Sie sind auf Seite 1von 17

Received: 12 August 2018 | Accepted: 22 August 2018

DOI: 10.1002/jcp.27411

REVIEW ARTICLE

Cyclooxygenase‐2 in cancer: A review

Nasser Hashemi Goradel1 | Masoud Najafi2 | Eniseh Salehi3 | Bagher Farhood4 |


Keywan Mortezaee5

1
Department of Medical Biotechnology,
School of Advanced Technologies in Medicine, Abstract
Tehran University of Medical Sciences, Cyclooxygenase‐2 (COX‐2) is frequently expressed in many types of cancers exerting
Tehran, Iran
2 a pleiotropic and multifaceted role in genesis or promotion of carcinogenesis and
Radiology and Nuclear Medicine Department,
School of Paramedical Sciences, Kermanshah cancer cell resistance to chemo‐ and radiotherapy. COX‐2 is released by cancer‐
University of Medical Sciences,
associated fibroblasts (CAFs), macrophage type 2 (M2) cells, and cancer cells to the
Kermanshah, Iran
3
Department of Anatomy, School of Medicine, tumor microenvironment (TME). COX‐2 induces cancer stem cell (CSC)‐like activity,
Tehran University of Medical Sciences, and promotes apoptotic resistance, proliferation, angiogenesis, inflammation, inva-
Tehran, Iran
4
sion, and metastasis of cancer cells. COX‐2 mediated hypoxia within the TME along
Departments of Medical Physics and
Radiology, Faculty of Paramedical Sciences, with its positive interactions with YAP1 and antiapoptotic mediators are all in favor
Kashan University of Medical Sciences,
of cancer cell resistance to chemotherapeutic drugs. COX‐2 exerts most of the
Kashan, Iran
5
Department of Anatomy, School of Medicine,
functions through its metabolite prostaglandin E2. In some and limited situations,
Kurdistan University of Medical Sciences, COX‐2 may act as an antitumor enzyme. Multiple signals are contributed to the
Sanandaj, Iran
functions of COX‐2 on cancer cells or its regulation. Members of mitogen‐activated
Correspondence protein kinase (MAPK) family, epidermal growth factor receptor (EGFR), and nuclear
Keywan Mortezaee, Department of Anatomy,
School of Medicine, Kurdistan University of
factor‐κβ are main upstream modulators for COX‐2 in cancer cells. COX‐2 also has
Medical Sciences, Sanandaj 6681853363, Iran. interactions with a number of hormones within the body. Inhibition of COX‐2
Email: keywan987@yahoo.com;
mortezaee.k@muk.ac.ir
provides a high possibility to exert therapeutic outcomes in cancer. Administration of
COX‐2 inhibitors in a preoperative setting could reduce the risk of metastasis in
cancer patients. COX‐2 inhibition also sensitizes cancer cells to treatments like radio‐
and chemotherapy. Chemotherapeutic agents adversely induce COX‐2 activity.
Therefore, choosing an appropriate chemotherapy drugs along with adjustment of the
type and does for COX‐2 inhibitors based on the type of cancer would be an effective
adjuvant strategy for targeting cancer.

KEYWORDS
cancer cell, COX‐2, NF‐κβ, prostaglandin

1 | INTRODUCTION inflammation (H.‐Y. Qiu et al., 2016). COX‐2 is a prostaglandin


(PG)–endoperoxide synthase 2 (X. Xu, Huang, et al., 2018) enzyme
Cyclooxygenases (COXs) have three isoforms: COX‐1, COX‐2, and responsible for generation of prostanoids like prostaglandin E2 (PGE2)
COX‐3 (B. Li, Lu, et al., 2017; Wong et al., 2017). COX‐2 is a that are contributed to the modulation of multiple procarcinogenic
membrane‐bound (W. Xu, Huang, et al., 2018), short‐living (Obermoser effects (Dannenberg, Lippman, Mann, Subbaramaiah, & DuBois, 2005;
et al., 2018), and rate‐limiting enzyme (W. Xu, Huang, et al., 2018) F. Li & Zhu, 2015). COX‐2 expression is negligible in normal cells
discovered in 1991 (Goswami & Sharma‐Walia, 2016) and has long (Gurram et al., 2018) in which its basal expression only occurs within
been known as a target for relief of pain and treatment of stomach (Su, Zhang, & Zhu, 2016), kidney, central nervous system, and

J Cell Physiol. 2018;1-17. wileyonlinelibrary.com/journal/jcp © 2018 Wiley Periodicals, Inc. | 1


2 | HASHEMI ET AL.

in organs of female reproduction (Obermoser et al., 2018). However, it the proteasome in association with endoplasmic reticulum (ER;
is expressed frequently at the tumorigenic nests in most types of Sample, Zhao, Qiang, & He, 2017). Due to existence of many
cancers (Gurram et al., 2018; Raj et al., 2018), including adenocarci- polymorphisms in the COX‐2 gene, susceptibility among individuals
noma, squamous cell carcinoma (SCC), cholangiocarcinoma, transi- to cancer is different (W. Xu, Huang, et al., 2018). Proteins
tional cell carcinoma, endometrial carcinoma (Dannenberg et al., specifically binding to the COX‐2 promotor would take an important
2005), and hepatocellular carcinoma (HCC; Mortezaee, 2018). In fact, role in promotion of tumorigenesis (Xuan et al., 2016). COX‐2
the tumor microenvironment (TME) is an inducer for COX‐2 over- promoter contains a 3‐untranslated region (3‐UTR) that could be a
expression (Ohtsuka et al., 2018). This overexpression is due to target for microRNAs (miRNAs) like miR‐144 for suppression of
dysregulated control over its transcriptional or post‐transcriptional cancer progression (Yao, Gu, Wang, & Xue, 2018). There are AU‐rich
levels (Y. Liu, Borchert, Surazynski, & Phang, 2008), so it could be a elements (AREs) in the 3‐UTR of COX‐2. These AREs could influence
promising marker for identification of the tumor from normal cells messenger RNA (mRNA) decay and protein translation (Dannenberg
(Gurram et al., 2018; Yue, Nguyen, Zellmer, Zhang, & Zorlutuna, 2018). et al., 2005) probably through interaction with Hu antigen‐R (HuR;
COX‐2 is also a constituent of the tumor‐derived exosomes Janakiraman et al., 2017). Stability in the COX‐2 mRNA is regulated
(Theodoraki, Hoffmann, & Whiteside, 2018). Oncogenic viruses by p38 mitogen‐activated protein kinase (MAPK; Hung et al., 2004)
(Charalambous, Maihöfner, Bhambra, Lightfoot, & Gooderham, and K‐ras (Araki et al., 2003).
2003), cancer promotors (Y. Xu, Yang, et al., 2018), radiation (Kirk Promoter of COX‐2 brings sites (response elements) for the
et al., 2018), and chemotherapy (Ikeya, Sakabe, Yamada, Naito, & followings:
Tokura, 2018) and proinflammatory cytokines (H.‐Y. Qiu et al., 2016)
are inducers of COX‐2 expression in cancer cells. COX‐2 over- • Two sites for nuclear factor‐κβ (NF‐κβ; Charalambous et al., 2003)
expression promotes carcinogenesis (Hung et al., 2004), increases the by which this nuclear factor can influence COX‐2 gene transcription
rate of cancer recurrence (Montezuma et al., 2018) and reduces (X. Zheng et al., 2018). P300 and NF‐κβ work as co‐ and
survival (Höing et al., 2018; Sharma et al., 2005; H. Sun et al., 2017) in transactivator for COX‐2 promoter, respectively (Mortezaee,
affected patients, and it is related to cancer cell resistance to chemo‐ 2018). Recruitment of p300 to the COX‐2 promoter is mediated
and radiotherapy (J. Li, Zhou, et al., 2017). by tonicity‐responsive enhancer binding protein (J.H. Lee et al.,
COX‐2 via its pleiotropic effects could evolve a wide range of 2018), while suppression of nuclear localization for this coactivator
activities (Noda et al., 2002). Expression of this rapid response gene is mediated by N‐myc (and STAT) interactor (NMI; J. Wang, Zou,
(Y. Hao et al., 2017) occurs as probably an early event (Charalambous et al., 2017). Factors like thyroid hormone receptor interactor 4
et al., 2003) promoting cancer cell survival, growth, and angiogenesis (TRIP4) have synergy with p300 for inducing COX‐2 promoter
(Noda et al., 2002), and that COX‐2 transgene has been recognized to directly (J. Hao et al., 2018). Oxidative stress is an inducer of the
be a sufficient initiator of cancers like HCC (H. Chen et al., 2017). inflammatory enhanceosome sites on the COX‐2 promoter (J.H. Lee
There is also evidence for the involvement of COX‐2 in cancer et al., 2018).
development (Dong et al., 2018) and invasion (Greenhough et al., • Sites for several cytokines like interleukin (IL)‐6 (J. Wang, Zou,
2009), and its contribution to tumor aggressiveness and poor patient et al., 2017).
prognosis (Esbona et al., 2018). • A site for cyclic AMP response element (CRE). CRE is a target for
The COX‐2 blockade has been shown to provide chemopre- protein binding (CREB) via activation by p38 MAPK, so this site is
ventive for a number of cancers (Harris, Beebe‐Donk, & Alshafie, for transcriptional activation of COX‐2 (Yadav et al., 2018). CRE is
2007), so targeting this enzyme might be a promising approach also a binding site for c‐Jun (Qin et al., 2015).
for cancer therapy. To write this review, we intended to • A site for TCF‐4‐binding element through which activation of COX‐
summarize roles for COX‐2 in cancer focusing on its upstream 2 promoter is mediated by β‐catenin (Araki et al., 2003).
or downstream signaling cascades and targeting for cancer • TEAD binding sites for interaction with factors like YAP
therapeutic approaches. PubMed database was searched for contributed to the regulation of COX‐2 transcription favoring
recent and novel articles in this context. The criteria for article drug resistance in cancer cells (W. Li, Cao, et al., 2017).
selection was based on the novelty, time of publication, quality of • Activator protein‐2 (AP‐2) and specificity protein binding sites
the journals, and number of citations. A total number of 1,100 are responsive to lipopolysaccharide (Wong et al., 2017). AP‐2
papers were scanned for this review with the keywords “COX‐2” is a transactivator for COX‐2 (X. Wang, Yu, et al., 2017;
and “cancer.” Figure 1).

2 | COX ‐2 S TRUCTURE, A CTIVATION, 3 | P Gs AR E I M P O RTA NT TAR GE T S F O R


AN D D EGRA DATION COX ‐2 I N TH E P RO M O TIO N O F CA NC ER

COX‐2 gene is located on the chromosome 1q25.2‐q25.3 in human It is accepted that COX‐2 contribution to carcinogenesis is mediated
subjects (W. Xu, Huang, et al., 2018). Degradation of COX‐2 occurs in through overproduction of PGs. Metabolites of these PGs released
HASHEMI ET AL. | 3

F I G U R E 1 Cyclooxygenase‐2 (COX‐2) promoter. AP‐2: activator protein‐2; ARE: AU‐rich elements; CRE: cyclic AMP response
element; CREB: CRE protein binding; HuR: Hu Antigen‐R; IL: interleukin; mRNA: messenger RNA; NF‐κβ: nuclear factor‐κβ; 3‐UTR,
3‐untranslated region; TBE: Tcf‐4‐binding element; TEAD: TEA domain

into the urine are now considered as approved biomarkers for 4 | CO NTRIBUTION OF
prediction of cancer risk (Cui et al., 2017). PGE2 is a cardinal COX ‐2‐ EX P R ES S I N G T U M O R‐A S S O C IA T E D
mediator of inflammation playing a key role in carcinogenesis (Cui MACROPHAGES (TAMs) TO T UMOR
et al., 2017). There is a positive‐feedback loop between COX‐2 PROGRESSION
expression and PGE2 (D. Wang, Buchanan, Wang, Dey, & DuBois,
2005). The effects of PGE2 are exerted through interaction with its TAMs are part of inflammatory circuits that exhibit antitumor (M1) or
putative transmembrane G‐protein‐coupled EP receptors (EP1–EP4), protumor (M2) features depending on the cytokine concentration in the
especially EP1 and EP4 (Majumder et al., 2018; Majumder, Landman, tumor milieu. COX‐2 is not a specific determinator of macrophage
Liu, Hess, & Lala, 2015; Pan et al., 2016). PGE2 is responsible for polarity toward either type. In human and animal intestinal adenomas, it
induction of COX‐2 expression through activation of ROS/AKT/ has been attested no conspicuous phonotypic alterations in macrophages
activator protein‐1 (E.‐H. Kim et al., 2008), Ras (D. Wang et al., 2005), expressing COX‐2 (Hull et al., 2017). Although it has been asserted that
and Ras subfamily RhoC (Lang et al., 2017). COX‐2 inhibition could make a switch in the TAM phenotype from M2 to
COX‐2 action on PGs especially PGE2 accounts for diverse M1 (Nakanishi et al., 2011), this is not persuasive to come to the
functions in favor of tumor promotion as follows: conclusion of COX‐2 acting as TAM switcher toward M2 cells. A point in
this context is that M1 is a proinflammatory phenotype, while M2 is an
(1) Apoptosis resistance via upregulation of Bcl‐2 (Hosseini et al., anti‐inflammatory cell type (Xiang et al., 2018). Another point is that
2018; Todoric, Antonucci, & Karin, 2016). polarity toward M1 cell type is stimulated by NF‐κβ that influences
(2) Angiogenesis through upregulation of vascular endothelial inflammatory states in macrophages in an active heterodimer form (p50‐
growth factor (VEGF; Hosseini et al., 2018; Todoric et al., 2016). p65; Genard et al., 2018). As it has been discussed, NF‐κβ is a
(3) Metastasis by upregulation of matrix metalloproteinases (MMPs) transactivator for COX‐2 promoter contributed to activation of COX‐2
type 2 and 9 (Hosseini et al., 2018; Todoric et al., 2016) and IL‐11 in both cancer cells (Charalambous et al., 2003) and TAMs (Hung et al.,
(Singh, Berry, Shoher, & Lucci, 2006) along with activation of 2004). There is also evidence for protumoral activity of COX‐2 in
oncogenic miR526b (Majumder et al., 2015). macrophages (Lu Gan et al., 2016). Taken together, it could be speculated
(4) Tumor growth via upregulation of epidermal growth factor that macrophage polarity is not influenced by COX‐2. Rather, COX‐2
receptor (EGFR; Hosseini et al., 2018; Todoric et al., 2016). could act as a promoter of macrophage protumoral activity.
(5) Maintenance of cancer stem cell (CSC)‐like activity via activation Upon irradiation, infiltration of the COX‐2 expressing TAMs to the
of WNT/β‐catenin/TCF (Greenhough et al., 2009) and miR526b tumor milieu occurs. This infiltration promotes tumor growth (Esbona
(Majumder et al., 2015), and suppression of dual specificity et al., 2018), reduces patient survival (Esbona et al., 2018), and
phosphatase 2 (Hou et al., 2017). influences normal epithelial cells around the tumor by relaying
(6) Cancer cell survival (Majumder et al., 2018) and inflammation protumorigenic signals (S.C. Ko et al., 2002). In addition, release of
(Ramu et al., 2018) through interaction with PI3K/AKT. COX‐2 from M2 cells is also contributed to tumor invasion (Tjiu et al.,
(7) Cancer cell invasion via upregulation of extracellular signal‐ 2009), angiogenesis (Rong et al., 2016), and metastasis (Lu Gan et al.,
regulated kinase (ERK; Majumder et al., 2018) and membrane 2016). COX‐2 expression in M2 cells induces protein kinase B (AKT)
proteases (C. Ko et al., 2017). pathway in cancer cells for secretion of MMPs (mediators of invasion
(8) Modulation of the immune system through increase of the and metastasis; Lu Gan et al., 2016; Tjiu et al., 2009). M2 macrophages
activity for regulatory T cells (Tregs; Sharma et al., 2005; via are also able to induce EP1 expression in cancer cells for the
induction of Nr4a expression; Hibino et al., 2018) and the activity promotion of angiogenesis (Rong et al., 2016). In addition, TAMs
for protumor M2 cells (Esbona et al., 2018; Figure 2). express sphingosine kinase‐1 (SphK1)/sphingosine 1‐phosphate (S1P;
4 | HASHEMI ET AL.

F I G U R E 2 Cyclooxygenase‐2 (COX‐2) acting as a procancer enzyme. AKT: protein kinase B; APAF: apoptosis protease‐activating factor;
CSC: cancer stem cell; CYP19: aromatase cytochrome P450; DGLA: dihomo‐γ‐linolenic acid; EGFR: epidermal growth factor receptor; EMT:
epithelial–mesenchymal transition; ERK1: extracellular signal‐regulated kinase‐1; HIF: hypoxia inducible factor; IDO1: indoleamine 2,3‐
dioxygenase 1; LKB1: liver kinase B1; MAPK: mitogen‐activated protein kinase; MMP: matrix metalloproteinases; mTOR: mechanistic target of
rapamycin; NF‐κβ: nuclear factor‐κβ; PG: prostaglandin; PI3K: phosphoinositide 3‐kinase; PKC: protein kinase C; PTEN: phosphatase and tensin
homologue; RLS: reactive lipid species; STAT3: signal transducer and activator of transcription‐3; TCF: T‐cell factor; TGF‐α: tumour growth
factor‐α; TGF‐β: tumour growth factor‐β; VEGF: vascular endothelial growth factor

F I G U R E 3 Interaction between cyclooxygenase‐2 (COX‐2) derived from cancer cells with cells in the tumor microenvironment.
5‐MTP: 5‐methoxytryptophan; ADAM17: A disintegrin and metalloproteinase 17; CAF: cancer‐associated fibroblast; CTL: cytotoxic T
lymphocyte; IFN: interferon; NF‐κβ: nuclear factor‐κβ; PGE2: prostaglandin E2; TEC: tumor endothelial cell; Sp1: specificity protein;
SASP: senescence‐associated secretory phenotype; Treg: regulatory T cell
HASHEMI ET AL. | 5

F I G U R E 4 Signaling pathways or factors contributed to positive or negative regulation of cyclooxygenase‐2 (COX‐2) in cancer cells. AhR:
arylhydrocarbon receptor; AKT: protein kinase B; AMPK: AMP‐activated protein kinase; ANXA5: annexin A; ARNT: aryl hydrocarbon receptor
nuclear translocator; CREB: cyclic AMP response element protein binding; DUSP2: dual specificity phosphatase 2; EGFR: epidermal growth
factor receptor; ER: endoplasmic reticulum; ERK: extracellular signal‐regulated kinase; GATA-4: GATA-binding protein 4; HGF: hepatocyte
growth factor; HIF: hypoxia inducible factor; IL: interleukin; JNK: c‐Jun NH2‐terminal kinase; LKB1: liver kinase B1; m: mutant; MGL:
monoglyceride lipase; NF‐κβ: nuclear factor‐κβ; NMI: N‐myc (and STAT) interactor; PI3K: phosphoinositide 3‐kinase; PKC: protein kinase C;
POX: proline oxidase; PTEN: phosphatase and tensin homologue; ROS: reactive oxygen species; SIRT: sirtuin; Sp1: specificity protein; TRIP4:
thyroid hormone receptor interactor 4; TNF‐α: tumour necrosis factor‐α; UV: ultraviolet; VRCZ: voriconazole; wt: wild type

Furuya et al., 2017) and a disintegrin and metalloproteinase 17 mediated by COX‐2 would remove a possible barrier made by normal
(ADAM17; Bohrer et al., 2016) that are known for their role in fibroblasts against tumor progression. For example, normal fibro-
regulation of COX‐2 expression in cancer cells (Figure 3). blasts release factors like 5‐methoxytryptophan acting as inhibitors
for COX‐2 expression in cancer cells (Cheng et al., 2016; Figure 3).

5 | CONT RIBUTIO N OF
COX ‐2 ‐E X P R E S S I N G C A N C E R ‐A S S O C I A T E D 6 | CO X‐ 2 A ND A UTO P HA GY I N CAN CER
FIBROBLAST S (CAFs) TO TUMORIGE NESIS
Autophagy is a process of waste disposal for degradation of damaged
There is a positive relation between COX‐2 and collagen synthesis in organelles/proteins. A dysfunction (dysregulation) in this process is
tumor cells attested by either pharmacologic inhibition of COX‐2 contributed to tumorigenesis (Mortezaee, 2018). Autophagy‐related
(Esbona et al., 2016; Krishnamachary et al., 2017) or using proline (ATG) gene ATG6 activates COX‐2 in a CREB‐depended mechanism
oxidases (Y. Liu et al., 2008), and that this relation is considered to be (Qiang et al., 2017). COX‐2 is also regulated by p62 (Sample et al.,
a driving force for reducing the time of cancer metastasis (Esbona 2017). P62 is an intracellular ubiquitin‐binding protein responsible
et al., 2018). CAFs are responsible for production of the high amount for the generation of autophagosome after interaction with LC3
of collagen in cancer cells (Krishnamachary et al., 2017). There is a (Mortezaee, 2018; Figure 4).
positive relation between COX‐2 expression with the number of
CAFs (Krishnamachary et al., 2017). CAFs are major producers of
COX‐2 to the TME (Su et al., 2016). COX‐2 release by cancer cells to 7 | CO X‐ 2 A ND O X I D AT IV E S TR ES S
this milieu could also influence CAFs into attaining a senescence‐ IN CA NC ER
associated secretory phenotype (SASP) called senescence CAFs that
are inducers of cancer cell motility (Kabir et al., 2016). Therefore, Reactive oxygen species (ROS) are among COX‐2 derived paracrine
COX‐2 provides a link between cancer cells with CAFs for the mediators for inducing mutation in cells (S.C. Ko et al., 2002).
promotion of cancer, and that increasing in the number of CAFs COX‐2 is involved in peroxidation of lipids (Y. Xu, Yang, et al., 2018)
6 | HASHEMI ET AL.

taking a role for the generation of reactive lipid species (RLS; Wagner, ways for COX‐2‐mediated cancer cell proliferation as follows: (a)
Mullally, & Fitzpatrick, 2006). Cyclopentenone PGs are an example for Induction of aromatase gene activity (aromatase cytochrome P450
these COX‐2‐derived RLS (Higdon, Diers, Oh, Landar, & Darley‐Usmar, [CYP19]; Esbona et al., 2018). Activation of CYP19 is contributed to
2012; Wagner et al., 2006) contributed to inhibition of the tumor‐ estrogen biosynthesis (Harris et al., 2007). COX‐2/PGE2 also induces
suppressing liver kinase B1 (LKB1; Wagner et al., 2006). ROS have two cytochrome P450 1B1 (CYP1B1) that is responsible for conversion of
diverse effects on COX‐2 in cancer cells by being either inducer (E.‐H. the estrogen into estrogen quinones (Esbona et al., 2018) involved in
Kim et al., 2008) or inhibitor (Jeong, Kim, Lee, & Kim, 2017; Y. Liu et al., tumor mitogenic and proliferative features (Harris et al., 2007). (b)
2008) of this enzyme. Induction of COX‐2 by ROS is mediated by Activation of neutrophils. COX‐2 derived from cancer cells is able to
activation of AKT (at Ser473), EGFR (at Tyr1068; J. Lee et al., 2017) and activate neutrophils for the release of elastase relying proliferative
ERK (J. Qiu, Shi, & Jiang, 2017), and inhibition of COX‐2 is mediated by signals on cancer cells (Hattar et al., 2014; Figure 3). (c) Activation of
the stimulatory effect of ROS on AMP‐activated protein kinase stromal CAFs. CAFs also send proliferative signals on cancer cells
(AMPK; Jeong et al., 2017). The ROS/ERK/COX‐2 is contributed to (Hull et al., 2017; Figure 3). Finally, (d) inactivation of adhesion
cancer cell migration and invasion (J. Qiu et al., 2017; Figure 4). molecules involved in regulation of cancer cell proliferation. An
CSCs and bulk cancer cells are vulnerable to alterations in the example for these molecules is E‐cadherin that it makes a complex
intracellular redox state (C. Lu, Laws, Eskandari, & Suntharalingam, with catenin (Noda et al., 2002).
2017) depended on the ROS concentration. Low ROS concentration
promotes cell proliferation via compensatory increase of the activity for
antioxidative enzymes, while high ROS concentration induces mito- 11 | COX ‐2 I S A N I N H I B I T O R
chondrial pathway of apoptosis through inducing release of proapopto- OF APOPTO SIS I N CANCE R CELLS
tic proteins from mitochondria into cytosol (Mortezaee, 2018).
COX‐2 is related to apoptosis suppression in many cancers. COX‐2
causes apoptosis resistance in cancer cells by making a delay in G1
8 | COX ‐2 I S A MEDIATOR OF phase (Gungor, Ilhan, & Eroksuz, 2018), induction of the expressions
I N F L A M M A T IO N I N C A N C E R for Survivin (Krysan, Dalwadi, Sharma, Põld, & Dubinett, 2004), Mcl‐2
(W. Chen et al., 2010) and Bcl‐2 (Hosseini et al., 2018; Todoric et al.,
Inflammation is an inducer of carcinogenesis in which an inflamma-
2016), and repression of caspase‐3 (Janakiraman et al., 2017;
tory microenvironment is considered as a promoter of cancer
Figure 2).
development and invasiveness (Echizen, Oshima, Nakayama, &
The apoptosis mediator wild‐type p53 is a suppressor for COX‐2
Oshima, 2018). COX‐2 is a proinflammatory enzyme (Pollock et al.,
expression (Mortezaee, 2018; Ohtsuka et al., 2018). In cancer cells,
2018) overexpressed at the inflammatory site of cancer (Raj et al.,
however, mutations in the p53 occurs, so a positive‐feedback loop
2018). There is a reciprocal positive feedback between COX‐2 and
between COX‐2 and the mutant p53 is an interesting event that
inflammatory mediators with either one inducing another (Desai,
might be a target for chemotherapeutic agents (Chin et al., 2018;
Prickril, & Rasooly, 2018; Hosseini et al., 2018; Figure 4).
Ohtsuka et al., 2018).

9 | COX ‐2 AN D T U M O R IM M U N I T Y
12 | COX ‐2 I S A M E D I A T O R O F T U M O R
COX‐2 is able to instruct an immune phenotype through inducing
MIGRATION, IN VA SION , AN D META STASIS
immune cell recruitment into the tumor tissue to induce an
COX‐2 has been identified as one of the key metastasis progression
immunosuppressive state in the tumor milieu (Lang et al., 2006) in
genes (Greenhough et al., 2009) involved in the metastasis into
favor of cancer cell activation (Höing et al., 2018). COX‐2/PGE2
lymph nodes (Höing et al., 2018), bone (Singh et al., 2006), liver
released from cancer cells to this milieu suppresses host immuno-
(Sorski et al., 2016), brain (Soto et al., 2016), and so forth. COX‐2
logical responses to tumor‐derived antigens through blockade of the
induces factors like IL‐11 that are related to cancer metastasis
activity for cytotoxic T lymphocytes (Miao et al., 2017). Another way
(Singh et al., 2006). Epithelial–mesenchymal transition (EMT) in
for COX‐2‐mediated immune resistance is through inducing indolea-
cancer cells is a promoter of cancer invasiveness (Mortezaee, 2018).
mine 2,3‐dioxygenase 1 in cancer cells (Hennequart et al., 2017;
COX‐2 induces EMT through increasing the activity for miR526b
Figure 3).
(Majumder et al., 2015) and factors like signal transducer and
activator of transcription‐3 (STAT3; Tong et al., 2017), and that
10 | COX ‐2 I S A ME DIATOR OF CANCER inhibition of COX‐2‐mediated EMT occurs after application of
CE LL GROW TH cannabinoids for cancer (Xian et al., 2016). COX‐2 also induces β1‐
integrin (Pan et al., 2016) and membrane proteases like matripase
There is a strong evidence for the contribution of COX‐2 in the that are responsible for cancer cell invasion (C. Ko et al.,
promotion of cancer cell growth (Raj et al., 2018). There are several 2017; Figure 2).
HASHEMI ET AL. | 7

13 | COX ‐2 I S A M E D IA TO R 15.3 | COX‐2/STAT3


OF AN G I O G E NES I S I N C A N CE R
COX‐2/STAT3 signaling is responsible for the promotion of an
immunosuppressive microenvironment (Maturu et al., 2017), EMT
COX‐2 is expressed in tumor endothelial cells (Hull et al., 2017) and it is
(Tong et al., 2017), and proliferation (Ramu et al., 2018) of cancer
related to an increase in the number of blood vessels in human cancers
cells. This signaling could be a part of the extended NF‐κβ/COX‐2/
(Montezuma et al., 2018) for cancer cell migratory purposes (Rüegg,
BCL‐2/IL‐6/STAT3 pathway (Das et al., 2017; Figure 2).
Dormond, & Mariotti, 2004; Figure 3). COX‐2 inhibitors like
c‐phycocyanin are able to suppress angiogenesis in cancer cells through
docking with VEGF (Jiang et al., 2018). Activation of COX‐2 for the 15.4 | PI3K/AKT/COX‐2
promotion of angiogenesis is induced by caspase‐3 (Feng et al., 2017;
COX‐2 takes a role for activation of PI3K/AKT pathway by
Figure 2).
suppressing phosphatase and tensin homologue (PTEN, an essential
tumor‐suppressing factor) in cancer cells either directly (Chang et al.,
14 | COX ‐2 AS A N I N D UCE R O F C S C 2018) or indirectly through suppression of TET‐1‐induced PTEN
AC TIVITY IN C ANC ER CE LLS activation (H. Chen et al., 2017). MiR‐221/222 is able to block PTEN
resulting in activation of the PI3K/AKT/NF‐κβ/COX‐2 pathway (B. Li,
COX‐2 is identified to be implicated in maintenance of tissue resistance Lu, et al., 2017). Phosphoinositide 3‐kinase (PI3K) acts as both a
CSC populations through promotion of CSC‐like properties including positive (Rundhaug & Fischer, 2008) or negative (W. Liu et al., 2003)
tumorosphere formation, stemness, and metastatic propensity (C. Lu, activator of COX‐2. Activation of COX‐2 by PI3K/AKT is mediated by
Eskandari, Cressey, & Suntharalingam, 2017). Inducible effect of COX‐2 induction of NF‐κβ (B. Li, Lu, et al., 2017; Ramu et al., 2018).
on TGF‐β is related to the enrichment of CSCs (Tian et al., 2017). Interestingly, PI3K also reduces binding activity of COX‐2 for NF‐κβ
Subcellular localization of COX‐2 within mitochondria favors mitochon- (W. Liu et al., 2003; Figures 2 and 4).
drial recruitment (translocation) of p53 that in turn induces the activity
for mitochondrial fission Drp1 for the promotion of stemness (Zhou
et al., 2017). In addition, COX‐2 induces Id1 that serves as a marker of
15.5 | MAPK/COX‐2
CSC self‐renewal (Cook et al., 2016). Members of MAPK family including p38, ERK1/2, and c‐Jun NH2‐
terminal kinase are contributed to induction of COX‐2 (W. Liu et al.,
15 | MECHANISMS FOR C OX ‐2 2003). ERK1/2 (Maturu et al., 2017) and p38 (Hu et al., 2017) are also
CONTRIBUTION TO CANCER downstream targets for COX‐2. These MAPK members are mediator
PROGRE SSION of PKC inducible effects on COX‐2 in both cancer cells (Chou et al.,
2015) and CAFs (F. Li & Zhu, 2015). In addition, MAPK activity
15.1 | Sirtuin (SIRT)/COX‐2 mediates inducible activity of EGFR on COX‐2 (Dannenberg et al.,
2005; Hu et al., 2017) in which a possible interaction between EGFR/
SIRT6 induces COX‐2 in cancer cells by suppressing AMPK (Yadav
p38 with COX‐2 favors angiogenesis in cancer cells (Hu et al., 2017).
et al., 2018). AMPK activation is a downstream target for the tumor
P38/COX‐2 is also involved in cancer cell resistance to apoptosis
suppressor LKB1 (Gao, Ge, & Ji, 2011; Figure 4).
(Semaan et al., 2016; Figure 4).

15.2 | NF‐κβ/COX‐2
15.6 | COX‐2/hypoxia inducible factor (HIF)
NF‐κβ is contributed to activation of COX‐2 in both cancer cells
(Charalambous et al., 2003) and TAMs (Hung et al., 2004). There are HIF‐1α is a marker of angiogenesis in cancer cells (Mortezaee, 2018)
five related NF‐κβ proteins including p50 (NF‐κβ1), p52 (NF‐κβ2), p65 and a target for COX‐2 (Maturu et al., 2017). An existing interplay
(RelA), RelB, and c‐Rel (Mortezaee, 2018), among them p65 has been between COX‐2 and HIF‐1α is contributed to induction of hypoxia
identified for its role in the activation of COX‐2 in cancer cells (Cai within the TME. In the hypoxic conditions, HIF‐1α acts as a positive
et al., 2017; J. Hao et al., 2018). IKK and IkB are upstream signaling inducer for COX‐2 expression (Greenhough et al., 2009). COX‐2/HIF‐
molecules for NF‐κβ acting in its nuclear translocation (Mortezaee, 2α is associated with cancer cell resistance to chemotherapeutic
2018). NF‐κβ/COX‐2 could be a target for chemotherapeutic agents for drugs in hypoxic conditions (Dong et al., 2018; Figure 2).
exerting their anticancer roles (H. Yang, Huang, et al., 2017). NF‐κβ/
COX‐2 signaling is induced by ERK1/2 (Wong et al., 2017), protein
15.7 | Arylhydrocarbon receptor (AhR)/ARNT/
kinase C (PKC; H.‐S. Baek et al., 2017), TRIP4 (J. Hao et al., 2018),
COX‐2
caspase‐3 (Feng et al., 2017), cytokines like interleukin 1β (IL‐1β;
Charalambous et al., 2003), and conditions like ER stress (Hung et al., AhR interaction with its nuclear transporter ARNT is contributed to
2004). Inhibition of this signaling is mediated by miR‐16 (X. Liu, Li, Li, the expression of COX‐2 and further production of PGE2 in cancer
et al., 2018) and annexin A5 (H.‐S. Baek et al., 2017; Figure 4). cells (Ikeya et al., 2018; Figure 4).
8 | HASHEMI ET AL.

15.8 | EGFR/COX‐2
There is a dual positive feedback between EGFR and COX‐2 in
cancer. This cross‐talk can amplify the process of carcinogenesis.
Tobacco smoking (Dannenberg et al., 2005; probably via its active
nicotine component; Dinicola et al., 2018) and ultraviolet light
(Fritsche et al., 2007) are among stimulators of this signaling. EGFR is
a downstream target for AhR signaling (Fritsche et al., 2007), and it is
also a part of the EGFR/COX‐2/Nodal pathway for cancer EMT and
CSC activity. Nodal is a member of TGF‐β superfamily (X. Wang,
Reyes, et al., 2017; Figures 2 and 4).

15.9 | SDF‐1/CXCR4/COX‐2
SDF‐1a is a member of CXC chemokine family that upon
interaction with its receptor CXCR4 and further stimulation of
COX‐2 is contributed to cancer cell invasion (N. Zheng, Chen, Liu,
et al., 2017) and metastasis (N. Zheng, Chen, Li, et al., 2017;
Figure 4).

F I G U R E 5 Cyclooxygenase‐2 (COX‐2) acting as an anticancer


enzyme. 8‐HOA: 8‐hydroxyoctanoic acid; DGLA: dihomo‐γ‐linolenic acid;
15.10 | COX‐2/β‐catenin ERK1/2: extracellular signal‐regulated kinase‐1/2; PPARγ: peroxisome
proliferator‐activated receptor γ; ROS: reactive oxygen species
β‐Catenin is another signaling for COX‐2/PGE2 to exert pro‐
oncogenic actions. This pathway is activated in lower stages of
cancer progression, and it is stimulated by factors like serpinB3 subsequent apoptosis in cancer cells (Chin et al., 2018). In addition,
(Terrin et al., 2017; Figure 2). resveratrol represses COX‐2 translocation to mitochondria to suppress
stemness in cacner cells (Zhou et al., 2017). Eventually, cytosolic
retention of COX‐2 leads to cancer cell proliferation mediated through
16 | COX ‐2 AS A N A NTICA NC ER ENZY M E inhibition of the attachment between ERK and COX‐2 (Chin et al., 2018;
Lin et al., 2017).
COX‐2 acts a double‐edged sword by being as a pro‐ or For some tumors, COX‐2 inhibition is considered as a
anticancer enzyme. COX‐2 is able to metabolize dihomo‐γ‐ disadvantage for cancer therapy. Overexpression of COX‐2 for
linolenic acid into two different agents with diverse functions: cancers like osteosarcoma (Z. Xu et al., 2006) favors the suppres-
PGs and 8‐hydroxyoctanoic acid (8‐HOA; X. Yang, Xu, et al., sion of tumor proliferation and induction of apoptosis. Activation of
2017). COX‐2 promotes tumor expansion through regulation of the P38/COX‐2 signaling (J.‐H. Baek et al., 2018) induces ROS
PGs (Gurram et al., 2018). On the other hand, high level of COX‐2 overproduction (Z. Xu et al., 2006) that is further contributed to
can inhibit tumor growth and migration through regulation of 8‐ promotion of apoptosis in cancers like osteosarcoma. For glioma,
HOA (Y. Xu, Yang, et al., 2018; X. Yang, Xu, et al., 2017). Products there are two opposite views for the roles of COX‐2 overexpression
from COX‐2 can also exert both anti‐ and protumorigenic with either enhancement of cancer promotion through interaction
functions. An example is the 15d‐PGJ2 acting as an anti‐ and between COX‐2 with NF‐κβ (X. Wang, Yu, et al., 2017; J. Zhao, Zhu,
proangiogenic ligand (Ciccone, Monti, Monzani, Casella, & et al., 2017), WNT (Wu et al., 2017), and EGFR (J. Li, Zhou, et al.,
Morbidelli, 2018; E.‐H. Kim & Surh, 2008). 15d‐PGJ2 is an 2017) for respective induction of inflammation, CSC activity and
inducer for the proapoptotic peroxisome proliferator‐activated survival in cancer cells, or suppression of tumor progression
receptor γ (PPARγ; Walther, Emmrich, Ramer, Mittag, & Hinz, through possible interaction between COX‐2 with the endocanna-
2016), while PGE2 is an inhibitor for this proapoptotic factor binoid system that further elicits proapoptotic actions in this system
(Rossi et al., 2018). (Hinz, Ramer, Eichele, Weinzierl, & Brune, 2004). Evidence for
Depending on accumulation in which subcellular component, COX‐2 antitumor activity of COX‐2 has been shown in the Figure 5. A point
could be either a promoter or suppressor of cancer progression. This in this context is that the protumor activity of COX‐2 is far more
would be best explained after application of resveratrol for cancer cells. justifiable than its antitumor activity. Combining the knowledge
Transresveratrol is a component of the polyphenol extract and a stilbene from Figures 6 to 8 in which COX‐2 overexpression is related to
analog (Regulski et al., 2016) that acts on ERK1/2 (Lin et al., 2017) for tumorigenesis for a wide variety of cancers (Figure 6), targeting
induction of nuclear accumulation (shuttling) of COX‐2. Nuclear shuttling COX‐2 expression in cancer cells is a common function of a variety
of ERK/COX‐2 complex favors p53 phosphorylation at Ser‐15 and the of chemotherapeutic agents (Figure 7), and COX‐2 induces multiple
HASHEMI ET AL. | 9

F I G U R E 6 Cancers known to overexpress cyclooxygenase‐2 (COX‐2). HNSCC: head and neck squamous cell carcinoma; LSCC: laryngeal
squamous cell carcinoma; NSCLC: non‐small‐cell lung cancer; OSCC: oral squamous cell carcinoma

F I G U R E 7 Agents identified for their chemopreventive roles against tumors working through cyclooxygenase‐2 (COX‐2) blockade in
cancer cells. ALG, α‐L‐guluronic acid; ALT: alantolactone; EA: Ellagic aicd; MC‐LR: microcystin‐LR; NSAIDs: nonsteroidal anti‐inflammatory
drugs; rcdtB: clostridium difficile toxin B; UA, uroslic acid

signaling in cancer cells for the promotion of cancer cell progression addition, the existence of a positive‐feedback loop between estrogen
(Figure 8), our justification toward the fate for COX‐2 would be and COX‐2 favors aggravation of tumor‐promoting effect for this
more into considering COX‐2 as a tumor promoter, rather than an hormone (Chung et al., 2017). Another hormone is epinephrine that
antitumor enzyme. has been identified for its role in induction of COX‐2 and mediation
of an immunosuppressive state in cancer cells (Muthuswamy
et al., 2017).
17 | HORMONES AFFE CT THE F ATE FOR
COX ‐2 EXP R E S S I O N D U RI N G TH E PROC ES S
TUMORIGENE SIS 18 | COX ‐2 I N H I B I T O R S AS A P R O M IS IN G
T H ER A P EU T I C AP P R O A C H F O R C A N C E R
A number of hormones are identified to have interactions with
COX‐2 for the promotion of tumorigenesis. Thyroxin (T4) stimulates Inhibition of COX‐2 could provide a high possibility to exert
PD‐L1 to mediate cytoplasmic retention of COX‐2 that further blocks therapeutic outcomes in cancer (Shi et al., 2018). Administration of
the formation of ERK/COX‐2 complex resulting in proproliferative COX‐2 inhibitors for organs like lung, breast, colon, and prostate has
features evolved in thyroid cancer cells (Chin et al., 2018; Lin et al., been attested to cause a reduction of cancer risk by about 68%
2017). Testosterone, DHT, suppresses ERK/COX‐2 and COX‐2/p53 (Brizzolara et al., 2017). Interestingly, although it has been estimated
complexes leading to proproliferative features in breast cancer cells that about half of the adenomatic cancers do not express COX‐2
(Lin et al., 2017). Androgen inducible genes and androgen recep- (Kashfi & Rigas, 2005; Noda et al., 2002), administration of COX‐2
tors are also positively affected by COX‐2 (Brizzolara et al., 2017). In inhibitors has shown promising results for suppression of adenoma
10 | HASHEMI ET AL.

copper‐bearing NSAIDs has been taken a special focus due to their


anti‐CSC properties (Eskandari et al., 2016).
Some NSAIDs like aspirin (Amaral, Nery, Leite, de Azevedo
Junior, & Campos, 2018) and celecoxib (J. Kim et al., 2018) depending
on the type of cancer could either inhibit or stimulate COX‐2
expression (Amaral et al., 2018), both of which favors apoptosis in
cancer cells. To explain, initial activation of COX‐2 by these inhibitors
may induce the proapoptotic factor PPARγ (Ramer et al., 2013), while
a late and extended deactivation of COX‐2 is probably required for
activation of PPARγ‐mediated apoptosis in cancer cells (Michael,
Badr, & Badawi, 2003; W.‐H. Sun et al., 2009). This deactivation could
blunt the inhibitory roles for COX‐2 metabolites like PGE2 on PPARγ
activity (Rossi et al., 2018). Therefore, COX‐2 inhibitors like celecoxib
although induce an initial expression for COX‐2 in cancers like lung,
this induction favors cancer cell apoptosis (Ramer et al., 2013).
Aspirin may also suppress growth of cancer cells via mechanisms
independent on COX‐2 (Henry et al., 2017).
COX‐2 could be a target for hormonal therapy. Melatonin, a
F I G U R E 8 A diagram revealing multitasking roles for secretory product of pineal gland with diverse pharmacological
cyclooxygenase‐2 (COX‐2) in promotion of cancer. CSC: cancer
effects including anticancer activity (Mortezaee & Khanlarkhani,
stem cell
2018; Mortezaee, Khanlarkhani, Beyer, & Zendedel, 2018), is an
example in this context showing promising results for suppression of
development (Hull et al., 2017; Noda et al., 2002). For these types of cancer progression by inhibiting NF‐κβ/COX‐2 signaling in cancer
cancers, there is probably a shift for COX‐2 expression in stromal cells (J.‐J. Lu et al., 2016; Mortezaee, 2018).
(rather than cancer) cells (Hull et al., 2017). Application of miRNAs could be another approach for suppres-
COX‐2 inhibitors are relatively inexpensive in comparison with sion of COX‐2 in cancer cells. MiRs 16 (X. Liu, Li, Li, et al., 2018), 101
the standard cancer therapies, having tolerable side effects, and that (Y. Liu, Li, Zhao, & Jia, 2018), 379 (O’Brien et al., 2018), and 144 (Yao
they are able to sensitize cancer cells to treatments like radiotherapy et al., 2018) are identified for their role in suppression of COX‐2 in
(Esbona et al., 2018) and chemotherapy (Shi et al., 2018) both of cancer cells. COX‐2 could be an inducer of oncogenic miRs like
which are recognized to induce COX‐2 expression in cancer cells miR655 and miR526b in cancers like breast (Majumder et al.,
(Ikeya et al., 2018; Kirk et al., 2018). In fact, COX‐2 inhibitors are able 2018; Figure 4).
to relieve COX‐2‐mediated expression of multidrug resistance Another strategy for targeting COX‐2 in cancer cells is to use
proteins in cancer cells (Guo et al., 2017), and when they are carriers delivering siRNA for this enzyme. Agents like dextran
administered in a perioperative setting, the inhibitors would reduce would do such work important for suppression of COX‐2 expres-
the risk of surgical related metastasis (Behrenbruch et al., 2018; sion in cancer cells (Z. Chen, Krishnamachary, Penet, & Bhujwalla,
Shaashua et al., 2017). COX‐2 could be targeted by many types of 2018).
chemotherapeutic drugs, as it has been shown in the Figure 7. COX‐2 inhibitors provide a common target for exerting
Nonsteroidal anti‐inflammatory drugs (NSAIDs) are among the synergistic therapeutic effects after combination with either
drugs used for prevention of cancer. COX‐2 inhibition by NASIDs is chemotherapeutic drugs (X. Xu et al., 2017) or chemoradiation
related to the reduction of cancer recurrence and increase of therapy (Das et al., 2017) against cancer cells. Chemotherapeutic
patient survival (Esbona et al., 2018). Elevation of ROS concentra- agents could adversely cause cancer cell repopulation through
tion by NSAIDS is possibly accounted for inhibition of COX‐2 in induction of caspase‐3‐mediated COX‐2 activity (Y. Zhao, Cui,
cancer cells (C. Lu, Laws, et al., 2017). Conventional NSAIDs are not et al., 2017). Irradiation is also an inducer of COX‐2‐mediated
specific for inhibition of COX‐2, as they are also an inhibitor for angiogenesis through activation of the same pathway (Feng et al.,
COX‐1, and that they irritate upper gastrointestinal (GI) system. 2017). Some chemotherapeutic agents adversely induce COX‐2
Example of these nonselective NSAIDs are ibuprofen (Liaras, expression indirectly via promotion of an inflammatory setting
Fesatidou, & Geronikaki, 2018), sulindac (Dannenberg et al., (Kirk et al., 2018) probably through developing CAFs into
2005), nimesulide (Barac et al., 2017), diclofenac, indomethacin, senescence CAFs by acquiring a proinflammatory SASP (Kabir
and aspirin (J. Qiu et al., 2017). By contrast, the rate of injury to GI et al., 2016). Therefore, it would be advisable to use COX‐2
using selective COX‐2 inhibitors is low (Dannenberg et al., 2005). inhibitors as an adjuvant with chemotherapy and/or radiotherapy.
Coxibs (Lang et al., 2006; J. Qiu et al., 2017), etodolac (Noda et al., Such combination has been reported to synergistically increases
2002), and NS‐398 (J. Qiu et al., 2017) are examples for these the antitumoral activity for chemotherapeutic agents like sorafe-
selective COX‐2 inhibitors (Lang et al., 2006; Figure 7). Recently, nib (Dong et al., 2018), 5‐fluorouracil, bleomycin, irinotecan (Shi
HASHEMI ET AL. | 11

et al., 2018), cisplatin (Barac et al., 2017; F. Wang et al., 2018; 21 | S I GN IF IC AN CE


C.‐C. Yang et al., 2016), paclitaxel, carboplatin (Altorki et al.,
2003), sunitinib (Q. Zhao, Guo, Wang, Chu, & Hu, 2017), and • Both activation and suppression of COX‐2 is related to tumorigenesis.
cetuximab (an EGFR blocker; Y. Lu, Shi, Qiu, & Fan, 2016) among • COX‐2 activation could be both pro‐ or antitumorigenic, but the
others. This combination also improves the rate of tolerance to evidence for its protumor activity is by far more strength.
chemoradiation (Araujo‐Mino et al., 2018) and overall response • COX‐2 could be either activated or deactivated by ROS. This is
rate for advance stages of cancers (Dai et al., 2018), especially probably dependent on the concentration of ROS.
when it is administered before radiotherapy (J. Qiu et al., 2017; • For most type of cancers, uncontrolled activation of COX‐2 is
Salehifar & Hosseinimehr, 2016). related to worse outcome and its blockade favors cancer
prophylaxis and regression.
• Most of the functions of COX‐2 on tumor cells is exerted through
19 | F AC T O RS IN F LU E NC I NG
engagement of PGE2/EP4, so targeting EP4 would provide an
COX ‐2 ‐T A R G E T E D T H E R A P Y FO R C A N C E R
optimistic approach for cancer therapy.
• Members of MAPK family, EGFR, and p65 NF‐κβ are major
These factors are as follows:
regulators of COX‐2 in cancer cells. ERK acting on COX‐2 is
contributed to both pro‐ and antitumorigenic functions of COX‐2.
(1) The type of cancer: Some types of cancers like schwannoma do not
• Nuclear translocation of COX‐2 induces apoptosis in cancer cells,
respond to COX‐2 inhibitors despite its high rate of expression
while mitochondrial translocation of COX‐2 promotes stemness in
(Wahle et al., 2018). In addition, cancer cells have their specific
cancer cells.
activators for COX‐2, as for gastric cancer that the activator is
• TME is a key for the promotion of COX‐2‐mediated tumorigenesis.
Helicobacter pylori (X. Liu et al., 2017) via JMJD2B (Han et al., 2016).
Production of COX‐2 occurs within this milieu. Macrophage type 2
(2) The type of COX‐2 inhibitor: As for NSAIDSs, the response would
cells, Tregs, and cancer cells are contributed to COX‐2 release to
be more potent after application of selective drugs compared to
the TME.
the non‐specific agents.
• Due to the complexity of signaling pathways contributed to the
(3) The dose for COX‐2 inhibitors: High doses for COX‐2 inhibitors exert
regulation of COX‐2 in cancer cells and cancer cell resistance to
opposite effects by showing anticancer activity (Shi et al., 2018). This
chemotherapy, it is preferred to use a combination of COX‐2
might be due to the effects of negative feedback loop or probably
inhibitor with a suitable chemotherapeutic agent to achieve
upregulation of COX‐2 isoforms (Noda et al., 2002). To remove this
appropriate responses.
burden (decreasing the doses demanded for COX‐2 inhibitors), these
• Existence of a negative feedback between COX‐2 and mediator of
inhibitors are preferred to be administered in combination with
apoptosis favors both apoptosis resistance and angiogenesis in
conventional anticancer agents (Shi et al., 2018).
cancer cells.
(4) The type of chemotherapy (a conventional antitumor drug
received by patient) used in combination with COX‐2 inhibitors.
Choosing a right type of chemotherapy as an adjuvant with COX‐ CON F LI CTS OF I NTERE ST
2 inhibitor is also important.
The authors declare that there are no conflicts of interest.
(5) The grade of tumor: Relation between the grade of tumor with
COX‐2 expression is still a controversial issue. For example, two
studies performed on ovarian cancer were reached opposite ORCI D
results with a one reported high expression for COX‐2 in the low Nasser Hashemi Goradel http://orcid.org/0000-0003-0713-1639
(mucus) grade (Beeghly‐Fadiel et al., 2018), but another the high Keywan Mortezaee http://orcid.org/0000-0003-2004-3465
rate for COX‐2 in the high (serous) grade of cancer (Beeghly‐
Fadiel et al., 2018). This controversy has also been marked for
R E F E R E N CE S
the breast cancer (Tury et al., 2016).
Altorki, N. K., Keresztes, R., Port, J., Libby, D., Korst, R., Flieder, D., …
Pasmantier, M. (2003). Celecoxib, a selective cyclo‐oxygenase‐2
20 | CONC LU SION AN D P ERSP ECTIVE inhibitor, enhances the response to preoperative paclitaxel and
carboplatin in early‐stage non–small‐cell lung cancer. Journal of
Clinical Oncology, 21(14), 2645–2650.
From what has been discussed for most of the cancer types, COX‐2 is
Amaral, M. E. A., Nery, L. R., Leite, C. E., de Azevedo Junior, W. F., &
a marker indicating worse prognosis and a stimulator for a variety of Campos, M. M. (2018). Pre‐clinical effects of metformin and aspirin on
cancers (Figure 6) by taking multitasking roles (Figure 8), so the cell lines of different breast cancer subtypes. Investigational New
suppression of COX‐2 could provide a promising approach for cancer Drugs, 1–15.
Araki, Y., Okamura, S., Hussain, S. P., Nagashima, M., He, P., Shiseki, M., …
therapy especially when it is used as an adjuvant with appropriate
Harris, C. C. (2003). Regulation of cyclooxygenase‐2 expression by the
chemotherapeutic agents. Wnt and ras pathways. Cancer Research, 63(3), 728–734.
12 | HASHEMI ET AL.

Araujo‐Mino, E. P., Patt, Y. Z., Murray‐Krezan, C., Hanson, J. A., Bansal, P., Chou, W. Y., Chuang, K. H., Sun, D., Lee, Y. H., Kao, P. H., Lin, Y. Y., … Wu,
Liem, B. J., … Lee, F. C. (2018). Phase II trial using a combination of Y. L. (2015). Inhibition of PKC‐induced COX‐2 and IL‐8 expression in
oxaliplatin, capecitabine, and celecoxib with concurrent radiation for human breast cancer cells by glucosamine. Journal of Cellular
newly diagnosed resectable rectal cancer. The Oncologist, 23(1), 2–e5. Physiology, 230(9), 2240–2251.
Baek, H.‐S., Park, N., Kwon, Y.‐J., Ye, D.‐J., Shin, S., & Chun, Y.‐J. (2017). Chung, H. H., Or, Y. Z., Shrestha, S., Loh, J. T., Lim, C. L., Ong, Z., … Lin, V. C.
Annexin A5 suppresses cyclooxygenase‐2 expression by downregu- L. (2017). Estrogen reprograms the activity of neutrophils to foster
lating the protein kinase C‐ζ–nuclear factor‐κB signaling pathway in protumoral microenvironment during mammary involution. Scientific
prostate cancer cells. Oncotarget, 8(43), 74263–74275. Reports, 7, 46485.
Baek, J.‐H., Yim, J.‐H., Song, J.‐Y., Um, H.‐D., Park, J. K., Park, I.‐C., … Ciccone, V., Monti, M., Monzani, E., Casella, L., & Morbidelli, L. (2018). The
Hwang, S. G. (2018). Knockdown of end‐binding protein 1 induces metal‐nonoate Ni (SalPipNONO) inhibits in vitro tumor growth,
apoptosis in radioresistant A549 lung cancer cells via p38 kinase‐ invasiveness and angiogenesis. Oncotarget, 9(17), 13353.
dependent COX‐2 upregulation. Oncology Reports, 39(4), Cook, P. J., Thomas, R., Kingsley, P. J., Shimizu, F., Montrose, D. C.,
1565–1572. Marnett, L. J., … Benezra, R. (2016). Cox‐2‐derived PGE2 induces Id1‐
Barac, A., Mitulovic, G., Hallstrom, S., Zehetmayer, S., Grasl, M., & Erovic, dependent radiation resistance and self‐renewal in experimental
B. (2017). Impact of combined treatment with nimesulide and cisplatin glioblastoma. Neuro‐Oncology, 18(10), 1379–1389.
on oral carcinoma cells. OncoTargets and Therapy, 10, 3607–3616. Cui, Y., Shu, X. O., Li, H. L., Yang, G., Wen, W., Gao, Y. T., … Zheng, W.
Beeghly‐Fadiel, A., Wilson, A. J., Keene, S., El Ramahi, M., Xu, S., Marnett, (2017). Prospective study of urinary prostaglandin E2 metabolite and
L. J., … Khabele, D. (2018). Differential cyclooxygenase expression pancreatic cancer risk. International Journal of Cancer, 141(12),
levels and survival associations in type I and type II ovarian tumors. 2423–2429.
Journal of Ovarian Research, 11(1), 17. Dai, P., Li, J., Ma, X.‐P., Huang, J., Meng, J.‐J., & Gong, P. (2018). Efficacy
Behrenbruch, C., Shembrey, C., Paquet‐Fifield, S., Mølck, C., Cho, H.‐J., and safety of COX‐2 inhibitors for advanced non‐small‐cell lung
Michael, M., … Hollande, F. (2018). Surgical stress response and cancer with chemotherapy: A meta‐analysis. OncoTargets and Therapy,
promotion of metastasis in colorectal cancer: A complex and 11, 721–730.
heterogeneous process. Clinical & Experimental Metastasis, 35(4), Dannenberg, A. J., Lippman, S. M., Mann, J. R., Subbaramaiah, K., & DuBois,
333–345. R. N. (2005). Cyclooxygenase‐2 and epidermal growth factor receptor:
Bohrer, L. R., Chaffee, T. S., Chuntova, P., Brady, N. J., Witschen, P. M., Pharmacologic targets for chemoprevention. Journal of Clinical
Kemp, S. E., … Schwertfeger, K. L. (2016). ADAM17 in tumor Oncology, 23(2), 254–266.
associated leukocytes regulates inflammatory mediators and pro- Das, U., Biswas, S., Chattopadhyay, S., Chakraborty, A., Dey sharma, R.,
motes mammary tumor formation. Genes & Cancer, 7(7–8), 240–253. Banerji, A., & Dey, S. (2017). Radiosensitizing effect of ellagic acid on
Brizzolara, A., Benelli, R., Venè, R., Barboro, P., Poggi, A., Tosetti, F., & growth of hepatocellular carcinoma cells: An in vitro study. Scientific
Ferrari, N. (2017). The ErbB family and androgen receptor signaling Reports, 7(1), 14043.
are targets of Celecoxib in prostate cancer. Cancer Letters, 400, 9–17. Desai, S. J., Prickril, B., & Rasooly, A. (2018). Mechanisms of Phytonutrient
Cai, T.‐T., Ye, S.‐B., Liu, Y.‐N., He, J., Chen, Q.‐Y., Mai, H.‐Q., … Li, J. (2017). Modulation of Cyclooxygenase‐2 (COX‐2) and Inflammation Related
LMP1‐mediated glycolysis induces myeloid‐derived suppressor cell to Cancer. Nutrition and Cancer, 70, 1–26.
expansion in nasopharyngeal carcinoma. PLoS Pathogens, 13(7), Dinicola, S., Masiello, M. G., Proietti, S., Coluccia, P., Fabrizi, G.,
e1006503. Catizone, A., … Cucina, A. (2018). Nicotine increases colon cancer
Chang, J., Tang, N., Fang, Q., Zhu, K., Liu, L., Xiong, X., … Tao, J. (2018). cell migration and invasion through epithelial to mesenchymal
Inhibition of COX‐2 and 5‐LOX regulates the progression of colorectal transition (EMT): COX‐2 involvement. Journal of Cellular Physiol-
cancer by promoting PTEN and suppressing PI3K/AKT pathway. ogy, 233(6), 4935–4948.
Biochemical and Biophysical Research Communications. https://doi.org/ Dong, Z.‐R., Dong, X.‐F., Liu, T.‐Q., Zhi, X.‐T., Zou, J., Zhong, J.‐T., …
10.1016/j.bbrc.2018.01.061 Guo, W.‐W. (2018). COX‐2/PGE2 axis regulates HIF‐2α activity to
Charalambous, M. P., Maihöfner, C., Bhambra, U., Lightfoot, T., & promote hepatocellular carcinoma hypoxic response and reduce
Gooderham, N. J. (2003). Upregulation of cyclooxygenase‐2 is the sensitivity of sorafenib treatment. Clinical Cancer Research, 24,
accompanied by increased expression of nuclear factor‐κB and IκB 3204–3216.
kinase‐α in human colorectal cancer epithelial cells. British Journal of Echizen, K., Oshima, H., Nakayama, M., & Oshima, M. (2018). The
Cancer, 88(10), 1598–1604. inflammatory microenvironment that promotes gastrointestinal can-
Chen, H., Cai, W., Chu, E. S. H., Tang, J., Wong, C. C., Wong, S. H., … Yu, J. cer development and invasion. Advances in Biological Regulation, 68,
(2017). Hepatic cyclooxygenase‐2 overexpression induced sponta- 39–45.
neous hepatocellular carcinoma formation in mice. Oncogene, 36(31), Esbona, K., Inman, D., Saha, S., Jeffery, J., Schedin, P., Wilke, L., & Keely, P.
4415–4426. (2016). COX‐2 modulates mammary tumor progression in response to
Chen, W., Bai, L., Wang, X., Xu, S., Belinsky, S. A., & Lin, Y. (2010). Acquired collagen density. Breast Cancer Research, 18(1), 35.
activation of the Akt/cyclooxygenase‐2/Mcl‐1 pathway renders lung Esbona, K., Yi, Y., Saha, S., Yu, M., Van Doorn, R. R., Conklin, M. W., …
cancer cells resistant to apoptosis. Molecular Pharmacology, 77(3), Keely, P. J. (2018). The presence of cyclooxygenase 2, tumor‐
416–423. associated macrophages, and collagen alignment as prognostic
Chen, Z., Krishnamachary, B., Penet, M.‐F., & Bhujwalla, Z. M. (2018). Acid‐ markers for invasive breast carcinoma patients. The American Journal
degradable dextran as an image guided siRNA carrier for COX‐2 of Pathology, 188(3), 559–573.
downregulation. Theranostics, 8(1), 1–12. Eskandari, A., Boodram, J. N., Cressey, P. B., Lu, C., Bruno, P. M., Hemann,
Cheng, H.‐H., Chu, L.‐Y., Chiang, L.‐Y., Chen, H.‐L., Kuo, C.‐C., & Wu, K. K. M. T., & Suntharalingam, K. (2016). The breast cancer stem cell
(2016). Inhibition of cancer cell epithelial mesenchymal transition by potency of copper (II) complexes bearing nonsteroidal anti‐inflamma-
normal fibroblasts via production of 5‐methoxytryptophan. Oncotar- tory drugs and their encapsulation using polymeric nanoparticles.
get, 7(21), 31243. Dalton Transactions, 45(44), 17867–17873.
Chin, Y.‐T., Wei, P.‐L., Ho, Y., Nana, A. W., Changou, C. A., Chen, Y.‐R., … Feng, X., Yu, Y., He, S., Cheng, J., Gong, Y., Zhang, Z., … Huang, Q. (2017).
Lin, H. Y. (2018). Thyroxine inhibits resveratrol‐caused apoptosis by Dying glioma cells establish a proangiogenic microenvironment
PD‐L1 in ovarian cancer cells. Endocrine‐Related Cancer, 25(5), through a caspase 3 dependent mechanism. Cancer Letters, 385,
533–545. 12–20.
HASHEMI ET AL. | 13

Fritsche, E., Schafer, C., Calles, C., Bernsmann, T., Bernshausen, T., Wurm, M., … Hibino, S., Chikuma, S., Kondo, T., Ito, M., Nakatsukasa, H., Omata‐Mise, S.,
Krutmann, J. (2007). Lightening up the UV response by identification of & Yoshimura, A. (2018). Inhibition of Nr4a receptors enhances anti‐
the arylhydrocarbon receptor as a cytoplasmatic target for ultraviolet B tumor immunity by breaking Treg‐mediated immune tolerance. Cancer
radiation. Proceedings of the National Academy of Sciences, 104(21), Research, 24, 3204–32016.
8851–8856. Higdon, A., Diers, A. R., Oh, J. Y., Landar, A., & Darley‐Usmar, V. M. (2012).
Furuya, H., Tamashiro, P. M., Shimizu, Y., Iino, K., Peres, R., Chen, R., … Cell signalling by reactive lipid species: New concepts and molecular
Kawamori, T. (2017). Sphingosine Kinase 1 expression in peritoneal mechanisms. Biochemical Journal, 442(3), 453–464.
macrophages is required for colon carcinogenesis. Carcinogenesis, 38 Hinz, B., Ramer, R., Eichele, K., Weinzierl, U., & Brune, K. (2004). Up‐
(12), 1218–1227. regulation of cyclooxygenase‐2 expression is involved in R (+)‐
Gan, L., Qiu, Z., Huang, J., Li, Y., Huang, H., Xiang, T., … Ren, G. (2016). methanandamide‐induced apoptotic death of human neuroglioma
Cyclooxygenase‐2 in tumor‐associated macrophages promotes meta- cells. Molecular Pharmacology, 66(6), 1643–1651.
static potential of breast cancer cells through Akt pathway. Interna- Hosseini, F., Mahdian‐Shakib, A., Jadidi‐Niaragh, F., Enderami, S. E.,
tional Journal of Biological Sciences, 12(12), 1533–1543. Mohammadi, H., Hemmatzadeh, M., … Mirshafiey, A. (2018). Anti‐
Gao, Y., Ge, G., & Ji, H. (2011). LKB1 in lung cancerigenesis: A inflammatory and anti‐tumor effects of α‐l‐guluronic acid (G2013) on
serine/threonine kinase as tumor suppressor. Protein & Cell, 2(2), cancer‐related inflammation in a murine breast cancer model.
99–107. Biomedicine & Pharmacotherapy, 98, 793–800.
Genard, G., Wera, A.‐C., Huart, C., Le Calve, B., Penninckx, S., Fattaccioli, A., … Hou, P.‐C., Li, Y.‐H., Lin, S.‐C., Lin, S.‐C., Lee, J.‐C., Lin, B.‐W., … Tsai, S. J.
Michiels, C. (2018). Proton irradiation orchestrates macrophage (2017). Hypoxia‐induced downregulation of DUSP‐2 phosphatase
reprogramming through NFκB signaling. Cell Death & Disease, 9(7), 728. drives colon cancer stemness. Cancer Research, 77(16), 4305–4316.
Goswami, S., & Sharma‐Walia, N. (2016). Crosstalk between osteoprote- Hu, H., Han, T., Zhuo, M., Wu, L., Yuan, C., Wu, L., … Wang, L.‐W. (2017).
gerin (OPG), fatty acid synthase (FASN) and, cycloxygenase‐2 (COX‐2) Elevated COX‐2 expression promotes angiogenesis through EGFR/
in breast cancer: Implications in carcinogenesis. Oncotarget, 7(37), p38‐MAPK/Sp1‐dependent signalling in pancreatic cancer. Scientific
58953. Reports, 7(1), 470.
Greenhough, A., Smartt, H. J. M., Moore, A. E., Roberts, H. R., Williams, A. Hull, M. A., Cuthbert, R. J., Ko, C. W. S., Scott, D. J., Cartwright, E. J.,
C., Paraskeva, C., & Kaidi, A. (2009). The COX‐2/PGE 2 pathway: Key Hawcroft, G., … Coletta, P. L. (2017). Paracrine cyclooxygenase‐2
roles in the hallmarks of cancer and adaptation to the tumour activity by macrophages drives colorectal adenoma progression in the
microenvironment. Carcinogenesis, 30(3), 377–386. Apc Min/+mouse model of intestinal tumorigenesis. Scientific Reports,
Gungor, H., Ilhan, N., & Eroksuz, H. (2018). The effectiveness of 7(1), 6074.
cyclooxygenase‐2 inhibitors and evaluation of angiogenesis in the Hung, J.‐H., Su, I.‐J., Lei, H.‐Y., Wang, H.‐C., Lin, W.‐C., Chang, W.‐T., … Lai,
model of experimental colorectal cancer. Biomedicine & Pharmacother- M. D. (2004). Endoplasmic reticulum stress stimulates the expression
apy, 102, 221–229. of cyclooxygenase‐2 through activation of NF‐κB and pp38 mitogen‐
Guo, Q., Liu, X., Lu, L., Yuan, H., Wang, Y., Chen, Z., … Zhou, Y. (2017). activated protein kinase. Journal of Biological Chemistry, 279(45),
Comprehensive evaluation of clinical efficacy and safety of celecoxib 46384–46392.
combined with chemotherapy in management of gastric cancer. Höing, B., Kanaan, O., Altenhoff, P., Petri, R., Thangavelu, K., Schlüter, A., …
Medicine, 96(51), e8857. Brandau, S. (2018). Stromal versus tumoral inflammation differentially
Gurram, B., Zhang, S., Li, M., Li, H., Xie, Y., Cui, H., … Peng, X. (2018). contribute to metastasis and poor survival in laryngeal squamous cell
Celecoxib conjugated fluorescent probe for identification and carcinoma. Oncotarget, 9(9), 8415.
discrimination of cyclooxygenase‐2 enzyme in cancer cells. Analytical Ikeya, S., Sakabe, J., Yamada, T., Naito, T., & Tokura, Y. (2018). Voriconazole‐
Chemistry, 90, 5187–5193. induced photocarcinogenesis is promoted by aryl hydrocarbon receptor‐
Han, F., Ren, J., Zhang, J., Sun, Y., Ma, F., Liu, Z., … Li, W. (2016). JMJD2B is dependent COX‐2 upregulation. Scientific Reports, 8(1), 5050.
required for Helicobacter pylori‐induced gastric carcinogenesis via Janakiraman, H., House, R. P., Talwar, S., Courtney, S. M., Hazard, E. S.,
regulating COX‐2 expression. Oncotarget, 7(25), 38626. Hardiman, G., … Palanisamy, V. (2017). Repression of caspase‐3 and
Hao, J., Xu, H., Luo, M., Yu, W., Chen, M., Liao, Y., … Deng, W. (2018). The RNA‐binding protein HuR cleavage by cyclooxygenase‐2 promotes
tumor‐promoting role of TRIP4 in melanoma progression and its drug resistance in oral squamous cell carcinoma. Oncogene, 36(22),
involvement in response to BRAF‐targeted therapy. Journal of 3137–3148.
Investigative Dermatology, 138(1), 159–170. Jeong, A., Kim, J.‐H., Lee, H.‐J., & Kim, S.‐H. (2017). Reactive oxygen
Hao, Y., Zhang, J., Shan, G., Zhang, N., Jin, W., & Nan, K. (2017). Establishment species dependent phosphorylation of the liver kinase B1/AMP
of optimal regulatory network of colorectal cancer based on p42. 3 activated protein kinase/acetyl‐CoA carboxylase signaling is critically
protein. Saudi Journal of Biological Sciences, 24, 1781–1786. involved in apoptotic effect of lambertianic acid in hepatocellular
Harris, R. E., Beebe‐Donk, J., & Alshafie, G. A. (2007). Cancer carcinoma cells. Oncotarget, 8(41), 70116.
chemoprevention by cyclooxygenase 2 (COX‐2) blockade: Results of Jiang, L., Wang, Y., Liu, G., Liu, H., Zhu, F., Ji, H., & Li, B. (2018). C‐
case control studies, Subcellular Biochemistry, 42, 193–212. phycocyanin exerts anti‐cancer effects via the MAPK signaling
Hattar, K., Franz, K., Ludwig, M., Sibelius, U., Wilhelm, J., Lohmeyer, J., … pathway in MDA‐MB‐231 cells. Cancer Cell International, 18(1), 12.
Grandel, U. (2014). Interactions between neutrophils and non‐small Kabir, T., Leigh, R., Tasena, H., Mellone, M., Coletta, R., Parkinson, E., …
cell lung cancer cells: Enhancement of tumor proliferation and Lambert, D. (2016). A miR‐335/COX‐2/PTEN axis regulates the
inflammatory mediator synthesis. Cancer Immunology, Immunotherapy, secretory phenotype of senescent cancer‐associated fibroblasts.
63(12), 1297–1306. Aging, 8(8), 1608–1635.
Hennequart, M., Pilotte, L., Cane, S., Hoffmann, D., Stroobant, V., Plaen, E. Kashfi, K., & Rigas, B. (2005). Is COX‐2 a ‘collateral’ target in cancer
D., & Eynde, B. J. V. (2017). Constitutive IDO1 expression in human prevention? Biochemical Society Transactions, 33(Pt 4), 724–727.
tumors is driven by cyclooxygenase‐2 and mediates intrinsic immune Kim, J., Hong, S.‐W., Kim, S., Kim, D., Hur, D. Y., Jin, D.‐H., … Kim, Y. S.
resistance. Cancer Immunology Research, 5, 695–709. (2018). Cyclooxygenase‐2 expression is induced by celecoxib treat-
Henry, W. S., Laszewski, T., Tsang, T., Beca, F., Beck, A. H., McAllister, S. S., & ment in lung cancer cells and is transferred to neighbor cells via
Toker, A. (2017). Aspirin suppresses growth in PI3K‐mutant breast exosomes. International Journal of Oncology, 52(2), 613–620.
cancer by activating AMPK and inhibiting mTORC1 signaling. Cancer Kim, E.‐H., Na, H.‐K., Kim, D.‐H., Park, S.‐A., Kim, H.‐N., Song, N.‐Y., & Surh,
Research, 77(3), 790–801. Y.‐J. (2008). 15‐Deoxy‐Δ 12, 14‐prostaglandin J 2 induces COX‐2
14 | HASHEMI ET AL.

expression through Akt‐driven AP‐1 activation in human breast apoptosis and inhibit tumor growth in colorectal cancers. Oncogene,
cancer cells: A potential role of ROS. Carcinogenesis, 29(4), 688–695. 27(53), 6729–6737.
Kim, E.‐H., & Surh, Y.‐J. (2008). The role of 15‐deoxy‐Δ12, 14‐ Liu, X., Ji, Q., Zhang, C., Liu, X., Liu, Y., Liu, N., … Li, Q. (2017). miR‐30a acts
prostaglandin J2, an endogenous ligand of peroxisome proliferator‐ as a tumor suppressor by double‐targeting COX‐2 and BCL9 in H.
activated receptor γ, in tumor angiogenesis. Biochemical Pharmacology, pylori gastric cancer models. Scientific Reports, 7(1), 7113.
76(11), 1544–1553. Liu, X., Li, S., Li, Y., Cheng, B., Tan, B., & Wang, G. (2018). Puerarin inhibits
Kirk, J., Shah, N., Noll, B., Stevens, C. B., Lawler, M., Mougeot, F. B., & proliferation and induces apoptosis by up‐regulation of miR‐16 in
Mougeot, J. L. C. (2018). Text mining‐based in silico drug discovery in bladder cancer cell line T24. Oncology Research.
oral mucositis caused by high‐dose cancer therapy. Supportive Care in Liu, Y., Li, H., Zhao, C., & Jia, H. (2018). MicroRNA‐101 inhibits
Cancer, 26, 1–11. angiogenesis via COX‐2 in endometrial carcinoma. Molecular and
Ko, C. J., Lan, S. W., Lu, Y. C., Cheng, T. S., Lai, P. F., Tsai, C. H., … Lee, M. S. Cellular Biochemistry, 1–9. https://doi.org/10.1007/s11010-018-
(2017). Inhibition of cyclooxygenase‐2‐mediated matriptase activa- 3313-0
tion contributes to the suppression of prostate cancer cell motility Liu, W., Reinmuth, N., Stoeltzing, O., Parikh, A. A., Tellez, C., Williams, S., …
and metastasis. Oncogene, 36(32), 4597–4609. Ellis, L. M. (2003). Cyclooxygenase‐2 is up‐regulated by interleukin‐1β
Ko, S. C., Chapple, K. S., Hawcroft, G., Coletta, P. L., Markham, A. F., & Hull, in human colorectal cancer cells via multiple signaling pathways.
M. A. (2002). Paracrine cyclooxygenase‐2‐mediated signalling by Cancer Research, 63(13), 3632–3636.
macrophages promotes tumorigenic progression of intestinal epithe- Lu, C., Eskandari, A., Cressey, P. B., & Suntharalingam, K. (2017). Cancer
lial cells. Oncogene, 21(47), 7175–7186. stem cell and bulk cancer cell active copper (II) complexes with
Krishnamachary, B., Stasinopoulos, I., Kakkad, S., Penet, M.‐F., Jacob, D., Vanillin Schiff base derivatives and naproxen. Chemistry‐A European
Wildes, F., … Bhujwalla, Z. M. (2017). Breast cancer cell cyclooxygenase‐2 Journal, 23, 11366–11374.
expression alters extracellular matrix structure and function and numbers Lu, C., Laws, K., Eskandari, A., & Suntharalingam, K. (2017). A reactive
of cancer associated fibroblasts. Oncotarget, 8(11), 17981. oxygen species‐generating, cyclooxygenase‐2 inhibiting, cancer stem
Krysan, K., Dalwadi, H., Sharma, S., Põld, M., & Dubinett, S. (2004). cell‐potent tetranuclear copper (II) cluster. Dalton Transactions, 46(38),
Cyclooxygenase 2‐dependent expression of survivin is critical for 12785–12789.
apoptosis resistance in non‐small cell lung cancer. Cancer Research, 64 Lu, J.‐J., Fu, L., Tang, Z., Zhang, C., Qin, L., Wang, J., … Xie, F. (2016).
(18), 6359–6362. Melatonin inhibits AP‐2β/hTERT, NF‐κB/COX‐2 and Akt/ERK and
Lang, S., Busch, H., Boerries, M., Brummer, T., Timme, S., Lassmann, S., … activates caspase/Cyto C signaling to enhance the antitumor activity
Schmidt, G. (2017). Specific role of RhoC in tumor invasion and of berberine in lung cancer cells. Oncotarget, 7(3), 2985.
metastasis. Oncotarget, 8(50), 87364. Lu, Y., Shi, C., Qiu, S., & Fan, Z. (2016). Identification and validation of
Lang, S., Picu, A., Hofmann, T., Andratschke, M., Mack, B., Moosmann, A., … COX‐2 as a co‐target for overcoming cetuximab resistance in
Zeidler, R. (2006). COX‐inhibitors relieve the immunosuppressive colorectal cancer cells. Oncotarget, 7(40), 64766.
effect of tumor cells and improve functions of immune effectors. Majumder, M., Dunn, L., Liu, L., Hasan, A., Vincent, K., Brackstone, M., …
International Journal of Immunopathology and Pharmacology, 19(2), Lala, P. K. (2018). COX‐2 induces oncogenic micro RNA miR655 in
409–419. human breast cancer. Scientific Reports, 8(1), 327.
Lee, J., Ha, S. J., Park, J., Kim, Y. H., Lee, N. H., Kim, Y. E., … Jung, S. K. Majumder, M., Landman, E., Liu, L., Hess, D., & Lala, P. K. (2015). COX‐2
(2017). 1, 8‐cineole prevents UVB‐induced skin carcinogenesis by elevates oncogenic miR‐526b in breast cancer by EP4 activation.
targeting the aryl hydrocarbon receptor. Oncotarget, 8(62), 105995. Molecular Cancer Research, 13(6), 1022–1033.
Lee, J. H., Suh, J. H., Choi, S. Y., Kang, H. J., Lee, H. H., Ye, B. J., … Kwon, H. Maturu, P., Jones, D., Ruteshouser, E. C., Hu, Q., Reynolds, J. M., Hicks, J.,
M. (2018). Tonicity‐responsive enhancer‐binding protein promotes … Overwijk, W. W. (2017). Role of cyclooxygenase‐2 pathway in
hepatocellular carcinogenesis, recurrence and metastasis. Gut. gutjnl‐ creating an immunosuppressive microenvironment and in initiation
2017‐315348. https://doi.org/10.1136/gutjnl-2017-315348 and progression of Wilms’ tumor. Neoplasia, 19(3), 237–249.
Li, W., Cao, Y., Xu, J., Wang, Y., Li, W., Wang, Q., … Sun, Y. (2017). YAP Miao, J., Lu, X., Hu, Y., Piao, C., Wu, X., Liu, X., … Liu, J. (2017).
transcriptionally regulates COX‐2 expression and GCCSysm‐4 (G‐4), a Prostaglandin E2 and PD‐1 mediated inhibition of antitumor CTL
dual YAP/COX‐2 inhibitor, overcomes drug resistance in colorectal responses in the human tumor microenvironment. Oncotarget, 8(52),
cancer. Journal of Experimental & Clinical Cancer Research, 36(1), 144. 89802.
Li, B., Lu, Y., Yu, L., Han, X., Wang, H., Mao, J., … Song, B. (2017). miR‐221/ Michael, M., Badr, M., & Badawi, A. (2003). Inhibition of cyclooxygenase‐2
222 promote cancer stem‐like cell properties and tumor growth of and activation of peroxisome proliferator‐activated receptor‐γ syner-
breast cancer via targeting PTEN and sustained Akt/NF‐κB/COX‐2 gistically induces apoptosis and inhibits growth of human breast
activation. Chemico‐Biological Interactions, 277, 33–42. cancer cells. International Journal of Molecular Medicine, 11(6),
Li, J., Zhou, Y., Wang, H., Gao, Y., Li, L., Hwang, S. H., … Hammock, B. D. 733–736.
(2017). COX‐2/sEH dual inhibitor PTUPB suppresses glioblastoma Montezuma, M. A. P., Fonseca, F. P., Benites, B. M., Soares, C. D., do
growth by targeting epidermal growth factor receptor and hyaluronan Amaral‐Silva, G. K., de Almeida, O. P., … Fregnani, E. R. (2018). COX‐2
mediated motility receptor. Oncotarget, 8(50), 87353. as a determinant of lower disease‐free survival for patients affected
Li, F., & Zhu, Y.‐T. (2015). HGF‐activated colonic fibroblasts mediates by ameloblastoma. Pathology, Research and Practice, 214, 907–913.
carcinogenesis of colonic epithelial cancer cells via PKC‐cMET‐ERK1/ Mortezaee, K. (2018). Human hepatocellular carcinoma: Protection by
2‐COX‐2 signaling. Cellular Signalling, 27(4), 860–866. melatonin. Journal of Cellular Physiology, 233, 6486–6508.
Liaras, K., Fesatidou, M., & Geronikaki, A. (2018). Thiazoles and Mortezaee, K., & Khanlarkhani, N. (2018). Melatonin application in
thiazolidinones as COX/LOX inhibitors. Molecules, 23(3), 685. targeting oxidative‐induced liver injuries: A review. Journal of Cellular
Lin, H. Y., Hsieh, M. T., Cheng, G. Y., Lai, H. Y., Chin, Y. T., Shih, Y. J., … Physiology, 233(5), 4015–4032.
Wang, K. (2017). Mechanisms of action of nonpeptide hormones on Mortezaee, K., Khanlarkhani, N., Beyer, C., & Zendedel, A. (2018).
resveratrol‐induced antiproliferation of cancer cells. Annals of the New Inflammasome: Its role in traumatic brain and spinal cord injury.
York Academy of Sciences, 1403, 92–100. Journal of Cellular Physiology, 233(7), 5160–5169.
Liu, Y., Borchert, G. L., Surazynski, A., & Phang, J. M. (2008). Proline Muthuswamy, R., Okada, N. J., Jenkins, F. J., McGuire, K., McAuliffe, P. F.,
oxidase, a p53‐induced gene, targets COX‐2/PGE 2 signaling to induce Zeh, H. J., … Kalinski, P. (2017). Epinephrine promotes COX‐2‐dependent
HASHEMI ET AL. | 15

immune suppression in myeloid cells and cancer tissues. Brain, Behavior, Rossi, E. L., Khatib, S. A., Doerstling, S. S., Bowers, L. W., Pruski, M., Ford,
and Immunity, 62, 78–86. N. A., … Hursting, S. D. (2018). Resveratrol inhibits obesity‐associated
Nakanishi, Y., Nakatsuji, M., Seno, H., Ishizu, S., Akitake‐Kawano, R., adipose tissue dysfunction and tumor growth in a mouse model of
Kanda, K., … Chiba, T. (2011). COX‐2 inhibition alters the phenotype postmenopausal claudin‐low breast cancer. Molecular Carcinogenesis,
of tumor‐associated macrophages from M2 to M1 in Apc Min/+ 57(3), 393–407.
mouse polyps. Carcinogenesis, 32(9), 1333–1339. Rüegg, C., Dormond, O., & Mariotti, A. (2004). Endothelial cell integrins
Noda, M., Tatsumi, Y., Tomizawa, M., Takama, T., Mitsufuji, S., Sugihara, H., and COX‐2: Mediators and therapeutic targets of tumor angiogenesis.
… Hattori, T. (2002). Effects of etodolac, a selective cyclooxygenase‐2 Biochimica et Biophysica Acta (BBA) – Reviews on Cancer, 1654(1),
inhibitor, on the expression of E‐cadherin‐catenin complexes in 51–67.
gastrointestinal cell lines. Journal of Gastroenterology, 37(11), Rundhaug, J. E., & Fischer, S. M. (2008). Cyclo‐oxygenase‐2 plays a critical
896–904. role in UV‐induced skin carcinogenesis. Photochemistry and Photobiol-
Obermoser, V., Baecker, D., Schuster, C., Braun, V., Kircher, B., & Gust, R. ogy, 84(2), 322–329.
(2018). Chlorinated cobalt alkyne complexes derived from acetylsa- Salehifar, E., & Hosseinimehr, S. J. (2016). The use of cyclooxygenase‐2
licylic acid as new specific antitumor agents. Dalton Transactions, 47 inhibitors for improvement of efficacy of radiotherapy in cancers.
(12), 4341–4351. Drug Discovery Today, 21(4), 654–662.
O’Brien, K. P., Khan, S., Gilligan, K. E., Zafar, H., Lalor, P., Glynn, C., … Sample, A., Zhao, B., Qiang, L., & He, Y.‐Y. (2017). Adaptor protein p62
Dwyer, R. M. (2018). Employing mesenchymal stem cells to support promotes skin tumor growth and metastasis and is induced by UVA
tumor‐targeted delivery of extracellular vesicle (EV)‐encapsulated radiation. Journal of Biological Chemistry, 292(36), 14786–14795.
microRNA‐379. Oncogene, 37, 1–2149. Semaan, J., Pinon, A., Rioux, B., Hassan, L., Limami, Y., Pouget, C., … Liagre, B.
Ohtsuka, J., Oshima, H., Ezawa, I., Abe, R., Oshima, M., & Ohki, R. (2018). (2016). Resistance to 3‐HTMC‐induced apoptosis through activation of
Functional loss of p53 cooperates with the in vivo microenvironment PI3K/Akt, MEK/ERK, and p38/COX‐2/PGE2 pathways in human HT‐29
to promote malignant progression of gastric cancers. Scientific Reports, and HCT116 colorectal cancer cells. Journal of Cellular Biochemistry, 117
8(1), 2291. (12), 2875–2885.
Pan, J., Yang, Q., Shao, J., Zhang, L., Ma, J., Wang, Y., … Bai, X. (2016). Shaashua, L., Shabat‐Simon, M., Haldar, R., Matzner, P., Zmora, O.,
Cyclooxygenase‐2 induced β1‐integrin expression in NSCLC and Shabtai, M., … Ben‐Eliyahu, S. (2017). Perioperative COX‐2 and β‐
promoted cell invasion via the EP1/MAPK/E2F‐1/FoxC2 signal adrenergic blockade improves metastatic biomarkers in breast
pathway. Scientific Reports, 6, 33823. cancer patients in a phase‐II randomized trial. Clinical Cancer
Pollock, J. K., Greene, L. M., Nathwani, S. M., Kinsella, P., O’Boyle, N. M., Research, 23, 4651–4661.
Meegan, M. J., & Zisterer, D. M. (2018). Involvement of NF‐κB in Sharma, S., Yang, S.‐C., Zhu, L., Reckamp, K., Gardner, B., Baratelli, F., …
mediating the anti‐tumour effects of combretastatins in T cells. Dubinett, S. M. (2005). Tumor cyclooxygenase‐2/prostaglandin E2–
Investigational New Drugs, 36, 1–13. dependent promotion of FOXP3 expression and CD4+CD25+T
Qiang, L., Sample, A., Shea, C. R., Soltani, K., Macleod, K. F., & He, Y.‐Y. regulatory cell activities in lung cancer. Cancer Research, 65(12),
(2017). Autophagy gene ATG7 regulates ultraviolet radiation‐induced 5211–5220.
inflammation and skin tumorigenesis. Autophagy, 13, 2086–2103. Shi, L., Xu, L., Wu, C., Xue, B., Jin, X., Yang, J., & Zhu, X. (2018). Celecoxib‐
Qin, G., Xu, F., Qin, T., Zheng, Q., Shi, D., Xia, W., … Wang, S. (2015). induced self‐assembly of smart albumin‐doxorubicin conjugate for
Palbociclib inhibits epithelial‐mesenchymal transition and metastasis enhanced cancer therapy. ACS Applied Materials & Interfaces, 10(10),
in breast cancer via c‐Jun/COX‐2 signaling pathway. Oncotarget, 6(39), 8555–8565.
41794. Singh, B., Berry, J. A., Shoher, A., & Lucci, A. (2006). COX‐2 induces IL‐11
Qiu, J., Shi, Z., & Jiang, J. (2017). Cyclooxygenase‐2 in glioblastoma production in human breast cancer cells. Journal of Surgical Research,
multiforme. Drug Discovery Today, 22(1), 148–156. 131(2), 267–275.
Qiu, H.‐Y., Wang, P.‐F., Li, Z., Ma, J.‐T., Wang, X.‐M., Yang, Y.‐H., & Zhu, H.‐ Sorski, L., Melamed, R., Matzner, P., Lavon, H., Shaashua, L., Rosenne, E., &
L. (2016). Synthesis of dihydropyrazole sulphonamide derivatives that Ben‐Eliyahu, S. (2016). Reducing liver metastases of colon cancer in
act as anti‐cancer agents through COX‐2 inhibition. Pharmacological the context of extensive and minor surgeries through β‐adrenocep-
Research, 104, 86–96. tors blockade and COX2 inhibition. Brain, Behavior, and Immunity, 58,
Raj, V., Bhadauria, A. S., Singh, A. K., Kumar, U., Rai, A., Keshari, A. K., … 91–98.
Saha, S. (2018). Novel 1, 3, 4‐thiadiazoles inhibit colorectal cancer via Soto, M. S., O’brien, E. R., Andreou, K., Scrace, S. F., Zakaria, R.,
blockade of IL‐6/COX‐2 mediated JAK2/STAT3 signals as evidenced Jenkinson, M. D., … Sibson, N. R. (2016). Disruption of tumour‐host
through data‐based mathematical modeling. Cytokine. https://doi.org/ communication by downregulation of LFA‐1 reduces COX‐2 and
10.1016/j.cyto.2018.03.026 e‐NOS expression and inhibits brain metastasis growth. Oncotarget,
Ramer, R., Walther, U., Borchert, P., Laufer, S., Linnebacher, M., & Hinz, B. 7(32), 52375.
(2013). Induction but not inhibition of COX‐2 confers human lung Su, C.‐W., Zhang, Y., & Zhu, Y.‐T. (2016). Stromal COX‐2 signaling are
cancer cell apoptosis by celecoxib. Journal of Lipid Research, 54(11), correlated with colorectal cancer: A review. Critical Reviews in
3116–3129. Oncology/Hematology, 107, 33–38.
Ramu, A., Kathiresan, S., Ramadoss, H., Nallu, A., Kaliyan, R., & Azamuthu, Sun, W.‐H., Chen, G.‐S., Ou, X.‐L., Yang, Y., Luo, C., Zhang, Y., … Ding, G. X.
T. (2018). Gramine attenuates EGFR‐mediated inflammation and cell (2009). Inhibition of COX‐2 and activation of peroxisome proliferator‐
proliferation in oral carcinogenesis via regulation of NF‐κB and STAT3 activated receptor γ synergistically inhibits proliferation and induces
signaling. Biomedicine & Pharmacotherapy, 98, 523–530. apoptosis of human pancreatic carcinoma cells. Cancer Letters, 275(2),
Regulski, M., Regulska, K., Prukała, W., Piotrowska, H., Stanisz, B., & 247–255.
Murias, M. (2016). COX‐2 inhibitors: A novel strategy in the Sun, H., Zhang, X., Sun, D., Jia, X., Xu, L., Qiao, Y., & Jin, Y. (2017). COX‐2
management of breast cancer. Drug Discovery Today, 21(4), 598–615. expression in ovarian cancer: An updated meta‐analysis. Oncotarget,
Rong, X., Huang, B., Qiu, S., Li, X., He, L., & Peng, Y. (2016). Tumor‐ 8(50), 88152.
associated macrophages induce vasculogenic mimicry of glioblastoma Terrin, L., Agostini, M., Ruvoletto, M., Martini, A., Pucciarelli, S., Bettin, C.,
multiforme through cyclooxygenase‐2 activation. Oncotarget, 7(51), … Pontisso, P. (2017). SerpinB3 upregulates the Cyclooxygenase‐2/β‐
83976. Catenin positive loop in colorectal cancer. Oncotarget, 8(9), 15732.
16 | HASHEMI ET AL.

Theodoraki, M. N., Hoffmann, T., & Whiteside, T. (2018). Separation of Xiang, W., Shi, R., Kang, X., Zhang, X., Chen, P., Zhang, L., … Miao, H.
plasma‐derived exosomes into CD3 (+) and CD3 (–) fractions allows (2018). Monoacylglycerol lipase regulates cannabinoid receptor 2‐
for association of immune cell and tumour cell markers with disease dependent macrophage activation and cancer progression. Nature
activity in HNSCC patients. Clinical & Experimental Immunology, 192, Communications, 9(1), 2574.
271–283. Xu, Z., Choudhary, S., Voznesensky, O., Mehrotra, M., Woodard, M.,
Tian, J., Hachim, M. Y., Hachim, I. Y., Dai, M., Lo, C., Raffa, F. A., … Lebrun, J. Hansen, M., … Pilbeam, C. (2006). Overexpression of COX‐2 in human
J. (2017). Cyclooxygenase‐2 regulates TGFβ‐induced cancer stemness osteosarcoma cells decreases proliferation and increases apoptosis.
in triple‐negative breast cancer. Scientific Reports, 7, 40258. Cancer Research, 66(13), 6657–6664.
Tjiu, J.‐W., Chen, J.‐S., Shun, C.‐T., Lin, S.‐J., Liao, Y.‐H., Chu, C.‐Y., … Jee, Xu, W., Huang, Y., Zhang, T., Zhao, L., Fan, J., & Li, L. (2018).
S. H. (2009). Tumor‐associated macrophage‐induced invasion and Cyclooxygenase‐2 gene polymorphisms and susceptibility to hepato-
angiogenesis of human basal cell carcinoma cells by cyclooxygen- cellular carcinoma: A meta‐analysis based on 10 case‐control studies.
ase‐2 induction. Journal of Investigative Dermatology, 129(4), Journal of Cancer Research and Therapeutics, 14(8), 105.
1016–1025. Xu, Y., Yang, X., Wang, T., Yang, L., He, Y.‐Y., Miskimins, K., & Qian, S. Y.
Todoric, J., Antonucci, L., & Karin, M. (2016). Targeting inflammation in cancer (2018). Knockdown delta‐5‐desaturase in breast cancer cells that
prevention and therapy. Cancer Prevention Research, 9(12), 895–905. overexpress COX‐2 results in inhibition of growth, migration and
Tong, D., Liu, Q., Liu, G., Xu, J., Lan, W., Jiang, Y., … Jiang, J. (2017). invasion via a dihomo‐γ‐linolenic acid peroxidation dependent
Metformin inhibits castration‐induced EMT in prostate cancer by mechanism. BMC Cancer, 18(1), 330.
repressing COX2/PGE2/STAT3 axis. Cancer Letters, 389, 23–32. Xu, X., Zhu, G.‐Q., Zhang, K., Zhou, Y.‐C., Li, X.‐L., Xu, W., … Sun, W.‐H.
Tury, S., Becette, V., Assayag, F., Vacher, S., Benoist, C., Kamal, M., … (2017). Cyclooxygenase‐2 mediated synergistic effect of ursolic acid
Callens, C. (2016). Combination of COX‐2 expression and PIK3CA in combination with paclitaxel against human gastric carcinoma.
mutation as prognostic and predictive markers for celecoxib treat- Oncotarget, 8(54), 92770.
ment in breast cancer. Oncotarget, 7(51), 85124. Xuan, Y., Wang, J., Ban, L., Lu, J.‐J., Yi, C., Li, Z., … Deng, W. (2016).
Wagner, T. M., Mullally, J. E., & Fitzpatrick, F. A. (2006). Reactive lipid hnRNPA2/B1 activates cyclooxygenase‐2 and promotes tumor
species from cyclooxygenase‐2 inactivate tumor suppressor LKB1/ growth in human lung cancers. Molecular Oncology, 10(4), 610–624.
STK11 cyclopentenone prostaglandins and 4‐hydroxy‐2‐nonenal Yadav, D. K., Kumar, S., Saloni, S., Misra, S., Yadav, L., Teli, M., … Kim, M.
covalently modify and inhibit the AMP‐kinase kinase that modulates (2018). Molecular insights into the interaction of RONS and thieno [3,
cellular energy homeostasis and protein translation. Journal of 2‐c] pyran analogs with SIRT6/COX‐2: A molecular dynamics study.
Biological Chemistry, 281(5), 2598–2604. Scientific Reports, 8(1), 4777.
Wahle, B. M., Hawley, E. T., He, Y., Smith, A. E., Yuan, J., Masters, A. R., … Yang, H., Huang, S., Wei, Y., Cao, S., Pi, C., Feng, T., … Ren, G. (2017).
Yates, C. W. (2018). Chemopreventative celecoxib fails to prevent Curcumin enhances the anticancer effect of 5‐fluorouracil against
schwannoma formation or sensorineural hearing loss in genetically gastric cancer through down‐regulation of COX‐2 and NF‐κB signaling
engineered murine model of neurofibromatosis type 2. Oncotarget, 9 pathways. Journal of Cancer, 8(18), 3697–3706.
(1), 718. Yang, C.‐C., Tu, H.‐F., Wu, C.‐H., Chang, H.‐C., Chiang, W.‐F., Shih, N.‐C., …
Walther, U., Emmrich, K., Ramer, R., Mittag, N., & Hinz, B. (2016). Chang, K.‐W. (2016). Up‐regulation of HB‐EGF by the COX‐2/PGE2
Lovastatin lactone elicits human lung cancer cell apoptosis via a COX‐ signaling associates with the cisplatin resistance and tumor recur-
2/PPARγ‐dependent pathway. Oncotarget, 7(9), 10345. rence of advanced HNSCC. Oral Oncology, 56, 54–61.
Wang, D., Buchanan, F. G., Wang, H., Dey, S. K., & DuBois, R. N. (2005). Yang, X., Xu, Y., Wang, T., Shu, D., Guo, P., Miskimins, K., & Qian, S. Y.
Prostaglandin E2 enhances intestinal adenoma growth via activation (2017). Inhibition of cancer migration and invasion by knocking down
of the Ras‐mitogen‐activated protein kinase cascade. Cancer Research, delta‐5‐desaturase in COX‐2 overexpressed cancer cells. Redox
65(5), 1822–1829. Biology, 11, 653–662.
Wang, X., Reyes, M. E., Zhang, D., Funakoshi, Y., Trape, A. P., Gong, Y., … Ueno, Yao, Q., Gu, A., Wang, Z., & Xue, Y. (2018). MicroRNA‐144 functions as a
N. T. (2017). EGFR signaling promotes inflammation and cancer stem‐like tumor suppressor in gastric cancer by targeting cyclooxygenase‐2.
activity in inflammatory breast cancer. Oncotarget, 8(40), 67904. Experimental and Therapeutic Medicine, 15(3), 3088–3095.
Wang, X., Yu, Z., Wang, C., Cheng, W., Tian, X., Huo, X., … Xing, J. (2017). Yue, X., Nguyen, T. D., Zellmer, V., Zhang, S., & Zorlutuna, P. (2018).
Alantolactone, a natural sesquiterpene lactone, has potent antitumor Stromal cell‐laden 3D hydrogel microwell arrays as tumor micro-
activity against glioblastoma by targeting IKKβ kinase activity and environment model for studying stiffness dependent stromal cell‐
interrupting NF‐κB/COX‐2‐mediated signaling cascades. Journal of cancer interactions. Biomaterials, 170, 37–48.
Experimental & Clinical Cancer Research, 36(1), 93. Zhao, Y., Cui, L., Pan, Y., Shao, D., Zheng, X., Zhang, F., … Chen, L. (2017).
Wang, F., Zhang, H., Ma, A.‐H., Yu, W., Zimmermann, M., Yang, J., … Pan, C. X. Berberine inhibits the chemotherapy‐induced repopulation by sup-
(2018). COX‐2/sEH Dual Inhibitor PTUPB Potentiates the Antitumor pressing the arachidonic acid metabolic pathway and phosphorylation
Efficacy of Cisplatin. Molecular Cancer Therapeutics, 17(2), 474–483. of FAK in ovarian cancer. Cell Proliferation, 50(6), e12393.
Wang, J., Zou, K., Feng, X., Chen, M., Li, C., Tang, R., … Deng, W. (2017). Zhao, Q., Guo, J., Wang, G., Chu, Y., & Hu, X. (2017). Suppression of
Downregulation of NMI promotes tumor growth and predicts poor immune regulatory cells with combined therapy of celecoxib and
prognosis in human lung adenocarcinomas. Molecular Cancer, 16(1), 158. sunitinib in renal cell carcinoma. Oncotarget, 8(1), 1668.
Wong, J.‐H., Ho, K.‐H., Nam, S., Hsu, W.‐L., Lin, C.‐H., Chang, C.‐M., … Zhao, J., Zhu, J., Lv, X., Xing, J., Liu, S., Chen, C., & Xu, Y. (2017). Curcumin
Chang, W.‐C. (2017). Store‐operated Ca 2+ entry facilitates the potentiates the potent antitumor activity of ACNU against glioblas-
lipopolysaccharide‐induced cyclooxygenase‐2 expression in gastric toma by suppressing the PI3K/AKT and NF‐κB/COX‐2 signaling
cancer cells. Scientific Reports, 7(1), 12813. pathways. OncoTargets and Therapy, 10, 5471–5482.
Wu, M., Guan, J., Li, C., Gunter, S., Nusrat, L., Ng, S., … Das, S. (2017). Zheng, N., Chen, J., Li, T., Liu, W., Liu, J., Chen, H., … Jia, L. (2017).
Aberrantly activated Cox‐2 and Wnt signaling interact to maintain Abortifacient metapristone (RU486 derivative) interrupts CXCL12/
cancer stem cells in glioblastoma. Oncotarget, 8(47), 82217. CXCR4 axis for ovarian metastatic chemoprevention. Molecular
Xian, X., Huang, L., Zhang, B., Wu, C., Cui, J., & Wang, Z. (2016). WIN Carcinogenesis, 56(8), 1896–1908.
55,212‐2 Inhibits the Epithelial Mesenchymal Transition of Gastric Zheng, N., Chen, J., Liu, W., Liu, J., Li, T., Chen, H., … Jia, L. (2017).
Cancer Cells via COX‐2 Signals. Cellular Physiology and Biochemistry, Mifepristone inhibits ovarian cancer metastasis by intervening in
39(6), 2149–2157. SDF‐1/CXCR4 chemokine axis. Oncotarget, 8(35), 59123.
HASHEMI ET AL. | 17

Zheng, X., Jia, B., Song, X., Kong, Q.‐Y., Wu, M.‐L., Qiu, Z.‐W., … Liu, J.
(2018). Preventive Potential of Resveratrol in Carcinogen‐Induced How to cite this article: Hashemi Goradel N, Najafi M, Salehi
Rat Thyroid Tumorigenesis. Nutrients, 10(3), 279. E, Farhood B, Mortezaee K. Cyclooxygenase‐2 in cancer: A
Zhou, T.‐J., Zhang, S.‐L., He, C.‐Y., Zhuang, Q.‐Y., Han, P.‐Y., Jiang, S.‐W., …
review. J Cell Physiol. 2018;1–17.
Lin, Z. N. (2017). Downregulation of mitochondrial cyclooxygenase‐2
inhibits the stemness of nasopharyngeal carcinoma by decreasing the https://doi.org/10.1002/jcp.27411
activity of dynamin‐related protein 1. Theranostics, 7(5), 1389–1406.

Das könnte Ihnen auch gefallen