Abstract
In the wake of health disasters associated with the chronic use of cyclooxygenase-2 (COX-2) inhibitor drugs, it has been widely proposed that modulation of downstream prostanoid synthases or receptors might provide more specificity than simply shutting down the entire COX cascade for anti-inflammatory benefits. The pathogenic actions of COX-2 have long been thought attributable to the prostaglandin E2 (PGE2) signaling through its Gαs-coupled EP2 receptor subtype; however, the truly selective EP2 antagonists did not emerge until 2011. These small molecules provide game-changing tools to better understand the EP2 receptor in inflammation-associated conditions. Their applications in preclinical models also reshape our knowledge of PGE2/EP2 signaling as a node of inflammation in health and disease. As we celebrate the 10-year anniversary of this breakthrough, the exploration of their potential as drug candidates for next-generation anti-inflammatory therapies has just begun. The first decade of EP2 antagonists passes, while their future looks brighter than ever.
Graphical Abstract

1. INTRODUCTION
Cyclooxygenase (COX) is the key enzyme in the two-step biosynthesis of prostanoids from arachidonic acid (AA), a 20-carbon fatty acid, after it is liberated from the cell membrane by cytoplasmic phospholipase A2 (cPLA2) (Figure 1A).1,2 COX has two currently known isoforms, namely, COX-1 and COX-2, which are encoded by ptgs1 and ptgs2, respectively. On the one hand, the COX-1 isozyme is constitutively expressed to play some essential roles, such as maintaining the integrity of the gastrointestinal tract lining and preserving normal renal and cardiovascular functions.1 COX-2, on the other hand, is usually expressed at low basal levels in most normal tissues and organs. However, it can be rapidly and robustly upregulated in response to inflammatory and mitogenic stimuli, thereby commonly being considered as an early response gene.1,2 Both COX-1 and COX-2 catalyze AA to prostaglandin H2 (PGH2), which is an intermediate molecule and can be further converted by tissue-specific prostanoid synthases to five forms of prostanoids (Figure 1A): prostaglandin D2 (PGD2), prostaglandin E2 (PGE2), prostaglandin F2α (PGF2α), prostacyclin or prostaglandin I2 (PGI2), and thromboxane A2 (TXA2).1 These short-lived lipid-like bioactive molecules in turn dynamically activate several membrane-bound G protein-coupled receptors (GPCRs): four receptors (EP1, EP2, EP3, and EP4) activated by PGE2; two (DP1 and DP2) activated by PGD2; each of the other three prostanoids (PGF2α, PGI2, and TXA2) acts on only one receptor, that is, FP, IP, and TP, respectively (Figure 1A).3,4
Figure 1.

COX-2/mPGES-1/PGE2/EP2 signaling pathways. (A) In response to inflammatory or injurious stimuli, AA, a 20-carbon fatty acid, is released from the cell membrane by cPLA2 and then converted to PGH2 by COX. The short-lived PGH2 is further catalyzed by tissue-specific prostanoid synthases to five types of prostanoids: PGD2, PGE2, PGF2α, PGI2, and TXA2. Three isozymes that are responsible for the conversion of PGH2 into PGE2 are mPGES-1, mPGES-2, and cPGES. Prostanoids act on a total of nine membrane-bound receptors as indicated for various physiological and pathological functions. (B) Activation of the EP2 receptor leads to both G protein-dependent and independent pathways, which often crosstalk with other pathways including the EGFR-mediated signaling. Targeting the EP2 receptor by selective small-molecule antagonists has been proposed to alleviate the downstream pathogenic functions that are mediated by the EP2 receptor.
Interestingly, despite their sequence and structural similarities, COX-1 and COX-2 have different major enzymatic products.5 Particularly, COX-2 is found to be functionally coupled to microsomal prostaglandin E synthase 1 (mPGES-1), which is also inducible and catalyzes COX-2-derived PGH2 into PGE2 in response to inflammatory or injurious stimuli.2 The mPGES-2 and cytosolic PGES (cPGES) are the other two PGES isoenzymes that synthase PGE2 from COX-1-produced PGH2 at basal physiological conditions (Figure 1A). PGE2 is known to produce pleiotropic effects throughout the body including immunogenicity, inflammation, vasodilation, platelet aggregation, and memory formation, and it is thought to perpetuate the COX-2 expression through a positive feedback loop.6–9 PGE2 regulates these physiological and pathological events via interactions with four GPCRs, EP1–EP4 (Figure 1A). The EP1 receptor is Gαq-coupled to regulate the mobilization of cytosolic Ca2+ and the activation of protein kinase C (PKC); EP2 and EP4 are linked to Gαs for cAMP-dependent pathways; the EP3 receptor is mainly coupled to Gαi to downregulate the cAMP signaling.10,11 PGE2 signaling via the EP2 subtype has been demonstrated to be involved in many of the immunoregulatory, inflammatory, neurotoxic, and other pathophysiological effects that are mediated by the COX-2/PGE2 signaling cascade under various disease conditions10,12–27
2. COX AND PGES INHIBITORS
Nonsteroidal anti-inflammatory drugs (NSAIDs) such as aspirin, ibuprofen, and naproxen are nonspecific inhibitors of both COX isoenzymes (Figure 1A). These drugs are widely used to reduce pain, fever, and inflammation, but their mechanisms of action were not fully elucidated until the early 1970s.28 NSAIDs act by inhibiting the production of prostanoids, which may initiate signaling pathways that are essential to some important physiological functions in many organs and tissues under normal conditions; thus, their adverse effects can be widespread. Depending on the specific drug and dose used, NSAIDs can damage the gastrointestinal tract and lead to kidney, cardiovascular, and liver dysfunctions, which are believed to be caused by inhibition of COX-1.29
In the hopes of mitigating the side effects associated with a nonselective COX inhibition, focus has been shifted to COX-2 for more specificity. The COX-2 enzyme was originally discovered and sequenced in 1988 and then confirmed in 1991,30 fostering the development of selective small-molecule inhibitors for COX-2 (coxibs) (Figure 1A). Less than eight years later, the first coxibs were developed and then quickly introduced to the market, with Vioxx (rofecoxib) and Celebrex (celecoxib) launched in 1999. These selective COX-2 inhibitors are very effective to alleviate pain and other inflammation-related complications without causing major gastrointestinal ulceration and bleeding or other nonselective COX inhibitors-associated side effects and, thus, were thought safer than the conventional NSAIDs.31 Unfortunately, it soon became evident that coxibs were associated with severe cardiac side effects in patients taking them as long-term medication,32–34 suggesting that a certain COX downstream prostanoid signaling pathway might function to protect the microvascular systems.35 Targeting the inducible mPGES-1 to inhibit the PGE2 synthesis from COX-2-derived PGH2 without affecting other types of prostanoids was thought more specific than inhibiting COX-2 itself. However, designing and testing compounds to inhibit this enzyme have proved more convoluted than previously thought due to the interspecies differences in the sequence and structure of the mPGES-1 enzyme.36,37 Thus, to circumvent these complications derived from inhibiting biosynthetic enzymes for PGE2, the downstream PGE2 receptor EP2 has been proposed as an alternative and hopefully more specific target owing to its leading role in the COX-2-mediated inflammatory signaling cascade.16
3. PGE2/EP2 SIGNALING
The prostaglandin receptor EP2 is coupled to a heterotrimeric Gs protein complex consisting of α, β, and γ subunits. The activation of EP2 receptor by PGE2 quickly separates the Gs complex into Gα and Gβγ, which in turn regulate diverse downstream signaling molecules that also can crosstalk with many other pathways (Figure 1B).11 First and foremost, Gαs activates adenyl cyclase (AC) to increase cellular levels of cAMP, thereby activating protein kinase A (PKA) and exchange factor directly activated by cAMP (EPAC). On the one hand, the cAMP-stimulated EPAC then, in turn, can activate its downstream effectors Rap GTP-binding proteins 1/2 (RAP1/2) to regulate many pathogenic events such as neuroinflammation and neurotoxicity (Figure 1A).10,38,39 On the other hand, the phosphorylated PKA acts on the transcription factor cAMP response element-binding protein (CREB), which then translocates to the nucleus, where it can regulate the transcription of responding genes.16 Additionally, EP2 receptor, through the dissociated Gβγ subunits upon PGE2 binding, activates the phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB or Akt) pathway, which causes the phosphorylation and inactivation of glycogen synthase kinase 3β (GSK-3β), eventually leading to the stabilization and nuclear translocation of β-catenin and the expression of growth-promoting genes and inflammation.40–42 Activated PI3K/Akt may also phosphorylate the transcription factor nuclear factor κB (NF-κB), which then translocates to the nucleus to regulate the transcription of a wide variety of genes involved in inflammation.43,44 Interestingly, PGE2/EP2 signaling can transactivate the epidermal growth factor receptor (EGFR) likely via PKA and Src, leading to the activation of the Ras/Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway and cell proliferation and invasion.44–46 The EP2 receptor also mediates a G protein-independent pathway via association with β-arrestin,47 resulting in the activation of a c-Jun N-terminal kinase (JNK)/profilin 1 (Pfn-1)/filamentous actin (F-actin) pathway for cell migration and proliferation.48 In addition, the transactivation of EGFR by PGE2/EP2 is likely mediated by β-arrestin, which can phosphorylate and activate Src (Figure 1B).49 Unlike other prostanoid receptors (e.g., EP4), the EP2 receptor usually does not undergo internalization and thus resists desensitization even upon a repeated stimulation by PGE2,2,50 allowing for a continuous receptor signaling and drug effects.
The EP2 receptor is widely expressed in the human body and has been primarily detected in the central nervous system (CNS), bones, reproductive system, leukocytes, and smooth muscle,51,52 where it regulates a variety of physiological functions, such as immunoregulation, ovulation and fertilization, vasodilation, osteoclasts, regulation of intraocular pressure, neuronal plasticity, learning, and memory. However, the deregulated PGE2 signaling via the EP2 receptor may contribute to the molecular and cellular mechanisms of a number of inflammation-associated pathogenic processes, such as tumorigenesis, oxidative stress, amyloid β (Aβ) accumulation, neurotoxicity, neurodegeneration, and neuroinflammation.16,17,19,38,53–57 The broad pathophysiological roles of the excessive PGE2/EP2 signaling taken together lend support to a translational strategy suppressing the EP2 receptor to harness the uncontrolled inflammation in these disease states.
4. EP2 ANTAGONISTS
Genetic ablation of EP2 (EP2−/−) was an inhibitory strategy available in early investigations to elucidate the physiological and pathological roles of the EP2 receptor. Though very useful, it was also complicated by the developmental and other homeostatic adjustments in mice that resulted in hypertension and reduced litter size.58–60 In the meantime, small-molecule agonists of the EP2 receptor that have been widely used include butaprost, CP-533536, CAY10399, ONO-AE1–259, and C-9 (Figure 2).16,61,62 However, butaprost is only ~18-fold selective for EP2 over EP3;63 CP-533536 (evatanepag) is ~64-fold selective over the EP4 receptor;64 CAY10399 and ONO-AE1–259 are highly selective on EP2 but have prostanoid-like structures; C-9 is quite selective against other PGE2 receptors but less than fourfold selective over the TP receptor.65 Positive allosteric modulators for EP2 with nonprostanoid structures have been reported and provide alternative chemical probes to study the receptor in the presence of PGE2;66,67 however, the uncertainty of their pharmacokinetic properties impedes their uses in vivo.
Figure 2.

Conventional ligands and compounds that have been used to study EP2 receptor. Derived from PGE2, the EP2 agonists including butaprost, ONO-AE1–259, and CAY10399 have prostanoid-like structures. CP-533536 (evatanepag) and C-9 are EP2 agonists with nonprostanoid structures. AH-6809 is a poorly selective antagonist that inhibits EP1, EP2, and DP1 receptors.
A critical reason to explain the frustration in the early investigations of the EP2 receptor in health and disease was the lack of a suitable EP2 receptor antagonist.16 Among all nine currently known prostanoid receptors, EP2 was the last one that did not have a truly selective small-molecule antagonist until 2011. Although the underlying reason is unclear, a recent study on the cryo-electron microscopy structure of the EP2-Gs complex revealed its unconventional conformation in the active state when compared to those of all currently known active type 1 GPCRs.68 Prior to 2011, a commonly used antagonist to study the EP2 receptor was AH-6809 (Figure 2). Unfortunately, AH-6809 is neither selective nor potent—it weakly inhibits both EP1 and EP2 without palpable preference—and is unsuitable for in vivo studies.63 To overcome these limitations, researchers from Pfizer, Emory, and Amgen independently developed the first truly selective EP2 antagonists with well-characterized pharmacokinetic and pharmacodynamic profiles that are suitable for both in vitro and in vivo testing.69–71 The rest of this review focuses on the discovery and optimization of these EP2 antagonists, followed by their evaluations in various preclinical models of peripheral and central inflammation-associated conditions.
4.1. Pfizer Compounds.
In 2011, Pfizer reported its first EP2-selective antagonist, compound 1 (Figure 3), which showed promise in both in vitro and in vivo assays.69 Compound 1 did not show any agonistic activity, nor did it exert any effects on EP2 without the presence of PGE2 or another agonist. These results indicate that it specifically counteracts the effects of PGE2 or other agonists acting on the EP2 receptor.69,72 Compound 1 also displayed promising pharmacokinetic profiles and good tissue-penetrating properties and, thus, allowed studies on EP2 antagonism in an in vivo setting. Moreover, it was able to induce a relaxation of mouse trachea, indicating its therapeutic potential in asthmatic diseases. Two years later, Pfizer reported its second EP2-selective antagonist, compound 2 (Figure 3), which was found ~10-fold more potent than the previously reported compound 1 for the antagonism of PGE2 (functional EP2 Ki: 0.63 vs 7.6 nM).73 Just like compound 1, the newly reported compound showed good selectivity on EP2 over other Gs-coupled prostanoid receptors. Both Pfizer compounds, but not EP4-selective antagonist CJ-042794, powerfully antagonized the PGE2-provoked cAMP signaling and the consequent inhibition of IgE-dependent histamine release in human lung mast cells in a competitive mechanism, although both EP2 and EP4 receptors were expressed in these cells.73 These interesting findings suggest that the principal mechanism whereby PGE2 inhibits a mediator release in lung mast cells is mainly through activation of the EP2 receptor, and thus, these EP2 antagonists likely have therapeutic potential to treat asthma and other respiratory diseases. The brain penetration of these two compounds has not been reported by Pfizer; however, a recent study revealed that compound 1 was unable to cross the blood-brain barrier,26 suggesting that its action is only limited in the periphery. Currently, there is no study on the structure–function relationships (SAR) of these Pfizer compounds, nor are there any new EP2 antagonists reported by Pfizer. Their current status for further development is unknown.
Figure 3.

Selective EP2 antagonists that were developed by Pfizer and tested in animal models. Only two compounds PF-04418948 (1) and PF-04852946 (2) have been reported. As EP2 antagonists, compound 2 is ~10-fold more potent than compound 1, as their functional Ki values are 0.63 and 7.6 nM, respectively.
4.2. Emory Compounds.
In 2008, utilizing a set of cell-based time-resolved fluorescence resonance energy transfer (TR-FRET) assays of cAMP formation, researchers at Emory University performed a high-throughput screening (HTS) of 262 371 compounds (https://pubchem.ncbi.nlm.nih.gov/bioassay/1422). Because of the lack of a truly selective EP2 antagonist back then, their goal was to develop compounds that selectively inhibit the EP2 receptor and thus can be used to study a prolonged seizure-induced brain inflammation. Consequently, a series of small molecules were identified as competitive antagonists of the human EP2 receptor. Among these, compound 3 (Figure 4A) is the most potent compound with a functional Schild KB of 2.4 nM for the antagonism of PGE2 and showed a plasma half-life (t1/2) of 0.6 h and a brain-to-plasma ratio of 0.3.70 They then performed the first SAR study based on the 3-aryl-acrylamide scaffold in compound 3 and disclosed that an indole moiety, ethylene linker, acrylamide, and methoxyphenyl or halogenphenyl promoted activities in the nanomolar level. Meanwhile, the introduction of fluorine atoms into the compound 3 scaffold improved the metabolic stability but slightly decreased the potency of the compounds, leading to the discovery of compound 4 (Figure 4A).14,70,74–77 Compared to compound 3, compound 4 is less potent with a functional Schild KB of 17.8 nM on human EP2 but has an acceptable extended plasma half-life (t1/2) of 1.6 h and a favorable brain-to-plasma ratio of 1.6. In addition, both compounds 3 and 4 demonstrated a similar potency on human, mouse, and rat EP2 receptors,14,70 justifying their use in both animal models and human conditions.78 Off-target profiling of compounds 3 and 4 displayed negligible effects on a panel of more than 40 critical ion channels, enzymes, receptors, and neurotransmitter transporters,14,15 attesting to their safety for in vivo uses.
Figure 4.

EP2 antagonists that were developed by Emory University and extensively evaluated in preclinical models. (A) TG4–155 (3) is a hit compound from an ultrahigh-throughput screening (uHTS). Introduction of fluorine atoms into the compound 3 scaffold led to the discovery of TG6–10–1 (4) with an improved metabolic stability but a reduced potency. (B) TG8–69 (5) and TG11–77·HCl (6) are representative second-generation EP2 antagonists with improved pharmacodynamic and pharmacokinetic properties. (C) TG6–129 (7) is another hit from the same uHTS but with a distinct chemical scaffold. The functional Schild KB values of compounds are listed to indicate their potency and selectivity.
Additional optimization and SAR studies on the core structure of compounds 3 and 4 led to the development of second-generation EP2 antagonists compound 5 and compound 6, in which the indol-1-yl is replaced with an indol-3-yl and the acrylamide linker is replaced with an amide (Figure 4B).79,80 These modifications enable compounds 5 and 6 to maintain the nanomolar efficacy and enhance the selectivity to EP2 receptor over DP1. In addition, an introduction of nitrogen-containing heterocycles greatly improves their water solubility, yielding the water-soluble EP2 antagonists. Further SAR study reveals that the tetrazole ring is good for EP2 potency, solubility, and metabolic stability.75 On the one hand, indeed, tetrazole ring-containing compound 5 has a functional EP2 KB of 48.5 nM and is highly soluble (500 μM in water), metabolically stable in vivo (plasma half-life in mice: 10.5 h), and peripherally restricted (brain-to-plasma ratio: 0.02).79 On the other hand, another SAR study on the middle phenyl ring reinforces the notion that nitrogen in the ring can enhance the aqueous solubility, leading to the discovery of compound 6, which has a Schild KB of 9.7 nM on EP2, a water solubility of 2.5 mM, a brain-to-plasma ratio of 0.4, and a plasma half-life of 2.4 h in mice.80 Overall, these two second-generation EP2 antagonists show much improved selectivity against the other three Gαs-coupled prostanoid receptors (DP1, EP4, and IP) when compared to their precursors.
Compound 7 was among the original hits from the Emory HTS and possesses a distinct chemical scaffold, a carbamothioylacrylamide backbone (Figure 4C). This compound has a functional EP2 KB of 8.8 nM, and with a systemic administration in mice it showed a plasma half-life of 2.7 h and a brain-to-plasma ratio of 0.02.81 The low brain penetration of compounds 5 and 7 enables these two compounds to be ideal tools to study the functions of the EP2 receptor in peripheral diseases associated with chronic inflammation such as rheumatoid arthritis and chronic obstructive pulmonary disease, in which EP2 appears to play essential pathogenic roles.82–84 Molecular docking using the recently solved cryo-electron microscopy (cryo-EM) structure of the human EP2 receptor and Gs protein complex (PDB code: 7CX3) revealed the simulated interactions between the EP2 receptor and Emory compounds using Schrödinger software,68,85 exemplified by compound 3 (Figure 5). Understanding the dynamic three-dimensional (3-D) interactions between these competitive antagonists and the EP2-Gs complex might help with better rational designs to develop the next-generation EP2 antagonists with more balanced potency and selectivity.
Figure 5.

Simulated antagonist-EP2-Gs protein complex. Competitive antagonist compound 3 was docked into the cryo-EM structure of the human EP2 receptor (PDB code: 7CX3) using Glide (Schrödinger Maestro 2021–2). The receptor and G proteins are shown as ribbons (cyan, green, dark blue, gray). The ligand compound 3 is depicted as the ball-and-stick model, where carbons are shown in purple, nitrogens in blue, and oxygens in red. Pink dashed lines represent π-π stacking, and yellow dashed lines indicate hydrogen bonds. Key interacting residues are displayed in orange, with residues labeled correspondingly. Each of the three residues identified here have been implicated as residues important for the binding capabilities of ligands within the active site of the EP2 receptor. A two-dimensional representation of the ligand interactions within the active site of the EP2 receptor is also displayed on the bottom. ECL2, extra cellular loop 2.
4.3. Amgen Compounds.
In 2015, an HTS of a small-molecule library by researchers at Amgen led to the identification of compound 8 (Figure 6) as a novel EP2 antagonist with a moderate to mild potency across the human, mouse, and rat receptors. In selectivity tests, it was more than 400-fold selective against the EP1 receptor, 300-fold selective against EP3, but only 50-fold selective over EP4, the other Gαs-coupled receptor for PGE2. Compound 8 had a low metabolic stability in liver microsomes and significantly inhibited both CYP3A4 and CYP2D6 and71 raised concerns of drug–drug interactions because these two CYP enzymes metabolize more than 50% of clinically important drugs. Compound 8 then underwent several lead-optimization steps to improve its metabolic stability, reduce its inhibition on CYP3A4 and CYP2D6, and increase the selectivity against the EP3 and EP4 receptors. Subsequent SAR studies revealed that the replacements for the benzothiophene ring, pyridine ring, and methyl ether of compound 8 could address all these liabilities (Figure 6) and, eventually, resulted in the discovery of compound 9, which instead has a fluoroindole ring, pyridone ring, and chlorobenzoxazepine (Figure 6). These changes afforded compound 9 a single-digit nanomolar binding affinity for the EP2 receptor and significantly improved microsomal stability. Importantly, compound 9 demonstrated a comparable potency across the human, mouse, and rat EP2 receptors, was greater than 660-fold selective in the EP2 cAMP assay against DP1, EP4, and IP receptors, and was devoid of CYP inhibition.71 With an oral dose, compound 9 had a plasma half-life of 3.4 h in CD-1 mice and brain-to-plasma ratios of 0.7 and 0.9 in C57BL/6 mice and Sprague–Dawley rats, respectively.71 The favorable in vivo half-life and high brain penetration as well as a capability of increasing the macrophage-mediated clearance of amyloid-beta plaques ex vivo raise the therapeutic potential of compound 9 for Alzheimer’s disease (AD) and other inflammation-associated CNS conditions.26,71
Figure 6.

EP2 antagonists developed by Amgen. Benzoxazepine-1 (8) is a hit from a small HTS. Benzoxazepine-52 (9) is a lead compound with dramatically improved pharmacodynamic and pharmacokinetic properties. The half-maximal inhibitory concentration (IC50) values from SPA binding assays are listed to indicate their potency and selectivity.
5. EPILEPTIC SEIZURES
A prolonged convulsive seizure or status epilepticus (SE) is a common life-threatening condition that can cause significant morbidity and mortality. Resulting from the failure of the mechanisms in charge of seizure termination, SE is traditionally defined as a continuous seizure lasting longer than 30 min or two or more separate seizures without a complete recovery of consciousness between any of them.86 Despite the introduction of a newer generation of antiseizure drugs (ASDs) during the past few decades, SE remains the second most common neurological emergency only after acute stroke. With more than 20% mortality across various populations, SE accounts for hundreds to thousands of annual deaths in the United States (U.S.) alone.87 SE in humans triggers a cascade of molecular, cellular, and systemic events in the brain that can eventually aggravate the occurrence of unprovoked seizures—that is, development of epilepsy—in survivors.88 Prolonged seizures induced by proconvulsive agents such as kainic acid, pilocarpine, and organophosphate in experimental rodents trigger similar reactions within brain tissue, such as pro-inflammatory processes engaging cytokines, reactive gliosis, blood-brain barrier breakdown, neuronal death, and changes in the synaptic efficacy.89–91 Given their recapitulation of many important features of human SE conditions, such as mortality, behavioral abnormality, and the development of life-long epilepsy,91 these animal models have been commonly used in preclinical studies to evaluate COX inhibitors, EP2-selective antagonists, and other anti-inflammatory compounds for therapeutic benefits,92 as both COX-2 and EP2 receptor in the brain are rapidly induced by SE.18,93,94
5.1. Pilocarpine-Induced SE.
As a commonly used proconvulsive agent, pilocarpine with systemic administration can quickly trigger a continuous seizure-like behavior in mice and rats, which closely simulates the SE observed in human. Without intervention, a pilocarpine-induced SE usually lasts for hours, making it a suitable model to study SE-related neuronal injury, oxidative stress, and inflammation in the brain.95–97 As the first EP2-selective antagonist that was ever tested in an animal seizure model (Table 1), compound 3 dramatically reduced a hippocampal injury when administered in mice beginning 1 h after the termination of a pilocarpine-induced SE.70 The neuroprotective effect of compound 3 was extended by broader beneficial effects of its analogue compound 4 in the same mouse model of SE (Table 1), where compound 4 reduced the mortality rate of pilocarpine-treated mice by 30% at the end of the first week and by 35% after two months.14,93 Moreover, the neuroprotective effect of compound 4 was accompanied by a substantial downregulation of pro-inflammatory mediators, such as oxidative stress enzymes, cytokines, and chemokines, in addition to blocking the feedback amplification of the COX-2 cascade upon EP2 activation.14,22,93,94 Taken together, these findings demonstrated that the EP2 inhibition completely recapitulated the multiple benefits of a conditional deletion of COX-2 from a restricted population of forebrain neurons in a mouse pilocarpine model98 and suggested that COX-2-mediated pathogenic effects after the SE should be mainly attributed to its downstream PGE2/EP2 signaling.14,99
Table 1.
Emory EP2 Antagonists That Were Tested in Preclinical Modelsa
| compound | animal model | major outcomes | reference |
|---|---|---|---|
| 3 | Pilocarpine-induced SE in mice | Compound 3 treatment after SE decreased acute neuronal death in the hippocampus. | Jiang et al., 2012 |
| 4 | Pilocarpine-induced SE in mice | Treatment with compound 4 after SE reduced delayed mortality, seizure-provoked weight loss, functional deficits, brain inflammation, RRR opening, and acute neuronal death in the hippocampus, but did modify SE. | Jiang et al., 2013; Jiang et al., 2015 |
| DFP-induced SE in rats | Treatment with compound 4 after DFP-induced SE in rats decreased the weight loss, acute neuronal death in the hippocampus, inflammatory cytokine burst, reactive microgliosis, and the BBB breakdown, and alleviated the DFP SE-induced memory impairment, but did not prevent the anxiety-like behavior. | Rojas et al., 2015; 2016; 2020 | |
| Retinitis pigmentosa in rd10 mice | Compound 4 treatment prevented activation of retinal microglia, suppressed excessive generation of prototypic proinflammatory cytokines, rescued retinal function and visual performance, and reduced degeneration of retinal photoreceptors. | Zabel et al., 2016 | |
| Mouse model of endometriosis-induced pain | Treatment with compound 4 reversed abdominal and paw hyperalgesia associated with endometriosis. | Greaves et al., 2017 | |
| Kainic acid-induced SE in mice | Treatment with compound 4 after SE reduced seizure-promoted functional deficits, inflammatory cytokine induction, reactive gliosis, BBB impairment, and hippocampal damage, but did not alter seizures. | Jiang et al., 2019; Varvel et al., 2021 | |
| Mouse xenograft model of glioblastoma | Oral treatment with compound 4 suppressed malignant glioma growth and increased survival rates of mice that harbored intracranial tumors formed by human glioblastoma cells. | Qiu et al., 2019 | |
| Mouse xenograft model of neuroblastoma | Treatment with compound 4 moderately impaired the growth of human neuroblastoma xenografts in nude mice. | Hou et al., 2020 | |
| LPS-induced systemic inflammation in mice | Compound 4 treatment after LPS-triggered systemic inflammation facilitated the recovery of body weight, mitigated brain inflammation and microgliosis, prevented the loss of synaptic proteins, and ameliorated the depression-like behavior and cognitive deficits. | Jiang et al., 2020 | |
| Mouse model of transient MCAO | Compound 4 treatment after MCAO improved overall neurological score, decreased lesion size, and downregulated gene expression of pro-inflammatory cytokines. | Li et al., 2020 | |
| Experimental febrile SE in rats | Administration of compound 4 with three doses after eFSE had minimal effects on the total number of aberrant EEG spikes recorded in the first 7–60 days after eFSE, nor did it alter the duration of each spike series provoked by eFSE. | Brennan et al., 2021 | |
| 7 | Mouse xenograft model of neuroblastoma | Treatment with compound 7 substantially impaired the growth of human neuroblastoma xenografts, decreased angiogenesis, and downregulated proinflammatory cytokine release. | Hou et al., 2020 |
| 10 | Pilocarpine-induced SE in rats | Compound 10 administered after pilocarpine-induced SE reduced hippocampal neuroinflammation and gliosis but did affect SE-triggered neuronal injury or BBB breakdown. | Rojas et al., 2021 |
Abbreviations: BBB, blood-brain barrier; DFP, diisopropyl fluorophosphate; eFSE, experimental febrile status epilepticus; MCAO, middle cerebral artery occlusion; rd10, retinal degeneration 10; SE, status epilepticus.
Continual efforts in medicinal chemistry and lead optimization to improve the drug-like properties of small-molecule EP2 antagonists 3 and 4 led to the discovery of a second-generation EP2 antagonist, TG8–260 (10). Unlike the first-generation EP2 antagonists (Figure 4A), compound 10 does not possess an acrylamide moiety; however, its structure was not fully disclosed until recently.100 Nonetheless, compound 10 was tested in a TR-FRET cAMP functional assay with a Schild KB of 13.2 nM and a water solubility of 238 μM. Strikingly, compound 10 showed a more than 500-fold selectivity to EP2 over other Gαs-coupled prostanoid receptors DP, EP4, and IP,100 which is a significant improvement compared to compounds 3 and 4. With an intraperitoneal administration in C57BL/6 mice, compound 10 showed a terminal plasma half-life of 2.8 h and an extremely low brain penetration with a brain-to-plasma ratio of 0.02, which is ~15-and 80-fold less than that of compounds 3 and 4, respectively. Interestingly, a 60 min episode of pilocarpine-induced SE in Sprague–Dawley rats was able to significantly increase the brain-to-plasma ratio of compound 10 from 0.03 to 0.05, suggesting that the SE-induced blood-brain barrier (BBB) opening facilitated its entry to the brain parenchyma.100 EP2 receptor inhibition by compound 10 administered in adult male Sprague–Dawley rats beginning 2 h after a pilocarpine-induced SE considerably reduced hippocampal neuroinflammation and gliosis (Table 1). However, SE-triggered neuronal injury and BBB breakdown were not mitigated by the treatment with compound 10,100 suggesting that the lack of brain penetration prevented the compound from providing favorable beneficial effects like what the first-generation EP2 antagonists did in the pilocarpine model of seizures. Nevertheless, both first-generation (compound 4) and second-generation (compound 10) EP2 antagonists exhibited powerful anti-inflammatory activities following pilocarpine-induced SE.
5.2. Kainic Acid-Induced SE.
As the two most commonly used chemoconvulsants in animal models, pilocarpine and kainic acid trigger prolonged seizures that share most fundamental commonalities that simulate human SE conditions, such as mortality, behavioral abnormality, and the development of life-long epilepsy.14,89,91,96,101 However, the proconvulsant effect of pilocarpine is caused by its direct activation of the muscarinic acetylcholine receptor subtype M1,102 while kainic acid induces experimental seizures through selectively acting on the glutamate receptor subtypes GluK1 in interneurons and GluK2 in principal neurons.103,104 Therefore, these two convulsive agents are likely to have different peripheral and central effects. As such, it is critical to use both models to test therapeutic agents to identify and exclude any model-specific outcomes. The beneficial effects of EP2 inhibition in pilocarpine-treated mice and rats encouraged studies to test EP2 antagonists in a kainic acid model of SE, and compound 4 was first selected owing to its adequate in vivo half-life and favorable brain penetration in rodents.14,74 It has been found that EP2 inhibition by systemic administration of compound 4 after a 1 h episode of kainic acid-induced SE in mice reduced seizure-triggered functional deficits, cytokine induction, reactive gliosis, BBB impairment, and hippocampal damage.18 Importantly, most of the beneficial effects by compound 4 were independently reproduced in a similar mouse kainic acid model of SE (Table 1).27 The common, broad, reproducible benefits from a post-SE treatment with compound 4 after pilocarpine- and kainic acid-induced seizures eliminate the possibility that the neuroprotective and anti-inflammatory effects of EP2 antagonism following SE is model-specific and reinforce the feasibility of blocking PGE2/EP2 signaling as an adjunctive strategy to treat prolonged seizures. In a recent study, BI1029539, a selective mPGES-1 inhibitor, prevented an upregulation of P-glycoprotein expression and transport activity in capillaries from humanized mPGES-1 mice after kainic acid-induced SE.105 Whether PGE2/EP2 is also involved in SE-promoted P-glycoprotein expression and transport activity in capillaries remains to be determined.
Evidence from mounting preclinical studies over the past two decades suggests that the irregular brain-derived neurotrophic factor (BDNF) signaling via its tropomyosin-related kinase receptor B (TrkB) is essential to acquired epilepsy of etiologies such as traumatic brain injury (TBI) and de novo SE.106–109 It thus has been proposed that blocking BDNF/TrkB signaling or the downstream effector phospholipase C γ1 (PLC-γ1) might provide promising strategies to interrupt acquired epileptogenesis.109–111 Interestingly, both COX-2 and BDNF in the hippocampus were rapidly elevated by kainic acid or pilocarpine-induced SE with the induction of COX-2 temporally leading that of BDNF.20 COX-2 inhibition by SC-58125, a potent selective COX-2 inhibitor,112 or EP2 antagonism by compound 4, prevented a BDNF elevation in the hippocampus following a pilocarpine-induced SE in rats and mice. Likewise, treatment with compound 4 after a kainic acid-induced SE in mice decreased the SE-triggered phosphorylation of the cAMP response element-binding protein (CREB) and the activation of the BDNF/TrkB signaling in the hippocampus.20 It is well-known that the CREB activation can upregulate the BDNF expression and TrkB activation,113,114 which in turn can maintain or increase the CREB phosphorylation through a Ca2+/calmodulin-dependent protein kinase (CaMK)-dependent mechanism.115 These interesting findings together suggest that the COX-2 via PGE2/EP2 signaling regulates the hippocampal BDNF/TrkB pathway following prolonged seizures (Figure 1). Therefore, EP2 inhibition by selective antagonists might also provide a novel strategy to suppress the aberrant CREB/BDNF/TrkB activity during an acquired epileptogenesis.
5.3. Diisopropyl Fluorophosphate-Induced SE.
Diisopropyl fluorophosphate (DFP) is an organophosphorus-based agent with a cholinergic toxicity that can provoke the onset of SE in rodents within minutes and thus is commonly used to model an exposure to nerve agents in humans. A DFP exposure in rats leads to early consequences of cholinesterase inhibition such as whole-body motor convulsions, muscle weakness, and SE, followed days later by neuronal death, neuroinflammation, gliosis, BBB breakdown, weight loss, muscle weakness, and gastrointestinal dysfunction. The long-term consequences of DFP exposure include anxiety behaviors, cognitive deficits, and unprovoked seizures.116,117 Treatment with compound 4 after a DFP-induced SE in rats with six doses beginning 80–150 min after the SE onset substantially decreased the weight loss, acute neuronal death in the hippocampus, inflammatory cytokine burst, reactive microgliosis, and the BBB breakdown in the days after the SE (Table 1).116,118 A selective inhibition of the EP2 receptor by compound 4 in rats did not prevent the anxiety-like behavior assessed by open-field and light-dark box tests four weeks following the DFP exposure; however, it did alleviate the DFP SE-induced memory impairment measured by a novel object recognition test 6–12 weeks after the SE.116,119
These substantial benefits on intermediate and long-term consequences of SE by treatment with EP2 antagonist compound 4 after a DFP exposure reinforce the therapeutic potential of EP2 inhibition in organophosphate-induced pathologies.
Note that a single systemic administration of compound 4 approximately 1 h prior to the seizure induction by pilocarpine, kainic acid, or DFP did not change the behavioral seizure progression, the latency to SE, or the duration of SE in these rodent SE models (Table 1).14,18,116,119 The absence of an effect on behavioral seizures by the EP2 antagonist compound 4 was further validated by an electroencephalography (EEG) recording in pilocarpine-treated mice14 as well as in DFP-treated rats.116,119 Thus, the neuroprotection, cognitive improvement, and other benefits observed in EP2 antagonist-treated mice and rats were not direct outcomes of an anticonvulsant effect; they instead likely resulted from the anti-inflammatory effects of EP2 receptor inhibition. The lack of an antiseizure effect of EP2 antagonists also rules out the likelihood of the EP2 inhibition as a monotherapy for SE. Rather, these preclinical studies together raise the feasibility that inhibition of the EP2 receptor might provide an adjuvant strategy for the current first-line antiseizure drugs to treat SE.22
5.4. Febrile Seizures.
Febrile seizures are caused by high body temperature and are the most common type of seizure in infants and young children. Retrospective studies reveal that persistent febrile seizures or febrile status epilepticus (FSE) in childhood is highly associated with an increased risk of temporal lobe epilepsy (TLE) in adulthood.120,121 Similar to the pilocarpine and kainic acid-induced seizures, the experimental febrile status epilepticus (eFSE) induced in immature rats caused strong inflammatory reactions with a rapid and long-lasting elevation of pro-inflammatory cytokines including interleukin 1β (IL-1β), tumor necrosis factor α (TNF-α), and IL-6, astrocytic, and microglial activation as well as the activation and upregulation of the COX-2/mPGES-1/PGE2/EP2 signaling axis.122,123 However, administration of the EP2 antagonist compound 4 with only three doses beginning 4 h after the eFSE had minimal effects on the total number of aberrant EEG spikes recorded in the first 7–60 d after the eFSE, and it did not significantly alter the duration of each spike series provoked by the eFSE (Table 1).123 However, whether a longer-term treatment with compound 4 can decrease the development of an abnormal hyperexcitability following eFSE remains to be determined.
The discrepancy of EP2 inhibition after pilocarpine/kainic acid-induced SE in adult rodents and eFSE in newborn rats might be at least partially caused by the difference between mature and infantile brains (Table 1). It is possible that the targeting of a specific inflammatory signaling in juvenile epilepsy might alternatively lead to the activation of other inflammatory pathways in response or may simply be inadequate to afford meaningful impacts on the epilepsy development. It is important to know that the EP2 is highly expressed in the developing mouse brain but declines to a very low level in an adult mouse brain, suggesting that EP2 receptor may play some fundamental roles in the brain during the early stages of its development when the EP2 expression remains high.19,25 Indeed, the genetic ablation of the EP2 receptor led to impaired cognition, emotional behaviors, and hippocampal long-term potentiation (LTP) and long-term depression (LTD).8,124,125 Thus, EP2 inhibition during the early development stages may impose more profound adversity in the immature brain than it does to the adult brain. All these potential unwanted effects of EP2 inhibition in newborn animals together may profoundly compromise its therapeutic benefits observed in adult SE models.
6. ISCHEMIC STROKE
Ischemic stroke accounts for ~87% of all stroke cases, but due to its complex pathophysiology and relatively narrow intervention window the current treatment remains mainly limited to intravenous thrombolysis.126 COX-2 is highly regulated by neuronal activities and is often rapidly and robustly induced within the brain by cerebral ischemia.127,128 A large number of early studies using various rodent stroke models have shown that the induced COX-2 by cerebral ischemia might contribute to the neuronal injury, whereas the administration of selective COX-2 inhibitors or a genetic ablation of COX-2 reduced infarct volumes.127–131 However, the therapeutical targeting of COX-2 has been increasingly dampened due to extensive complications of cerebrovascular and cardiovascular systems caused by chronic uses of COX-2 inhibitors as medication. Strategies therapeutically targeting the downstream prostanoid receptors that are responsible for the deleterious effects of the COX cascade have been proposed.23,25,53,132
Several early studies utilizing global congenital EP2 knockout mice found that EP2 receptor activation might play some neuroprotective role following an ischemic stroke, because EP2 deficiency exacerbated the infarction after a middle cerebral artery occlusion (MCAO) with or without reperfusion.133–135 However, in a recent animal study using more sophisticated and more specific knockout strategies, it was found that the postnatal deletion of EP2 or the conditional ablation of its neuronal form, but not the endothelial or myeloid form, decreased the cortical infarction and neurological deficit in mice after a 45 min episode of MCAO followed by reperfusion.19 The contradiction between these early and recent studies can be explained by the broad complications and neurological dysfunctions that are caused by the global congenital deletion of EP2, as the receptor plays some essential functions in the brain, such as synaptic transmission, sensorimotor gating, synaptic plasticity, and cognitive functions.8,124,125 The postnatal and conditional deletion of EP2 would preserve its normal neurophysiological functions during the early stages of brain development, and it thus would avoid the cognitive and behavioral impairments observed in the global congenital EP2 knockout mice.19 As such, the conditional and postnatally induced deletion of EP2 provided more specific and reliable strategies to investigate the EP2 receptor-mediated neuroinflammation following ischemic stroke in the latest study.19
Nonetheless, because of the significant contribution of PGE2/EP2 signaling to COX-2 cascade-mediated neuronal inflammation and injury in excitotoxic and ischemic injuries,18,19 several EP2-selective small-molecule antagonists were evaluated for therapeutic potential in animal models of an ischemic stroke with the Amgen compound 9 being the first one (Table 2). Systemic treatment with compound 9 at 4.5 h and again at 24 h after ischemia start improved the neurological scores as well as decreased the weight loss and infarct volumes in mice after an episode of 45 min of MCAO followed by 72 h of reperfusion.19 Likewise, intraperitoneal injection of compound 4 with three doses that were administered at 4.5, 12, and 24 h following an MCAO for 45 min significantly improved the overall neurological score and histological lesion size in male mice (Table 1).21 Moreover, treatment with compound 4 was shown to downregulate the expression of prototypical inflammatory cytokines IL-1β, IL-6, and TNF-α in injured sites after MCAO. Similar results derived from studies using two different EP2 antagonists suggest that the beneficial effects of these compounds are unlikely related to a specific chemical structure; they rather are caused by their inhibition on EP2 receptor, that is, the prospective mechanisms of drug action. These proof-of-concept preclinical studies consistently support EP2 antagonism by brain-permeable EP2 antagonists such as compounds 4 and 9 as an emerging strategy to treat an ischemic stroke.
Table 2.
Pfizer and Amgen EP2 Antagonists That Were Tested in Preclinical Modelsa
| compound | animal model | major outcomes | reference |
|---|---|---|---|
| 1 | Mouse AOM/DSS model of colon cancer | Daily treatment with compound 1 for 80 d suppressed colon tumorigenesis in a dose-dependent manner. | Ma et al., 2015 |
| Intracranial aneurysm induced in rats | Treatment with compound 1 suppressed intracranial aneurysm in a dose-dependent manner, and prevented NF-κB activation, COX-2 induction, MCP-1 expression, and reduced macrophage infiltration. | Aoki et al., 2017 | |
| Mouse model of endometriosis-induced pain Aged mice (22–24 months old) |
Compound 1 reversed abdominal and paw hyperalgesia associated with endometriosis and reduced allodynia in a time-dependent manner. | Greaves et al., 2017 | |
| Daily treatment with compound 1 restored cellular bioenergetics and microglia/macrophage phagocytosis, and improved hippocampal synaptic plasticity as well as spatial memory in aged mice. | Minhas et al., 2021 | ||
| 8 | Mouse model of transient MCAO | Treatment with compound 8 after ischemia improved the neurological scores and decreased the weight loss and infarct volumes. | Liu et al., 2019 |
| Aged mice (22–24 months old) | Treatment with compound 8 restored cellular bioenergetics and microglia/macrophage phagocytosis, and improved hippocampal synaptic plasticity as well as spatial memory. | Minhas et al., 2021 |
Abbreviations: AOM, azoxymethane; COX-2, cyclooxygenase 2; DSS, dextran sodium sulfate; MCAO, middle cerebral artery occlusion; MCP-1, monocyte chemoattractant protein 1; NF-κB, nuclear factor κB.
7. ALZHEIMER’S DISEASE AND COGNITIVE AGING
The pathology of AD is characterized by the development of amyloid β (Aβ) plaques and neurofibrillary tangles.136 At the onset of AD, activated microglia are designated to contribute to the clearance of the neurotoxic Aβ plaques through phagocytosis.137 As the plaques aggregate, however, microglia activated by a long-term exposure to Aβ can be skewed from the phagocytic phenotype to a neuroinflammatory phenotype, and this transition during the disease progression has been proposed to be mediated by PGE2 signaling. It has been shown that PGE2 induces an inhibitory effect on a microglia/macrophages-mediated phagocytosis through the EP2 receptor.138 An upregulation of the EP2 receptor is associated with an increased pro-inflammatory gene expression and a decreased clearance of Aβ plaques in which microglia are involved.57 It has been noted that the EP2 signaling appears to suppress pretty much all beneficial aspects of activated microglia including phagocytosis, termination of pro-inflammatory responses, release of neurotrophic factors, and lysosomal function.139 When the microglial EP2 receptor is ablated, microglial phagocytotic activity is enhanced, and the aggregation of Aβ is mitigated.140 Thus, pharmacologically targeting the EP2 receptor might provide a novel therapeutic strategy to restore the beneficial functions of healthy microglial and prevent or even reverse the progression to AD and cognitive aging.
Among the newly discovered selective EP2 antagonists, compound 9 was evaluated in an ex vivo assay using brain slices that were harvested from 18-month-old Tg2576 mice, a common experimental model for AD.71 It was observed that treatment with compound 9 increased the macrophage-mediated Aβ phagocytosis in a concentration-dependent manner.71 In a more recent study, two EP2 antagonists were evaluated in the context of cognitive aging: brain permeable compound 9 and brain impermeable compound 1 (Table 2). Interestingly, both Amgen and Pfizer compounds restored cellular bioenergetics and microglia/macrophage phagocytosis and improved the hippocampal synaptic plasticity as well as the spatial memory in aged mice.26 Together, the improved bioenergetics and immune shifting induced by the EP2 antagonism contributed to the recovery of hippocampal synaptic protein levels and the long-term potentiation (LTP) in the CA1 region. Interestingly, compound 1—though it was unable to penetrate the brain parenchyma—attenuated pro-inflammatory responses not only in the blood but also in the CNS.26 It is postulated that the peripheral EP2 inhibition by compound 1 may cause as-yet-unidentified alterations in the blood that beneficially influence the aging cerebrovascular endothelium or cross the BBB to directly improve the neuronal function.26 It is also possible that the compromised BBB integrity in an aged brain enables compound 1 to cross the BBB as compound 10 did in rats undergoing a pilocarpine-induced SE.100
8. INFLAMMATORY PAIN AND HYPERALGESIA
Synaptic plasticity in nociceptive spinal pathways has been proposed as an important mechanism that might contribute to the amplification of nociceptive signaling under conditions such as acute postoperative pain and chronic pain associated with a peripheral inflammation.141 COX-2 has been found constitutively expressed in both spinal neurons and radial glia under naive conditions;142,143 however, it also can be substantially upregulated in the spinal dorsal horn after injury,144,145 suggesting that a COX-2/PGE2 cascade might be involved in the regulation of synaptic LTP within the superficial dorsal horn of the spinal cord. Indeed, both phospholipase A2 (PLA2) selective inhibitor arachidonyl trifluoromethyl ketone (AACOCF3) and COX-2 selective inhibitor nimesulide were able to prevent spike timing-dependent LTP (or tLTP) at sensory synapses onto spinoparabrachial neurons.143 Interestingly, selective EP2 antagonist compound 1 fully recapitulated the inhibitory effects of AACOCF3 and nimesulide on tLTP, suggesting that the amplification of ascending nociceptive transmission by the spinal superficial dorsal horn network is likely governed by PLA2/COX-2/PGE2/EP2 signaling axis.
Endometriosis is a chronic gynecological disease that currently does not have a cure and is often characterized by devastating chronic pain and infertility. Changes in pain perception in women are believed to engage a number of pro-inflammatory mediators in efferent peripheral nerve endings. As a well-known mediator of inflammation and nociception in inflammatory and neuropathic pain,146–148 PGE2 has been found within endometriosis lesions where COX-2 expression is substantially induced.149–151 Interestingly, in a recent study on a preclinical mouse model of endometriosis, COX-2, EP2, and EP4 were found upregulated in endometriosis lesions, dorsal root ganglia, spinal cord, thalamus, and forebrain, suggesting an amplification process along the pain neuroaxis caused by endometriosis.152
Intraperitoneal injection of the EP2-selective antagonists compound 1 and compound 4, but not EP4 antagonist L-161982, significantly reversed abdominal or paw hyperalgesia in this model of endometriosis (Tables 1 and 2). Further study showed that the oral administration of compound 1 in mice with endometriosis also resulted in dramatically reduced allodynia in both abdomen and hind-paw tests in a time-dependent manner (Table 2).152 Taken together, these interesting findings using two different EP2-selective antagonists demonstrated the feasibility of targeting the EP2 receptor as an emerging strategy to treat inflammatory pain and hyperalgesia.
9. INTRACRANIAL ANEURYSMS
Intracranial aneurysm or brain aneurysm is a weak or thin spot on a cerebral artery or vein that balloons out and fills with blood. More than 50% of intracranial aneurysm patients are unaware of their conditions and thus are left untreated, increasing the chances of aneurysm rupture and fatal subarachnoid hemorrhage.153 Inflammatory and immunological reactions have long been known in unruptured cerebral aneurysms and may be related to the formation and rupture of aneurysms.154 Examining the expression of COXs, PGESs, and EP receptors revealed that COX-2, mPGES-1, and EP2 were induced in endothelial cells in the walls of a cerebral aneurysm in both human patients and rats with an induced cerebral aneurysm.155 Interestingly, the incidence of cerebral aneurysm in mice was significantly prevented by the COX-2 inhibitor celecoxib and the genetic ablation of the EP2 receptor but not by that of EP1, EP3, or EP4. The deficiency of the EP2 receptor also suppressed NF-κB-mediated chronic inflammation in cerebral aneurysm lesions.155 These findings revealed an essential role of COX-2/mPGES-1/PGE2/EP2/NF-κB signaling axis in the development and enlargement of a cerebral aneurysm. Oral administration of the EP2 antagonist compound 1 suppressed intracranial aneurysm that was induced in rats in a dose-dependent manner as well as prevented NF-κB activation, COX-2 induction, and the expression of monocyte chemoattractant protein 1 (MCP-1), leading to the reduction in macrophage infiltration into intracranial aneurysm lesions (Table 2).43 Thus, targeting the EP2 receptor by small-molecule antagonists could be a novel pharmacological strategy to treat developing intracranial aneurysms and reduce the potential risk of a fatal subarachnoid hemorrhage.
10. RETINITIS PIGMENTOSA
Retinitis pigmentosa is a group of diseases that involve the breakdown and loss of cells in the retina and is characterized by a progressive degeneration of photoreceptors in the retina. Consisting of a class of heterogenous inherited eye disorders, retinitis pigmentosa can be caused by a host of gene mutations and develop into partial or complete blindness, as currently there is no effective treatment available for patients. Neuroinflammation mediated by activated microglia within the retina is thought to facilitate the progression of this disorder.156 In retinal degeneration 10 (rd10) mice, an experimental model for retinitis pigmentosa, COX-1 expression, was found to be upregulated within retinal microglia, compared to the wild-type peers.157 The genetic ablation or pharmacological inhibition of COX-1 alleviated microglial activation as well as preserved the retinal photoreceptor, retinal function, and visual performance in these rd10 mice, suggesting the involvement of COX/PGE2 signaling in the pathophysiology of the disease. Interestingly, pharmacological inhibition of the EP2 receptor by compound 4 twice daily largely prevented the activation of retinal microglia and suppressed the excessive generation of prototypic pro-inflammatory cytokines including IL-1β and TNF-α. Furthermore, a reduction in the degeneration of retinal photoreceptors in rd10 mice that were treated by compound 4 was also observed. Consequently, the retinal function and visual performance were largely preserved in rd10 mice by a compound 4 treatment.157 Taken together, the EP2 inhibition precisely recapitulated the beneficial effects of COX-1 deletion or inhibition in the rd10 retina (Table 1), highlighting the potential and feasibility of EP2 antagonists in treating retinitis pigmentosa.
11. SYSTEMIC INFLAMMATION AND SEPSIS
Sepsis arises from the dysfunction of the regulatory mechanisms that govern the immune system during the response to various infections. Notably, almost 70% of patients with sepsis will develop sepsis-associated encephalopathy, which induces neuroinflammation, a key feature associated with long-term cognitive deficits. The COX-2/PGE2/EP2 signaling pathway has been implicated in playing a pivotal role in neuroinflammation through the elevation of pro-inflammatory cytokines, chemokines, and COX-2 expression within the brain. The systemic administration of the selective EP2 antagonist compound 4 after LPS-triggered systemic inflammation facilitated the recovery of body weight, mitigated brain inflammation as evaluated by pro-inflammatory mediators such as IL-1β, IL-6, TNF-α, chemokine (C–C motif) ligand 2 (CCL2), and COX-2 as well as microgliosis, and prevented the loss of synaptic proteins such as the postsynaptic density protein 95 (PSD-95) and synaptophysin. In a panel of behavioral tests that were performed approximately one month after an LPS injection in mice, compound 4 treatment ameliorated the depression-like behavior in a sucrose preference test and cognitive deficits in a novel object recognition test (Table 1).158 These results recognize EP2 receptor inhibition as a new strategy to alleviate neuroinflammation and long-term affective and cognitive issues of sepsis survivors.
12. CANCERS
COX-2 is commonly expressed in a variety of types of cancers and has been well-known to promote inflammatory microenvironments, angiogenesis, immune evasion, and treatment resistance.9,159–161 Most of these pro-tumor effects of COX-2 are believed to be largely attributed to the PGE2 signaling mediated by the EP2 receptor, because the genetic deletion of EP2 led to the suppression of tumor development and the progression of various backgrounds in early studies.15,44,162–168 However, considering other significant complications in EP2−/− mice, the results from the EP2 ablation were unable to be validated by a reliable pharmacological approach due to the lack of truly selective EP2 antagonists with adequate pharmacokinetic and pharmacodynamic profiles. The discovery of brain-permeable and brain-impermeable EP2 antagonists enable pharmacological strategies to study multifaceted roles of EP2 in tumor development and progression, chronic inflammation, metastasis, angiogenesis, and multidrug resistance in animal models of cancers of both the periphery and brain.
12.1. Colon Cancer.
As the third most prevalent cancer worldwide, colon cancer constitutes the fourth most common cause of cancer-related death.169 It has long been observed that a regular consumption of aspirin or other NSAIDs is highly correlated with a reduction in the mortality rates of sporadic colorectal cancer patients. In addition, NSAIDs have been shown to facilitate the self-regression of familial colon polyposis, a precancerous condition.170–172 Results from these epidemiological analyses suggest that the pathogenesis of colon cancer is highly associated with inflammatory responses in the colon, and thus suppressing tumor inflammation might prevent the development and progression of colon cancer. Although the mechanism whereby COX cascade-mediated inflammation promotes colon cancer largely remains elusive, the downstream PGE2/EP2 signaling pathway has been thought to contribute to tumor formation and progression.168,173,174
Interestingly, a deficiency of the EP2 receptor (EP2−/−), but not EP1 or EP3, selectively decreased tumor formation in azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colon tumorigenesis, a colitis-associated cancer model, accompanied by downregulated pro-inflammatory genes such as TNF-α, IL-6, chemokine (C-X-C motif) ligand 1 (CXCL1), and COX-2.175 These findings are consistent with results from early studies on mouse models of intestinal polyp formation,162 mammary hyperplasia,164 skin tumor,165 and lung tumorigenesis, in which the genetic ablation of the EP2 receptor in mice relieved tumor-associated inflammation and suppressed tumor development. Importantly, in the colitis-associated colorectal cancer model, systemic administration of EP2 antagonist compound 1 daily for 80 d potently suppressed colon tumorigenesis induced by AOM/DSS in a dose-dependent manner (Table 2).175 Results from this study also suggest that EP2 in tumor-associated fibroblasts and neutrophils may facilitate colon tumorigenesis by amplifying the inflammation and shaping the tumor microenvironment.168,175 Given that the PGE2/EP2 signaling likely serves as a key node of chronic inflammation in the colon tumor microenvironment, EP2 antagonists might represent promising candidates of NSAIDs-alternative for chemoprevention of colon cancer.
12.2. Malignant Glioma.
Gliomas constitute ~80% of all primary malignant brain tumors in humans, and more than 80% of these cases are classified by the World Health Organization (WHO) as grade IV tumor–glioblastoma. With the current standard treatment, namely, surgical resection followed by concurrent radiotherapy and chemotherapy with Temozolomide, the prognosis of glioblastoma remains poor with a median overall survival below 15 months, and less than 10% of patients can survive more than five years.176–178 Among several comprehensive factors that render malignant glioma particularly difficult to treat is that most antitumor agents including many immunotherapeutic drugs are unable to reach the tumor sites due to their poor brain penetration.179 The development of new therapeutics with an adequate efficacy for this most devastating and deadly type of brain cancer is certainly an urgent unmet need.180,181
Inflammation within the brain emerged as a key contributor to many forms of brain cancer.182 COX-2, as a chief pro-inflammatory mediator, is often induced in intracranial tumors,183,184 and it has been shown to promote the growth, migration, angiogenesis, and immune evasion of malignant gliomas.185–187 It appears that COX-2/mPGES-1/PGE2 signaling axis is highly associated with the aggressiveness of human gliomas, and the EP2 receptor is a key Gαs-coupled receptor that mediates COX-2/PGE2-initiated cAMP signal pathways in human malignant glioma cells.188 Interestingly, inhibition of the EP2 receptor by the antagonist compound 4 reduced the COX-2 activity-driven glioblastoma cell proliferation, invasion, and migration, and it caused cell-cycle arrest at G0-G1 and apoptosis of the brain tumor cells.188 Moreover, oral administration of compound 4 twice daily for four consecutive weeks suppressed malignant glioma growth and increased survival rates of mice that harbored intracranial tumors formed by human glioblastoma cells (Table 1).188 These results together suggest that PGE2 signaling via EP2 receptor contributes to the malignancy of human glioma cells and that the selective EP2 antagonists with adequate brain penetration such as compounds 3 and 4 might represent promising candidates as new therapeutic agents for malignant glioma.189
12.3. Neuroblastoma.
Neuroblastoma is the third-most common type of pediatric cancers and accounts for nearly 15% of cancer-related deaths in young children.160 Despite marked advances in tumor diagnosis and management during the past decades, the five-year survival rates for patients with high-risk neuroblastoma remain below 50%.190 COX, a conventional inflammatory executor, has recently been demonstrated to provide the essential driving force for neuroblastoma pathogenesis.191 PGE2 highly presents in various tumor tissues including those of neuroblastoma, where the COX expression is elevated.192,193 A daily oral intake of COX inhibitors decreased the burden of tumors with an amplification of the MYCN oncogene (encoding a transcription factor),160 reduced the presence of tumor-associated innate immune cells,194 and delayed the progression of 11q-deleted neuroblastoma.193 Likewise, the inhibition of PGE2 direct synthase mPGES-1 by Compound III (CIII) or 934 was found to suppress neuroblastoma with the MYCN amplification or 11q deletion,195,196 attesting to an essential role for PGE2 in the high-risk neuroblastoma. Interestingly, correlation analyses on neuroblastoma patient data sets revealed a positive relationship between the COX/PGE2/EP2 signaling axis and the aggressiveness of a human neuroblastoma.197 A cell-based TR-FRET assay method identified EP2 as the primary Gαs-coupled receptor that mediates PGE2-initiated cAMP signaling in neuroblastoma cells with various risk factors.197 A preliminary study using compounds 4 and 7 demonstrated that pharmacological inhibition of the EP2 receptor in mice substantially impaired the growth of human neuroblastoma xenografts and the associated angiogenesis and downregulated pro-inflammatory cytokines in tumor tissues (Table 1).197 Collectively, these preliminary results suggest that PGE2 via EP2 receptor increases the growth and malignant potential of human neuroblastoma cells; selective EP2 antagonists might provide novel alternative therapeutic strategies to COX or mPGES-1 inhibition for this devastating type of pediatric cancer.
13. CONCLUDING REMARKS AND FUTURE DIRECTIONS
Over the past decade, significant progress has been made in developing small-molecule antagonists selectively targeting the EP2 receptor for the treatment of peripheral and central inflammation-associated conditions. Compared to the first-generation EP2 antagonists, the second-generation analogues in general are more selective and possess drug-like properties with diverse pharmacokinetic profiles. Some of these compounds such as compounds 4 and 9 have a favorable brain penetration and thus showed excellent therapeutic potential in preclinical models of epilepsy, cerebral ischemia, and brain tumor; others like TG6–129 and compound 1 cannot cross the BBB at all and demonstrated tremendous efficacy in managing peripheral cancers including colon cancer and neuroblastoma. Despite the challenges in early years, the development of novel small molecules that precisely target the PGE2/EP2 signaling pathway has promising therapeutic potential to afford profound anti-inflammatory effects in a broad range of inflammation-associated conditions (Tables 1 and 2). Currently, there is no human clinical trial on EP2-selective antagonists; however, a dual antagonist TPST-1495 targeting both EP2 and EP4 is in a current phase 1a/1b study for solid tumors (https://clinicaltrials.gov/ct2/show/NCT04344795). The recently solved cryo-EM structures of the human EP2 receptor and Gs protein complex are expected to play some fundamental roles in rational designs to develop the next-generation EP2 antagonists with further improved pharmacokinetic and pharmacodynamic properties for clinical applications (Figure 5).68 In addition, the use of nanoparticles encapsulated by macrocycle-based compounds such as calixarenes for the controlled release of these potential therapeutic agents might also help in targeted therapy.198
13.1. Harnessing the Power of Neuroinflammation.
Many CNS disorders are now considered as inflammation-associated neurological conditions, highlighting the commonality and essentiality of inflammatory signaling pathways in the pathophysiological mechanisms of these diseases. The brain disorders associated with profound neuroinflammatory causes and pathologies can be acute conditions including strokes 199–201 and epileptic seizures,92,202,203 or chronic diseases, for example, inflammatory and neuropathic pain204,205 and neurodegenerative diseases such as AD, Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS).206–209 Accumulating evidence from recent studies suggests that the COX-2/mPGES-1/PGE2/EP2 signaling axis may act as a key culprit of the maladaptive immune and inflammatory responses in nearly all these CNS conditions (Tables 1 and 2).17,19,21–23,26,57,143,210,211 It is worth noting that the activation of the EP2 receptor leads to the elevation of pro-inflammatory cytokines and NF-κB, which can further induce COX-2 and thus sustain and even amplify the inflammatory chain reactions. Specifically blocking the PGE2/EP2 signaling, a key node of the inflammatory networks, by brain-permeable EP2 selective antagonists is designated to break this vicious self-reinforcing feedback loop of neuroinflammation but does not affect pathways mediated by other prostanoids that may play physiological and beneficial functions under normal conditions.
13.2. Suppressing Tumor Microenvironment and Inflammation.
Cancer biology research has been shifting from the focus on cancer cells alone to a more inclusive concept of the cancer microenvironment comprised of cancer cells, cancer-associated fibroblasts, vascular cells, and infiltrating immune cells.212 Tumor-elicited inflammation has been increasingly recognized for its essential roles in shaping the plasticity of the cancer microenvironment by independently amplifying immune responses and the release of cytokines and many other inflammatory mediators and growth factors that act together to facilitate tumor initiation, growth, progression, and metastasis.213 The COX-2/PGE2/EP2 signaling pathway has been shown to be commonly upregulated in multiple cancer types and to be involved in cancer-related chronic inflammation, immune suppression, angiogenesis, tumor invasion and metastasis, and multidrug resistance.44 More importantly, elevated PGE2/EP2 signaling was identified both within the tumor and its microenvironment. In addition, PGE2 signaling via the EP2 receptor can also crosstalk with the epidermal growth factor (EGF)/EGF receptor (EGFR) transduction axis, which in turn can activate several other signal pathways, such as mitogen-activated protein kinase (MAPK), PI3K/Akt, signal transducer and activator of transcription (STAT), and PLC, thereby facilitating the proliferation, migration, differentiation, and survival of tumor cells (Figure 1B).44 Therefore, targeting the EP2 receptor by these EP2 antagonists has the potential to serve as a comprehensive treatment strategy to directly impede the proliferation of cancer cells (Tables 1 and 2). They may also suppress the cancer-nourishing molecular and cellular components within the cancer microenvironment and, thus, can potentially be developed as adjunct therapies for current standard anticancer drugs. To determine this possibility in the future, it is important to evaluate combined treatment engaging both EP2 antagonists and front-line chemotherapy or immunotherapy drugs in animal models.
13.3. Quenching the Cytokine Storm in SARS-COV-2 Infection.
The ongoing pandemic of the coronavirus disease 2019 (COVID-19) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has once again demonstrated the importance of effective immune responses to a host and the devastating consequences when they are dysregulated to become out of control. Cytokine storm as a term was first used to describe the engraftment complications of acute graft versus host disease (GVHD) after allogeneic bone marrow transplantation, where inflammatory cytokines, such as IL-1, IL-2, interferon γ (IFN-γ), and TNF-α, may play critical and unique roles.214,215 Despite the term’s long history, there is still no single or widely accepted definition likely due to its wide array of causes and outcomes.216 Generally, it is thought of as an overactivation of the immune system characterized by an extreme elevation of systemic cytokine circulation and hyperactive immune cells, both of which can have detrimental effects on multiorgan systems.216 COX-2/PGE2/EP2 signaling has been shown to play a considerable role in regulating the immune response, specifically through an induction of a wide array of pro-inflammatory cytokines and chemokines including IL-1β, IL-6, TNF-α, and CCL2 (MCP-1) in various disease conditions.18,21,26,38,55,157,197 Interestingly, spike proteins of the COVID-19 virus have been shown to induce a COX-2 expression, which could implicate its involvement in facilitating the cytokine storm in COVID-19.217,218 NSAIDs are primary medicines that are widely used to alleviate pain, fever, and inflammation, which are the common symptoms of COVID-19 patients.219 Indeed, treatment with NSAIDs has been demonstrated to stifle the inflammatory response to COVID-19, but it also altered the host’s ability to produce protective antibodies.220 Given that there is no clear evidence of risk or benefit for the use of NSAIDs in COVID-19 patients,221 further research is needed to better define the role of NSAIDs and particularly COX-2 inhibitors in patients with COVID-19 infection.222
Both EP2 and EP4 receptors were detected in human lung mast cells, but it is the EP2 that mediates the cAMP signaling and inhibitory effects of PGE2 on IgE-dependent histamine release in these cells,73 suggesting a potential use of EP2 antagonists to alleviate the inflammation in asthma and other respiratory diseases. Whether the EP2 receptor is also involved in excessive inflammatory reactions and cytokine storm that are caused by SARS-CoV-2 would be a very interesting question to investigate. Therefore, future studies should also be directed to determine whether the selective EP2 antagonists are able to provide more therapeutic efficacy and specificity than COX-targeting drugs as a novel strategy to combat the uncontrollable cytokine storm following COVID-19 infection.
ACKNOWLEDGMENTS
This work was supported by the National Institutes of Health (NIH)/National Institute of Neurological Disorders and Stroke (NINDS) Grant Nos. R01NS100947 (J.J.) and R21NS109687 (J.J.). We are very grateful to Dr. Laura Wilt from St. Jude Children’s Research Hospital for her tremendous help with the molecular docking work.
ABBREVIATIONS USED
- AACOCF3
arachidonyl trifluoromethyl ketone
- Aβ
amyloid β
- AC
adenylate cyclase
- AD
Alzheimer’s disease
- ALS
amyotrophic lateral sclerosis
- AOM
azoxymethane
- APC
Adenomatous polyposis coli
- ASDs
antiseizure drugs
- BBB
blood-brain barrier
- BDNF
brain-derived neurotropic factor
- CaMK
Ca2+/calmodulin-dependent protein kinase
- cAMP
cyclic AMP
- CCL2
chemokine (C–C motif) ligand 2
- COVID-19
coronavirus disease 2019
- COX
cyclooxygenase
- Coxibs
COX-2 specific inhibitors
- cPLA2
cytoplasmic phospholipase A2
- CREB
cAMP response element-binding protein
- cPGES
cytosolic PGES
- CXCL1
chemokine (C-X-C motif) ligand 1
- DFP
Diisopropyl fluorophosphates
- DSS
dextran sodium sulfate
- EEG
electroencephalography
- EGF
epidermal growth factor
- eFSE
experimental febrile status epilepticus
- EGFR
epidermal growth factor receptor
- EPAC
exchange protein activated by cAMP
- ERK
extracellular signal-regulated kinase
- F-actin
filamentous actin
- FSE
febrile status epileptics
- GPCR
G protein-coupled receptor
- GSK-3β
glycogen synthase kinase 3β
- GVHD
graft versus host disease
- IFN-γ
interferon γ
- IL-1
interleukin 1
- IL-1β
interleukin 1β
- IL-1R
interleukin 1 receptor
- IL-2
interleukin 2
- IL-6
interleukin 6
- JNK
c-Jun N-terminal kinase
- LPS
lipopolysaccharide
- LTD
long-term depression
- LTP
long-term potentiation
- MAPK
mitogen-activated protein kinase
- MCAO
middle cerebral artery occlusion
- MCP-1
monocyte chemoattractant protein 1
- MEK
mitogen-activated protein kinase kinase
- mPGES-1
microsomal prostaglandin E synthase 1
- mPGES-2
microsomal prostaglandin E synthase 2
- NF-κB
nuclear factor κB
- NSAIDs
nonsteroidal anti-inflammatory drugs
- PD
Parkinson’s disease
- Pfn-1
profilin 1
- PGD2
prostaglandin D2
- PGE2
prostaglandin E2
- PGES
Prostaglandin E synthase
- PGF2α
prostaglandin F2α
- PGH2
prostaglandin H2
- PGI2
prostaglandin I2
- PI3K
phosphoinositide 3-kinase
- PKA
protein kinase A
- PKB
protein kinase B or Akt
- PLC-γ1
phospholipase C γ1
- PSD-95
postsynaptic density protein 95
- rd10
retinal degeneration 10
- SAR
structure–activity relationship
- SARS-COV-2
severe acute respiratory syndrome coronavirus 2
- SE
status epilepticus
- STAT
signal transducer and activator of transcription
- TBI
traumatic brain injury
- TLE
temporal lobe epilepsy
- tLTP
spike timing-dependent LTP
- TNF-α
tumor necrosis factor α
- TR-FRET
time-resolved fluorescence resonance energy transfer
- TrkB
tropomyosin-related kinase receptor B
- TXA2
thromboxane A2
- WHO
world health organization
Biographies
Biographies
Madison Sluter received her B.A. in Psychology from Ithaca College in 2019. During her undergraduate career she worked on researching the influence of alcohol on the neuroimmune response to traumatic brain injury under the guidance of Dr. Tamara Fitzwater. In addition, she worked in an organic chemistry lab synthesizing novel antimalaria compounds under the direction of Dr. D. J. Robinson. Currently she is pursuing her Ph.D. in Molecular and Systems Pharmacology at the University of Tennessee Health Science Center under the training of Dr. Jianxiong Jiang. She employs biochemical, pharmacological, and genetic techniques to explore new potential drug targets for CNS cancers.
Ruida Hou received his M.D. from Jilin University in China in 2016. After three years of residency training in Urology at China-Japan Union Hospital of Jilin University, he decided to pursue his Ph.D. at the University of Tennessee Health Science Center under the training of Dr. Jianxiong Jiang in 2019. His research is focused on targeting the inflammatory PGE2 signaling cascade in neuroblastoma.
Lexiao Li received his Ph.D. from Heidelberg University in Germany in 2017 under the supervision of Prof. Hugo Marti in Physiology. He started his postdoctoral training in 2018 at the University of Tennessee Health Science Center in Dr. Tauheed Ishrat’s lab in Neurobiology and then joined Dr. Jianxiong Jiang’s lab in Pharmaceutical Sciences in 2019. His research focuses on experimental strokes from the aspects of endogenous hypoxic adaptation, hyperglycemic stroke, and microglia-mediated neuroinflammation.
Nelufar Yasmen received her B. Pharm. from Jessore University of Science and Technology in Bangladesh in 2017. During her undergraduate study she worked on the anti-inflammatory and analgesic activities of different crude plant extracts. After graduation, she spent two years as a Quality Control Executive in a pharmaceutical company in Bangladesh. Currently she is pursuing her Ph.D. under the supervision of Dr. Jianxiong Jiang at the University of Tennessee Health Science Center with research focusing on neuroinflammation in epilepsy.
Ying Yu is a research scientist at the University of Tennessee Health Science Center College of Pharmacy. Her current research work focuses on neuroinflammation and neurodegeneration in CNS conditions such as epileptic seizures. She obtained her B.S. in Biochemistry from East China University of Science and Technology in Shanghai and Ph.D. in Chemistry (Biomolecular) from Emory University. She received postdoctoral training at Emory University School of Medicine and worked as a research associate at Cincinnati Children’s Hospital Medical Center.
Jiawang Liu received his Ph.D. in Medicinal Chemistry from Peking University in 2007. He has been engaged in the design, synthesis, and evaluation of small molecules as anticancer and antithrombotic agents since 2002. As a research scientist at Xavier RCMI Cancer Research Center from 2015 to 2017, he participated in the development of oral SERD ZB716, which is in a phase I/II clinical trial for ER+/HER2-locally advanced or metastatic breast cancer. Since September 2017, he has been serving as the director of Medicinal Chemistry Core and assistant professor in the Department of Pharmaceutical Sciences at the University of Tennessee Health Science Center.
Jianxiong Jiang received his Ph.D. in 2008 from Auburn University under the guidance of Dr. Marie Wooten. He then spent nearly seven years in postdoctoral research training with Dr. Ray Dingledine at Emory University School of Medicine, where he developed the first-generation EP2-selective small-molecule antagonists in collaboration with medicinal chemist Dr. Thota Ganesh and the Emory Chemical Biology Discovery Center. In January 2015, he started his first independent academic position at the University of Cincinnati College of Pharmacy. He then joined the University of Tennessee Health Science Center College of Pharmacy in July 2018. His research has led to ~50 peer-reviewed publications and five patents about EP2 antagonists. He is a recipient of the NIH Pathway to Independence Award and NARSAD Young Investigator Award.
Footnotes
Complete contact information is available at: https://pubs.acs.org/10.1021/acs.jmedchem.1c00816
The authors declare no competing financial interest.
Contributor Information
Madison N. Sluter, Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
Ruida Hou, Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
Lexiao Li, Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
Nelufar Yasmen, Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
Ying Yu, Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
Jiawang Liu, Department of Pharmaceutical Sciences, College of Pharmacy and Medicinal Chemistry Core, Office of Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
Jianxiong Jiang, Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States.
REFERENCES
- (1).Vane JR; Bakhle YS; Botting RM Cyclooxygenases 1 and 2. Annu. Rev. Pharmacol. Toxicol 1998, 38, 97–120. [DOI] [PubMed] [Google Scholar]
- (2).Korbecki J; Baranowska-Bosiacka I; Gutowska I; Chlubek D Cyclooxygenase pathways. Acta Biochim Pol 2014, 61, 639–649. [PubMed] [Google Scholar]
- (3).Breyer RM; Bagdassarian CK; Myers SA; Breyer MD Prostanoid receptors: subtypes and signaling. Annu. Rev. Pharmacol. Toxicol 2001, 41, 661–690. [DOI] [PubMed] [Google Scholar]
- (4).Hirata T; Narumiya S Prostanoid receptors. Chem. Rev 2011, 111, 6209–6230. [DOI] [PubMed] [Google Scholar]
- (5).Fitzpatrick FA Cyclooxygenase enzymes: regulation and function. Curr. Pharm. Des 2004, 10, 577–588. [DOI] [PubMed] [Google Scholar]
- (6).Yang H; Chen C Cyclooxygenase-2 in synaptic signaling. Curr. Pharm. Des 2008, 14, 1443–1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (7).Hein AM; O’Banion MK Neuroinflammation and memory: the role of prostaglandins. Mol. Neurobiol 2009, 40, 15–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (8).Yang H; Zhang J; Breyer RM; Chen C Altered hippocampal long-term synaptic plasticity in mice deficient in the PGE2 EP2 receptor. J. Neurochem 2009, 108, 295–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (9).Hashemi Goradel N; Najafi M; Salehi E; Farhood B; Mortezaee K Cyclooxygenase-2 in cancer: A review. J. Cell. Physiol 2019, 234, 5683–5699. [DOI] [PubMed] [Google Scholar]
- (10).Jiang J; Qiu J; Li Q; Shi Z Prostaglandin E2 Signaling: Alternative Target for Glioblastoma? Trends Cancer 2017, 3, 75–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (11).Yu Y; Nguyen DT; Jiang JG protein-coupled receptors in acquired epilepsy: Druggability and translatability. Prog. Neurobiol 2019, 183, 101682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Cimino PJ; Keene CD; Breyer RM; Montine KS; Montine TJ Therapeutic targets in prostaglandin E2 signaling for neurologic disease. Curr. Med. Chem 2008, 15, 1863–1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (13).Dudek FE Mechanisms of Seizure-Induced Inflammation of the Brain: Many Possible Roles for Neuronal COX-2. Epilepsy Curr. 2012, 12, 115–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (14).Jiang J; Quan Y; Ganesh T; Pouliot WA; Dudek FE; Dingledine R Inhibition of the prostaglandin receptor EP2 following status epilepticus reduces delayed mortality and brain inflammation. Proc. Natl. Acad. Sci. U. S. A 2013, 110, 3591–3596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (15).Jiang J; Dingledine R Role of prostaglandin receptor EP2 in the regulations of cancer cell proliferation, invasion, and inflammation. J. Pharmacol. Exp. Ther 2013, 344, 360–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (16).Jiang J; Dingledine R Prostaglandin receptor EP2 in the crosshairs of anti-inflammation, anti-cancer, and neuroprotection. Trends Pharmacol. Sci 2013, 34, 413–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Kang X; Qiu J; Li Q; Bell KA; Du Y; Jung DW; Lee JY; Hao J; Jiang J Cyclooxygenase-2 contributes to oxidopamine-mediated neuronal inflammation and injury via the prostaglandin E2 receptor EP2 subtype. Sci. Rep 2017, 7, 9459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (18).Jiang J; Yu Y; Kinjo ER; Du Y; Nguyen HP; Dingledine R Suppressing pro-inflammatory prostaglandin signaling attenuates excitotoxicity-associated neuronal inflammation and injury. Neuropharmacology 2019, 149, 149–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (19).Liu Q; Liang X; Wang Q; Wilson EN; Lam R; Wang J; Kong W; Tsai C; Pan T; Larkin PB; Shamloo M; Andreasson KI PGE2 signaling via the neuronal EP2 receptor increases injury in a model of cerebral ischemia. Proc. Natl. Acad. Sci. U. S. A 2019, 116, 10019–10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (20).Yu Y; Jiang J COX-2/PGE2 axis regulates hippocampal BDNF/TrkB signaling via EP2 receptor after prolonged seizures. Epilepsia Open 2020, 5, 418–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Li L; Yu Y; Hou R; Hao J; Jiang J Inhibiting the PGE2 Receptor EP2Mitigates Excitotoxicity and Ischemic Injury. ACS Pharmacol Transl Sci. 2020, 3, 635–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (22).Nagib MM; Yu Y; Jiang J Targeting prostaglandin receptor EP2 for adjunctive treatment of status epilepticus. Pharmacol. Ther 2020, 209, 107504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Li L; Sluter MN; Yu Y; Jiang J Prostaglandin E receptors as targets for ischemic stroke: Novel evidence and molecular mechanisms of efficacy. Pharmacol. Res 2021, 163, 105238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (24).Hou R; Yu Y; Jiang J PGE2 receptors in detrusor muscle: Drugging the undruggable for urgency. Biochem. Pharmacol 2021, 184, 114363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (25).Jiang J; Yu Y Small molecules targeting cyclooxygenase/prostanoid cascade in experimental brain ischemia: Do they translate? Med. Res. Rev 2021, 41, 828–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (26).Minhas PS; Latif-Hernandez A; McReynolds MR; Durairaj AS; Wang Q; Rubin A; Joshi AU; He JQ; Gauba E; Liu L; Wang C; Linde M; Sugiura Y; Moon PK; Majeti R; Suematsu M; Mochly-Rosen D; Weissman IL; Longo FM; Rabinowitz JD; Andreasson KI Restoring metabolism of myeloid cells reverses cognitive decline in ageing. Nature 2021, 590, 122–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Varvel NH; Espinosa-Garcia C; Hunter-Chang S; Chen D; Biegel A; Hsieh A; Blackmer-Raynolds L; Ganesh T; Dingledine R Peripheral Myeloid Cell EP2 Activation Contributes to the Deleterious Consequences of Status Epilepticus. J. Neurosci 2021, 41, 1105–1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (28).DuBois RN; Abramson SB; Crofford L; Gupta RA; Simon LS; Putte LBA; Lipsky PE Cyclooxygenase in biology and disease. FASEB J. 1998, 12, 1063–1073. [PubMed] [Google Scholar]
- (29).Rainsford KD Profile and mechanisms of gastrointestinal and other side effects of nonsteroidal anti-inflammatory drugs (NSAIDs). Am. J. Med 1999, 107, 27–35 discussion 35S–36S.. [DOI] [PubMed] [Google Scholar]
- (30).Xie WL; Chipman JG; Robertson DL; Erikson RL; Simmons DL Expression of a mitogen-responsive gene encoding prostaglandin synthase is regulated by mRNA splicing. Proc. Natl. Acad. Sci. U. S. A 1991, 88, 2692–2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (31).Langman MJ; Jensen DM; Watson DJ; Harper SE; Zhao PL; Quan H; Bolognese JA; Simon TJ Adverse upper gastrointestinal effects of rofecoxib compared with NSAIDs. JAMA 1999, 282, 1929–1933. [DOI] [PubMed] [Google Scholar]
- (32).Dogne JM; Supuran CT; Pratico D Adverse cardiovascular effects of the coxibs. J. Med. Chem 2005, 48, 2251–2257. [DOI] [PubMed] [Google Scholar]
- (33).Marnett LJ The COXIB experience: a look in the rearview mirror. Annu. Rev. Pharmacol. Toxicol 2009, 49, 265–290. [DOI] [PubMed] [Google Scholar]
- (34).Harirforoosh S; Asghar W; Jamali F Adverse effects of nonsteroidal antiinflammatory drugs: an update of gastrointestinal, cardiovascular and renal complications. J. Pharm. Pharm. Sci 2013, 16, 821–847. [DOI] [PubMed] [Google Scholar]
- (35).Grosser T; Fries S; FitzGerald GA Biological basis for the cardiovascular consequences of COX-2 inhibition: therapeutic challenges and opportunities. J. Clin. Invest 2005, 116, 4–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (36).Koeberle A; Werz O Perspective of microsomal prostaglandin E2 synthase-1 as drug target in inflammation-related disorders. Biochem. Pharmacol 2015, 98, 1–15. [DOI] [PubMed] [Google Scholar]
- (37).Bergqvist F; Morgenstern R; Jakobsson PJ A review on mPGES-1 inhibitors: From preclinical studies to clinical applications. Prostaglandins Other Lipid Mediators 2020, 147, 106383. [DOI] [PubMed] [Google Scholar]
- (38).Quan Y; Jiang J; Dingledine R EP2 receptor signaling pathways regulate classical activation of microglia. J. Biol. Chem 2013, 288, 9293–9302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (39).Chang MC; Lin SI; Lin LD; Chan CP; Lee MS; Wang TM; Jeng PY; Yeung SY; Jeng JH Prostaglandin E2 Stimulates EP2, Adenylate Cyclase, Phospholipase C, and Intracellular Calcium Release to Mediate Cyclic Adenosine Monophosphate Production in Dental Pulp Cells. J. Endod 2016, 42, 584–588. [DOI] [PubMed] [Google Scholar]
- (40).Castellone MD; Teramoto H; Williams BO; Druey KM; Gutkind JS Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-beta-catenin signaling axis. Science 2005, 310, 1504–1510. [DOI] [PubMed] [Google Scholar]
- (41).Singh N; Bansal M; Pal S; Alam S; Jagdale P; Ayanur A; Ansari KM COX-2/EP2-EP4/beta-catenin signaling regulates patulin-induced intestinal cell proliferation and inflammation. Toxicol. Appl. Pharmacol 2018, 356, 224–234. [DOI] [PubMed] [Google Scholar]
- (42).Hsu HH; Lin YM; Shen CY; Shibu MA; Li SY; Chang SH; Lin CC; Chen RJ; Viswanadha VP; Shih HN; Huang CY Prostaglandin E2-Induced COX-2 Expressions via EP2 and EP4 Signaling Pathways in Human LoVo Colon Cancer Cells. Int. J. Mol. Sci 2017, 18, 1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (43).Aoki T; Frosen J; Fukuda M; Bando K; Shioi G; Tsuji K; Ollikainen E; Nozaki K; Laakkonen J; Narumiya S Prostaglandin E2-EP2-NF-kappaB signaling in macrophages as a potential therapeutic target for intracranial aneurysms. Sci. Signaling 2017, 10, No. eaah6037. [DOI] [PubMed] [Google Scholar]
- (44).Sun X; Li Q Prostaglandin EP2 receptor: Novel therapeutic target for human cancers (Review). Int. J. Mol. Med 2018, 42, 1203–1214. [DOI] [PubMed] [Google Scholar]
- (45).Donnini S; Finetti F; Solito R; Terzuoli E; Sacchetti A; Morbidelli L; Patrignani P; Ziche M EP2 prostanoid receptor promotes squamous cell carcinoma growth through epidermal growth factor receptor transactivation and iNOS and ERK1/2 pathways. FASEB J. 2007, 21, 2418–2430. [DOI] [PubMed] [Google Scholar]
- (46).Fernandez-Martinez AB; Lucio-Cazana FJ Transactivation of EGFR by prostaglandin E2 receptors: a nuclear story? Cell. Mol. Life Sci 2015, 72, 2187–2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (47).Chu CH; Chen SH; Wang Q; Langenbach R; Li H; Zeldin D; Chen SL; Wang S; Gao H; Lu RB; Hong JS PGE2 Inhibits IL-10 Production via EP2-Mediated beta-Arrestin Signaling in Neuroinflammatory Condition. Mol. Neurobiol 2015, 52, 587–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (48).Yun SP; Ryu JM; Jang MW; Han HJ Interaction of profilin-1 and F-actin via a beta-arrestin-1/JNK signaling pathway involved in prostaglandin E(2)-induced human mesenchymal stem cells migration and proliferation. J. Cell. Physiol 2011, 226, 559–571. [DOI] [PubMed] [Google Scholar]
- (49).Chun KS; Lao HC; Trempus CS; Okada M; Langenbach R The prostaglandin receptor EP2 activates multiple signaling pathways and beta-arrestin1 complex formation during mouse skin papilloma development. Carcinogenesis 2009, 30, 1620–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (50).Desai S; April H; Nwaneshiudu C; Ashby B Comparison of agonist-induced internalization of the human EP2 and EP4 prostaglandin receptors: role of the carboxyl terminus in EP4 receptor sequestration. Mol. Pharmacol 2000, 58, 1279–1286. [DOI] [PubMed] [Google Scholar]
- (51).Woodward DF; Jones RL; Narumiya S International Union of Basic and Clinical Pharmacology. LXXXIII: classification of prostanoid receptors, updating 15 years of progress. Pharmacol. Rev 2011, 63, 471–538. [DOI] [PubMed] [Google Scholar]
- (52).Markovic T; Jakopin Z; Dolenc MS; Mlinaric-Rascan I Structural features of subtype-selective EP receptor modulators. Drug Discovery Today 2017, 22, 57–71. [DOI] [PubMed] [Google Scholar]
- (53).Andreasson K Emerging roles of PGE2 receptors in models of neurological disease. Prostaglandins Other Lipid Mediators 2010, 91, 104–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (54).Ganesh T Prostanoid receptor EP2 as a therapeutic target. J. Med. Chem 2014, 57, 4454–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (55).Fu Y; Yang MS; Jiang J; Ganesh T; Joe E; Dingledine R EP2 Receptor Signaling Regulates Microglia Death. Mol. Pharmacol 2015, 88, 161–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (56).Johansson JU; Pradhan S; Lokteva LA; Woodling NS; Ko N; Brown HD; Wang Q; Loh C; Cekanaviciute E; Buckwalter M; Manning-Bog AB; Andreasson KI Suppression of inflammation with conditional deletion of the prostaglandin E2 EP2 receptor in macrophages and brain microglia. J. Neurosci 2013, 33, 16016–16032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (57).Johansson JU; Woodling NS; Wang Q; Panchal M; Liang X; Trueba-Saiz A; Brown HD; Mhatre SD; Loui T; Andreasson KI Prostaglandin signaling suppresses beneficial microglial function in Alzheimer’s disease models. J. Clin. Invest 2015, 125, 350–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (58).Hizaki H; Segi E; Sugimoto Y; Hirose M; Saji T; Ushikubi F; Matsuoka T; Noda Y; Tanaka T; Yoshida N; Narumiya S; Ichikawa A Abortive expansion of the cumulus and impaired fertility in mice lacking the prostaglandin E receptor subtype EP(2). Proc. Natl. Acad. Sci U. S. A 1999, 96, 10501–10506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (59).Kennedy CR; Zhang Y; Brandon S; Guan Y; Coffee K; Funk CD; Magnuson MA; Oates JA; Breyer MD; Breyer RM Salt-sensitive hypertension and reduced fertility in mice lacking the prostaglandin EP2 receptor. Nat. Med 1999, 5, 217–220. [DOI] [PubMed] [Google Scholar]
- (60).Tilley SL; Audoly LP; Hicks EH; Kim HS; Flannery PJ; Coffman TM; Koller BH Reproductive failure and reduced blood pressure in mice lacking the EP2 prostaglandin E2 receptor. J. Clin. Invest 1999, 103, 1539–1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (61).Alexander SPH; Cidlowski JA; Kelly E; Mathie A; Peters JA; Veale EL; Armstrong JF; Faccenda E; Harding SD; Pawson AJ; Sharman JL; Southan C; Davies JA; Coons L; Fuller P; Korach KS; Young M THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: G protein-coupled receptors. Br. J. Pharmacol 2019, 176, S21–S141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (62).Breyer RM; Clapp L; Coleman RA; Giembycz M; Heinemann A; Hills R; Jones RL; Narumiya S; Norel X; Pettipher R; et al. Prostanoid receptors (version 2020.4) in the IUPHAR/BPS Guide to Pharmacology Database. IUPHAR/BPS Guide to Pharmacology CITE 2020, 2020, 2020. [Google Scholar]
- (63).Abramovitz M; Adam M; Boie Y; Carriere M; Denis D; Godbout C; Lamontagne S; Rochette C; Sawyer N; Tremblay NM; Belley M; Gallant M; Dufresne C; Gareau Y; Ruel R; Juteau H; Labelle M; Ouimet N; Metters KM The utilization of recombinant prostanoid receptors to determine the affinities and selectivities of prostaglandins and related analogs. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2000, 1483, 285–293. [DOI] [PubMed] [Google Scholar]
- (64).Cameron KO; Lefker BA; Ke HZ; Li M; Zawistoski MP; Tjoa CM; Wright AS; DeNinno SL; Paralkar VM; Owen TA; Yu L; Thompson DD Discovery of CP-533536: an EP2 receptor selective prostaglandin E2 (PGE2) agonist that induces local bone formation. Bioorg. Med. Chem. Lett 2009, 19, 2075–2078. [DOI] [PubMed] [Google Scholar]
- (65).Belley M; Gallant M; Roy B; Houde K; Lachance N; Labelle M; Trimble LA; Chauret N; Li C; Sawyer N; Tremblay N; Lamontagne S; Carriere MC; Denis D; Greig GM; Slipetz D; Metters KM; Gordon R; Chan CC; Zamboni RJ Structure-activity relationship studies on ortho-substituted cinnamic acids, a new class of selective EP(3) antagonists. Bioorg. Med. Chem. Lett 2005, 15, 527–530. [DOI] [PubMed] [Google Scholar]
- (66).Jiang J; Ganesh T; Du Y; Thepchatri P; Rojas A; Lewis I; Kurtkaya S; Li L; Qui M; Serrano G; Shaw R; Sun A; Dingledine R Neuroprotection by selective allosteric potentiators of the EP2 prostaglandin receptor. Proc. Natl. Acad. Sci. U. S. A 2010, 107, 2307–2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (67).Jiang J; Van TM; Ganesh T; Dingledine R Discovery of 2-Piperidinyl Phenyl Benzamides and Trisubstituted Pyrimidines as Positive Allosteric Modulators of the Prostaglandin Receptor EP2. ACS Chem. Neurosci 2018, 9, 699–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (68).Qu C; Mao C; Xiao P; Shen Q; Zhong YN; Yang F; Shen DD; Tao X; Zhang H; Yan X; Zhao RJ; He J; Guan Y; Zhang C; Hou G; Zhang PJ; Hou G; Li Z; Yu X; Chai RJ; Guan YF; Sun JP; Zhang Y Ligand recognition, unconventional activation, and G protein coupling of the prostaglandin E2 receptor EP2 subtype. Sci. Adv 2021, 7, No. eabf1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (69).af Forselles KJ; Root J; Clarke T; Davey D; Aughton K; Dack K; Pullen N In vitro and in vivo characterization of PF-04418948, a novel, potent and selective prostaglandin EP(2) receptor antagonist. Br. J. Pharmacol 2011, 164, 1847–1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (70).Jiang J; Ganesh T; Du Y; Quan Y; Serrano G; Qui M; Speigel I; Rojas A; Lelutiu N; Dingledine R Small molecule antagonist reveals seizure-induced mediation of neuronal injury by prostaglandin E2 receptor subtype EP2. Proc. Natl. Acad. Sci. U. S. A 2012, 109, 3149–3154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (71).Fox BM; Beck HP; Roveto PM; Kayser F; Cheng Q; Dou H; Williamson T; Treanor J; Liu H; Jin L; Xu G; Ma J; Wang S; Olson SH A selective prostaglandin E2 receptor subtype 2 (EP2) antagonist increases the macrophage-mediated clearance of amyloid-beta plaques. J. Med. Chem 2015, 58, 5256–5273. [DOI] [PubMed] [Google Scholar]
- (72).Birrell MA; Nials AT At last, a truly selective EP(2) receptor antagonist. Br. J. Pharmacol 2011, 164, 1845–1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (73).Kay LJ; Gilbert M; Pullen N; Skerratt S; Farrington J; Seward EP; Peachell PT Characterization of the EP receptor subtype that mediates the inhibitory effects of prostaglandin E2 on IgE-dependent secretion from human lung mast cells. Clin. Exp. Allergy 2013, 43, 741–751. [DOI] [PubMed] [Google Scholar]
- (74).Ganesh T; Jiang J; Yang MS; Dingledine R Lead optimization studies of cinnamic amide EP2 antagonists. J. Med. Chem 2014, 57, 4173–4184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (75).Ganesh T; Jiang J; Dingledine R Development of second generation EP2 antagonists with high selectivity. Eur. J. Med. Chem 2014, 82, 521–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (76).Jiang J; Ganesh T; Du Y; Thepchatri P; Quan Y; Dingledine RJ Prostaglandin receptor EP2 antagonists, derivatives, compositions, and uses related thereto. U.S. Patent 10040783, 2018.
- (77).Ganesh T; Jiang J; Dingledine RJ Prostaglandin receptor EP2 antagonists, derivatives, compositions, and uses related thereto. U.S. Patent 10568889, 2020.
- (78).Norel X; Sugimoto Y; Ozen G; Abdelazeem H; Amgoud Y; Bouhadoun A; Bassiouni W; Goepp M; Mani S; Manikpurage HD; Senbel A; Longrois D; Heinemann A; Yao C; Clapp LH International Union of Basic and Clinical Pharmacology. CIX. Differences and Similarities between Human and Rodent Prostaglandin E2 Receptors (EP1–4) and Prostacyclin Receptor (IP): Specific Roles in Pathophysiologic Conditions. Pharmacol. Rev 2020, 72, 910–968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (79).Ganesh T; Banik A; Dingledine R; Wang W; Amaradhi R Peripherally Restricted, Highly Potent, Selective, Aqueous-Soluble EP2 Antagonist with Anti-Inflammatory Properties. Mol. Pharmaceutics 2018, 15, 5809–5817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (80).Amaradhi R; Banik A; Mohammed S; Patro V; Rojas A; Wang W; Motati DR; Dingledine R; Ganesh T Potent, Selective, Water Soluble, Brain-Permeable EP2 Receptor Antagonist for Use in Central Nervous System Disease Models. J. Med. Chem 2020, 63, 1032–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (81).Ganesh T; Jiang J; Shashidharamurthy R; Dingledine R Discovery and characterization of carbamothioylacrylamides as EP2 selective antagonists. ACS Med. Chem. Lett 2013, 4, 616–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (82).Michalski J; Kanaji N; Liu X; Nogel S; Wang X; Basma H; Nakanishi M; Sato T; Gunji Y; Fahrid M; Nelson A; Muller KC; Holz O; Magnussen H; Rabe KF; Toews ML; Rennard SI Attenuation of inhibitory prostaglandin E2 signaling in human lung fibroblasts is mediated by phosphodiesterase 4. Am. J. Respir. Cell Mol. Biol 2012, 47, 729–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (83).Dagouassat M; Gagliolo JM; Chrusciel S; Bourin MC; Duprez C; Caramelle P; Boyer L; Hue S; Stern JB; Validire P; Longrois D; Norel X; Dubois-Rande JL; Le Gouvello S; Adnot S; Boczkowski J The cyclooxygenase-2-prostaglandin E2 pathway maintains senescence of chronic obstructive pulmonary disease fibroblasts. Am. J. Respir. Crit. Care Med 2013, 187, 703–714. [DOI] [PubMed] [Google Scholar]
- (84).Jia XY; Chang Y; Sun XJ; Dai X; Wei W The role of prostaglandin E2 receptor signaling of dendritic cells in rheumatoid arthritis. Int. Immunopharmacol 2014, 23, 163–169. [DOI] [PubMed] [Google Scholar]
- (85).Schrödinger, Release 2021–2; Maestro, Schrödinger, LLC: New York, NY, 2021. [Google Scholar]
- (86).Trinka E; Cock H; Hesdorffer D; Rossetti AO; Scheffer ID; Shinnar S; Shorvon S; Lowenstein DH A definition and classification of status epilepticus–Report of the ILAE Task Force on Classification of Status Epilepticus. Epilepsia 2015, 56, 1515–1523. [DOI] [PubMed] [Google Scholar]
- (87).Betjemann JP; Lowenstein DH Status epilepticus in adults. Lancet Neurol. 2015, 14, 615–624. [DOI] [PubMed] [Google Scholar]
- (88).Rossetti AO; Lowenstein DH Management of refractory status epilepticus in adults: still more questions than answers. Lancet Neurol. 2011, 10, 922–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (89).Reddy DS; Kuruba R Experimental models of status epilepticus and neuronal injury for evaluation of therapeutic interventions. Int. J. Mol. Sci 2013, 14, 18284–18318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (90).Varvel NH; Jiang J; Dingledine R Candidate drug targets for prevention or modification of epilepsy. Annu. Rev. Pharmacol. Toxicol 2015, 55, 229–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (91).Loscher W Animal Models of Seizures and Epilepsy: Past, Present, and Future Role for the Discovery of Antiseizure Drugs. Neurochem. Res 2017, 42, 1873–1888. [DOI] [PubMed] [Google Scholar]
- (92).Dey A; Kang X; Qiu J; Du Y; Jiang J Anti-Inflammatory Small Molecules To Treat Seizures and Epilepsy: From Bench to Bedside. Trends Pharmacol. Sci 2016, 37, 463–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (93).Jiang J; Yang MS; Quan Y; Gueorguieva P; Ganesh T; Dingledine R Therapeutic window for cyclooxygenase-2 related antiinflammatory therapy after status epilepticus. Neurobiol. Dis 2015, 76, 126–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (94).Du Y; Kemper T; Qiu J; Jiang J Defining the therapeutic time window for suppressing the inflammatory prostaglandin E2 signaling after status epilepticus. Expert Rev. Neurother 2016, 16, 123–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (95).Fujikawa DG The temporal evolution of neuronal damage from pilocarpine-induced status epilepticus. Brain Res. 1996, 725, 11–22. [DOI] [PubMed] [Google Scholar]
- (96).Covolan L; Mello LE Temporal profile of neuronal injury following pilocarpine or kainic acid-induced status epilepticus. Epilepsy Res. 2000, 39, 133–152. [DOI] [PubMed] [Google Scholar]
- (97).Freitas RM; Vasconcelos SM; Souza FC; Viana GS; Fonteles MM Oxidative stress in the hippocampus after pilocarpine-induced status epilepticus in Wistar rats. FEBS J. 2005, 272, 1307–1312. [DOI] [PubMed] [Google Scholar]
- (98).Serrano GE; Lelutiu N; Rojas A; Cochi S; Shaw R; Makinson CD; Wang D; FitzGerald GA; Dingledine R Ablation of cyclooxygenase-2 in forebrain neurons is neuroprotective and dampens brain inflammation after status epilepticus. J. Neurosci 2011, 31, 14850–14860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (99).Rojas A; Jiang J; Ganesh T; Yang MS; Lelutiu N; Gueorguieva P; Dingledine R Cyclooxygenase-2 in epilepsy. Epilepsia 2014, 55, 17–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (100).Rojas A; Amaradhi R; Banik A; Jiang C; Abreu-Melon J; Wang S; Dingledine R; Ganesh T A Novel Second-Generation EP2 Receptor Antagonist Reduces Neuroinflammation and Gliosis After Status Epilepticus in Rats. Neurotherapeutics 2021. DOI: 10.1007/s13311-020-00969-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- (101).Yu Y; Li L; Nguyen DT; Mustafa SM; Moore BM; Jiang J Inverse Agonism of Cannabinoid Receptor Type 2 Confers Anti-inflammatory and Neuroprotective Effects Following Status Epilepticus. Mol. Neurobiol 2020, 57, 2830–2845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (102).Bymaster FP; Carter PA; Yamada M; Gomeza J; Wess J; Hamilton SE; Nathanson NM; McKinzie DL; Felder CC Role of specific muscarinic receptor subtypes in cholinergic parasympathomimetic responses, in vivo phosphoinositide hydrolysis, and pilocarpine-induced seizure activity. European journal of neuroscience 2003, 17, 1403–1410. [DOI] [PubMed] [Google Scholar]
- (103).Mulle C; Sailer A; Perez-Otano I; Dickinson-Anson H; Castillo PE; Bureau I; Maron C; Gage FH; Mann JR; Bettler B; Heinemann SF Altered synaptic physiology and reduced susceptibility to kainate-induced seizures in GluR6-deficient mice. Nature 1998, 392, 601–605. [DOI] [PubMed] [Google Scholar]
- (104).Ben-Ari Y. Kainate and Temporal Lobe Epilepsies: 3 decades of progress. In Jasper’s Basic Mechanisms of the Epilepsies; Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, Eds.; National Center for Biotechnology Information: Bethesda, MD, 2012. [PubMed] [Google Scholar]
- (105).Soldner ELB; Hartz AMS; Akanuma SI; Pekcec A; Doods H; Kryscio RJ; Hosoya KI; Bauer B Inhibition of human microsomal PGE2 synthase-1 reduces seizure-induced increases of P-glycoprotein expression and activity at the blood-brain barrier. FASEB J. 2019, 33, 13966–13981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (106).Binder DK; Croll SD; Gall CM; Scharfman HE BDNF and epilepsy: too much of a good thing? Trends Neurosci. 2001, 24, 47–53. [DOI] [PubMed] [Google Scholar]
- (107).McNamara JO; Scharfman HE Temporal Lobe Epilepsy and the BDNF Receptor, TrkB. In Jasper’s Basic Mechanisms of the Epilepsies; Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, Eds.; National Center for Biotechnology Information: Bethesda, MD, 2012. [PubMed] [Google Scholar]
- (108).Thomas AX; Cruz Del Angel Y; Gonzalez MI; Carrel AJ; Carlsen J; Lam PM; Hempstead BL; Russek SJ; Brooks-Kayal AR Rapid Increases in proBDNF after Pilocarpine-Induced Status Epilepticus in Mice Are Associated with Reduced proBDNF Cleavage Machinery. eNeuro 2016, 3, No. e0020–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (109).Lin TW; Harward SC; Huang YZ; McNamara JO Targeting BDNF/TrkB pathways for preventing or suppressing epilepsy. Neuropharmacology 2020, 167, 107734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (110).Liu G; Gu B; He XP; Joshi RB; Wackerle HD; Rodriguiz RM; Wetsel WC; McNamara JO Transient inhibition of TrkB kinase after status epilepticus prevents development of temporal lobe epilepsy. Neuron 2013, 79, 31–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (111).Gu B; Huang YZ; He XP; Joshi RB; Jang W; McNamara JO A Peptide Uncoupling BDNF Receptor TrkB from Phospholipase Cgamma1 Prevents Epilepsy Induced by Status Epilepticus. Neuron 2015, 88, 484–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (112).Seibert K; Zhang Y; Leahy K; Hauser S; Masferrer J; Perkins W; Lee L; Isakson P Pharmacological and biochemical demonstration of the role of cyclooxygenase 2 in inflammation and pain. Proc. Natl. Acad. Sci. U. S. A 1994, 91, 12013–12017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (113).Lund IV; Hu Y; Raol YH; Benham RS; Faris R; Russek SJ; Brooks-Kayal AR BDNF selectively regulates GABAA receptor transcription by activation of the JAK/STAT pathway. Sci. Signaling 2008, 1 , No. ra9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (114).Xue W; Wang W; Gong T; Zhang H; Tao W; Xue L; Sun Y; Wang F; Chen G PKA-CREB-BDNF signaling regulated long lasting antidepressant activities of Yueju but not ketamine. Sci. Rep 2016, 6, 26331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (115).Finkbeiner S; Tavazoie SF; Maloratsky A; Jacobs KM; Harris KM; Greenberg ME CREB: a major mediator of neuronal neurotrophin responses. Neuron 1997, 19, 1031–1047. [DOI] [PubMed] [Google Scholar]
- (116).Rojas A; Ganesh T; Wang W; Wang J; Dingledine R A rat model of organophosphate-induced status epilepticus and the beneficial effects of EP2 receptor inhibition. Neurobiol. Dis 2020, 133, 104399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (117).Rojas A; McCarren HS; Wang J; Wang W; Abreu-Melon J; Wang S; McDonough JH; Dingledine R Comparison of neuropathology in rats following status epilepticus induced by diisopropylfluorophosphate and soman. NeuroToxicology 2021, 83, 14–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (118).Rojas A; Ganesh T; Lelutiu N; Gueorguieva P; Dingledine R Inhibition of the prostaglandin EP2 receptor is neuroprotective and accelerates functional recovery in a rat model of organophosphorus induced status epilepticus. Neuropharmacology 2015, 93, 15–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (119).Rojas A; Ganesh T; Manji Z; O’Neill T; Dingledine R Inhibition of the prostaglandin E2 receptor EP2 prevents status epilepticus-induced deficits in the novel object recognition task in rats. Neuropharmacology 2016, 110, 419–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (120).Dube CM; Brewster AL; Richichi C; Zha Q; Baram TZ Fever, febrile seizures and epilepsy. Trends Neurosci. 2007, 30, 490–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (121).Yokoi S; Kidokoro H; Yamamoto H; Ohno A; Nakata T; Kubota T; Tsuji T; Morishita M; Kawabe T; Naiki M; Maruyama K; Itomi K; Kato T; Ito K; Natsume J Hippocampal diffusion abnormality after febrile status epilepticus is related to subsequent epilepsy. Epilepsia 2019, 60, 1306–1316. [DOI] [PubMed] [Google Scholar]
- (122).Patterson KP; Brennan GP; Curran M; Kinney-Lang E; Dube C; Rashid F; Ly C; Obenaus A; Baram TZ Rapid, Coordinate Inflammatory Responses after Experimental Febrile Status Epilepticus: Implications for Epileptogenesis. eNeuro 2015, 2, No. e0034–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (123).Brennan GP; Garcia-Curran MM; Patterson KP; Luo R; Baram TZ Multiple Disruptions of Glial-Neuronal Networks in Epileptogenesis That Follows Prolonged Febrile Seizures. Front Neurol 2021, 12, 615802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (124).Sang N; Zhang J; Marcheselli V; Bazan NG; Chen C Postsynaptically synthesized prostaglandin E2 (PGE2) modulates hippocampal synaptic transmission via a presynaptic PGE2 EP2 receptor. J. Neurosci 2005, 25, 9858–9870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (125).Savonenko A; Munoz P; Melnikova T; Wang Q; Liang X; Breyer RM; Montine TJ; Kirkwood A; Andreasson K Impaired cognition, sensorimotor gating, and hippocampal long-term depression in mice lacking the prostaglandin E2 EP2 receptor. Exp. Neurol 2009, 217, 63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (126).Catanese L; Tarsia J; Fisher M Acute Ischemic Stroke Therapy Overview. Circ. Res 2017, 120, 541–558. [DOI] [PubMed] [Google Scholar]
- (127).Nogawa S; Zhang F; Ross ME; Iadecola C Cyclooxygenase-2 gene expression in neurons contributes to ischemic brain damage. J. Neurosci 1997, 17, 2746–2755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (128).Nakayama M; Uchimura K; Zhu RL; Nagayama T; Rose ME; Stetler RA; Isakson PC; Chen J; Graham SH Cyclooxygenase-2 inhibition prevents delayed death of CA1 hippocampal neurons following global ischemia. Proc. Natl. Acad. Sci. U. S. A 1998, 95, 10954–10959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (129).Iadecola C; Niwa K; Nogawa S; Zhao X; Nagayama M; Araki E; Morham S; Ross ME Reduced susceptibility to ischemic brain injury and N-methyl-D-aspartate-mediated neurotoxicity in cyclooxygenase-2-deficient mice. Proc. Natl. Acad. Sci. U. S. A 2001, 98, 1294–1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (130).Dore S; Otsuka T; Mito T; Sugo N; Hand T; Wu L; Hurn PD; Traystman RJ; Andreasson K Neuronal overexpression of cyclooxygenase-2 increases cerebral infarction. Ann. Neurol 2003, 54, 155–162. [DOI] [PubMed] [Google Scholar]
- (131).Sugimoto K; Iadecola C Delayed effect of administration of COX-2 inhibitor in mice with acute cerebral ischemia. Brain Res. 2003, 960, 273–276. [DOI] [PubMed] [Google Scholar]
- (132).Frankowski JC; DeMars KM; Ahmad AS; Hawkins KE; Yang C; Leclerc JL; Dore S; Candelario-Jalil E Detrimental role of the EP1 prostanoid receptor in blood-brain barrier damage following experimental ischemic stroke. Sci. Rep 2015, 5, 17956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (133).McCullough L; Wu L; Haughey N; Liang X; Hand T; Wang Q; Breyer RM; Andreasson K Neuroprotective function of the PGE2 EP2 receptor in cerebral ischemia. J. Neurosci 2004, 24, 257–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (134).Liu D; Wu L; Breyer R; Mattson MP; Andreasson K Neuroprotection by the PGE2 EP2 receptor in permanent focal cerebral ischemia. Ann. Neurol 2005, 57, 758–761. [DOI] [PubMed] [Google Scholar]
- (135).Ahmad M; Saleem S; Shah Z; Maruyama T; Narumiya S; Dore S The PGE2 EP2 receptor and its selective activation are beneficial against ischemic stroke. Exp. Transl. Stroke Med 2010, 2, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (136).Braak H; Braak E Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol. Aging 1997, 18, 351–357. [DOI] [PubMed] [Google Scholar]
- (137).Simard AR; Soulet D; Gowing G; Julien JP; Rivest S Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer’s disease. Neuron 2006, 49, 489–502. [DOI] [PubMed] [Google Scholar]
- (138).Aronoff DM; Canetti C; Peters-Golden M Prostaglandin E2 inhibits alveolar macrophage phagocytosis through an E-prostanoid 2 receptor-mediated increase in intracellular cyclic AMP. J. Immunol 2004, 173, 559–565. [DOI] [PubMed] [Google Scholar]
- (139).Woodling NS; Andreasson KI Untangling the Web: Toxic and Protective Effects of Neuroinflammation and PGE2 Signaling in Alzheimer’s Disease. ACS Chem. Neurosci 2016, 7, 454–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (140).Keene CD; Chang RC; Lopez-Yglesias AH; Shalloway BR; Sokal I; Li X; Reed PJ; Keene LM; Montine KS; Breyer RM; Rockhill JK; Montine TJ Suppressed accumulation of cerebral amyloid {beta} peptides in aged transgenic Alzheimer’s disease mice by transplantation with wild-type or prostaglandin E2 receptor subtype 2-null bone marrow. Am. J. Pathol 2010, 177, 346–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (141).Ruscheweyh R; Wilder-Smith O; Drdla R; Liu XG; Sandkuhler J Long-term potentiation in spinal nociceptive pathways as a novel target for pain therapy. Mol. Pain 2011, 7, 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (142).Ghilardi JR; Svensson CI; Rogers SD; Yaksh TL; Mantyh PW Constitutive spinal cyclooxygenase-2 participates in the initiation of tissue injury-induced hyperalgesia. J. Neurosci 2004, 24, 2727–2732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (143).Li J; Serafin E; Baccei ML Prostaglandin Signaling Governs Spike Timing-Dependent Plasticity at Sensory Synapses onto Mouse Spinal Projection Neurons. J. Neurosci 2018, 38, 6628–6639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (144).Beiche F; Scheuerer S; Brune K; Geisslinger G; Goppelt-Struebe M Up-regulation of cyclooxygenase-2 mRNA in the rat spinal cord following peripheral inflammation. FEBS Lett. 1996, 390, 165–169. [DOI] [PubMed] [Google Scholar]
- (145).Ichitani Y; Shi T; Haeggstrom JZ; Samuelsson B; Hokfelt T Increased levels of cyclooxygenase-2 mRNA in the rat spinal cord after peripheral inflammation: an in situ hybridization study. NeuroReport 1997, 8, 2949–2952. [DOI] [PubMed] [Google Scholar]
- (146).SCHAIBLE H-G; EBERSBERGER A; BANCHET GS Mechanisms of pain in arthritis. Ann. N. Y. Acad. Sci 2002, 966, 343–354. [DOI] [PubMed] [Google Scholar]
- (147).Ma W; Quirion R Does COX2-dependent PGE2 play a role in neuropathic pain? Neurosci. Lett 2008, 437, 165–169. [DOI] [PubMed] [Google Scholar]
- (148).Schaible HG; Richter F; Ebersberger A; Boettger MK; Vanegas H; Natura G; Vazquez E; Segond von Banchet G Joint pain. Exp. Brain Res 2009, 196, 153–162. [DOI] [PubMed] [Google Scholar]
- (149).Noble LS; Takayama K; Zeitoun KM; Putman JM; Johns DA; Hinshelwood MM; Agarwal VR; Zhao Y; Carr BR; Bulun SE Prostaglandin E2 stimulates aromatase expression in endometriosis-derived stromal cells. J. Clin. Endocrinol. Metab 1997, 82, 600–606. [DOI] [PubMed] [Google Scholar]
- (150).Chishima F; Hayakawa S; Sugita K; Kinukawa N; Aleemuzzaman S; Nemoto N; Yamamoto T; Honda M Increased expression of cyclooxygenase-2 in local lesions of endometriosis patients. Am. J. Reprod. Immunol 2002, 48, 50–56. [DOI] [PubMed] [Google Scholar]
- (151).Banu SK; Lee J; Speights VO Jr.; Starzinski-Powitz A; Arosh JA Cyclooxygenase-2 regulates survival, migration, and invasion of human endometriotic cells through multiple mechanisms. Endocrinology 2008, 149, 1180–1189. [DOI] [PubMed] [Google Scholar]
- (152).Greaves E; Horne AW; Jerina H; Mikolajczak M; Hilferty L; Mitchell R; Fleetwood-Walker SM; Saunders PT EP2 receptor antagonism reduces peripheral and central hyperalgesia in a preclinical mouse model of endometriosis. Sci. Rep 2017, 7, 44169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (153).Investigators UJ; Morita A; Kirino T; Hashi K; Aoki N; Fukuhara S; Hashimoto N; Nakayama T; Sakai M; Teramoto A; Tominari S; Yoshimoto T The natural course of unruptured cerebral aneurysms in a Japanese cohort. N. Engl. J. Med 2012, 366, 2474–2482. [DOI] [PubMed] [Google Scholar]
- (154).Chyatte D; Bruno G; Desai S; Todor DR Inflammation and intracranial aneurysms. Neurosurgery 1999, 45, 1137–46 discussion 1146–7;. [DOI] [PubMed] [Google Scholar]
- (155).Aoki T; Nishimura M; Matsuoka T; Yamamoto K; Furuyashiki T; Kataoka H; Kitaoka S; Ishibashi R; Ishibazawa A; Miyamoto S; Morishita R; Ando J; Hashimoto N; Nozaki K; Narumiya S PGE(2) -EP(2) signalling in endothelium is activated by haemodynamic stress and induces cerebral aneurysm through an amplifying loop via NF-kappaB. Br. J. Pharmacol 2011, 163, 1237–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (156).Zabel MK; Zhao L; Zhang Y; Gonzalez SR; Ma W; Wang X; Fariss RN; Wong WT Microglial phagocytosis and activation underlying photoreceptor degeneration is regulated by CX3CL1-CX3CR1 signaling in a mouse model of retinitis pigmentosa. Glia 2016, 64, 1479–1491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (157).Yang W; Xiong G; Lin B Cyclooxygenase-1 mediates neuroinflammation and neurotoxicity in a mouse model of retinitis pigmentosa. J. Neuroinflammation 2020, 17, 306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (158).Jiang C; Caskurlu A; Ganesh T; Dingledine R Inhibition of the prostaglandin EP2 receptor prevents long-term cognitive impairment in a model of systemic inflammation. Brain, Behavior, & Immunity - Health 2020, 8, 100132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (159).Zelenay S; van der Veen AG; Bottcher JP; Snelgrove KJ; Rogers N; Acton SE; Chakravarty P; Girotti MR; Marais R; Quezada SA; Sahai E; Reis e Sousa C Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity. Cell 2015, 162, 1257–1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (160).Matthay KK; Maris JM; Schleiermacher G; Nakagawara A; Mackall CL; Diller L; Weiss WA Neuroblastoma. Nat. Rev. Dis Primers 2016, 2, 16078. [DOI] [PubMed] [Google Scholar]
- (161).Pannunzio A; Coluccia M Cyclooxygenase-1 (COX-1) and COX-1 Inhibitors in Cancer: A Review of Oncology and Medicinal Chemistry Literature. Pharmaceuticals 2018, 11, 101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (162).Sonoshita M; Takaku K; Sasaki N; Sugimoto Y; Ushikubi D; Narumiya S; Oshima M; Taketo MM Acceleration of intestinal polyposis through prostaglandin receptor EP2 in Apc(Delta 716) knockout mice. Nat. Med 2001, 7, 1048–1051. [DOI] [PubMed] [Google Scholar]
- (163).Yang L; Yamagata N; Yadav R; Brandon S; Courtney RL; Morrow JD; Shyr Y; Boothby M; Joyce S; Carbone DP; Breyer RM Cancer-associated immunodeficiency and dendritic cell abnormalities mediated by the prostaglandin EP2 receptor. J. Clin. Invest 2003, 111, 727–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (164).Chang SH; Ai Y; Breyer RM; Lane TF; Hla T The prostaglandin E2 receptor EP2 is required for cyclooxygenase 2-mediated mammary hyperplasia. Cancer Res. 2005, 65, 4496–4499. [DOI] [PubMed] [Google Scholar]
- (165).Sung YM; He G; Fischer SM Lack of expression of the EP2 but not EP3 receptor for prostaglandin E2 results in suppression of skin tumor development. Cancer Res. 2005, 65, 9304–9311. [DOI] [PubMed] [Google Scholar]
- (166).Keith RL; Geraci MW; Nana-Sinkam SP; Breyer RM; Hudish TM; Meyer AM; Malkinson AM; Dwyer-Nield LD Prostaglandin E2 receptor subtype 2 (EP2) null mice are protected against murine lung tumorigenesis. Anticancer Res. 2006, 26, 2857–2861. [PubMed] [Google Scholar]
- (167).Greenhough A; Smartt HJ; Moore AE; Roberts HR; Williams AC; Paraskeva C; Kaidi A The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 2009, 30, 377–386. [DOI] [PubMed] [Google Scholar]
- (168).Aoki T; Narumiya S Prostaglandin E2-EP2 signaling as a node of chronic inflammation in the colon tumor microenvironment. Inflamm Regen 2017, 37, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (169).Brenner H; Kloor M; Pox CP Colorectal cancer. Lancet 2014, 383, 1490–1502. [DOI] [PubMed] [Google Scholar]
- (170).Clevers H Colon cancer–understanding how NSAIDs work. N. Engl. J. Med 2006, 354, 761–763. [DOI] [PubMed] [Google Scholar]
- (171).Rothwell PM; Fowkes FG; Belch JF; Ogawa H; Warlow CP; Meade TW Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet 2011, 377, 31–41. [DOI] [PubMed] [Google Scholar]
- (172).Bosetti C; Rosato V; Gallus S; Cuzick J; La Vecchia C Aspirin and cancer risk: a quantitative review to 2011. Ann. Oncol 2012, 23, 1403–1415. [DOI] [PubMed] [Google Scholar]
- (173).Wang D; DuBois RN An inflammatory mediator, prostaglandin E2, in colorectal cancer. Cancer J. 2013, 19, 502–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (174).Karpisheh V; Nikkhoo A; Hojjat-Farsangi M; Namdar A; Azizi G; Ghalamfarsa G; Sabz G; Yousefi M; Yousefi B; Jadidi-Niaragh F Prostaglandin E2 as a potent therapeutic target for treatment of colon cancer. Prostaglandins Other Lipid Mediators 2019, 144, 106338. [DOI] [PubMed] [Google Scholar]
- (175).Ma X; Aoki T; Tsuruyama T; Narumiya S Definition of Prostaglandin E2-EP2 Signals in the Colon Tumor Microenvironment That Amplify Inflammation and Tumor Growth. Cancer Res. 2015, 75, 2822–2832. [DOI] [PubMed] [Google Scholar]
- (176).Stupp R; Hegi ME; Mason WP; van den Bent MJ; Taphoorn MJ; Janzer RC; Ludwin SK; Allgeier A; Fisher B; Belanger K; Hau P; Brandes AA; Gijtenbeek J; Marosi C; Vecht CJ; Mokhtari K; Wesseling P; Villa S; Eisenhauer E; Gorlia T; Weller M; Lacombe D; Cairncross JG; Mirimanoff RO Effects of radiotherapy with concomitant and adjuvant Temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [DOI] [PubMed] [Google Scholar]
- (177).Omuro A; DeAngelis LM Glioblastoma and other malignant gliomas: a clinical review. JAMA 2013, 310, 1842–1850. [DOI] [PubMed] [Google Scholar]
- (178).Alexander BM; Cloughesy TF Adult Glioblastoma. J. Clin. Oncol 2017, 35, 2402–2409. [DOI] [PubMed] [Google Scholar]
- (179).Aldape K; Brindle KM; Chesler L; Chopra R; Gajjar A; Gilbert MR; Gottardo N; Gutmann DH; Hargrave D; Holland EC; Jones DTW; Joyce JA; Kearns P; Kieran MW; Mellinghoff IK; Merchant M; Pfister SM; Pollard SM; Ramaswamy V; Rich JN; Robinson GW; Rowitch DH; Sampson JH; Taylor MD; Workman P; Gilbertson RJ Challenges to curing primary brain tumours. Nat. Rev. Clin. Oncol 2019, 16, 509–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (180).Mellinghoff IK; Gilbertson RJ Brain Tumors: Challenges and Opportunities to Cure. J. Clin. Oncol 2017, 35, 2343–2345. [DOI] [PubMed] [Google Scholar]
- (181).Tan AC; Ashley DM; Lopez GY; Malinzak M; Friedman HS; Khasraw M Management of glioblastoma: State of the art and future directions. Ca-Cancer J. Clin 2020, 70, 299–312. [DOI] [PubMed] [Google Scholar]
- (182).Sowers JL; Johnson KM; Conrad C; Patterson JT; Sowers LC The role of inflammation in brain cancer. Adv. Exp. Med. Biol 2014, 816, 75–105. [DOI] [PubMed] [Google Scholar]
- (183).Joki T; Heese O; Nikas DC; Bello L; Zhang J; Kraeft SK; Seyfried NT; Abe T; Chen LB; Carroll RS; Black PM Expression of cyclooxygenase 2 (COX-2) in human glioma and in vitro inhibition by a specific COX-2 inhibitor, NS-398. Cancer Res. 2000, 60, 4926–4931. [PubMed] [Google Scholar]
- (184).Patti R; Gumired K; Reddanna P; Sutton LN; Phillips PC; Reddy CD Overexpression of cyclooxygenase-2 (COX-2) in human primitive neuroectodermal tumors: effect of celecoxib and rofecoxib. Cancer Lett. 2002, 180, 13–21. [DOI] [PubMed] [Google Scholar]
- (185).Xu K; Shu HK Transcription factor interactions mediate EGF-dependent COX-2 expression. Mol. Cancer Res 2013, 11, 875–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (186).Xu K; Wang L; Shu HK COX-2 overexpression increases malignant potential of human glioma cells through Id1. Oncotarget 2014, 5, 1241–1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (187).Qiu J; Shi Z; Jiang J Cyclooxygenase-2 in glioblastoma multiforme. Drug Discovery Today 2017, 22, 148–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (188).Qiu J; Li Q; Bell KA; Yao X; Du Y; Zhang E; Yu JJ; Yu Y; Shi Z; Jiang J Small-molecule inhibition of prostaglandin E receptor 2 impairs cyclooxygenase-associated malignant glioma growth. Br. J. Pharmacol 2019, 176, 1680–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (189).Jiang J Methods and compositions for the treatment of gliomas. U.S. Patent 10758516, 2020.
- (190).Maris JM Recent advances in neuroblastoma. N. Engl. J. Med 2010, 362, 2202–2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (191).Carlson LM; Kogner P Neuroblastoma-related inflammation: May small doses of aspirin be suitable for small cancer patients? Oncoimmunology 2013, 2, No. e24658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (192).Johnsen JI; Lindskog M; Ponthan F; Pettersen I; Elfman L; Orrego A; Sveinbjornsson B; Kogner P Cyclooxygenase-2 is expressed in neuroblastoma, and nonsteroidal anti-inflammatory drugs induce apoptosis and inhibit tumor growth in vivo. Cancer Res. 2004, 64, 7210–7215. [DOI] [PubMed] [Google Scholar]
- (193).Larsson K; Kock A; Idborg H; Arsenian Henriksson M; Martinsson T; Johnsen JI; Korotkova M; Kogner P; Jakobsson PJ COX/mPGES-1/PGE2 pathway depicts an inflammatory-dependent high-risk neuroblastoma subset. Proc. Natl. Acad. Sci. U. S. A 2015, 112, 8070–8075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (194).Carlson LM; Rasmuson A; Idborg H; Segerstrom L; Jakobsson PJ; Sveinbjornsson B; Kogner P Low-dose aspirin delays an inflammatory tumor progression in vivo in a transgenic mouse model of neuroblastoma. Carcinogenesis 2013, 34, 1081–1088. [DOI] [PubMed] [Google Scholar]
- (195).Kock A; Larsson K; Bergqvist F; Eissler N; Elfman LHM; Raouf J; Korotkova M; Johnsen JI; Jakobsson PJ; Kogner P Inhibition of Microsomal Prostaglandin E Synthase-1 in Cancer-Associated Fibroblasts Suppresses Neuroblastoma Tumor Growth. EBioMedicine 2018, 32, 84–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (196).Kock A; Bergqvist F; Steinmetz J; Elfman LHM; Korotkova M; Johnsen JI; Jakobsson PJ; Kogner P; Larsson K Establishment of an in vitro 3D model for neuroblastoma enables preclinical investigation of combined tumor-stroma drug targeting. FASEB J. 2020, 34, 11101–11114. [DOI] [PubMed] [Google Scholar]
- (197).Hou R; Yu Y; Nguyen DT; Sluter MN; Li L; Yang J; Jiang J Prostaglandin receptor EP2 is a novel molecular target for high-risk neuroblastoma. BioRxiv 2020. DOI: 10.1101/2020.02.24.963108. [DOI] [Google Scholar]
- (198).Dawn A; Yao X; Yu Y; Jiang J; Kumari H Assessment of the in vitro toxicity of calixarenes and a metal-seamed calixarene: a chemical pathway for clinical application. Supramol. Chem 2019, 31, 425–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (199).Anrather J; Iadecola C Inflammation and Stroke: An Overview. Neurotherapeutics 2016, 13, 661–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (200).Hartings JA; York J; Carroll CP; Hinzman JM; Mahoney E; Krueger B; Winkler MKL; Major S; Horst V; Jahnke P; Woitzik J; Kola V; Du Y; Hagen M; Jiang J; Dreier JP Subarachnoid blood acutely induces spreading depolarizations and early cortical infarction. Brain 2017, 140, 2673–2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (201).Jayaraj RL; Azimullah S; Beiram R; Jalal FY; Rosenberg GA Neuroinflammation: friend and foe for ischemic stroke. J. Neuroinflammation 2019, 16, 142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (202).Aronica E; Bauer S; Bozzi Y; Caleo M; Dingledine R; Gorter JA; Henshall DC; Kaufer D; Koh S; Löscher W; Louboutin JP; Mishto M; Norwood BA; Palma E; Poulter MO; Terrone G; Vezzani A; Kaminski RM Neuroinflammatory targets and treatments for epilepsy validated in experimental models. Epilepsia 2017, 58, 27–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (203).Rana A; Musto AE The role of inflammation in the development of epilepsy. J. Neuroinflammation 2018, 15, 144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (204).Myers RR; Campana WM; Shubayev VI The role of neuroinflammation in neuropathic pain: mechanisms and therapeutic targets. Drug Discovery Today 2006, 11, 8–20. [DOI] [PubMed] [Google Scholar]
- (205).Ellis A; Bennett DL Neuroinflammation and the generation of neuropathic pain. Br. J. Anaesth 2013, 111, 26–37. [DOI] [PubMed] [Google Scholar]
- (206).Hirsch EC; Hunot S Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009, 8, 382–397. [DOI] [PubMed] [Google Scholar]
- (207).Philips T; Robberecht W Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease. Lancet Neurol. 2011, 10, 253–263. [DOI] [PubMed] [Google Scholar]
- (208).Heneka MT; Carson MJ; Khoury J; Landreth GE; Brosseron F; Feinstein DL; Jacobs AH; Wyss-Coray T; Vitorica J; Ransohoff RM; Herrup K; Frautschy SA; Finsen B; Brown GC; Verkhratsky A; Yamanaka K; Koistinaho J; Latz E; Halle A; Petzold GC; Town T; Morgan D; Shinohara ML; Perry VH; Holmes C; Bazan NG; Brooks DJ; Hunot S; Joseph B; Deigendesch N; Garaschuk O; Boddeke E; Dinarello CA; Breitner JC; Cole GM; Golenbock DT; Kummer MP Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (209).Chen WW; Zhang X; Huang WJ Role of neuroinflammation in neurodegenerative diseases (Review). Mol. Med. Rep 2016, 13, 3391–3396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (210).Jin J; Shie FS; Liu J; Wang Y; Davis J; Schantz AM; Montine KS; Montine TJ; Zhang J Prostaglandin E2 receptor subtype 2 (EP2) regulates microglial activation and associated neurotoxicity induced by aggregated alpha-synuclein. J. Neuroinflammation 2007, 4, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (211).Liang X; Wang Q; Shi J; Lokteva L; Breyer RM; Montine TJ; Andreasson K The prostaglandin E2 EP2 receptor accelerates disease progression and inflammation in a model of amyotrophic lateral sclerosis. Ann. Neurol 2008, 64, 304–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (212).Nagarsheth N; Wicha MS; Zou W Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol 2017, 17, 559–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (213).Greten FR; Grivennikov SI Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (214).Ferrara JL; Abhyankar S; Gilliland DG Cytokine storm of graft-versus-host disease: a critical effector role for interleukin-1. Transplant Proc. 1993, 25, 1216–1217. [PubMed] [Google Scholar]
- (215).Ferrara JL Cytokine dysregulation as a mechanism of graft versus host disease. Curr. Opin. Immunol 1993, 5, 794–799. [DOI] [PubMed] [Google Scholar]
- (216).Fajgenbaum DC; June CH Cytokine Storm. N. Engl. J. Med 2020, 383, 2255–2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (217).Liu M; Gu C; Wu J; Zhu Y Amino acids 1 to 422 of the spike protein of SARS associated coronavirus are required for induction of cyclooxygenase-2. Virus Genes 2006, 33, 309–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (218).Baghaki S; Yalcin CE; Baghaki HS; Aydin SY; Daghan B; Yavuz E COX2 inhibition in the treatment of COVID-19: Review of literature to propose repositioning of celecoxib for randomized controlled studies. Int. J. Infect. Dis 2020, 101, 29–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (219).Robb CT; Goepp M; Rossi AG; Yao C Non-steroidal anti-inflammatory drugs, prostaglandins, and COVID-19. Br. J. Pharmacol 2020, 177, 4899–4920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (220).Chen JS; Alfajaro MM; Chow RD; Wei J; Filler RB; Eisenbarth SC; Wilen CB Non-steroidal anti-inflammatory drugs dampen the cytokine and antibody response to SARS-CoV-2 infection. J. Virol 2021, 95, No. e00014–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (221).FitzGerald GA Misguided drug advice for COVID-19. Science 2020, 367, 1434. [DOI] [PubMed] [Google Scholar]
- (222).Capuano A; Scavone C; Racagni G; Scaglione F NSAIDs in patients with viral infections, including Covid-19: Victims or perpetrators? Pharmacol. Res 2020, 157, 104849. [DOI] [PMC free article] [PubMed] [Google Scholar]
