Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jun 10.
Published in final edited form as: Prostaglandins Other Lipid Mediat. 2011 Aug 16;96(0):27–36. doi: 10.1016/j.prostaglandins.2011.08.004

Regulation of Inflammation in Cancer by Eicosanoids

Emily R Greene 1,2, Sui Huang 3, Charles N Serhan 4, Dipak Panigrahy 1,2
PMCID: PMC4051344  NIHMSID: NIHMS322718  PMID: 21864702

Abstract

Inflammation in the tumour microenvironment is now recognized as one of the hallmarks of cancer. Endogenously produced lipid autacoids, locally acting small molecule lipid mediators, play a central role in inflammation and tissue homeostasis, and have recently been implicated in cancer. A well-studied group of autacoid mediators that are the products of arachidonic acid metabolism include: the prostaglandins, leukotrienes, lipoxins and cytochrome P450 (CYP) derived bioactive products. These lipid mediators are collectively referred to as eicosanoids and are generated by distinct enzymatic systems initiated by cyclooxygenase (COX 1 and 2), lipoxygenases (5-LOX, 12-LOX, 15-LOXa, 15-LOXb), and cytochrome P450s, respectively. These pathways are the target of approved drugs for the treatment of inflammation, pain, asthma, allergies, and cardiovascular disorders. Beyond their potent anti-inflammatory and anti-cancer effects, non-steroidal anti-inflammatory drugs (NSAIDs) and COX-2 specific inhibitors have been evaluated in both preclinical tumor models and clinical trials. Eicosanoid biosynthesis and actions can also be directly influenced by nutrients in the diet, as evidenced by the emerging role of omega-3 fatty acids in cancer prevention and treatment. Most research dedicated to using eicosanoids to inhibit tumor-associated inflammation has focused on the COX and LOX pathways. Novel experimental approaches that demonstrate the anti-tumor effects of inhibiting cancer-associated inflammation currently include: eicosanoid receptor antagonism, overexpression of eicosanoid metabolizing enzymes, and the use of endogenous anti-inflammatory lipid mediators. Here we review the actions of eicosanoids on inflammation in the context of tumorigenesis. Eicosanoids may represent a missing link between inflammation and cancer and thus could serve as therapeutic target(s) for inhibiting tumor growth.

Keywords: Eicosanoids, Inflammation, Cancer, Metastasis, Tumor Microenvironment

1. Introduction

Inflammation is now recognized to be a critical component for tumor progression and one of the recently added “hallmarks of cancer” (1, 2). Inflammation is a biological response of vascularized tissues to noxious stimuli, such as chemical irritants or microbial pathogens. Epidemiological and genetic studies support the link between chronic inflammation and tumor progression (3). Inflammation and chronic infection are associated with tumorigenesis in one of every five cancer patients throughout the world (4). People with chronic inflammatory diseases are at increased risk of developing cancer of the respective inflamed tissue (5, 6) indicating that inflammation is, at least in part, cause and not an effect of cancer development. For example, chronic inflammation of the colon (ulcerative colitis) markedly increases the risk of developing colon cancer later in life (7, 8). Conversely, anti-inflammatory drugs decrease the risk of developing certain cancers. For instance, non-steroidal anti-inflammatory drugs (NSAIDs) reduce the risk of developing colon, breast, lung, and prostate cancer by reducing tumor associated-inflammation (9, 10). Selective anti-inflammatory COX-2 inhibitors can also decrease cancer incidence. While experimental evidence supports the causal relationship between inflammation and cancer, the molecular mechanisms and pathways linking inflammation and cancer remain not well understood. Eicosanoid generating enzymes, such as cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX), are over-expressed in several cancers including breast, lung, and pancreas (11). In addition, cancer patients have increased circulating markers of inflammation such as C-reactive protein (CRP), interleukin-6 (IL-6), and soluble tumour necrosis factor (TNF) receptors (12-14). Thus, novel biological therapies which target pro-inflammatory mediators including IL-6 and TNF-α are in clinical trials for cancer therapy (15).

Eicosanoids, including prostaglandins and leukotrienes, are products of local cell type specific arachidonic acid metabolism and can be potent mediators of inflammation (16-18). These lipid mediators play critical roles in diverse physiological and pathological processes, such as pulmonary fibrosis and cancer. The enzyme families cyclooxygenase (COX) and lipoxygenase (LOX) are responsible for the metabolism of arachidonic acid; this metabolism leads to the production of prostaglandins and leukotrienes, respectively. These enzymes are the target of approved drugs for the treatment of pain, inflammation, asthma, and allergies (18). Both of these pathways have been studied and implicated in cancer (11, 19). There is however, another prominent enzymatic pathway for which arachidonic acid is also the substrate, the cytochrome P450 (CYP) system. This eicosanoid pathway consists of two main branches: ω-hydroxylases which converts arachidonic acid into hydroxyeicosatetraenoic acids (HETEs) and epoxygenases which converts it to epoxyeicosatrienoic acids (EETs). The CYP pathway is particularly relevant in tissues that express low COX and LOX activities. This CYP pathway, originally studied in conjunction with inflammatory and cardiovascular disease, appears to also have a role in tumor growth (20). While the COX and LOX-derived eicosanoids have already been intensely studied in tumor biology, investigation of cytochrome P450-derived eicosanoids has focused on their effects on inflammation and cardiovascular functions rather than cancer (16, 18, 20-23).

2. History of Inflammation and Cancer

Treating inflammation may have started around 400 BC with Hippocrates, a Greek physician, who used salicylates in willow tree bark to alleviate pain and fever, to symptoms associated with inflammation (24). In 1863 Rudolph Virchow was the first to notice leukocytes in neoplastic tissues and established the association between inflammation and cancer (3). In 1897 Felix Hoffman, a chemist with Friedrich Bayer & Co. in Germany, patented aspirin (acetyl-salicylic acid). In the late 1800s aspirin was introduced to the market to treat ailments such as arthritis. Much later, in 1971 Sir John Vane discovered the inhibition of prostaglandin biosynthesis as a mechanism of action for aspirin-like drugs (25). Vane was awarded the Nobel Prize for Medicine in 1982 with Bengt Samuelsson and Sune Bergstrom for their work on the identification of prostaglandins and the mechanism of aspirin (26, 27). In the 1990’s two highly selective cyclooxygenase-2 (COX-2) inhibitors, the coxibs (celecoxib and rofecoxib) were developed to treat inflammation and pain. Many studies have now shown that these anti-inflammatory drugs (aspirin and COX-2 inhibitors) suppress tumor growth (11).

Over the past century, the anti-inflammatory mechanisms have focused on suppressing production of pro-inflammatory mediators, enzymes, or pathways. However, a new direction of inflammation research, spearheaded by the Serhan laboratory, has emerged with the discovery of novel autacoids such as lipoxins and resolvins as active, endogenous mediators of resolution (reviewed by Serhan) (28). Lipoxins, anti-inflammatory mediators which signal the resolution of inflammation, were first described by Serhan, Hamberg, and Samuelsson in 1984 (29). Resolvins, initially discovered by the Serhan laboratory, are novel anti-inflammatory and pro-resolving endogenous lipid mediators derived from the omega-3 fatty acids eicosapentaenoic acid (EPA) and docohexaenoic acid (DHA). These potent mediators are biosynthesized by LOX, cytochrome P450, and COX-2 dependent routes. Resolvins can be produced in the presence or absence of aspirin and in the presence of statins (30, 31).

3. Overview of biochemistry and pharmacology of eicosanoids

Eicosanoids consist of several relatively large families derived from polyunsaturated fatty acids, (omega-6 fatty acids or omega-3 fatty acids). The term eicosanoid is derived from the Greek “eicosa” for twenty and is the collective term for oxygenated 20-carbon essential fatty acids. The subscript of a particular eicosanoid represents the number of double bonds; for example arachidonic acid (AA)-derived prostaglandins have two double bonds (e.g. PGE2). Eicosanoids are generated via the oxidation of 20-carbon chain fatty acids (18). The omega-6 family of eicosanoids encompasses the well characterized prostanoids and leukotrienes generated from cyclooxygenases and lipoxygenases (16) (Figure 1A). The omega-3 family of eicosanoids includes: EPA- and DHA-derived resolvins and protectins (32) (Figure 1B). In addition, the omega-3 derived inflammatory exudates mediators appear to be biosynthesized in inflammatory exudates via their direct flow from peripherial blood into evolving exudates (33). Eicosanoids act locally and have a relatively short half-life, ranging from seconds to minutes (34). The mode of action for eicosanoids includes binding to a specific G protein-coupled receptor which has allowed for the development of specific receptor agonists and antagonists (34). Eicosanoids may initiate both the inflammatory response as well as mediate resolution of inflammation (28, 35). The biochemistry and biosynthesis of these diverse types of eicosanoids are out of the scope of this review and we refer to the recent reviews by Serhan (17), Zeldin (16) and others (11). Here we focus on the biological effects of several eicosanoid pathways, studied to date, related to inflammation and cancer.

Figure 1A. Bioactive eicosanoids derived from the omega-6 family.

Figure 1A

Arachidonic acid is metabolized by distinct enzymatic systems initiated by cyclooxygenase (COX 1 and 2), lipoxygenases (5-LOX and 15-LOX), and cytochrome (CYP) P450s. Schematic overview of major mediators and their metabolites (blue); enzymes (black, boxed), biological role (green). Inhibitors (red ovals). HETE, Hydroxyeicosatetraenoic acids; EETs, epoxyeicosatrienoic acids; CYP, cytochrome P450 enzymes. The sEH inhibitor (soluble epoxide hydrolase inhibitors) increase EET levels hence act as agonist of the EET pathway. PGE2, prostaglandin E2; PGI2, prostacyclin; LXA4, lipoxin A4; LTA4, leukotriene A4; 15-epi-LXA4, aspirin-triggered lipoxin A4; DHET, dihydroxyeicosatrienoic acid;15-PDGH, 15-hydroxyprostaglandin dehydrogenase.

Figure 1B. Bioactive eicosanoids derived from the omega-3 family.

Figure 1B

The omega-3 family of eicosanoids includes: eicosapentaenoic acid (EPA)- and docosahexaenoic acid (DHA)-derived resolvins and protectins. EPA and DHA are metabolized by distinct enzymatic systems initiated by cyclooxygenase (COX-2) and lipoxygenases (5-LOX and 15-LOX). Schematic overview of major mediators and their metabolites (blue); enzymes (black, boxed), biological role (green).

3. Inflammation in cancer

Epidemiological data supports that about 20% of cancer deaths are linked to unabated inflammation and chronic infections (3, 36). Examples include inflammatory bowel diseases, leading to colorectal cancer; bronchitis, leading to lung cancer; and prostatitis, leading to prostate cancer (as reviewed by Agarwal) (36). Most precancerous and cancerous tissue have signs of inflammation including innate immune cells, cytokines, and chemokines (37). Many studies support that cancer-associated inflammation stimulates tumor progression (as reviewed by Mantovani) (37). Colon carcinoma and cervical carcinoma are among the best examples of inflammation-associated cancer (37). Coussens et al described, in a seminal publication, a genetically engineered mouse model demonstrating inflammation is critical for carcinogenesis (38). We have previously demonstrated that anti-inflammatory drugs such as PPAR ligands and COX-2 inhibitors can inhibit tumor growth by suppressing inflammation and angiogenesis (39-43).

In 1971 Folkman shifted the focus of cancer research from tumor cells toward stromal cells such as endothelial cells (44). Since then, the contributions of non-cancerous cells to the growth of tumors has extended to non-local cells, including bone-marrow derived macrophages, neutrophils, mast cells, and mesenchymal stem cells, contributing to the invasiveness and metastatic ability of neoplastic epithelial cells (45, 46).

Macrophages in inflammatory diseases, for example in inflammatory bowel disease, are phenotypically different from normal macrophages in that they can secrete many cytokines such as IL-1, IL-6, and TNF-α (47). These pro-inflammatory cytokines can promote metastasis (48) and contribute to tumor growth by inducing chemokines (49). Pro-inflammatory cytokines induce adhesion molecules and metalloproteinases, resulting in tumor invasion and metastasis (50). Pro-inflammatory mediators, including IL-6, TNF-α, and versican, act as macrophage activators to enhance metastatic growth and angiogenesis (51). Based on these and other studies, cancer-related inflammation has been reported to be one of the newest hallmarks of cancer (1). The following cell types can modulate tumor growth, metastasis, and tumor angiogenesis: tumor associated macrophages, neutrophils, myeloid-derived suppressor cells and dendritic cells (3, 52). Inflammation has been shown to have both pro- and anti-tumorigenic activity. Blocking inflammation has been associated with the stimulation of cancer while inflammatory infiltration of lymphocytic/monocytic cells has been shown to inhibit tumor growth (11, 53-55). Therefore, inflammation in the tumor bed can either stimulate or inhibit tumor growth (41, 54, 56). Thus, pharmacological modulation of inflammation in cancer treatments must be evaluated with the notion that inflammation may be a double-edged sword in this field.

It has been recognized that tumor growth is a complex process involving many cell types. The intercellular communication that takes place between these cells is conducted by an array of soluble factors such as: proteinaceous growth factors and chemokines, VEGF, FGF2, TGF-β, TNF-α, IL-1, and oxygen radicals (57). Minimal attention has been paid to small molecule mediators (i.e. lipid autacoids) whose role in cancer has only recently emerged (58). Levels of eicosanoids increase progressively in patients with benign pelvic disease compared to those with epithelial ovarian cancer (58). In another example, endogenous levels of arachidonic acid and certain eicosanoids are elevated in human colorectal cancer in comparison to normal colon mucosae (59).

4. Cyclooxygenase-derived eicosanoids in cancer

Most research on eicosanoids in inflammation and cancer has focused on the COX pathways. The COX pathways produce five major prostanoids (prostaglandin D2, prostaglandin E2, prostaglandin F, prostaglandin I2 and thromboxane A2) which have important roles in tissue homeostasis (60, 61). COX-2 is a critical enzyme in the regulation of inflammation and plays an important role in cancer-associated inflammation, tumor progression, and metastasis (61, 62). The DuBois laboratory was the first to report that COX-2 is up-regulated in human colorectal adenomas and adenocarcinomas (63) and identified a key role for COX-2 in inflammatory bowel disease and colorectal cancer (64). Therefore, COX-2 has been targeted in many cancers including: colon cancer, colorectal cancer, breast cancer, gliomas, prostate cancer, esophageal carcinoma, pancreatic cancer, lung carcinoma, gastric carcinoma, ovarian cancer, Kaposi’s sarcoma and melanoma (36, 65). Cancer patients with tumors having increased COX-2 expression exhibit decreased survival rates (11). The role of COX-2 and PGE2 in inflammation and cancer has been extensively reviewed (12, 36, 62, 64).

COX-2 derived prostaglandins are important in the early phases of the inflammatory response and the resolution of inflammation (66, 67). Aspirin and other NSAIDs inhibit COX enzymes which decrease prostaglandin levels resulting in anti-inflammatory and anti-tumor activity (11). Targeting downstream prostanoids has provided a strategy to inhibit tumor progression (61). The decreased incidence of esophageal, colorectal, bladder, lung, and gastric cancer treated with aspirin and other NSAIDs demonstrates the importance of COX-2 in many cancers (36). While NSAIDs have potent anti-cancer activity clinically, their cardiovascular and gastrointestinal side effects have curtailed their long term use (11, 60).

While elevated levels of pro-carcinogenic prostanglandins are found in several human malignancies, PGE2 is the most common prostaglandin found in different human cancers including colon, lung, breast, and head and neck cancers. The up-regulation of PGE2 is associated with a poor prognosis (11, 68-71). In patients with head and neck cancers there was an inverse correlation between PGE2 levels and tumor stage (71). Serum levels of PGE2 were significantly decreased after successful treatment. Many studies on enzymes and receptors demonstrate that cyclooxygenase-2 (COX)-derived PGE2 promotes colon carcinoma invasion and growth in several mouse models of cancer (72-74). Consistent with the pro-tumorigenic role of PGE2, mice deficient in PGE2 production exhibit decreased tumorigenesis. Genetic deletion of microsomal PGE2 synthase 1 (mPGES-1), which generates PGE2, results in suppression of intestinal cancer growth by 66-95% (75). Mice genetically deficient in phospholipase A2 (PLA2), an enzyme that releases arachidonic acid from the cell membrane, exhibit decreased lung tumorigenesis that may be mediated by decreased prostaglandin synthesis (76).

Another crucial mechanism for the potent pro-tumorigenic activity of PGE2 is the local suppression of the immune responses (49). PGE2 inhibits macrophage, T cell, and natural killer cell activation, resulting in immunosuppressive and pro-tumorigenic activity (77, 78). Increased pro-inflammatory cytokines are required for tumor invasion, angiogenesis, and tumor growth (79, 80). The pro-inflammatory cytokine IL-1β induces the formation of myeloid-derived suppressor cells (MDSC), which are expressed in various cancers and suppress anti-tumor immune responses thereby allowing the proliferation of cancer cells (81). PGE2 promotes tumor progression by inducing MDSC; this provides a therapeutic approach for anti-cancer therapy (82).

In addition to blocking prostaglandin production, another strategy to inhibit experimental tumor growth is overexpression of eicosanoid metabolizing enzymes, such as 15-hydroxyprostaglandin dehydrogenase (15-PGDH), which is responsible for the enzymatic degradation of PGE2. 15-PGDH activity is down-regulated in human colorectal carcinomas, breast cancer, non-small cell lung cancer, gastric cancer, and bladder cancer, suggesting a tumor suppressive role of 15-PGDH (83-88).

The discovery of eicosanoid receptors has allowed for the inhibition of cancer-associated inflammation and tumor growth by inhibiting their activity (11, 61, 89). Mice deficient of PGE2 receptors (EP1, EP2 or EP4) exhibit decreased tumorigenesis (60, 90, 91). Conversely, knockout mice of PGE2 receptor exhibit increased tumorigenesis, suggesting a protective role in cancer (92). In addition to genetic disruption of PGE2 receptors, administration of PGE2 receptor antagonists may inhibit tumor growth or even act as chemo-preventative agents (90, 93, 94). While PGE2 stimulates cancer migration, the PGE2 EP1 and EP4 receptor antagonists inhibited tumor cell migration and tumor metastasis (95, 96). Administration of soluble EP2 has been shown to inhibit osteolytic tumor growth by neutralizing PGE2 cell signaling (97). Anti-EP therapy and anti-COX-2 may have therapeutic efficacy in Kaposi’s Sarcoma by further reducing chronic inflammation (98).

In contrast to PGE2, prostaglandin D2 (PGD2) exhibits both pro- and anti-tumorigenic activity in various experimental models (11, 78). PGD2 exhibits anti-cancer effects as hematopoietic prostaglandin D synthase, which synthesizes PGD2, suppresses intestinal adenomas by 80%. Disruption of the gene for hematopoietic PGD synthase leads to 50% more intestinal adenomas as compared to controls (99). Hematopoietic PGD synthase was detected mainly in macrophages and monocytes of the gut mucosa (99). In contrast treatment of a colitis model in rats with a selective PGD2 receptor (DP1) antagonist suggest that prolonged elevation of PGD2 synthesis after resolution of colitis may contribute to colitis-associated colorectal cancer (100). Prostaglandin D2 and its receptor (DP1) were recently demonstrated to be up-regulated in individuals in long-term remission from ulcerative colitis (101). In contrast patients with active ulcerative colitis demonstrated increased expression of several pro-inflammatory cytokines and PGE2 synthesis (101).

We and others have demonstrated the anti-tumor activity of COX-2 inhibitors in inflammation-associated experimental tumor models including transplantable and genetically engineered models of cancer (36, 43, 102-104). COX-2 inhibitors initially did not exhibit the toxicity associated with aspirin, such as gastrointestinal bleeding. Over a decade ago, celecoxib was shown to significantly reduce adenoma size and number with no toxicity in patients with familial adenomatous polyposis (FAP) (105). Subsequently three major clinical trials demonstrated significant inhibition of adenoma growth in patients at high risk for colorectal carcinoma, all three trials were terminated because of cardiovascular toxicity and death as well as gastrointestinal complications (106-108). COX-2 inhibitors increase cardiovascular risk and are not recommended for patients, especially those with a history of atherosclerotic disease (11, 109, 110). Celecoxib remains approved by the FDA for the treatment of colon and rectal polyps in patients with familial adenomatous polyposis (11). However, the toxicities of COX-2 inhibitors underscore the importance of developing novel classes of anti-inflammatory agents for cancer therapy.

Aspirin has emerged as the best candidate for use in chemoprevention because of its known cardiovascular benefit, safety, and efficacy data (110, 111). Aspirin promotes the formation of antiproliferative aspirin-triggered lipoxins (ATLs, or 15-epi-lipoxin) (112). ATLs were generated by a human lung adenocarcinoma cell line (A549)-neutrophil interactions (17). These endogenous anti-inflammatory aspirin-triggered lipoxins may mediate part of the anti-inflammatory and anti-cancer activity of aspirin (112).

5. Lipoxygenase-derived eicosanoids and cancer

While the cyclooxygenase pathways have been the most extensively studied pathways in eicosanoids related to cancer, the lipoxygenase (LOX) pathways also have an important role in tumor progression and survival (113). The principal lipoxygenases expressed in humans are 5-lipoxygenase (5-LOX), 12-lipoxygense (12-LOX), and 15-lipoxygenase (15-LOX; that exists in at least 2 main forms Type 1 and 2). 5-LOX and 12-LOX stimulate angiogenesis and tumor growth (114-116), 15-LOX-2 has an anti-tumorigenic role while 15-LOX-1 can have both pro-tumorigenic and anti-tumorigenic activity (113). Both leukotrienes and lipoxins have an emerging role in cancer. Indeed, lipoxygenases were recently demonstrated to mediate invasion of intrametastatic lymphatic vessels and lymph node metastasis in ductal mammary carcinoma (117).

The 5-lipoxygenase (5-LOX) pathway is implicated in the development and progression of human cancers. 5-LOX, whose crystal structure was recently identified (118), is a key enzyme in metabolizing arachidonic acid to leukotrienes. 5-LOX can be induced by pro-inflammatory stimuli and is expressed in epithelial cancers including lung, prostate, breast, and colon (113). Hence, 5-LOX inhibitors have been targeted for their chemopreventive effects. Inhibition of 5-LOX activity is shown to block prostate cancer cell proliferation as well as carcinogen-induced lung tumorigenesis (119, 120).

Inflammation-associated cancers express both 5-LOX and COX-2 proteins (121). The combination of a 5-LOX inhibitor and a COX-2 inhibitor is more potent in several tumor models (lung, colon, skin, pancreas, and esophageal cancer) than inhibiting either eicosanoid pathway alone (114, 119, 122-124). This inhibition of tumor growth was accompanied by the downregulation of PGE2 and LTB4 (122). Dual COX/5-LOX inhibitors are being developed and can induce tumor cell apoptosis independent of the arachidonic acid pathway (125, 126).

12-lipoxygenase (LOX), the main human 12-HETE generating enzyme, has also been shown to be involved in both cancer cell proliferation and survival. Inhibition of 12-LOX blocks cell proliferation and induces apoptosis in carcinoma cells. Guo et al recently demonstrated that 12-lipoxygenase is involved in the regulation of ovarian cancer cell growth and survival and is a therapeutic target for ovarian cancer patients (127). These studies show that 12-LOX expression is higher in high-grade serous ovarian carcinoma compared to normal epithelium and inhibition of 12-LOX by siRNA reduced tumor cell growth (127). Another lipoxygenase that contributes to inflammation and subsequent cancer development is 15-lipoxygenase-1 (15-LOX-1). 15-LOX-1 is expressed in cancers such as Hodgkin lymphoma and colorectal cancer and 15-LOX-1 derived metabolites contribute to the tumor-associated inflammation of Hodgkin lymphoma (128, 129).

The lipoxygenase (LOX) pathways convert arachidonic acid to leukotrienes. In contrast to prostaglandins, which act quickly and degraded more rapidly, leukotrienes have a half-life around 4 hours (12). Inflammatory cells including leukocytes, macrophages, and mast cells can synthesize leukotrienes such as leukotriene A4 (LTA4) and leukotriene B4 (LTB4) (130). The levels of LTB4, the main product of the 5-LOX pathway, were higher in prostate cancer samples than pericancerous samples (131). LTB4 can promote the growth of inflammation-induced melanoma (132). Furthermore, a LTB4 receptor antagonist suppressed inflammation-associated tumor growth (132). The LTB4 receptor antagonist, LY293111, inhibited colon cancer tumor growth and induced apoptosis in vitro (133). The combination of the LTB4 receptor antagonist, LY293111, and gemcitabine was most effective on tumor growth and metastases in an orthotopic model of pancreatic cancer (134). However, this combination did not improve progression-free survival in Phase II clinical trial of pancreatic cancer or non-small-cell lung cancer (135). Another pro-inflammatory mediator derived from 5-LOX metabolism of the arachidonic acid cascade is leukotriene D4 (LTD4). LTD4 is overexpressed in several cancers and levels of circulating LTD4 are elevated in patients with hepatocellular carcinoma and chronic hepatitis B (136).

Both 5-HETE and 12-HETE are also products of lipoxygenase and are involved in tumor progression (12). Exogenous 5-HETE can stimulate the proliferation of prostate cancer cells and act as a survival factor (137, 138). These results require relatively high concentrations (at a concentration of 10 μM). Blocking the formation of 5-HETE, by inhibiting 5-lipoxygenase, results in massive apoptosis of human prostate cancer cells (139). 12-HETE also has a critical role in tumor metastasis, including tumor cell motility, tumor cell-vasculature interactions, invasion, and angiogenesis (140). 12-lipoxygenase is widely expressed in tumor cell lines, providing a target for cancer therapy (141). In recent studies, Kerjaschki et al demonstrates the presence of metastatic tumor cells in sentinel lymph nodes of human mammary carcinomas that express 15-lipoxygenase-1 (ALOX15) and 12S-HETE. By inhibiting 15-lipoxygenase-1, lymph node metastasis was suppressed (117). While 5-HETE, 12-HETE, and 20-HETE have mitogenic and pro-tumorigenic activity, HETEs such as 8-HETE and 11-HETE are reported to have anti-mitogenic and anti-tumor activity (142). An antitumorigenic, prodifferentiating, and tumor suppressing role has been identified for 8S-LOX, and its metabolite 8-HETE, in skin carcinogenesis (143). 8-HETE induces growth inhibition in premalignant epithelial cells (144). Levels of 5-HETE and 12-HETE increase progressively in patients with pelvic disease to those with epithelial ovarian cancer while 15-HETE may disrupt cancer progression (58).

Conversely, lipoxins, which are also lipoxygenase-derived eicosanoids, are formed during the resolution of inflammation and are considered to be anti-tumorgenic (29). Lipoxin A4 (LXA4) and lipoxin B4 (LXB4) are the two principal lipoxins generated and have been shown to be potent mediators of the resolution of inflammation. Lipoxins have potent endogenous activity in stimulating monocytes non-phlogistically, i.e. not causing the release of pro-inflammatory mediators or reactive oxygen species (28). Lipoxins stimulate macrophages to phagocytos apoptotic neutrophils (145). Macrophages also appear to be important as they clear up apoptotic neutrophils and cellular debris as part of the resolution of inflammation (28). For more biochemical details please refer to Serhan et al (17). Of interest, inhibitors of the LTA4 hydrolase or soluble epoxide hydrolase lead to an increase in endogenous lipoxin levels (146, 147) that regulate inflammation and could be a useful strategy for certain cancer treatments.

A novel approach to cancer therapy is the administration of endogenous anti-inflammatory lipid mediators such as lipoxins. Lipoxin A4 (LXA4) reduces cell proliferation, inhibits tumor cell invasion, and suppresses experimental tumor growth (112, 148-150). LXA4 exhibits anti-inflammatory actions in human astrocytoma cells by inhibiting pro-inflammatory chemokine IL-8 and the adhesion molecule ICAM-1 (151).

6. Cytochrome P450-derived eicosanoids and cancer

Cytochrome P450 (CYP)-dependent metabolism of arachidonic acid occurs in several tissues including liver, kidney, and the cardiovascular system. The CYP enzymes relevant to arachidonic acid metabolism include two distinct pathways: the ω-hydroxylases and epoxygenases. The ω-hydroxylases of the 4A and 4F gene families of cytochrome P450 (CYP4A and CYP4F) convert arachidonic acid to autacoids such as hydroxyeicosatetraenoic acids (HETEs). The epoxygenase pathway is encoded predominantly by the CYP2C and CYP2J genes and generates epoxyeicosatrienoic acids (EETs), which have demonstrated anti-inflammatory activity (16, 152, 153). EETs are then further metabolized mainly by soluble epoxide hydrolase (sEH) to the dihydroxyeicosatrienoic acids (DHETs), which have traditionally been considered to be less active than EETs (154, 155).

The role of LOX-derived and CYP-derived HETEs in cancer biology has been extensively reviewed by Moreno (142). In 2008, U251 glioblastoma cells were genetically altered (transfected with rat CYP4A1 cDNA) to increase the formation of 20-HETE (156). This stimulated tumor cell proliferation in culture. When these U251 glioblastoma cells were implanted into the brain of rats, a 10-fold increase in tumor volume versus animals receiving mock-transfected U251 cells was observed (156). In contrast to the strategy to increase 20-HETE, Guo et al demonstrated that HET0016 significantly inhibited human U251 glioblastoma cell proliferation in a dose-dependent manner (157). Subsequently, the same group demonstrated that 9L gliosarcoma proliferation and tumor growth in rats were suppressed by HET0016 (158). Systemic administration of HET0016 inhibited the tumor growth of 9L gliosarcomas by 80%, and tumor angiogenesis by roughly 50%. In a separate study, HET0016 and a 20-HETE antagonist (WIT002) both inhibited the proliferation of a renal adenocarcinoma. This cell type expressed CYP4F isoforms and produced 20-HETE (159). Recent studies show that CYP ω-hydroxylase (via CYP4A11 transfection of tumor cells) promoted angiogenesis and metastasis associated with an increase of VEGF and MMP9 in non-small cell lung cancer cells (160). Little is known about 20-HETE in cancer patients. In one study, 12-HETE and 20-HETE concentrations were shown to be elevated in the urine of patients with benign prostatic hypertrophy and prostate cancer patients as compared to normal subjects (161). Further analysis did not establish a correlation between the concentrations of HETEs and prostatic specific antigen (PSA) level, gland size, or tumor grade (161).

Pioneering studies from the Hammock laboratory demonstrate that soluble epoxide hydrolase is a therapeutic target for inflammation (18, 147). In contrast to COX/LOX inhibitors, which directly inhibit the production of pro-inflammatory mediators, soluble epoxide hydrolase inhibitors promote the formation of pro-resolution mediators such as lipoxin A4 (147). Soluble epoxide hydrolase inhibitors, such as 12-(3-adamantan-1-yl-ureido)-dodecanoic acid butyl ester (AUDA-BE), stabilize anti-inflammatory EETs, which indirectly reduce the expression of COX-2, 5-LOX, iNOS, and VCAM-1 (162, 163). AUDA-BE decreased protein expression of COX-2 without altering COX-1 expression and decreased inflammatory eicosanoids such as PGE2 levels in lipopolysaccharide (LPS)-challenged mice (162). When AUDA-BE was used in combination with low doses of indomethacin, celecoxib, or rofecoxib, PGE2 concentrations were reduced with no effects on prostacyclin and thromboxane levels. Thus, soluble epoxide hydrolase inhibitors synergize with NSAIDs and COX-2 inhibitors in suppressing inflammation (162).

Administration of a soluble epoxide hydrolase inhibitor in a mouse colitis model resulted in decreased ulcer incidence (163). Based on the anti-inflammatory effects of soluble epoxide hydrolase inhibitors, one may expect these agents to have anti-cancer effects based solely on their anti-inflammatory activity. However, soluble epoxide hydrolase inhibitors exhibit pro-angiogenic activity (164) since tumor growth is angiogenesis-dependent (165), soluble epoxide hydrolase inhibitors may have a bi-phasic effect on tumor growth.

The expression of sEH, the main metabolizing enzyme of EETs, has been investigated in cancer. The loss of sEH, has been reported in hepatocellular carcinoma and hepatoma cells (166, 167). Enayetallah et al further confirmed that sEH is down-regulated in renal and hepatic tumors, in principle increasing the levels of EETs in the tumor tissue (168). These studies support a potential role for EETs in cancer due to its metabolizing enzyme being down-regulated. However, the expression of sEH has been shown to be up-regulated in seminoma, cholangiocarcinoma, and advanced ovarian cancer when compared to normal tissues or early stage cancer (168). In preliminary studies on human tissues, sEH expression was increased in ulcerative colitis, ulcerative-induced dysplasia, and ulcerative colitis-induced carcinoma (163). Thus, there is no consistent finding in the expression of sEH in tumors. If EETs promotes tumor growth, sEH would be expected to be down-regulated in tumors – which is observed only in certain tumors.

7. Omega-3 fatty acids and cancer

Eicosanoid biosynthesis and actions can be directly influenced by nutrients in the diet, as evidenced by the emerging role of omega-3 fatty acids in cancer prevention and treatment. Dietary fat may contribute to cancer-associated inflammation through abnormal arachidonic acid metabolism (11). A Western-style diet can increase risk of tumorigenesis via macrophage recruitment and subsequent activation of TNF-α, PGE2, NF-κβ and Wnt inflammatory pathways (169). Arachidonic acid can make up to 40% of the fatty acid composition of cancer cell membranes (12). The anti-tumorigenic effects of omega-3 polyunsaturated fatty acids (PUFAs) may be partly mediated by their anti-inflammatory effects (170). Omega-3 PUFAs inhibit carcinogenesis associated with a reduction in pro-inflammatory cytokines (171). FAT-1 transgenic mice, which have increased endogenous omega-3 PUFAs, exhibit decreased tumorigenesis associated with reduced pro-inflammatory factors such as TNF-α (172). The role of omega-3 fatty acids in cancer has been recently reviewed (173, 174)

Resolvins of the E series are novel endogenous anti-inflammatory and pro-resolving lipid mediators derived from the omega-3 polyunsaturated fatty acid eicosapentaenoic acid (EPA) (175). The E-series resolvins include Resolvin E1 (RvE1) and Resolvin E2 (RvE2) (176). Serhan et al first identified resolvins from murine resolving exudates in vivo (30). These resolvins display potent efficacy in treating diseases driven by inflammation. When aspirin is added to EPA, aspirin irreversibly acetylates the COX enzyme, generating COX-2 dependent RvE1 (177). Cytochrome P450 metabolism of EPA can also generate the precursors of resolvins of the E series (178).

Resolvins of the D series are a novel family of endogenous lipid autacoids that are derived from the omega-3 fatty acid docosahexaenoic acid (DHA) by the sequential actions enzymes 5-LOX and 15-LOX or by an acetylated COX-2 (179). Docosahexaenoic acid (DHA) is an omega-3 fatty acid which result in decreased formation of arachidonic acid-derived PGE2 (180). Omega-3 PUFA counter-regulate arachidonic acid-derived eicosanoids in humans, animals, and cells (180). Resolvins have been shown to play a pivotal role in the resolution of inflammation (179). For example, the combination of DHA and a COX-2 inhibitor, two anti-inflammatory agents, resulted in synergistic cytotoxicity in neuroblastoma cells (173). Similarly, the combination of DHA and COX-2 inhibitors inhibited prostate cancer and melanoma cell growth (181, 182).

A new direction of inflammation research, spearheaded by the Serhan laboratory, has emerged with the stimulation of resolution and the discovery of endogenous non-phlogistic chemical mediators of resolution (28). Endogenous anti-inflammatory mechanisms may have a role in carcinogenesis (15, 78). In contract to normal neural cells, neuroblastoma tumor cells do not generate the anti-inflammatory and endogenous lipid mediators, resolvins and protectins (183). The potential role of resolvins and protectins in cancer remains of interest.

8. Outlook

Many classes of drugs which were originally found to modulate inflammation and cardiovascular diseases have been shown to also have substantial effects on cancer growth. This appears to also be the case with eicosanoids, such as prostaglandins. Other eicosanoid mediators of inflammation, including leukotrienes, lipoxins, and HETEs, have received new attention as targets in cancer therapy. Therefore, additional eicosanoid pathways should also be evaluated for their potential impact in specific cancers as new therapeutic approaches. Future studies will be needed to determine if targeting products of the arachidonic acid cascade (using eicosanoid receptor antagonists or manipulating their metabolizing enzymes) or modulating endogenous lipid mediators (leukotrienes or lipoxins) directly can be used to control tumor-associated inflammation in cancer patients. Multiple inflammatory pathways should be targeted; a strategy now commonly used for cancer therapy in order to conquer the inherent resilience of tumor cells to selective single-target therapies. The design of such multi-prong approaches will be facilitated by our rapidly increasing knowledge of the many pathways that underlie the regulation of eicosanoid production in inflammation. On the other hand, we are still in the beginning of unraveling the multifaceted biological activities of eicosanoids in tumor-associated inflammatory processes. Sorting out the various modes of action of eicosanoids and their cellular targets in the tumor microenvironment is crucial to understanding the complex local actions of these lipid mediators in cancer. This will be paramount if the potent biological activities of eicosanoids are to be harnessed for the development of novel cancer therapies.

Highlights.

  1. Inflammation in the tumor stroma is now recognized as a hallmark of cancer.

  2. Eicosanoids may inhibit cancer growth by reducing tumor-associated inflammation.

  3. Eicosanoids may represent a missing link between inflammation and cancer.

  4. Drugs targeting eicosanoid pathways exhibit potent anti-inflammatory effects.

  5. Most research on eicosanoids in cancer has focused on the COX and LOX pathways.

Acknowledgements

We thank Arja Kaipainen and Gabrielle Fredman for suggestions in preparing the manuscript; Danielle Stanton for preparation of the figure. This work was supported by National Cancer Institute grant RO1CA148633-O1A1 (DP).

Abbreviations

AA

arachidonic acid

CYP and P450

cytochrome P450

COX

cyclooxygenase

LOX

lipoxygenase

NSAID

non-steroidal anti-inflammatory drug

CRP

C-reactive protein

IL-6

interleukin-6

TNF

tumour necrosis factor

EET

epoxyeicosatrienoic acid

HETE

hydroxyeicosatetraenoic acid

(EPA)

eicosapentaenoic acid

(DHA)

docosahexaenoic acid

LXA4

lipoxin A4

LTB4

leukotriene B4;

NF-κB

nuclear factor-kappaB

sEH

soluble epoxide hydrolase

DHET

dihydroxyeicosatrienoic acid

PGE2

prostaglandin E2

PGI2

prostacyclin

VEGF

vascular endothelial growth factor

VEGF;FGF-2

fibroblast growth factor-2

TGF-β

transforming growth factor-beta

mPGES-1

microsomal PGE2 synthase 1

PLA2

phospholipase A2

MDSC

myeloid-derived suppressor cells

15-PDGH

15-hydroxyprostaglandin dehydrogenase

EP2 receptor

prostaglandin E2 receptor

IFNγ

interferon-gamma

FDA

Food and Drug Administration

ATL

aspirin-triggered lipoxin

15-epi-LXA4,

aspirin-triggered lipoxin A4

ICAM

intracellular adhesion molecule

iNOS

inducible nitric oxide synthase

VCAM-1

vascular cell adhesion molecule-1

AUDA-BE

12-(3-adamantan-1-yl-ureido)-dodecanoic acid butyl ester

LPS

lipopolysaccharide

RvE1

Resolvin E1

PUFA

polyunsaturated fatty acid

Footnotes

Disclosure of conflict of interest

EG, SH, and DP have no conflict of interest. CNS is inventor on patents assigned to Brigham and Women’s Hospital and Partners HealthCare on the composition of matter, uses, and clinical development of anti-inflammatory and pro-resolving mediators. These include lipoxins and resolvins and related compounds that are licensed for clinical development. The lipoxins are licensed for clinical development. The lipoxins are licensed to Bayer Health Care. The resolvins are licensed to Resolvyx Pharmaceutical. CNS retains founder stock in Resolvyx Pharmaceuticals.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Mantovani A. Cancer: Inflaming metastasis. Nature. 2009;457:36–37. doi: 10.1038/457036b. [DOI] [PubMed] [Google Scholar]
  • 2.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 3.Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539–545. doi: 10.1016/S0140-6736(00)04046-0. [DOI] [PubMed] [Google Scholar]
  • 4.Balkwill F, Mantovani A. Cancer and inflammation: implications for pharmacology and therapeutics. Clin Pharmacol Ther. 2010;87:401–406. doi: 10.1038/clpt.2009.312. [DOI] [PubMed] [Google Scholar]
  • 5.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Vakkila J, Lotze MT. Inflammation and necrosis promote tumour growth. Nat Rev Immunol. 2004;4:641–648. doi: 10.1038/nri1415. [DOI] [PubMed] [Google Scholar]
  • 7.Ekbom A, Helmick C, Zack M, Adami HO. Ulcerative colitis and colorectal cancer. A population-based study. N Engl J Med. 1990;323:1228–1233. doi: 10.1056/NEJM199011013231802. [DOI] [PubMed] [Google Scholar]
  • 8.Kulaylat MN, Dayton MT. Ulcerative colitis and cancer. J Surg Oncol. 2010;101:706–712. doi: 10.1002/jso.21505. [DOI] [PubMed] [Google Scholar]
  • 9.Harris RE, Chlebowski RT, Jackson RD, Frid DJ, Ascenseo JL, Anderson G, Loar A, Rodabough RJ, White E, McTiernan A. Breast cancer and nonsteroidal anti-inflammatory drugs: prospective results from the Women’s Health Initiative. Cancer Res. 2003;63:6096–6101. [PubMed] [Google Scholar]
  • 10.Harris RE. Cyclooxygenase-2 (cox-2) blockade in the chemoprevention of cancers of the colon, breast, prostate, and lung. Inflammopharmacology. 2009;17:55–67. doi: 10.1007/s10787-009-8049-8. [DOI] [PubMed] [Google Scholar]
  • 11.Wang D, Dubois RN. Eicosanoids and cancer. Nat Rev Cancer. 2010;10:181–193. doi: 10.1038/nrc2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wallace JM. Nutritional and botanical modulation of the inflammatory cascade--eicosanoids, cyclooxygenases, and lipoxygenases--as an adjunct in cancer therapy. Integr Cancer Ther. 2002;1:7–37. doi: 10.1177/153473540200100102. discussion 37. [DOI] [PubMed] [Google Scholar]
  • 13.McMillan DC, Sattar N, Talwar D, O’Reilly DS, McArdle CS. Changes in micronutrient concentrations following anti-inflammatory treatment in patients with gastrointestinal cancer. Nutrition. 2000;16:425–428. doi: 10.1016/s0899-9007(00)00270-7. [DOI] [PubMed] [Google Scholar]
  • 14.Barber MD, Fearon KC, Ross JA. Relationship of serum levels of interleukin-6, soluble interleukin-6 receptor and tumour necrosis factor receptors to the acute-phase protein response in advanced pancreatic cancer. Clin Sci (Lond) 1999;96:83–87. [PubMed] [Google Scholar]
  • 15.Lawrence T. Inflammation and cancer: a failure of resolution? Trends Pharmacol Sci. 2007;28:162–165. doi: 10.1016/j.tips.2007.02.003. [DOI] [PubMed] [Google Scholar]
  • 16.Zeldin DC. Epoxygenase pathways of arachidonic acid metabolism. J Biol Chem. 2001;276:36059–36062. doi: 10.1074/jbc.R100030200. [DOI] [PubMed] [Google Scholar]
  • 17.Serhan CN, Haeggstrom JZ, editors. Lipid Mediators in Acute Inflammation and Resolution: Eicosanoids, PAF, Resolvins, and Protectins. Cambridge University Press; 2009. [Google Scholar]
  • 18.Imig JD, Hammock BD. Soluble epoxide hydrolase as a therapeutic target for cardiovascular diseases. Nat Rev Drug Discov. 2009;8:794–805. doi: 10.1038/nrd2875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Krishnamoorthy S, Honn KV. Eicosanoids in tumor progression and metastasis. Subcell Biochem. 2008;49:145–168. doi: 10.1007/978-1-4020-8831-5_6. [DOI] [PubMed] [Google Scholar]
  • 20.Panigrahy D, Kaipainen A, Greene ER, Huang S. Cytochrome P450- derived eicosanoids: the neglected pathway in cancer. Cancer Metastasis Rev. 2010;29:723–735. doi: 10.1007/s10555-010-9264-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Roman RJ. P-450 metabolites of arachidonic acid in the control of cardiovascular function. Physiol Rev. 2002;82:131–185. doi: 10.1152/physrev.00021.2001. [DOI] [PubMed] [Google Scholar]
  • 22.Fleming I. Epoxyeicosatrienoic acids, cell signaling and angiogenesis. Prostaglandins Other Lipid Mediat. 2007;82:60–67. doi: 10.1016/j.prostaglandins.2006.05.003. [DOI] [PubMed] [Google Scholar]
  • 23.Spector AA, Norris AW. Action of epoxyeicosatrienoic acids on cellular function. Am J Physiol Cell Physiol. 2007;292:C996–1012. doi: 10.1152/ajpcell.00402.2006. [DOI] [PubMed] [Google Scholar]
  • 24.Rainsford KD. Anti-inflammatory drugs in the 21st century. Subcell Biochem. 2007;42:3–27. doi: 10.1007/1-4020-5688-5_1. [DOI] [PubMed] [Google Scholar]
  • 25.Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat New Biol. 1971;231:232–235. doi: 10.1038/newbio231232a0. [DOI] [PubMed] [Google Scholar]
  • 26.Vane JR. Adventures and excursions in bioassay: the stepping stones to prostacyclin. In: Wiksell A, editor. Les Prix Nobel: Nobel Prizes, Presentations, Biographies and Lectures. Stockholm: 1982. pp. 181–206. [Google Scholar]
  • 27.Samuelsson B. From studies of biochemical mechanisms to novel biological mediators: prostaglandin endoperoxides, thromboxanes and leukotrienes. In: Wiksell A, editor. Les Prix Nobel: Nobel Prizes, Presentations, Biographies and Lectures. Stockholm: 1982. pp. 153–174. [DOI] [PubMed] [Google Scholar]
  • 28.Serhan CN. The resolution of inflammation: the devil in the flask and in the details. FASEB J. 2011;25:1441–1448. doi: 10.1096/fj.11-0502ufm. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Serhan CN, Hamberg M, Samuelsson B. Lipoxins: novel series of biologically active compounds formed from arachidonic acid in human leukocytes. Proc Natl Acad Sci U S A. 1984;81:5335–5339. doi: 10.1073/pnas.81.17.5335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang N, Gronert K. Novel functional sets of lipid-derived mediators with antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal antiinflammatory drugs and transcellular processing. J Exp Med. 2000;192:1197–1204. doi: 10.1084/jem.192.8.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, Moussignac RL. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med. 2002;196:1025–1037. doi: 10.1084/jem.20020760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Serhan CN, Krishnamoorthy S, Recchiuti A, Chiang N. Novel anti-inflammatory--pro-resolving mediators and their receptors. Curr Top Med Chem. 2011;11:629–647. doi: 10.2174/1568026611109060629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kasuga K, Yang R, Porter TF, Agrawal N, Petasis NA, Irimia D, Toner M, Serhan CN. Rapid appearance of resolvin precursors in inflammatory exudates: novel mechanisms in resolution. J Immunol. 2008;181:8677–8687. doi: 10.4049/jimmunol.181.12.8677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wymann MP, Schneiter R. Lipid signalling in disease. Nat Rev Mol Cell Biol. 2008;9:162–176. doi: 10.1038/nrm2335. [DOI] [PubMed] [Google Scholar]
  • 35.Gilroy DW, Newson J, Sawmynaden P, Willoughby DA, Croxtall JD. A novel role for phospholipase A2 isoforms in the checkpoint control of acute inflammation. FASEB J. 2004;18:489–498. doi: 10.1096/fj.03-0837com. [DOI] [PubMed] [Google Scholar]
  • 36.Agarwal S, Reddy GV, Reddanna P. Eicosanoids in inflammation and cancer: the role of COX-2. Expert Rev Clin Immunol. 2009;5:145–165. doi: 10.1586/1744666X.5.2.145. [DOI] [PubMed] [Google Scholar]
  • 37.Mantovani A. Cancer: inflammation by remote control. Nature. 2005;435:752–753. doi: 10.1038/435752a. [DOI] [PubMed] [Google Scholar]
  • 38.Coussens LM, Tinkle CL, Hanahan D, Werb Z. MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell. 2000;103:481–490. doi: 10.1016/s0092-8674(00)00139-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Panigrahy D, Singer S, Shen LQ, Butterfield CE, Freedman DA, Chen EJ, Moses MA, Kilroy S, Duensing S, Fletcher C, et al. PPARgamma ligands inhibit primary tumor growth and metastasis by inhibiting angiogenesis. J Clin Invest. 2002;110:923–932. doi: 10.1172/JCI15634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Panigrahy D, Kaipainen A, Huang S, Butterfield CE, Barnes CM, Fannon M, Laforme AM, Chaponis DM, Folkman J, Kieran MW. PPARalpha agonist fenofibrate suppresses tumor growth through direct and indirect angiogenesis inhibition. Proc Natl Acad Sci U S A. 2008;105:985–990. doi: 10.1073/pnas.0711281105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Panigrahy D, Kaipainen A, Kieran MW, Huang S. PPARs: A Double-Edged Sword in Cancer Therapy? PPAR Res. 2008;2008:350351. doi: 10.1155/2008/350351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kieran MW, Turner CD, Rubin JB, Chi SN, Zimmerman MA, Chordas C, Klement G, Laforme A, Gordon A, Thomas A, et al. A feasibility trial of antiangiogenic (metronomic) chemotherapy in pediatric patients with recurrent or progressive cancer. J Pediatr Hematol Oncol. 2005;27:573–581. doi: 10.1097/01.mph.0000183863.10792.d4. [DOI] [PubMed] [Google Scholar]
  • 43.Panigrahy D, Kaipainen A, Butterfield C, Chaponis DM, Laforme A, Folkman J, Kieran M. Inhibition of Tumor Angiogenesis by Oral Etoposide. Experimental and Therapeutic Medicine. 2010;1:739–746. doi: 10.3892/etm.2010.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182–1186. doi: 10.1056/NEJM197111182852108. [DOI] [PubMed] [Google Scholar]
  • 45.Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:335–348. doi: 10.1016/j.cell.2005.02.034. [DOI] [PubMed] [Google Scholar]
  • 46.Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9:239–252. doi: 10.1038/nrc2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mahida YR. The key role of macrophages in the immunopathogenesis of inflammatory bowel disease. Inflamm Bowel Dis. 2000;6:21–33. doi: 10.1097/00054725-200002000-00004. [DOI] [PubMed] [Google Scholar]
  • 48.Solinas G, Marchesi F, Garlanda C, Mantovani A, Allavena P. Inflammation-mediated promotion of invasion and metastasis. Cancer Metastasis Rev. 2010;29:243–248. doi: 10.1007/s10555-010-9227-2. [DOI] [PubMed] [Google Scholar]
  • 49.Dinarello CA. The paradox of pro-inflammatory cytokines in cancer. Cancer Metastasis Rev. 2006;25:307–313. doi: 10.1007/s10555-006-9000-8. [DOI] [PubMed] [Google Scholar]
  • 50.Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marme D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood. 1996;87:3336–3343. [PubMed] [Google Scholar]
  • 51.Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, Luo JL, Karin M. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009;457:102–106. doi: 10.1038/nature07623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Noonan DM, De Lerma Barbaro A, Vannini N, Mortara L, Albini A. Inflammation, inflammatory cells and angiogenesis: decisions and indecisions. Cancer Metastasis Rev. 2008;27:31–40. doi: 10.1007/s10555-007-9108-5. [DOI] [PubMed] [Google Scholar]
  • 53.Clevers H. At the crossroads of inflammation and cancer. Cell. 2004;118:671–674. doi: 10.1016/j.cell.2004.09.005. [DOI] [PubMed] [Google Scholar]
  • 54.Aggarwal BB, Shishodia S, Sandur SK, Pandey MK, Sethi G. Inflammation and cancer: how hot is the link? Biochem Pharmacol. 2006;72:1605–1621. doi: 10.1016/j.bcp.2006.06.029. [DOI] [PubMed] [Google Scholar]
  • 55.Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
  • 56.Kaipainen A, Kieran MW, Huang S, Butterfield C, Bielenberg D, Mostoslavsky G, Mulligan R, Folkman J, Panigrahy D. PPARalpha deficiency in inflammatory cells suppresses tumor growth. PLoS One. 2007;2:e260. doi: 10.1371/journal.pone.0000260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ono M. Molecular links between tumor angiogenesis and inflammation: inflammatory stimuli of macrophages and cancer cells as targets for therapeutic strategy. Cancer Sci. 2008;99:1501–1506. doi: 10.1111/j.1349-7006.2008.00853.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Freedman RS, Wang E, Voiculescu S, Patenia R, Bassett RL, Jr., Deavers M, Marincola FM, Yang P, Newman RA. Comparative analysis of peritoneum and tumor eicosanoids and pathways in advanced ovarian cancer. Clin Cancer Res. 2007;13:5736–5744. doi: 10.1158/1078-0432.CCR-07-0583. [DOI] [PubMed] [Google Scholar]
  • 59.Mal M, Koh PK, Cheah PY, Chan EC. Ultra-pressure liquid chromatography/tandem mass spectrometry targeted profiling of arachidonic acid and eicosanoids in human colorectal cancer. Rapid Commun Mass Spectrom. 2011;25:755–764. doi: 10.1002/rcm.4926. [DOI] [PubMed] [Google Scholar]
  • 60.Smyth EM, Grosser T, Wang M, Yu Y, FitzGerald GA. Prostanoids in health and disease. J Lipid Res. 2009;50(Suppl):S423–428. doi: 10.1194/jlr.R800094-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wang MT, Honn KV, Nie D. Cyclooxygenases, prostanoids, and tumor progression. Cancer Metastasis Rev. 2007;26:525–534. doi: 10.1007/s10555-007-9096-5. [DOI] [PubMed] [Google Scholar]
  • 62.Dubois RN, Abramson SB, Crofford L, Gupta RA, Simon LS, Van De Putte LB, Lipsky PE. Cyclooxygenase in biology and disease. FASEB J. 1998;12:1063–1073. [PubMed] [Google Scholar]
  • 63.Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S, DuBois RN. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology. 1994;107:1183–1188. doi: 10.1016/0016-5085(94)90246-1. [DOI] [PubMed] [Google Scholar]
  • 64.Wang D, Dubois RN. The role of COX-2 in intestinal inflammation and colorectal cancer. Oncogene. 2010;29:781–788. doi: 10.1038/onc.2009.421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Howe LR. Inflammation and breast cancer. Cyclooxygenase/prostaglandin signaling and breast cancer. Breast Cancer Res. 2007;9:210. doi: 10.1186/bcr1678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Rajakariar R, Yaqoob MM, Gilroy DW. COX-2 in inflammation and resolution. Mol Interv. 2006;6:199–207. doi: 10.1124/mi.6.4.6. [DOI] [PubMed] [Google Scholar]
  • 67.Ricciotti E, Fitzgerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31:986–1000. doi: 10.1161/ATVBAHA.110.207449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rigas B, Goldman IS, Levine L. Altered eicosanoid levels in human colon cancer. J Lab Clin Med. 1993;122:518–523. [PubMed] [Google Scholar]
  • 69.Wang D, Dubois RN. Cyclooxygenase-2: a potential target in breast cancer. Semin Oncol. 2004;31:64–73. doi: 10.1053/j.seminoncol.2004.01.008. [DOI] [PubMed] [Google Scholar]
  • 70.McLemore TL, Hubbard WC, Litterst CL, Liu MC, Miller S, McMahon NA, Eggleston JC, Boyd MR. Profiles of prostaglandin biosynthesis in normal lung and tumor tissue from lung cancer patients. Cancer Res. 1988;48:3140–3147. [PubMed] [Google Scholar]
  • 71.Hambek M, Baghi M, Wagenblast J, Schmitt J, Baumann H, Knecht R. Inverse correlation between serum PGE2 and T classification in head and neck cancer. Head Neck. 2007;29:244–248. doi: 10.1002/hed.20503. [DOI] [PubMed] [Google Scholar]
  • 72.Wang D, Dubois RN. Prostaglandins and cancer. Gut. 2006;55:115–122. doi: 10.1136/gut.2004.047100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cha YI, DuBois RN. NSAIDs and cancer prevention: targets downstream of COX-2. Annu Rev Med. 2007;58:239–252. doi: 10.1146/annurev.med.57.121304.131253. [DOI] [PubMed] [Google Scholar]
  • 74.Wang D, Wang H, Shi Q, Katkuri S, Walhi W, Desvergne B, Das SK, Dey SK, DuBois RN. Prostaglandin E(2) promotes colorectal adenoma growth via transactivation of the nuclear peroxisome proliferator-activated receptor delta. Cancer Cell. 2004;6:285–295. doi: 10.1016/j.ccr.2004.08.011. [DOI] [PubMed] [Google Scholar]
  • 75.Nakanishi M, Montrose DC, Clark P, Nambiar PR, Belinsky GS, Claffey KP, Xu D, Rosenberg DW. Genetic deletion of mPGES-1 suppresses intestinal tumorigenesis. Cancer Res. 2008;68:3251–3259. doi: 10.1158/0008-5472.CAN-07-6100. [DOI] [PubMed] [Google Scholar]
  • 76.Meyer AM, Dwyer-Nield LD, Hurteau GJ, Keith RL, O’Leary E, You M, Bonventre JV, Nemenoff RA, Malkinson AM. Decreased lung tumorigenesis in mice genetically deficient in cytosolic phospholipase A2. Carcinogenesis. 2004;25:1517–1524. doi: 10.1093/carcin/bgh150. [DOI] [PubMed] [Google Scholar]
  • 77.Walker W, Rotondo D. Prostaglandin E2 is a potent regulator of interleukin-12- and interleukin-18-induced natural killer cell interferon-gamma synthesis. Immunology. 2004;111:298–305. doi: 10.1111/j.1365-2567.2004.01810.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Rodriguez-Vita J, Lawrence T. The resolution of inflammation and cancer. Cytokine Growth Factor Rev. 2010;21:61–65. doi: 10.1016/j.cytogfr.2009.11.006. [DOI] [PubMed] [Google Scholar]
  • 79.Voronov E, Shouval DS, Krelin Y, Cagnano E, Benharroch D, Iwakura Y, Dinarello CA, Apte RN. IL-1 is required for tumor invasiveness and angiogenesis. Proc Natl Acad Sci U S A. 2003;100:2645–2650. doi: 10.1073/pnas.0437939100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Saijo Y, Tanaka M, Miki M, Usui K, Suzuki T, Maemondo M, Hong X, Tazawa R, Kikuchi T, Matsushima K, et al. Proinflammatory cytokine IL-1 beta promotes tumor growth of Lewis lung carcinoma by induction of angiogenic factors: in vivo analysis of tumor-stromal interaction. J Immunol. 2002;169:469–475. doi: 10.4049/jimmunol.169.1.469. [DOI] [PubMed] [Google Scholar]
  • 81.Bunt SK, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S. Inflammation induces myeloid-derived suppressor cells that facilitate tumor progression. J Immunol. 2006;176:284–290. doi: 10.4049/jimmunol.176.1.284. [DOI] [PubMed] [Google Scholar]
  • 82.Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67:4507–4513. doi: 10.1158/0008-5472.CAN-06-4174. [DOI] [PubMed] [Google Scholar]
  • 83.Backlund MG, Mann JR, Holla VR, Buchanan FG, Tai HH, Musiek ES, Milne GL, Katkuri S, DuBois RN. 15-Hydroxyprostaglandin dehydrogenase is down-regulated in colorectal cancer. J Biol Chem. 2005;280:3217–3223. doi: 10.1074/jbc.M411221200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wolf I, O’Kelly J, Rubinek T, Tong M, Nguyen A, Lin BT, Tai HH, Karlan BY, Koeffler HP. 15-hydroxyprostaglandin dehydrogenase is a tumor suppressor of human breast cancer. Cancer Res. 2006;66:7818–7823. doi: 10.1158/0008-5472.CAN-05-4368. [DOI] [PubMed] [Google Scholar]
  • 85.Hughes D, Otani T, Yang P, Newman RA, Yantiss RK, Altorki NK, Port JL, Yan M, Markowitz SD, Mazumdar M, et al. NAD+-dependent 15- hydroxyprostaglandin dehydrogenase regulates levels of bioactive lipids in non-small cell lung cancer. Cancer Prev Res (Phila) 2008;1:241–249. doi: 10.1158/1940-6207.CAPR-08-0055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Thiel A, Ganesan A, Mrena J, Junnila S, Nykanen A, Hemmes A, Tai HH, Monni O, Kokkola A, Haglund C, et al. 15-hydroxyprostaglandin dehydrogenase is down-regulated in gastric cancer. Clin Cancer Res. 2009;15:4572–4580. doi: 10.1158/1078-0432.CCR-08-2518. [DOI] [PubMed] [Google Scholar]
  • 87.Tseng-Rogenski S, Gee J, Ignatoski KW, Kunju LP, Bucheit A, Kintner HJ, Morris D, Tallman C, Evron J, Wood CG, et al. Loss of 15-hydroxyprostaglandin dehydrogenase expression contributes to bladder cancer progression. Am J Pathol. 2010;176:1462–1468. doi: 10.2353/ajpath.2010.090875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tatsuwaki H, Tanigawa T, Watanabe T, Machida H, Okazaki H, Yamagami H, Shiba M, Watanabe K, Tominaga K, Fujiwara Y, et al. Reduction of 15-hydroxyprostaglandin dehydrogenase expression is an independent predictor of poor survival associated with enhanced cell proliferation in gastric adenocarcinoma. Cancer Sci. 2010;101:550–558. doi: 10.1111/j.1349-7006.2009.01390.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wu WK, Sung JJ, Lee CW, Yu J, Cho CH. Cyclooxygenase-2 in tumorigenesis of gastrointestinal cancers: an update on the molecular mechanisms. Cancer Lett. 2010;295:7–16. doi: 10.1016/j.canlet.2010.03.015. [DOI] [PubMed] [Google Scholar]
  • 90.Watanabe K, Kawamori T, Nakatsugi S, Ohta T, Ohuchida S, Yamamoto H, Maruyama T, Kondo K, Ushikubi F, Narumiya S, et al. Role of the prostaglandin E receptor subtype EP1 in colon carcinogenesis. Cancer Res. 1999;59:5093–5096. [PubMed] [Google Scholar]
  • 91.Shoji Y, Takahashi M, Takasuka N, Niho N, Kitamura T, Sato H, Maruyama T, Sugimoto Y, Narumiya S, Sugimura T, et al. Prostaglandin E receptor EP3 deficiency modifies tumor outcome in mouse two-stage skin carcinogenesis. Carcinogenesis. 2005;26:2116–2122. doi: 10.1093/carcin/bgi193. [DOI] [PubMed] [Google Scholar]
  • 92.Shoji Y, Takahashi M, Kitamura T, Watanabe K, Kawamori T, Maruyama T, Sugimoto Y, Negishi M, Narumiya S, Sugimura T, et al. Downregulation of prostaglandin E receptor subtype EP3 during colon cancer development. Gut. 2004;53:1151–1158. doi: 10.1136/gut.2003.028787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Mutoh M, Watanabe K, Kitamura T, Shoji Y, Takahashi M, Kawamori T, Tani K, Kobayashi M, Maruyama T, Kobayashi K, et al. Involvement of prostaglandin E receptor subtype EP(4) in colon carcinogenesis. Cancer Res. 2002;62:28–32. [PubMed] [Google Scholar]
  • 94.Kawamori T, Uchiya N, Nakatsugi S, Watanabe K, Ohuchida S, Yamamoto H, Maruyama T, Kondo K, Sugimura T, Wakabayashi K. Chemopreventive effects of ONO-8711, a selective prostaglandin E receptor EP(1) antagonist, on breast cancer development. Carcinogenesis. 2001;22:2001–2004. doi: 10.1093/carcin/22.12.2001. [DOI] [PubMed] [Google Scholar]
  • 95.Piazuelo E, Jimenez P, Strunk M, Santander S, Garcia A, Esteva F, Lanas A. Effects of selective PGE2 receptor antagonists in esophageal adenocarcinoma cells derived from Barrett’s esophagus. Prostaglandins Other Lipid Mediat. 2006;81:150–161. doi: 10.1016/j.prostaglandins.2006.09.002. [DOI] [PubMed] [Google Scholar]
  • 96.Yang L, Huang Y, Porta R, Yanagisawa K, Gonzalez A, Segi E, Johnson DH, Narumiya S, Carbone DP. Host and direct antitumor effects and profound reduction in tumor metastasis with selective EP4 receptor antagonism. Cancer Res. 2006;66:9665–9672. doi: 10.1158/0008-5472.CAN-06-1271. [DOI] [PubMed] [Google Scholar]
  • 97.Takahashi T, Uehara H, Bando Y, Izumi K. Soluble EP2 neutralizes prostaglandin E2-induced cell signaling and inhibits osteolytic tumor growth. Mol Cancer Ther. 2008;7:2807–2816. doi: 10.1158/1535-7163.MCT-08-0153. [DOI] [PubMed] [Google Scholar]
  • 98.Paul A. George, Sharma-Walia N, Kerur N, White C, Chandran B. Piracy of prostaglandin E2/EP receptor-mediated signaling by Kaposi’s sarcoma-associated herpes virus (HHV-8) for latency gene expression: strategy of a successful pathogen. Cancer Res. 2010;70:3697–3708. doi: 10.1158/0008-5472.CAN-09-3934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Park JM, Kanaoka Y, Eguchi N, Aritake K, Grujic S, Materi AM, Buslon VS, Tippin BL, Kwong AM, Salido E, et al. Hematopoietic prostaglandin D synthase suppresses intestinal adenomas in ApcMin/+ mice. Cancer Res. 2007;67:881–889. doi: 10.1158/0008-5472.CAN-05-3767. [DOI] [PubMed] [Google Scholar]
  • 100.Zamuner SR, Bak AW, Devchand PR, Wallace JL. Predisposition to colorectal cancer in rats with resolved colitis: role of cyclooxygenase-2-derived prostaglandin d2. Am J Pathol. 2005;167:1293–1300. doi: 10.1016/S0002-9440(10)61216-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Vong L, Ferraz JG, Panaccione R, Beck PL, Wallace JL. A pro-resolution mediator, prostaglandin D(2), is specifically up-regulated in individuals in long-term remission from ulcerative colitis. Proc Natl Acad Sci U S A. 2010;107:12023–12027. doi: 10.1073/pnas.1004982107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Oba M, Miwa K, Fujimura T, Harada S, Sasaki S, Oyama K, Ohta T, Hattori T. A selective cyclooxygenase-2 inhibitor prevents inflammation-related squamous cell carcinogenesis of the forestomach via duodenogastric reflux in rats. Cancer. 2009;115:454–464. doi: 10.1002/cncr.23990. [DOI] [PubMed] [Google Scholar]
  • 103.Gupta S, Adhami VM, Subbarayan M, MacLennan GT, Lewin JS, Hafeli UO, Fu P, Mukhtar H. Suppression of prostate carcinogenesis by dietary supplementation of celecoxib in transgenic adenocarcinoma of the mouse prostate model. Cancer Res. 2004;64:3334–3343. doi: 10.1158/0008-5472.can-03-2422. [DOI] [PubMed] [Google Scholar]
  • 104.Kiguchi K, Ruffino L, Kawamoto T, Franco E, Kurakata S, Fujiwara K, Hanai M, Rumi M, DiGiovanni J. Therapeutic effect of CS-706, a specific cyclooxygenase-2 inhibitor, on gallbladder carcinoma in BK5.ErbB-2 mice. Mol Cancer Ther. 2007;6:1709–1717. doi: 10.1158/1535-7163.MCT-07-0015. [DOI] [PubMed] [Google Scholar]
  • 105.Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB, Wakabayashi N, Saunders B, Shen Y, Fujimura T, et al. The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med. 2000;342:1946–1952. doi: 10.1056/NEJM200006293422603. [DOI] [PubMed] [Google Scholar]
  • 106.Bresalier RS, Sandler RS, Quan H, Bolognese JA, Oxenius B, Horgan K, Lines C, Riddell R, Morton D, Lanas A, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med. 2005;352:1092–1102. doi: 10.1056/NEJMoa050493. [DOI] [PubMed] [Google Scholar]
  • 107.Solomon SD, McMurray JJ, Pfeffer MA, Wittes J, Fowler R, Finn P, Anderson WF, Zauber A, Hawk E, Bertagnolli M. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med. 2005;352:1071–1080. doi: 10.1056/NEJMoa050405. [DOI] [PubMed] [Google Scholar]
  • 108.Baron JA, Sandler RS, Bresalier RS, Quan H, Riddell R, Lanas A, Bolognese JA, Oxenius B, Horgan K, Loftus S, et al. A randomized trial of rofecoxib for the chemoprevention of colorectal adenomas. Gastroenterology. 2006;131:1674–1682. doi: 10.1053/j.gastro.2006.08.079. [DOI] [PubMed] [Google Scholar]
  • 109.Funk CD, FitzGerald GA. COX-2 inhibitors and cardiovascular risk. J Cardiovasc Pharmacol. 2007;50:470–479. doi: 10.1097/FJC.0b013e318157f72d. [DOI] [PubMed] [Google Scholar]
  • 110.Cuzick J, Otto F, Baron JA, Brown PH, Burn J, Greenwald P, Jankowski J, La Vecchia C, Meyskens F, Senn HJ, et al. Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention: an international consensus statement. Lancet Oncol. 2009;10:501–507. doi: 10.1016/S1470-2045(09)70035-X. [DOI] [PubMed] [Google Scholar]
  • 111.Rothwell PM, Wilson M, Elwin CE, Norrving B, Algra A, Warlow CP, Meade TW. Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet. 2010;376:1741–1750. doi: 10.1016/S0140-6736(10)61543-7. [DOI] [PubMed] [Google Scholar]
  • 112.Claria J, Lee MH, Serhan CN. Aspirin-triggered lipoxins (15-epi-LX) are generated by the human lung adenocarcinoma cell line (A549)-neutrophil interactions and are potent inhibitors of cell proliferation. Mol Med. 1996;2:583–596. [PMC free article] [PubMed] [Google Scholar]
  • 113.Pidgeon GP, Lysaght J, Krishnamoorthy S, Reynolds JV, O’Byrne K, Nie D, Honn KV. Lipoxygenase metabolism: roles in tumor progression and survival. Cancer Metastasis Rev. 2007;26:503–524. doi: 10.1007/s10555-007-9098-3. [DOI] [PubMed] [Google Scholar]
  • 114.Chen X, Wang S, Wu N, Sood S, Wang P, Jin Z, Beer DG, Giordano TJ, Lin Y, Shih WC, et al. Overexpression of 5-lipoxygenase in rat and human esophageal adenocarcinoma and inhibitory effects of zileuton and celecoxib on carcinogenesis. Clin Cancer Res. 2004;10:6703–6709. doi: 10.1158/1078-0432.CCR-04-0838. [DOI] [PubMed] [Google Scholar]
  • 115.Ye YN, Liu ES, Shin VY, Wu WK, Cho CH. Contributory role of 5-lipoxygenase and its association with angiogenesis in the promotion of inflammation-associated colonic tumorigenesis by cigarette smoking. Toxicology. 2004;203:179–188. doi: 10.1016/j.tox.2004.06.004. [DOI] [PubMed] [Google Scholar]
  • 116.Nie D, Hillman GG, Geddes T, Tang K, Pierson C, Grignon DJ, Honn KV. Platelet-type 12-lipoxygenase in a human prostate carcinoma stimulates angiogenesis and tumor growth. Cancer Res. 1998;58:4047–4051. [PubMed] [Google Scholar]
  • 117.Kerjaschki D, Bago-Horvath Z, Rudas M, Sexl V, Schneckenleithner C, Wolbank S, Bartel G, Krieger S, Kalt R, Hantusch B, et al. Lipoxygenase mediates invasion of intrametastatic lymphatic vessels and propagates lymph node metastasis of human mammary carcinoma xenografts in mouse. J Clin Invest. 2011;121:2000–2012. doi: 10.1172/JCI44751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Gilbert NC, Bartlett SG, Waight MT, Neau DB, Boeglin WE, Brash AR, Newcomer ME. The structure of human 5-lipoxygenase. Science. 2011;331:217–219. doi: 10.1126/science.1197203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Rioux N, Castonguay A. Inhibitors of lipoxygenase: a new class of cancer chemopreventive agents. Carcinogenesis. 1998;19:1393–1400. doi: 10.1093/carcin/19.8.1393. [DOI] [PubMed] [Google Scholar]
  • 120.Gunning WT, Kramer PM, Steele VE, Pereira MA. Chemoprevention by lipoxygenase and leukotriene pathway inhibitors of vinyl carbamate-induced lung tumors in mice. Cancer Res. 2002;62:4199–4201. [PubMed] [Google Scholar]
  • 121.Mobius C, Aust G, Wiedmann M, Wittekind C, Mossner J, Hauss J, Witzigmann H. Prognostic value of eicosanoid pathways in extrahepatic cholangiocarcinoma. Anticancer Res. 2008;28:873–878. [PubMed] [Google Scholar]
  • 122.Ye YN, Wu WK, Shin VY, Bruce IC, Wong BC, Cho CH. Dual inhibition of 5-LOX and COX-2 suppresses colon cancer formation promoted by cigarette smoke. Carcinogenesis. 2005;26:827–834. doi: 10.1093/carcin/bgi012. [DOI] [PubMed] [Google Scholar]
  • 123.Fegn L, Wang Z. Topical chemoprevention of skin cancer in mice, using combined inhibitors of 5-lipoxygenase and cyclo-oxygenase-2. J Laryngol Otol. 2009;123:880–884. doi: 10.1017/S0022215109004617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wenger FA, Kilian M, Bisevac M, Khodadayan C, von Seebach M, Schimke I, Guski H, Muller JM. Effects of Celebrex and Zyflo on liver metastasis and lipidperoxidation in pancreatic cancer in Syrian hamsters. Clin Exp Metastasis. 2002;19:681–687. doi: 10.1023/a:1021387826867. [DOI] [PubMed] [Google Scholar]
  • 125.Menna C, Olivieri F, Catalano A, Procopio A. Lipoxygenase inhibitors for cancer prevention: promises and risks. Curr Pharm Des. 2010;16:725–733. doi: 10.2174/138161210790883822. [DOI] [PubMed] [Google Scholar]
  • 126.Tavolari S, Bonafe M, Marini M, Ferreri C, Bartolini G, Brighenti E, Manara S, Tomasi V, Laufer S, Guarnieri T. Licofelone, a dual COX/5-LOX inhibitor, induces apoptosis in HCA-7 colon cancer cells through the mitochondrial pathway independently from its ability to affect the arachidonic acid cascade. Carcinogenesis. 2008;29:371–380. doi: 10.1093/carcin/bgm265. [DOI] [PubMed] [Google Scholar]
  • 127.Guo AM, Liu X, Al-Wahab Z, Maddippati KR, Ali-Fehmi R, Scicli AG, Munkarah AR. Role of 12-lipoxygenase in regulation of ovarian cancer cell proliferation and survival. Cancer Chemother Pharmacol. 2011 doi: 10.1007/s00280-011-1595-y. [DOI] [PubMed] [Google Scholar]
  • 128.Claesson HE. On the biosynthesis and biological role of eoxins and 15- lipoxygenase-1 in airway inflammation and Hodgkin lymphoma. Prostaglandins Other Lipid Mediat. 2009;89:120–125. doi: 10.1016/j.prostaglandins.2008.12.003. [DOI] [PubMed] [Google Scholar]
  • 129.Ikawa H, Kamitani H, Calvo BF, Foley JF, Eling TE. Expression of 15-lipoxygenase-1 in human colorectal cancer. Cancer Res. 1999;59:360–366. [PubMed] [Google Scholar]
  • 130.Funk CD. Prostaglandins and leukotrienes: advances in eicosanoid biology. Science. 2001;294:1871–1875. doi: 10.1126/science.294.5548.1871. [DOI] [PubMed] [Google Scholar]
  • 131.Larre S, Tran N, Fan C, Hamadeh H, Champigneulles J, Azzouzi R, Cussenot O, Mangin P, Olivier JL. PGE2 and LTB4 tissue levels in benign and cancerous prostates. Prostaglandins Other Lipid Mediat. 2008;87:14–19. doi: 10.1016/j.prostaglandins.2008.05.001. [DOI] [PubMed] [Google Scholar]
  • 132.Bachi AL, Kim FJ, Nonogaki S, Carneiro CR, Lopes JD, Jasiulionis MG, Correa M. Leukotriene B4 creates a favorable microenvironment for murine melanoma growth. Mol Cancer Res. 2009;7:1417–1424. doi: 10.1158/1541-7786.MCR-09-0038. [DOI] [PubMed] [Google Scholar]
  • 133.Hennig R, Ding XZ, Tong WG, Witt RC, Jovanovic BD, Adrian TE. Effect of LY293111 in combination with gemcitabine in colonic cancer. Cancer Lett. 2004;210:41–46. doi: 10.1016/j.canlet.2004.02.023. [DOI] [PubMed] [Google Scholar]
  • 134.Hennig R, Ventura J, Segersvard R, Ward E, Ding XZ, Rao SM, Jovanovic BD, Iwamura T, Talamonti MS, Bell RH, Jr., et al. LY293111 improves efficacy of gemcitabine therapy on pancreatic cancer in a fluorescent orthotopic model in athymic mice. Neoplasia. 2005;7:417–425. doi: 10.1593/neo.04559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Adrian TE, Hennig R, Friess H, Ding X. The Role of PPARgamma Receptors and Leukotriene B(4) Receptors in Mediating the Effects of LY293111 in Pancreatic Cancer. PPAR Res. 2008;2008:827096. doi: 10.1155/2008/827096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Zhou Y, Guo D, Li H, Jie S. Circulating LTD4 in patients with hepatocellular carcinoma. Tumour Biol. 2011;32:139–144. doi: 10.1007/s13277-010-0107-8. [DOI] [PubMed] [Google Scholar]
  • 137.Ghosh J, Myers CE. Arachidonic acid stimulates prostate cancer cell growth: critical role of 5-lipoxygenase. Biochem Biophys Res Commun. 1997;235:418–423. doi: 10.1006/bbrc.1997.6799. [DOI] [PubMed] [Google Scholar]
  • 138.Myers CE, Ghosh J. Lipoxygenase inhibition in prostate cancer. Eur Urol. 1999;35:395–398. doi: 10.1159/000019915. [DOI] [PubMed] [Google Scholar]
  • 139.Ghosh J, Myers CE. Inhibition of arachidonate 5-lipoxygenase triggers massive apoptosis in human prostate cancer cells. Proc Natl Acad Sci U S A. 1998;95:13182–13187. doi: 10.1073/pnas.95.22.13182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Tang K, Honn KV. 12(S)-HETE in cancer metastasis. Adv Exp Med Biol. 1999;447:181–191. doi: 10.1007/978-1-4615-4861-4_17. [DOI] [PubMed] [Google Scholar]
  • 141.Honn KV, Tang DG, Gao X, Butovich IA, Liu B, Timar J, Hagmann W. 12-lipoxygenases and 12(S)-HETE: role in cancer metastasis. Cancer Metastasis Rev. 1994;13:365–396. doi: 10.1007/BF00666105. [DOI] [PubMed] [Google Scholar]
  • 142.Moreno JJ. New aspects of the role of hydroxyeicosatetraenoic acids in cell growth and cancer development. Biochem Pharmacol. 2009;77:1–10. doi: 10.1016/j.bcp.2008.07.033. [DOI] [PubMed] [Google Scholar]
  • 143.Kim E, Rundhaug JE, Benavides F, Yang P, Newman RA, Fischer SM. An antitumorigenic role for murine 8S-lipoxygenase in skin carcinogenesis. Oncogene. 2005;24:1174–1187. doi: 10.1038/sj.onc.1208269. [DOI] [PubMed] [Google Scholar]
  • 144.Schweiger D, Furstenberger G, Krieg P. Inducible expression of 15- lipoxygenase-2 and 8-lipoxygenase inhibits cell growth via common signaling pathways. J Lipid Res. 2007;48:553–564. doi: 10.1194/jlr.M600311-JLR200. [DOI] [PubMed] [Google Scholar]
  • 145.Godson C, Mitchell S, Harvey K, Petasis NA, Hogg N, Brady HR. Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. J Immunol. 2000;164:1663–1667. doi: 10.4049/jimmunol.164.4.1663. [DOI] [PubMed] [Google Scholar]
  • 146.Rao NL, Dunford PJ, Xue X, Jiang X, Lundeen KA, Coles F, Riley JP, Williams KN, Grice CA, Edwards JP, et al. Anti-inflammatory activity of a potent, selective leukotriene A4 hydrolase inhibitor in comparison with the 5-lipoxygenase inhibitor zileuton. J Pharmacol Exp Ther. 2007;321:1154–1160. doi: 10.1124/jpet.106.115436. [DOI] [PubMed] [Google Scholar]
  • 147.Schmelzer KR, Kubala L, Newman JW, Kim IH, Eiserich JP, Hammock BD. Soluble epoxide hydrolase is a therapeutic target for acute inflammation. Proc Natl Acad Sci U S A. 2005;102:9772–9777. doi: 10.1073/pnas.0503279102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Schottelius AJ, Giesen C, Asadullah K, Fierro IM, Colgan SP, Bauman J, Guilford W, Perez HD, Parkinson JF. An aspirin-triggered lipoxin A4 stable analog displays a unique topical anti-inflammatory profile. J Immunol. 2002;169:7063–7070. doi: 10.4049/jimmunol.169.12.7063. [DOI] [PubMed] [Google Scholar]
  • 149.Zhou XY, Li YS, Wu P, Wang HM, Cai ZY, Xu FY, Ye DY. Lipoxin A(4) inhibited hepatocyte growth factor-induced invasion of human hepatoma cells. Hepatol Res. 2009;39:921–930. doi: 10.1111/j.1872-034X.2009.00520.x. [DOI] [PubMed] [Google Scholar]
  • 150.Chen Y, Hao H, He S, Cai L, Li Y, Hu S, Ye D, Hoidal J, Wu P, Chen X. Lipoxin A4 and its analogue suppress the tumor growth of transplanted H22 in mice: the role of antiangiogenesis. Mol Cancer Ther. 2010;9:2164–2174. doi: 10.1158/1535-7163.MCT-10-0173. [DOI] [PubMed] [Google Scholar]
  • 151.Decker Y, McBean G, Godson C. Lipoxin A4 inhibits IL-1beta-induced IL-8 and ICAM-1 expression in 1321N1 human astrocytoma cells. Am J Physiol Cell Physiol. 2009;296:C1420–1427. doi: 10.1152/ajpcell.00380.2008. [DOI] [PubMed] [Google Scholar]
  • 152.Fisslthaler B, Popp R, Kiss L, Potente M, Harder DR, Fleming I, Busse R. Cytochrome P450 2C is an EDHF synthase in coronary arteries. Nature. 1999;401:493–497. doi: 10.1038/46816. [DOI] [PubMed] [Google Scholar]
  • 153.Kaspera R, Totah RA. Epoxyeicosatrienoic acids: formation, metabolism and potential role in tissue physiology and pathophysiology. Expert Opin Drug Metab Toxicol. 2009;5:757–771. doi: 10.1517/17425250902932923. [DOI] [PubMed] [Google Scholar]
  • 154.Zeldin DC, Kobayashi J, Falck JR, Winder BS, Hammock BD, Snapper JR, Capdevila JH. Regio- and enantiofacial selectivity of epoxyeicosatrienoic acid hydration by cytosolic epoxide hydrolase. J Biol Chem. 1993;268:6402–6407. [PubMed] [Google Scholar]
  • 155.Yu Z, Xu F, Huse LM, Morisseau C, Draper AJ, Newman JW, Parker C, Graham L, Engler MM, Hammock BD, et al. Soluble epoxide hydrolase regulates hydrolysis of vasoactive epoxyeicosatrienoic acids. Circ Res. 2000;87:992–998. doi: 10.1161/01.res.87.11.992. [DOI] [PubMed] [Google Scholar]
  • 156.Guo AM, Sheng J, Scicli GM, Arbab AS, Lehman NL, Edwards PA, Falck JR, Roman RJ, Scicli AG. Expression of CYP4A1 in U251 human glioma cell induces hyperproliferative phenotype in vitro and rapidly growing tumors in vivo. J Pharmacol Exp Ther. 2008;327:10–19. doi: 10.1124/jpet.108.140889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Guo M, Roman RJ, Falck JR, Edwards PA, Scicli AG. Human U251 glioma cell proliferation is suppressed by HET0016 [N-hydroxy-N’-(4-butyl-2-methylphenyl)formamidine], a selective inhibitor of CYP4A. J Pharmacol Exp Ther. 2005;315:526–533. doi: 10.1124/jpet.105.088567. [DOI] [PubMed] [Google Scholar]
  • 158.Guo M, Roman RJ, Fenstermacher JD, Brown SL, Falck JR, Arbab AS, Edwards PA, Scicli AG. 9L gliosarcoma cell proliferation and tumor growth in rats are suppressed by N-hydroxy-N’-(4-butyl-2-methylphenol) formamidine (HET0016), a selective inhibitor of CYP4A. J Pharmacol Exp Ther. 2006;317:97–108. doi: 10.1124/jpet.105.097782. [DOI] [PubMed] [Google Scholar]
  • 159.Alexanian A, Rufanova VA, Miller B, Flasch A, Roman RJ, Sorokin A. Down-regulation of 20-HETE synthesis and signaling inhibits renal adenocarcinoma cell proliferation and tumor growth. Anticancer Res. 2009;29:3819–3824. [PMC free article] [PubMed] [Google Scholar]
  • 160.Yu W, Chen L, Yang YQ, Falck JR, Guo AM, Li Y, Yang J. Cytochrome P450 omega-hydroxylase promotes angiogenesis and metastasis by upregulation of VEGF and MMP-9 in non-small cell lung cancer. Cancer Chemother Pharmacol. 2010 doi: 10.1007/s00280-010-1521-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Nithipatikom K, Isbell MA, See WA, Campbell WB. Elevated 12- and 20-hydroxyeicosatetraenoic acid in urine of patients with prostatic diseases. Cancer Lett. 2006;233:219–225. doi: 10.1016/j.canlet.2005.03.025. [DOI] [PubMed] [Google Scholar]
  • 162.Schmelzer KR, Inceoglu B, Kubala L, Kim IH, Jinks SL, Eiserich JP, Hammock BD. Enhancement of antinociception by coadministration of nonsteroidal anti-inflammatory drugs and soluble epoxide hydrolase inhibitors. Proc Natl Acad Sci U S A. 2006;103:13646–13651. doi: 10.1073/pnas.0605908103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Norwood S, Liao J, Hammock BD, Yang GY. Epoxyeicosatrienoic acids and soluble epoxide hydrolase: potential therapeutic targets for inflammation and its induced carcinogenesis. Am J Transl Res. 2010;2:447–457. [PMC free article] [PubMed] [Google Scholar]
  • 164.Pozzi A, Macias-Perez I, Abair T, Wei S, Su Y, Zent R, Falck JR, Capdevila JH. Characterization of 5,6- and 8,9-epoxyeicosatrienoic acids (5,6- and 8,9-EET) as potent in vivo angiogenic lipids. J Biol Chem. 2005;280:27138–27146. doi: 10.1074/jbc.M501730200. [DOI] [PubMed] [Google Scholar]
  • 165.Folkman J. What is the evidence that tumors are angiogenesis-dependent? J. Natl. Cancer Inst. 1990;82:4–6. doi: 10.1093/jnci/82.1.4. [DOI] [PubMed] [Google Scholar]
  • 166.Yang MD, Wu CC, Chiou SH, Chiu CF, Lin TY, Chiang IP, Chow KC. Reduction of dihydrodiol dehydrogenase expression in resected hepatocellular carcinoma. Oncol Rep. 2003;10:271–276. [PubMed] [Google Scholar]
  • 167.Roques M, Bagrel D, Magdalou J, Siest G. Expression of arylhydrocarbon hydroxylase, epoxide hydrolases, glutathione S-transferase and UDP-glucuronosyltransferases in H5-6 hepatoma cells. Gen Pharmacol. 1991;22:677–684. doi: 10.1016/0306-3623(91)90077-j. [DOI] [PubMed] [Google Scholar]
  • 168.Enayetallah AE, French RA, Grant DF. Distribution of soluble epoxide hydrolase, cytochrome P450 2C8, 2C9 and 2J2 in human malignant neoplasms. J Mol Histol. 2006;37:133–141. doi: 10.1007/s10735-006-9050-9. [DOI] [PubMed] [Google Scholar]
  • 169.Kim IW, Myung SJ, Do MY, Ryu YM, Kim MJ, Do EJ, Park S, Yoon SM, Ye BD, Byeon JS, et al. Western-style diets induce macrophage infiltration and contribute to colitis-associated carcinogenesis. J Gastroenterol Hepatol. 2010;25:1785–1794. doi: 10.1111/j.1440-1746.2010.06332.x. [DOI] [PubMed] [Google Scholar]
  • 170.Jia Q, Lupton JR, Smith R, Weeks BR, Callaway E, Davidson LA, Kim W, Fan YY, Yang P, Newman RA, et al. Reduced colitis-associated colon cancer in Fat-1 (n-3 fatty acid desaturase) transgenic mice. Cancer Res. 2008;68:3985–3991. doi: 10.1158/0008-5472.CAN-07-6251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Lou YR, Peng QY, Li T, Medvecky CM, Lin Y, Shih WJ, Conney AH, Shapses S, Wagner GC, Lu YP. Effects of high fat diets rich in either omega-3 or omega-6 fatty acids on UVB-induced skin carcinogenesis in SKH-1 mice. Carcinogenesis. 2011 doi: 10.1093/carcin/bgr074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Weylandt KH, Krause LF, Gomolka B, Chiu CY, Bilal S, Nadolny A, Waechter SF, Fischer A, Rothe M, Kang JX. Suppressed liver tumorigenesis in fat-1 mice with elevated omega-3 fatty acids is associated with increased omega-3 derived lipid mediators and reduced TNF-{alpha} Carcinogenesis. 2011 doi: 10.1093/carcin/bgr049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Gleissman H, Johnsen JI, Kogner P. Omega-3 fatty acids in cancer, the protectors of good and the killers of evil? Exp Cell Res. 2010 doi: 10.1016/j.yexcr.2010.02.039. [DOI] [PubMed] [Google Scholar]
  • 174.Cockbain AJ, Toogood GJ, Hull MA. Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer. Gut. 2011 doi: 10.1136/gut.2010.233718. [DOI] [PubMed] [Google Scholar]
  • 175.Serhan CN. Novel Lipid Mediators and Resolution Mechanisms in Acute Inflammation. To Resolve or Not? Am J Pathol. 2010 doi: 10.2353/ajpath.2010.100322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Levy BD. Resolvins and protectins: natural pharmacophores for resolution biology. Prostaglandins Leukot Essent Fatty Acids. 2010;82:327–332. doi: 10.1016/j.plefa.2010.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Serhan CN, Arita M, Hong S, Gotlinger K. Resolvins, docosatrienes, and neuroprotectins, novel omega-3-derived mediators, and their endogenous aspirin-triggered epimers. Lipids. 2004;39:1125–1132. doi: 10.1007/s11745-004-1339-7. [DOI] [PubMed] [Google Scholar]
  • 178.Arita M, Clish CB, Serhan CN. The contributions of aspirin and microbial oxygenase to the biosynthesis of anti-inflammatory resolvins: novel oxygenase products from omega-3 polyunsaturated fatty acids. Biochem Biophys Res Commun. 2005;338:149–157. doi: 10.1016/j.bbrc.2005.07.181. [DOI] [PubMed] [Google Scholar]
  • 179.Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. 2008;8:349–361. doi: 10.1038/nri2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Gleissman H, Johnsen JI, Kogner P. Omega-3 fatty acids in cancer, the protectors of good and the killers of evil? Exp Cell Res. 2010;316:1365–1373. doi: 10.1016/j.yexcr.2010.02.039. [DOI] [PubMed] [Google Scholar]
  • 181.Narayanan NK, Narayanan BA, Reddy BS. A combination of docosahexaenoic acid and celecoxib prevents prostate cancer cell growth in vitro and is associated with modulation of nuclear factor-kappaB, and steroid hormone receptors. Int J Oncol. 2005;26:785–792. [PubMed] [Google Scholar]
  • 182.Chiu LC, Tong KF, Ooi VE. Cytostatic and cytotoxic effects of cyclooxygenase inhibitors and their synergy with docosahexaenoic acid on the growth of human skin melanoma A-375 cells. Biomed Pharmacother. 2005;59(Suppl 2):S293–297. doi: 10.1016/s0753-3322(05)80049-6. [DOI] [PubMed] [Google Scholar]
  • 183.Gleissman H, Yang R, Martinod K, Lindskog M, Serhan CN, Johnsen JI, Kogner P. Docosahexaenoic acid metabolome in neural tumors: identification of cytotoxic intermediates. FASEB J. 2010;24:906–915. doi: 10.1096/fj.09-137919. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES