). As NSAIDs may alter ABC transporter expression or activity in cancer cell lines, this mechanism may also be involved in vivo. Awara et al (87) reported that the inhibition of P-gp activity by NSAIDs is likely responsible for the enhanced antitumour effects of doxorubicin. It was suggested that NSAIDs exert their growth-inhibitory functions and synergistic effects with chemotherapeutics through multiple pathways. Neoangiogenesis plays a key role in tumour promotion and progression. Certain studies demonstrated the ability of NSAIDs, particularly selective COX-2 inhibitors, to suppress tumour growth by inhibiting angiogenesis and cell proliferation (131, 132). Although the suppression of angiogenesis that occurs with NSAID treatment alone may not be sufficient to inhibit tumour growth, NSAIDs may enhance the antiangiogenic and antiproliferative effects of certain antitumour drugs (123, 125, 127). As shown by Irie et al (125), celecoxib alone did not significantly inhibit tumour growth, although it did exhibit a certain antiangiogenic activity. However, in combination with 5-FU, celecoxib enhanced the antitumour effect of 5-FU and significantly suppressed angiogenesis and tumour growth, likely via the inhibition of VEGF and the induction of IFN-γ (125). Treatment with celecoxib in combination with doxorubicin and irinotecan was also found to be effective in decreasing tumour growth through the inhibition of cell proliferation and the suppression of tumour vasculature (127). A number of intracellular signalling proteins are involved in cell proliferation, survival and apoptosis. Several lines of evidence suggest that COX-2 may elevate the levels of the antiapoptotic proteins Bcl-2 and Mcl-1 through mitogen-activated protein kinase activation, which results in an inhibition of the cytochrome c pathway (133135). Moreover, a study by Zhang et al (129) revealed an improved therapeutic benefit of 5-FU via celecoxib addition, which occurred through the induction of the cytochrome c-dependent apoptotic pathway, as well as a possible role for 5-FU in the celecoxib-mediated inhibition of COX-2 expression. As previously mentioned, the antiproliferative, antiangiogenic and antitumour effects of NSAIDs may be, to a certain extent, COX-2-independent. Consistent with these findings, piroxicam was able to exert its effect via a COX/PGE2-independent mechanism (128). Moreover, piroxicam enhanced cisplatin-induced cytotoxicity via the upregulation of endogenous drug effectors and the inhibition of certain cell growth regulators. In vitro studies demonstrated that NSAIDs may mediate their antitumour effects through modulation of the NF-κB signalling pathway (99, 136). NF-κB, with its dual anti- and proapoptotic functions, plays an important role in regulating cellular proliferation and apoptotic cell death. The inhibition of NF-κB activity may be responsible for the celecoxib-induced doxorubicin cytotoxicity that results in decreased tumour volume (99). By contrast, certain studies suggested that NSAIDs may activate NF-κB, thereby inducing apoptosis (137, 138). Apart from enhancing the cytotoxicity of chemotherapeutic drugs, the addition of NSAIDs may also reduce the severity of chemotherapy-associated adverse effects, such as late diarrhoea and cachexia (139).

Table II

Effects of NSAIDs on cytotoxic drug efficacy in animal models.

Table II

Effects of NSAIDs on cytotoxic drug efficacy in animal models.

NSAIDs/chemotherapeuticsExperimental modelEffectMechanism of actionAuthor (Refs.)
Celecoxib/doxorubicinMouse SECSynInhibition of P-gpAwara et al (87)
Rat SH-SY5YSynInhibition of angiogenesis and cell proliferationPonthan et al (127)
Celecoxib/irinotecanMouse MCASynUnknownTrifan et al (139)
Rat SH-SY5YSynInhibition of angiogenesis and cell proliferationPonthan et al (127)
Celecoxib/5-FUMouse colon 26 cellsSynInhibition of tumour growth and angiogenesisIrie et al (125)
Mouse HT-29SynCytochrome c-dependent apoptotic pathwaysZhang et al (129)
Piroxicam/cisplatinDog TCCSynUnknownKnapp et al (126)
Mouse MSTO-211HSynEnhanced expression of intracellular drug effectorsSpugnini et al (128)
Aspirin/doxorubicinMouse HepG2SynReduction of tumour growth and weightHossain et al (124)
JTE-522/docetaxel, vinorelbineMouse ACC-LC-319SynInhibition of tumour growthHida et al (123)

[i] Syn, synergiSTIc effect of combinatory therapy; SEC, solid Ehrlich carcinoma; SH-SY5Y, neuroblastoma cell line; HT-29, human colorectal cancer CELL LINE; MCA, mouse colorectal adenocarcinoma; colon 26 cells, colon cancer cell line; TCC, transitional cell carcinoma; MSTO-211H, human mesothelioma cell line; HepG2, human hepatocellular carcinoma cell line; ACC-LC-319, human lung adenocarcinoma cell line; 5-FU, 5-fluorouracil; P-gp, permeability glycoprotein.

Combination of NSAIDs with chemotherapeutic drugs in clinical trials

Due to the limited effectiveness of certain cancer treatments, it is necessary to establish a novel treatment strategy that improves patient response to chemotherapy. A large number of studies have demonstrated that COX-2 may be involved in the development of several cancer types. COX-2 may positively affect multiple processes, including tumour cell growth, migration and invasiveness, but may also downregulate apoptosis and angiogenic stimulation (3538). Moreover, the overexpression of COX-2 may also reduce the response of cancer cells to cytotoxic therapy (140). Preclinical studies suggested that treatment with NSAIDs, particularly COX-2 inhibitors, may affect the outcome of chemotherapy through various mechanisms, including the inhibition of neoangiogenesis and the induction of apoptosis (130, 131, 141). Despite promising preclinical results with NSAIDs in combination with antitumour drugs, little is known regarding the effects of this combination on humans. The currently available clinical results are contradictory and mainly disappointing (142145) (Table III). For example, several combinations did not appear to improve therapy outcome, including celecoxib and docetaxel (144, 146); celecoxib and 5-FU (142, 143); rofecoxib, 5-FU and leucovorin (147); celecoxib and transtuzumab (148); rofecoxib, cisplatin and gemcitabine (149); celecoxib, docetaxel and carboplatin (150); and celecoxib and platinum derivates (151). However, certain phase II studies have yielded encouraging results. In the case of non-small-cell lung carcinoma (NSCLC), the combination of celecoxib and chemotherapy was associated with increased overall survival (152, 153). In the case of heavily pretreated recurrent ovarian cancer, the administration of celecoxib in combination with carboplatin-based chemotherapy also yielded promising results (154). The discrepancy in various results may be due to multiple factors, such as complex pharmacodynamic interactions between NSAIDs and the cytotoxic drugs and the varying levels of intratumoural COX-2 and ABC transporters (155159). The role of COX-2 expression in the response of cancer cells to combined NSAID and antitumour drug therapy was demonstrated by Edelman et al and has been supported by other studies (155, 160). Patients with COX-2-overexpressing tumours who did not receive combined celecoxib/chemotherapy treatment exhibited a significantly worse outcome. Possible adverse effects in patients with COX-2-non-expressing tumours that received celecoxib treatment were also demonstrated (160).

Table III

Effects of NSAIDs on cytotoxic drug efficacy in clinical trials.

Table III

Effects of NSAIDs on cytotoxic drug efficacy in clinical trials.

NSAIDs/chemotherapeuticsCancer typeEffectMechanism of actionAuthor (Refs.)
Celecoxib/paclitaxel, carboplatinNSCLCSynInhibition of COX-2Altorki et al (152)
Celecoxib/docetaxelNSCLCNone-Schneider et al (144)
NSCLCSynInhibition of COX-2Nugent et al (153)
Celecoxib/FOLFIRICRCNone-Maiello et al (143)
Celecoxib/FOLFIRI, CAPIRICRCNone-Kohne et al (142)
Celecoxib/platin derivates, gemcitabine, vinorelbinNSCLCNoneVarious COX-2 expression levelsKoch et al (151)
Celecoxib/carboplatin, docetaxelNSCLCNone-Groen et al (150)
Celecoxib/carboplatinOCSynPotential inhibition of COX-2Legge et al (154)
Celecoxib/transtuzumabBCNone-Dang et al (148)
Rofecoxib/5-FUCRCNone-Becerra et al (147)

[i] Syn, synergiSTIc effect of combinatory therapy; NSCLC, non-small-cell lung carcinoma; CRC, colorectal adenocarcinoma; OC, ovarian cancer; BC, breast adenocarcinoma; SCLC, small-cell lung carcinoma; FOLFIRI, leucovorin + 5-FU + irinotecan; CAPIRI, capecitabine + irinotecan; COX, cyclooxygenase; 5-FU, 5-fluorouracil; -, inadequate dosing.

Conclusion

NSAIDs are potent antitumour drugs, capable of inhibiting tumour angiogenesis, proliferation, invasion and motility, as well as of inducing apoptosis. Furthermore, a number of experimental and preclinical studies indicated that combining NSAIDs with antitumour drugs may improve outcome. The exact mechanism underlying this synergistic effect has not yet been fully elucidated, but may involve several diverse processes, including the inhibition of COX-2 expression, ABC transporter activity or NF-κB. However, the results of combined therapy in clinical trials are mainly disappointing. Despite significant efforts to determine the exact mechanism through which NSAIDs modulate the efficacy of anticancer drugs, there remain several unanswered questions.

Acknowledgements

This study was supported by the Cancer Research Foundation (contact no. O-12-102/0001-00).

References

1 

Hanif R, Pittas A, Feng Y, Koutsos MI, Qiao L, Staiano-Coico L, Shiff SI and Rigas B: Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway. Biochem Pharmacol. 52:237–245. 1996. View Article : Google Scholar : PubMed/NCBI

2 

Souza RF, Shewmake K, Beer DG, Cryer B and Spechler SJ: Selective inhibition of cyclooxygenase-2 suppresses growth and induces apoptosis in human esophageal adenocarcinoma cells. Cancer Res. 60:5767–5772. 2000.PubMed/NCBI

3 

Dai Y and Wang WH: Non-steroidal anti-inflammatory drugs in prevention of gastric cancer. World J Gastroenterol. 12:2884–2889. 2006.PubMed/NCBI

4 

DuBois RN and Smalley WE: Cyclooxygenase, NSAIDs, and colorectal cancer. J Gastroenterol. 31:898–906. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Rao CV and Reddy BS: NSAIDs and chemoprevention. Curr Cancer Drug Targets. 4:29–42. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Winde G, Schmid KW, Brandt B, Muller O and Osswald H: Clinical and genomic influence of sulindac on rectal mucosa in familial adenomatous polyposis. Dis Colon Rectum. 40:1156–1169. 1997. View Article : Google Scholar : PubMed/NCBI

7 

Maclagan T: The treatment of acute rheumatism by salicin and salicylic acid. Lancet. 113:875–877. 1879. View Article : Google Scholar

8 

Dugowson CE and Gnanashanmugam P: Nonsteroidal anti-inflammatory drugs. Phys Med Rehabil Clin N Am. 17347–354. (vi)2006. View Article : Google Scholar

9 

Vane JR and Botting RM: The mechanism of action of aspirin. Thromb Res. 110:255–258. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Flower RJ: Drugs which inhibit prostaglandin biosynthesis. Pharmacol Rev. 26:33–67. 1974.

11 

Vane JR and Botting RM: Anti-inflammatory drugs and their mechanism of action. Inflamm Res. 47 (Suppl 2):S78–S87. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Marnett LJ, Rowlinson SW, Goodwin DC, Kalgutkar AS and Lanzo CA: Arachidonic acid oxygenation by COX-1 and COX-2. Mechanisms of catalysis and inhibition. J Biol Chem. 274:22903–22906. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Rao P and Knaus EE: Evolution of nonsteroidal anti-inflammatory drugs (NSAIDs): cyclooxygenase (COX) inhibition and beyond. J Pharm Pharm Sci. 11:S81–S110. 2008.PubMed/NCBI

14 

Dubois RN, Abramson SB, Crofford L, et al: Cyclooxygenase in biology and disease. FASEB J. 12:1063–1073. 1998.PubMed/NCBI

15 

Rigas B and Shiff SJ: Nonsteroidal anti-inflammatory drugs (NSAIDs), cyclooxygenases, and the cell cycle. Their interactions in colon cancer. Adv Exp Med Biol. 470:119–126. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Wang D, Mann JR and DuBois RN: The role of prostaglandins and other eicosanoids in the gastrointestinal tract. Gastroenterology. 128:1445–1461. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Capdevila JH, Falck JR and Harris RC: Cytochrome P450 and arachidonic acid bioactivation. Molecular and functional properties of the arachidonate monooxygenase. J Lipid Res. 41:163–181. 2000.PubMed/NCBI

18 

Gerritsen ME: Physiological and pathophysiological roles of eicosanoids in the microcirculation. Cardiovasc Res. 32:720–732. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Harder DR, Campbell WB and Roman RJ: Role of cytochrome P-450 enzymes and metabolites of arachidonic acid in the control of vascular tone. J Vasc Res. 32:79–92. 1995. View Article : Google Scholar : PubMed/NCBI

20 

Harizi H, Corcuff JB and Gualde N: Arachidonic-acid-derived eicosanoids: roles in biology and immunopathology. Trends Mol Med. 14:461–469. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Chen YQ, Duniec ZM, Liu B, et al: Endogenous 12(S)-HETE production by tumor cells and its role in metastasis. Cancer Res. 54:1574–1579. 1994.PubMed/NCBI

22 

Gao X, Grignon DJ, Chbihi T, et al: Elevated 12-lipoxygenase mRNA expression correlates with advanced stage and poor differentiation of human prostate cancer. Urology. 46:227–237. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Honn KV, Tang DG, Gao X, et al: 12-lipoxygenases and 12(S)-HETE: role in cancer metastasis. Cancer Metastasis Rev. 13:365–396. 1994. View Article : Google Scholar : PubMed/NCBI

24 

Tang DG and Honn KV: 12-Lipoxygenase, 12(S)-HETE, and cancer metastasis. Ann N Y Acad Sci. 744:199–215. 1994. View Article : Google Scholar : PubMed/NCBI

25 

Timar J, Raso E, Fazakas ZS, Silletti S, Raz A and Honn KV: Multiple use of a signal transduction pathway in tumor cell invasion. Anticancer Res. 16:3299–3306. 1996.PubMed/NCBI

26 

Kage K, Fujita N, Oh-hara T, Ogata E, Fujita T and Tsuruo T: Basic fibroblast growth factor induces cyclooxygenase-2 expression in endothelial cells derived from bone. Biochem Biophys Res Commun. 254:259–263. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Fong CY, Pang L, Holland E and Knox AJ: TGF-beta1 stimulates IL-8 release, COX-2 expression, and PGE(2) release in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 279:L201–L207. 2000.PubMed/NCBI

28 

Saha D, Datta PK, Sheng H, et al: Synergistic induction of cyclooxygenase-2 by transforming growth factor-beta1 and epidermal growth factor inhibits apoptosis in epithelial cells. Neoplasia. 1:508–517. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Diaz A, Chepenik KP, Korn JH, Reginato AM and Jimenez SA: Differential regulation of cyclooxygenases 1 and 2 by interleukin-1 beta, tumor necrosis factor-alpha, and transforming growth factor-beta 1 in human lung fibroblasts. Exp Cell Res. 241:222–229. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Chandrasekharan NV, Dai H, Roos KL, et al: COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: cloning, structure, and expression. Proc Natl Acad Sci USA. 99:13926–13931. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Cui JG, Kuroda H, Chandrasekharan NV, et al: Cyclooxygenase-3 gene expression in Alzheimer hippocampus and in stressed human neural cells. Neurochem Res. 29:1731–1737. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Kis B, Snipes JA and Busija DW: Acetaminophen and the cyclooxygenase-3 puzzle: sorting out facts, fictions, and uncertainties. J Pharmacol Exp Ther. 315:1–7. 2005. View Article : Google Scholar

33 

Cerella C, Sobolewski C, Chateauvieux S, et al: COX-2 inhibitors block chemotherapeutic agent-induced apoptosis prior to commitment in hematopoietic cancer cells. Biochem Pharmacol. 82:1277–1290. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Surh YJ and Kundu JK: Signal transduction network leading to COX-2 induction: a road map in search of cancer chemopreventives. Arch Pharm Res. 28:1–15. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S and DuBois RN: Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology. 107:1183–1188. 1994.PubMed/NCBI

36 

Sano H, Kawahito Y, Wilder RL, et al: Expression of cyclooxygenase-1 and −2 in human colorectal cancer. Cancer Res. 55:3785–3789. 1995.

37 

Ristimaki A, Honkanen N, Jankala H, Sipponen P and Harkonen M: Expression of cyclooxygenase-2 in human gastric carcinoma. Cancer Res. 57:1276–1280. 1997.PubMed/NCBI

38 

Yip-Schneider MT, Barnard DS, Billings SD, et al: Cyclo­oxygenase-2 expression in human pancreatic adenocarcinomas. Carcinogenesis. 21:139–146. 2000.

39 

Wilson KT, Fu S, Ramanujam KS and Meltzer SJ: Increased expression of inducible nitric oxide synthase and cyclooxygenase-2 in Barrett's esophagus and associated adenocarcinomas. Cancer Res. 58:2929–2934. 1998.

40 

Wolff H, Saukkonen K, Anttila S, Karjalainen A, Vainio H and Ristimaki A: Expression of cyclooxygenase-2 in human lung carcinoma. Cancer Res. 58:4997–5001. 1998.PubMed/NCBI

41 

Hwang D, Scollard D, Byrne J and Levine E: Expression of cyclooxygenase-1 and cyclooxygenase-2 in human breast cancer. J Natl Cancer Inst. 90:455–460. 1998. View Article : Google Scholar : PubMed/NCBI

42 

Cao Y and Prescott SM: Many actions of cyclooxygenase-2 in cellular dynamics and in cancer. J Cell Physiol. 190:279–286. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Hida T, Kozaki K, Muramatsu H, et al: Cyclooxygenase-2 inhibitor induces apoptosis and enhances cytotoxicity of various anticancer agents in non-small cell lung cancer cell lines. Clin Cancer Res. 6:2006–2011. 2000.PubMed/NCBI

44 

O'Kane SL, Eagle GL, Greenman J, Lind MJ and Cawkwell L: COX-2 specific inhibitors enhance the cytotoxic effects of pemetrexed in mesothelioma cell lines. Lung Cancer. 67:160–165. 2010.PubMed/NCBI

45 

Sinha-Datta U, Taylor JM, Brown M and Nicot C: Celecoxib disrupts the canonical apoptotic network in HTLV-I cells through activation of Bax and inhibition of PKB/Akt. Apoptosis. 13:33–40. 2008. View Article : Google Scholar

46 

Totzke G, Schulze-Osthoff K and Janicke RU: Cyclooxygenase-2 (COX-2) inhibitors sensitize tumor cells specifically to death receptor-induced apoptosis independently of COX-2 inhibition. Oncogene. 22:8021–8030. 2003. View Article : Google Scholar

47 

Elder DJ, Halton DE, Hague A and Paraskeva C: Induction of apoptotic cell death in human colorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti-inflammatory drug: independence from COX-2 protein expression. Clin Cancer Res. 3:1679–1683. 1997.

48 

Grosch S, Tegeder I, Niederberger E, Brautigam 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

49 

Zhang X, Morham SG, Langenbach R and Young DA: Malignant transformation and antineoplastic actions of nonsteroidal antiinflammatory drugs (NSAIDs) on cyclooxygenase-null embryo fibroblasts. J Exp Med. 190:451–459. 1999. View Article : Google Scholar

50 

Pidgeon GP, Lysaght J, Krishnamoorthy S, et al: Lipoxygenase metabolism: roles in tumor progression and survival. Cancer Metastasis Rev. 26:503–524. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Radmark O, Werz O, Steinhilber D and Samuelsson B: 5-Lipoxygenase: regulation of expression and enzyme activity. Trends Biochem Sci. 32:332–341. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Werz O and Steinhilber D: Therapeutic options for 5-lipoxygenase inhibitors. Pharmacol Ther. 112:701–718. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Wasilewicz MP, Kolodziej B, Bojulko T, et al: Overexpression of 5-lipoxygenase in sporadic colonic adenomas and a possible new aspect of colon carcinogenesis. Int J Colorectal Dis. 25:1079–1085. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Metzger K, Angres G, Maier H and Lehmann WD: Lipoxygenase products in human saliva: patients with oral cancer compared to controls. Free Radic Biol Med. 18:185–194. 1995. View Article : Google Scholar : PubMed/NCBI

55 

Chen Y, Hu Y, Zhang H, Peng C and Li S: Loss of the Alox5 gene impairs leukemia stem cells and prevents chronic myeloid leukemia. Nat Genet. 41:783–792. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Hennig R, Ventura J, Segersvard R, et al: LY293111 improves efficacy of gemcitabine therapy on pancreatic cancer in a fluore­scent orthotopic model in athymic mice. Neoplasia. 7:417–425. 2005.PubMed/NCBI

57 

Paruchuri S, Broom O, Dib K and Sjolander A: The pro-inflammatory mediator leukotriene D4 induces phosphatidylinositol 3-kinase and Rac-dependent migration of intestinal epithelial cells. J Biol Chem. 280:13538–13544. 2005. View Article : Google Scholar

58 

Hayashi T, Nishiyama K and Shirahama T: Inhibition of 5-lipoxy­genase pathway suppresses the growth of bladder cancer cells. Int J Urol. 13:1086–1091. 2006.

59 

Meng Z, Cao R, Yang Z, Liu T, Wang Y and Wang X: Inhibitor of 5-lipoxygenase, zileuton, suppresses prostate cancer meta­stasis by upregulating E-cadherin and paxillin. Urology. 82(1452): e7–e14. 2013.PubMed/NCBI

60 

Schroeder CP, Yang P, Newman RA and Lotan R: Simultaneous inhibition of COX-2 and 5-LOX activities augments growth arrest and death of premalignant and malignant human lung cell lines. J Exp Ther Oncol. 6:183–192. 2007.

61 

Shin VY, Jin HC, Ng EK, Sung JJ, Chu KM and Cho CH: Activation of 5-lipoxygenase is required for nicotine mediated epithelial-mesenchymal transition and tumor cell growth. Cancer Lett. 292:237–245. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Hagmann W, Gao X, Zacharek A, Wojciechowski LA and Honn KV: 12-Lipoxygenase in Lewis lung carcinoma cells: molecular identity, intracellular distribution of activity and protein, and Ca2+-dependent translocation from cytosol to membranes. Prostaglandins. 49:49–62. 1995.PubMed/NCBI

63 

Pidgeon GP, Kandouz M, Meram A and Honn KV: Mechanisms controlling cell cycle arrest and induction of apoptosis after 12-lipoxygenase inhibition in prostate cancer cells. Cancer Res. 62:2721–2727. 2002.PubMed/NCBI

64 

Tang DG, Chen YQ and Honn KV: Arachidonate lipoxygenases as essential regulators of cell survival and apoptosis. Proc Natl Acad Sci USA. 93:5241–5246. 1996. View Article : Google Scholar : PubMed/NCBI

65 

Wong BC, Wang WP, Cho CH, et al: 12-Lipoxygenase inhibition induced apoptosis in human gastric cancer cells. Carcinogenesis. 22:1349–1354. 2001. View Article : Google Scholar : PubMed/NCBI

66 

Terada N, Shimizu Y, Kamba T, et al: Identification of EP4 as a potential target for the treatment of castration-resistant prostate cancer using a novel xenograft model. Cancer Res. 70:1606–1615. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Jiang WG, Watkins G, Douglas-Jones A and Mansel RE: Reduction of isoforms of 15-lipoxygenase (15-LOX)-1 and 15-LOX-2 in human breast cancer. Prostaglandins Leukot Essent Fatty Acids. 74:235–245. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Shappell SB, Boeglin WE, Olson SJ, Kasper S and Brash AR: 15-lipoxygenase-2 (15-LOX-2) is expressed in benign prostatic epithelium and reduced in prostate adenocarcinoma. Am J Pathol. 155:235–245. 1999. View Article : Google Scholar : PubMed/NCBI

69 

Shureiqi I, Wu Y, Chen D, et al: The critical role of 15-lipoxygenase-1 in colorectal epithelial cell terminal differentiation and tumorigenesis. Cancer Res. 65:11486–11492. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Tang DG, Bhatia B, Tang S and Schneider-Broussard R: 15-Lipoxygenase 2 (15-LOX2) is a functional tumor suppressor that regulates human prostate epithelial cell differentiation, senescence, and growth (size). Prostaglandins Other Lipid Mediat. 82:135–146. 2007. View Article : Google Scholar

71 

Bhatia B, Tang S, Yang P, et al: Cell-autonomous induction of functional tumor suppressor 15-lipoxygenase 2 (15-LOX2) contributes to replicative senescence of human prostate progenitor cells. Oncogene. 24:3583–3595. 2005. View Article : Google Scholar

72 

Tang S, Bhatia B, Maldonado CJ, et al: Evidence that arachidonate 15-lipoxygenase 2 is a negative cell cycle regulator in normal prostate epithelial cells. J Biol Chem. 277:16189–16201. 2002. View Article : Google Scholar : PubMed/NCBI

73 

Brown CM, Reisfeld B and Mayeno AN: Cytochromes P450: a structure-based summary of biotransformations using representative substrates. Drug Metab Rev. 40:1–100. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Cheranov SY, Karpurapu M, Wang D, Zhang B, Venema RC and Rao GN: An essential role for SRC-activated STAT-3 in 14,15-EET-induced VEGF expression and angiogenesis. Blood. 111:5581–5591. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Jiang JG, Ning YG, Chen C, et al: Cytochrome p450 epoxygenase promotes human cancer metastasis. Cancer Res. 67:6665–6674. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Webler AC, Michaelis UR, Popp R, et al: Epoxyeicosatrienoic acids are part of the VEGF-activated signaling cascade leading to angiogenesis. Am J Physiol Cell Physiol. 295:C1292–C1301. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Webler AC, Popp R, Korff T, et al: Cytochrome P450 2C9-induced angiogenesis is dependent on EphB4. Arterioscler Thromb Vasc Biol. 28:1123–1129. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Yan G, Chen S, You B and Sun J: Activation of sphingosine kinase-1 mediates induction of endothelial cell proliferation and angiogenesis by epoxyeicosatrienoic acids. Cardiovasc Res. 78:308–314. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Gottesman MM: Mechanisms of cancer drug resistance. Annu Rev Med. 53:615–627. 2002. View Article : Google Scholar : PubMed/NCBI

80 

Turk D and Szakacs G: Relevance of multidrug resistance in the age of targeted therapy. Curr Opin Drug Discov Devel. 12:246–252. 2009.PubMed/NCBI

81 

Arico S, Pattingre S, Bauvy C, et al: Celecoxib induces apoptosis by inhibiting 3-phosphoinositide-dependent protein kinase-1 activity in the human colon cancer HT-29 cell line. J Biol Chem. 277:27613–27621. 2002. View Article : Google Scholar : PubMed/NCBI

82 

Arunasree KM, Roy KR, Anilkumar K, Aparna A, Reddy GV and Reddanna P: Imatinib-resistant K562 cells are more sensitive to celecoxib, a selective COX-2 inhibitor: role of COX-2 and MDR-1. Leuk Res. 32:855–864. 2008. View Article : Google Scholar : PubMed/NCBI

83 

Roy KR, Reddy GV, Maitreyi L, et al: Celecoxib inhibits MDR1 expression through COX-2-dependent mechanism in human hepatocellular carcinoma (HepG2) cell line. Cancer Chemother Pharmacol. 65:903–911. 2010. View Article : Google Scholar : PubMed/NCBI

84 

Yu L, Wu WK, Li ZJ, Liu QC, Li HT, Wu YC and Cho CH: Enhancement of doxorubicin cytotoxicity on human esophageal squamous cell carcinoma cells by indomethacin and 4-[5-(4-chlorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfonamide (SC236) via inhibiting P-glycoprotein activity. Mol Pharmacol. 75:1364–1373. 2009.

85 

Zatelli MC, Luchin A, Tagliati F, et al: Cyclooxygenase-2 inhibitors prevent the development of chemoresistance phenotype in a breast cancer cell line by inhibiting glycoprotein p-170 expression. Endocr Relat Cancer. 14:1029–1038. 2007. View Article : Google Scholar : PubMed/NCBI

86 

de Vries EF, Doorduin J, Vellinga NA, van Waarde A, Dierckx RA and Klein HC: Can celecoxib affect P-glycoprotein-mediated drug efflux? A microPET study. Nucl Med Biol. 35:459–466. 2008.PubMed/NCBI

87 

Awara WM, El-Sisi AE, El-Sayad ME and Goda AE: The potential role of cyclooxygenase-2 inhibitors in the treatment of experimentally-induced mammary tumour: does celecoxib enhance the anti-tumour activity of doxorubicin? Pharmacol Res. 50:487–498. 2004. View Article : Google Scholar : PubMed/NCBI

88 

Yan YX, Li WZ, Huang YQ and Liao WX: The COX-2 inhibitor celecoxib enhances the sensitivity of KB/VCR oral cancer cell lines to vincristine by down-regulating P-glycoprotein expression and function. Prostaglandins Other Lipid Mediat. 97:29–35. 2011. View Article : Google Scholar

89 

Zrieki A, Farinotti R and Buyse M: Cyclooxygenase inhibitors down regulate P-glycoprotein in human colorectal Caco-2 cell line. Pharm Res. 25:1991–2001. 2008. View Article : Google Scholar : PubMed/NCBI

90 

Ye CG, Wu WK, Yeung JH, et al: Indomethacin and SC236 enhance the cytotoxicity of doxorubicin in human hepatocellular carcinoma cells via inhibiting P-glycoprotein and MRP1 expression. Cancer Lett. 304:90–96. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Kang HK, Lee E, Pyo H and Lim SJ: Cyclooxygenase-independent down-regulation of multidrug resistance-associated protein-1 expression by celecoxib in human lung cancer cells. Mol Cancer Ther. 4:1358–1363. 2005. View Article : Google Scholar : PubMed/NCBI

92 

Ko SH, Choi GJ, Lee JH, Han YA, Lim SJ and Kim SH: Differential effects of selective cyclooxygenase-2 inhibitors in inhibiting proliferation and induction of apoptosis in oral squamous cell carcinoma. Oncol Rep. 19:425–433. 2008.

93 

Natarajan K, Xie Y, Baer MR and Ross DD: Role of breast cancer resistance protein (BCRP/ABCG2) in cancer drug resistance. Biochem Pharmacol. 83:1084–1103. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Elahian F, Kalalinia F and Behravan J: Evaluation of indomethacin and dexamethasone effects on BCRP-mediated drug resistance in MCF-7 parental and resistant cell lines. Drug Chem Toxicol. 33:113–119. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Chen M, Yu L, Gu C, Zhong D, Wu S and Liu S: Celecoxib antagonizes the cytotoxic effect of cisplatin in human gastric cancer cells by decreasing intracellular cisplatin accumulation. Cancer Lett. 329:189–196. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Sugiura T, Saikawa Y, Kubota T, et al: Combination chemotherapy with JTE-522, a novel selective cyclooxygenase-2 inhibitor, and cisplatin against gastric cancer cell lines in vitro and in vivo. In Vivo. 17:229–233. 2003.

97 

Kim SH, Kim SH, Song YC and Song YS: Celecoxib potentiates the anticancer effect of cisplatin on vulvar cancer cells independently of cyclooxygenase. Ann N Y Acad Sci. 1171:635–641. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Li WZ, Wang XY, Li ZG, Zhang JH and Ding YQ: Celecoxib enhances the inhibitory effect of cisplatin on Tca8113 cells in human tongue squamous cell carcinoma in vivo and in vitro. J Oral Pathol Med. 39:579–584. 2010.PubMed/NCBI

99 

van Wijngaarden J, van Beek E, van Rossum G, et al: Celecoxib enhances doxorubicin-induced cytotoxicity in MDA-MB231 cells by NF-kappaB-mediated increase of intracellular doxorubicin accumulation. Eur J Cancer. 43:433–442. 2007.

100 

Xia W, Zhao T, Lv J, et al: Celecoxib enhanced the sensitivity of cancer cells to anticancer drugs by inhibition of the expression of P-glycoprotein through a COX-2-independent manner. J Cell Biochem. 108:181–194. 2009. View Article : Google Scholar : PubMed/NCBI

101 

Pereira MA, Tao L, Wang W, et al: Modulation by celecoxib and difluoromethylornithine of the methylation of DNA and the estrogen receptor-alpha gene in rat colon tumors. Carcinogenesis. 25:1917–1923. 2004. View Article : Google Scholar : PubMed/NCBI

102 

Ellinger J, Bastian PJ, Jurgan T, et al: CpG island hypermethylation at multiple gene sites in diagnosis and prognosis of prostate cancer. Urology. 71:161–167. 2008. View Article : Google Scholar : PubMed/NCBI

103 

Enokida H, Shiina H, Igawa M, et al: CpG hypermethylation of MDR1 gene contributes to the pathogenesis and progression of human prostate cancer. Cancer Res. 64:5956–5962. 2004. View Article : Google Scholar

104 

Qiu YY, Mirkin BL and Dwivedi RS: MDR1 hypermethylation contributes to the progression of neuroblastoma. Mol Cell Biochem. 301:131–135. 2007. View Article : Google Scholar : PubMed/NCBI

105 

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

106 

Cianchi F, Cortesini C, Magnelli L, et al: Inhibition of 5-lipoxygenase by MK886 augments the antitumor activity of celecoxib in human colon cancer cells. Mol Cancer Ther. 5:2716–2726. 2006. View Article : Google Scholar : PubMed/NCBI

107 

El-Awady RA, Saleh EM, Ezz M and Elsayed AM: Interaction of celecoxib with different anti-cancer drugs is antagonistic in breast but not in other cancer cells. Toxicol Appl Pharmacol. 255:271–286. 2011. View Article : Google Scholar : PubMed/NCBI

108 

Chen C, Shen HL, Yang J, Chen QY and Xu WL: Preventing chemoresistance of human breast cancer cell line, MCF-7 with celecoxib. J Cancer Res Clin Oncol. 137:9–17. 2011. View Article : Google Scholar : PubMed/NCBI

109 

Fosslien E: Molecular pathology of cyclooxygenase-2 in neoplasia. Ann Clin Lab Sci. 30:3–21. 2000.PubMed/NCBI

110 

Bol DK, Rowley RB, Ho CP, et al: Cyclooxygenase-2 overexpression in the skin of transgenic mice results in suppression of tumor development. Cancer Res. 62:2516–2521. 2002.PubMed/NCBI

111 

Nakopoulou L, Mylona E, Papadaki I, et al: Overexpression of cyclooxygenase-2 is associated with a favorable prognostic phenotype in breast carcinoma. Pathobiology. 72:241–249. 2005. View Article : Google Scholar : PubMed/NCBI

112 

Xu Z, Choudhary S, Voznesensky O, et al: Overexpression of COX-2 in human osteosarcoma cells decreases proliferation and increases apoptosis. Cancer Res. 66:6657–6664. 2006. View Article : Google Scholar : PubMed/NCBI

113 

Patel VA, Dunn MJ and Sorokin A: Regulation of MDR-1 (P-glycoprotein) by cyclooxygenase-2. J Biol Chem. 277:38915–38920. 2002. View Article : Google Scholar : PubMed/NCBI

114 

Saikawa Y, Sugiura T, Toriumi F, et al: Cyclooxygenase-2 gene induction causes CDDP resistance in colon cancer cell line, HCT-15. Anticancer Res. 24:2723–2728. 2004.PubMed/NCBI

115 

Surowiak P, Materna V, Matkowski R, et al: Relationship between the expression of cyclooxygenase 2 and MDR1/P-glycoprotein in invasive breast cancers and their prognostic significance. Breast Cancer Res. 7:R862–R870. 2005. View Article : Google Scholar : PubMed/NCBI

116 

Surowiak P, Pawelczyk K, Maciejczyk A, et al: Positive correlation between cyclooxygenase 2 and the expression of ABC transporters in non-small cell lung cancer. Anticancer Res. 28:2967–2974. 2008.PubMed/NCBI

117 

Ziemann C, Schafer D, Rudell G, Kahl GF and Hirsch-Ernst KI: The cyclooxygenase system participates in functional MDR1b overexpression in primary rat hepatocyte cultures. Hepatology. 35:579–588. 2002. View Article : Google Scholar : PubMed/NCBI

118 

Liu B, Qu L and Tao H: Cyclo-oxygenase 2 up-regulates the effect of multidrug resistance. Cell Biol Int. 34:21–25. 2010.PubMed/NCBI

119 

Jendzelovsky R, Mikes J, Koval J, et al: Drug efflux transporters, MRP1 and BCRP, affect the outcome of hypericin-mediated photodynamic therapy in HT-29 adenocarcinoma cells. Photochem Photobiol Sci. 8:1716–1723. 2009. View Article : Google Scholar

120 

Kleban J, Mikes J, Horvath V, et al: Mechanisms involved in the cell cycle and apoptosis of HT-29 cells pre-treated with MK-886 prior to photodynamic therapy with hypericin. J Photochem Photobiol B. 93:108–118. 2008. View Article : Google Scholar : PubMed/NCBI

121 

Kleban J, Mikes J, Szilardiova B, et al: Modulation of hypericin photodynamic therapy by pretreatment with 12 various inhibitors of arachidonic acid metabolism in colon adenocarcinoma HT-29 cells. Photochem Photobiol. 83:1174–1185. 2007. View Article : Google Scholar : PubMed/NCBI

122 

Kleban J, Szilardiova B, Mikes J, et al: Pre-treatment of HT-29 cells with 5-LOX inhibitor (MK-886) induces changes in cell cycle and increases apoptosis after photodynamic therapy with hypericin. J Photochem Photobiol B. 84:79–88. 2006. View Article : Google Scholar

123 

Hida T, Kozaki K, Ito H, et al: Significant growth inhibition of human lung cancer cells both in vitro and in vivo by the combined use of a selective cyclooxygenase 2 inhibitor, JTE-522, and conventional anticancer agents. Clin Cancer Res. 8:2443–2447. 2002.

124 

Hossain MA, Kim DH, Jang JY, et al: Aspiri. induces apoptosis in vitro and inhibits tumor growth of human hepatocellular carcinoma cells in a nude mouse xenograft model. Int J Oncol. 40:1298–1304. 2012.PubMed/NCBI

125 

Irie T, Tsujii M, Tsuji S, et al: Synergistic antitumor effects of celecoxib with 5-fluorouracil depend on IFN-gamma. Int J Cancer. 121:878–883. 2007. View Article : Google Scholar : PubMed/NCBI

126 

Knapp DW, Glickman NW, Widmer WR, et al: Cisplatin versus cisplatin combined with piroxicam in a canine model of human invasive urinary bladder cancer. Cancer Chemother Pharmacol. 46:221–226. 2000. View Article : Google Scholar : PubMed/NCBI

127 

Ponthan F, Wickstrom M, Gleissman H, et al: Celecoxib prevents neuroblastoma tumor development and potentiates the effect of chemotherapeutic drugs in vitro and in vivo. Clin Cancer Res. 13:1036–1044. 2007. View Article : Google Scholar : PubMed/NCBI

128 

Spugnini EP, Cardillo I, Verdina A, et al: Piroxicam and cisplatin in a mouse model of peritoneal mesothelioma. Clin Cancer Res. 12:6133–6143. 2006. View Article : Google Scholar : PubMed/NCBI

129 

Zhang DQ, Guo Q, Zhu JH and Chen WC: Increase of cyclooxygenase-2 inhibition with celecoxib combined with 5-FU enhances tumor cell apoptosis and antitumor efficacy in a subcutaneous implantation tumor model of human colon cancer. World J Surg Oncol. 11(16)2013. View Article : Google Scholar

130 

Tsujii M, Kawano S, Tsuji S, Sawaoka H, Hori M and DuBois RN: Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell. 93:705–716. 1998. View Article : Google Scholar : PubMed/NCBI

131 

Leahy KM, Ornberg RL, Wang Y, Zweifel BS, Koki AT and Masferrer JL: Cyclooxygenase-2 inhibition by celecoxib reduces proliferation and induces apoptosis in angiogenic endothelial cells in vivo. Cancer Res. 62:625–631. 2002.PubMed/NCBI

132 

Patel MI, Subbaramaiah K, Du B, et al: Celecoxib inhibits prostate cancer growth: evidence of a cyclooxygenase-2-independent mechanism. Clin Cancer Res. 11:1999–2007. 2005. View Article : Google Scholar : PubMed/NCBI

133 

Sakamoto T, Uozaki H, Kondo K, et al: Cyclooxygenase-2 regulates the degree of apoptosis by modulating bcl-2 protein in pleomorphic adenoma and mucoepidermoid carcinoma of the parotid gland. Acta Otolaryngol. 125:191–195. 2005. View Article : Google Scholar : PubMed/NCBI

134 

Tjiu JW, Liao YH, Lin SJ, et al: Cyclooxygenase-2 overexpression in human basal cell carcinoma cell line increases antiapoptosis, angiogenesis, and tumorigenesis. J Invest Dermatol. 126:1143–1151. 2006. View Article : Google Scholar : PubMed/NCBI

135 

Wang F, Sun GP, Zou YF, et al: Expression of COX-2 and Bcl-2 in primary fallopian tube carcinoma: correlations with clinicopathologic features. Folia Histochem Cytobiol. 49:389–397. 2011. View Article : Google Scholar : PubMed/NCBI

136 

Stark LA, Din FV, Zwacka RM and Dunlop MG: Aspirin-induced activation of the NF-kappaB signaling pathway: a novel mechanism for aspirin-mediated apoptosis in colon cancer cells. FASEB J. 15:1273–1275. 2001.

137 

Park IS, Jo JR, Hong H, et al: Aspirin induces apoptosis in YD-8 human oral squamous carcinoma cells through activation of caspases, down-regulation of Mcl-1, and inactivation of ERK-1/2 and AKT. Toxicol In Vitro. 24:713–720. 2010. View Article : Google Scholar : PubMed/NCBI

138 

Stark LA, Reid K, Sansom OJ, et al: Aspirin activates the NF-kappaB signalling pathway and induces apoptosis in intestinal neoplasia in two in vivo models of human colorectal cancer. Carcinogenesis. 28:968–976. 2007. View Article : Google Scholar

139 

Trifan OC, Durham WF, Salazar VS, et al: Cyclooxygenase-2 inhibition with celecoxib enhances antitumor efficacy and reduces diarrhea side effect of CPT-11. Cancer Res. 62:5778–5784. 2002.PubMed/NCBI

140 

Altorki NK, Port JL, Zhang F, et al: Chemotherapy induces the expression of cyclooxygenase-2 in non-small cell lung cancer. Clin Cancer Res. 11:4191–4197. 2005. View Article : Google Scholar : PubMed/NCBI

141 

Masferrer JL, Leahy KM, Koki AT, et al: Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res. 60:1306–1311. 2000.PubMed/NCBI

142 

Kohne CH, De Greve J, Hartmann JT, et al: Irinotecan combined with infusional 5-fluorouracil/folinic acid or capecitabine plus celecoxib or placebo in the first-line treatment of patients with metastatic colorectal cancer. EORTC study 40015. Ann Oncol. 19:920–926. 2008. View Article : Google Scholar

143 

Maiello E, Giuliani F, Gebbia V, et al: Gruppo Oncologico dell'Italia Meridionale, FOLFIRI with or without celecoxib in advanced colorectal cancer: a randomized phase II study of the Gruppo Oncologico dell'Italia Meridionale (GOIM). Ann Oncol. 17 (Suppl 7):vii55–59. 2006.

144 

Schneider BJ, Kalemkerian GP, Kraut MJ, et al: Phase II study of celecoxib and docetaxel in non-small cell lung cancer (NSCLC) patients with progression after platinum-based therapy. J Thorac Oncol. 3:1454–1459. 2008. View Article : Google Scholar : PubMed/NCBI

145 

Skapek SX, Anderson JR, Hill DA, et al: Safety and efficacy of high-dose tamoxifen and sulindac for desmoid tumor in children: results of a Children's Oncology Group (COG) phase II study. Pediatr Blood Cancer. 60:1108–1112. 2013.PubMed/NCBI

146 

Csiki I, Morrow JD, Sandler A, et al: Targeting cyclooxygenase-2 in recurrent non-small cell lung cancer: a phase II trial of celecoxib and docetaxel. Clin Cancer Res. 11:6634–6640. 2005. View Article : Google Scholar : PubMed/NCBI

147 

Becerra CR, Frenkel EP, Ashfaq R and Gaynor RB: Increased toxicity and lack of efficacy of rofecoxib in combination with chemotherapy for treatment of metastatic colorectal cancer: a phase II study. Int J Cancer. 105:868–872. 2003. View Article : Google Scholar : PubMed/NCBI

148 

Dang CT, Dannenberg AJ, Subbaramaiah K, et al: Phase II study of celecoxib and trastuzumab in metastatic breast cancer patients who have progressed after prior trastuzumab-based treatments. Clin Cancer Res. 10:4062–4067. 2004. View Article : Google Scholar : PubMed/NCBI

149 

Gridelli C, Gallo C, Ceribelli A, et al: Factorial phase III randomised trial of rofecoxib and prolonged constant infusion of gemcitabine in advanced non-small-cell lung cancer: the GEmcitabine-COxib in NSCLC (GECO) study. Lancet Oncol. 8:500–512. 2007. View Article : Google Scholar : PubMed/NCBI

150 

Groen HJ, Sietsma H, Vincent A, et al: Randomized, placebo-controlled phase III study of docetaxel plus carboplatin with celecoxib and cyclooxygenase-2 expression as a biomarker for patients with advanced non-small-cell lung cancer: the NVALT-4 study. J Clin Oncol. 29:4320–4326. 2011. View Article : Google Scholar : PubMed/NCBI

151 

Koch A, Bergman B, Holmberg E, et al Swedish Lung Cancer Study Group: Effect of celecoxib on survival in patients with advanced non-small cell lung cancer: a double blind randomised clinical phase III trial (CYCLUS study) by the Swedish Lung Cancer Study Group. Eur J Cancer. 47:1546–1555. 2011. View Article : Google Scholar : PubMed/NCBI

152 

Altorki NK, Keresztes RS, Port JL, et al: Celecoxib, a selective cyclo-oxygenase-2 inhibitor, enhances the response to preoperative paclitaxel and carboplatin in early-stage non-small-cell lung cancer. J Clin Oncol. 21:2645–2650. 2003. View Article : Google Scholar : PubMed/NCBI

153 

Nugent FW, Mertens WC, Graziano S, et al: Docetaxel and cyclooxygenase-2 inhibition with celecoxib for advanced non-small cell lung cancer progressing after platinum-based chemotherapy: a multicenter phase II trial. Lung Cancer. 48:267–273. 2005. View Article : Google Scholar : PubMed/NCBI

154 

Legge F, Paglia A, D'Asta M, Fuoco G, Scambia G and Ferrandina G: Phase II study of the combination carboplatin plus celecoxib in heavily pre-treated recurrent ovarian cancer patients. BMC Cancer. 11(214)2011. View Article : Google Scholar : PubMed/NCBI

155 

Altorki NK, Christos P, Port JL, et al: Preoperative taxane-based chemotherapy and celecoxib for carcinoma of the esophagus and gastroesophageal junction: results of a phase 2 trial. J Thorac Oncol. 6:1121–1127. 2011. View Article : Google Scholar : PubMed/NCBI

156 

An Y and Ongkeko WM: ABCG2: the key to chemoresistance in cancer stem cells? Expert Opin Drug Metab Toxicol. 5:1529–1542. 2009. View Article : Google Scholar : PubMed/NCBI

157 

Huang WZ, Fu JH, Wang DK, et al: Overexpression of cyclooxygenase-2 is associated with chemoradiotherapy resistance and prognosis in esophageal squamous cell carcinoma patients. Dis Esophagus. 21:679–684. 2008. View Article : Google Scholar : PubMed/NCBI

158 

Robey RW, To KK, Polgar O, et al: ABCG2: a perspective. Adv Drug Deliv Rev. 61:3–13. 2009. View Article : Google Scholar

159 

Szczuraszek K, Materna V, Halon A, et al: Positive correlation between cyclooxygenase-2 and ABC-transporter expression in non-Hodgkin's lymphomas. Oncol Rep. 22:1315–1323. 2009.PubMed/NCBI

160 

Edelman MJ, Watson D, Wang X, et al: Eicosanoid modulation in advanced lung cancer: cyclooxygenase-2 expression is a positive predictive factor for celecoxib + chemotherapy - Cancer and Leukemia Group B Trial 30203. J Clin Oncol. 26:848–855. 2008.PubMed/NCBI

Related Articles

Journal Cover

January-February 2015
Volume 3 Issue 1

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hiľovská L, Jendželovský R and Fedoročko P: Potency of non-steroidal anti-inflammatory drugs in chemotherapy (Review). Mol Clin Oncol 3: 3-12, 2015
APA
Hiľovská, L., Jendželovský, R., & Fedoročko, P. (2015). Potency of non-steroidal anti-inflammatory drugs in chemotherapy (Review). Molecular and Clinical Oncology, 3, 3-12. https://doi.org/10.3892/mco.2014.446
MLA
Hiľovská, L., Jendželovský, R., Fedoročko, P."Potency of non-steroidal anti-inflammatory drugs in chemotherapy (Review)". Molecular and Clinical Oncology 3.1 (2015): 3-12.
Chicago
Hiľovská, L., Jendželovský, R., Fedoročko, P."Potency of non-steroidal anti-inflammatory drugs in chemotherapy (Review)". Molecular and Clinical Oncology 3, no. 1 (2015): 3-12. https://doi.org/10.3892/mco.2014.446