Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Activation of prostanoid EP2 receptor exacerbates neuroinflammatory and neurodegenerative pathology in central nervous system diseases such as epilepsy, Alzheimer's disease, and cerebral aneurysms. A selective and brain-permeable EP2 antagonist will be useful to attenuate the inflammatory consequences of EP2 activation and to reduce the severity of these chronic diseases. We recently developed a brain-permeable EP2 antagonist 1 (TG6-10-1), which displayed anti-inflammatory and neuroprotective actions in rodent models of status epilepticus. However, this compound exhibited moderate selectivity to EP2, a short plasma half-life in rodents (1.7 h) and low aqueous solubility (27 μM), limiting its use in animal models of chronic disease. With lead-optimization studies, we have developed several novel EP2 antagonists with improved water solubility, brain penetration, high EP2 potency, and selectivity. These novel inhibitors suppress inflammatory gene expression induced by EP2 receptor activation in a microglial cell line, reinforcing the use of EP2 antagonists as anti-inflammatory agents.

Free full text 


Logo of nihpaAbout Author manuscriptsSubmit a manuscriptHHS Public Access; Author Manuscript; Accepted for publication in peer reviewed journal;
J Med Chem. Author manuscript; available in PMC 2021 Feb 13.
Published in final edited form as:
PMCID: PMC7394479
NIHMSID: NIHMS1607948
PMID: 31904232

Potent, selective, water soluble, brain-permeable EP2 receptor antagonist for use in central nervous system disease models

Abstract

Activation of prostanoid EP2 receptor exacerbates neuroinflammatory and neurodegenerative pathology in central nervous system diseases such as epilepsy, Alzheimer’s disease and cerebral aneurysms. A selective and brain-permeable EP2 antagonist will be useful to attenuate the inflammatory consequences of EP2 activation and to reduce the severity of these chronic diseases. We recently developed a brain-permeable EP2 antagonist 1 (TG6-10-1), which displayed anti-inflammatory and neuroprotective actions in rodent models of status epilepticus. However, this compound exhibited moderate selectivity to EP2, a short plasma half-life in rodents (1.7 h) and low aqueous solubility (27 μM), limiting its use in animal models of chronic disease. With lead optimization studies, we have developed several novel EP2 antagonists with improved water solubility, brain-penetration, high EP2 potency and selectivity. These novel inhibitors suppress inflammatory gene expression induced by EP2 receptor activation in a microglial cell line, reinforcing the use of EP2 antagonists as anti-inflammatory agents.

Keywords: Neuroinflammation, PGE2-receptors, Schild KB, competitive antagonist, lead optimization, SAR, blood-brain barrier, aqueous solubility

INTRODUCTION

Neuroinflammation has emerged as a key feature that exacerbates chronic neurodegenerative pathology in several central nervous system diseases including epilepsy, Alzheimer’s disease (AD) and Parkinson’s disease.14 Neuroinflammation is also an early event that plays a deleterious role in status epilepticus and traumatic brain injury.5, 6 Therefore, anti-inflammatory agents should offer a benefit to patients with these central nervous system (CNS) diseases. Production of inflammatory cytokines (interleukins) and chemokines, activation of glia, and induction of cyclooxygenase-2 are the early features of neuroinflammation.7, 8 Thus, there is an excellent opportunity for therapeutic discovery targeting the mediators of neuroinflammatory signaling.

A significant body of evidence suggests that neuroinflammation precedes the appearance of amyloid-β plaques and neurofibrillary tangles, which themselves precede clinical signs of AD in animal models and patients.9 Among the neuroinflammatory inducers, COX-2 has been very well studied thus far in AD patients by using small molecule inhibitors of COX-2.1012 Although retrospective epidemiological studies using non-selective COX-2 inhibitors suggested that chronic use of COX-2 inhibitors might prevent or delay the onset of Alzheimer’s disease in patients, prospective studies concluded that selective COX-2 drugs do not provide a clear benefit for Alzheimer’s patients.13 Reconciliation of the epidemiologic studies with prospective clinical trial results might be found in the clinical trial design, in which the treatment of patients was initiated when the disease is already at advanced stages in terms of classical pathological markers β-amyloid plaques and neurofibrillary tangles, or insufficient duration of treatment. The prevailing hypothesis emerging from clinical trials of a COX-2 drug is that treatment of patients starting at the prodromal stage of disease could offer benefit, but not treatment starting at late stages of the disease. Testing this proof of concept requires administration of a drug for several months (in animals) to years (in human) before clinical dementia symptoms are diagnosed. Regrettably, the COX-2 inhibitors cause adverse cardiovascular effects upon chronic use, by blunting PGI2 synthesis.14, 15 Two selective COX-2 inhibitors rofecoxib (Vioxx) and valdecoxib (Bextra) were withdrawn from the USA market, but a less selective COX-2 inhibitor, celecoxib (Celebrex), is in the clinic with black-box warning for thrombotic cardiotoxicity.16 Thus, future anti-inflammatory therapy within the COX-2 pathway should be targeted through a specific proinflammatory prostanoid synthase or a receptor downstream of COX-2, rather than blocking the entire COX-2 signaling pathways.17, 18

COX-2 catalyzes the synthesis of five prostanoids, PGD2, PGE2, PGF2, PGI2 and TXA2. These prostanoids activate nine receptors, DP1, DP2, EP1, EP2, EP3, EP4, FP, IP and TP respectively. Each of these receptors can play protective as well as deleterious roles in CNS and peripheral pathophysiologies.17, 18 The EP2 receptor has emerged as a predominantly proinflammatory receptor that mediates much of the COX-2 driven inflammatory consequences.19, 20 When activated by PGE2, EP2 receptors stimulate adenylate cyclase resulting in elevation of cytoplasmic cAMP concentration, which initiates downstream events via cell signaling pathways mediated by protein kinase A or exchange protein activated by cAMP (Epac).17, 18

Genetic ablation of the EP2 receptor reduces oxidative damage and amyloid-β burden in a mouse model of AD.21 EP2 deletion also attenuates neurotoxicity and reduces α-synuclein aggregation in a mouse model of PD.22 Moreover, EP2 deletion improves motor strength and survival of the ALS mouse.23 Furthermore, PGE2 signaling via EP2 receptor increases injury in models of cerebral ischemia,24 and mice lacking EP2 receptors have shown less cerebral oxidative damage produced by the activation of innate immunity.25 EP2 activation promotes inflammation and neurotoxicity in models of status epilepticus induced by pilocarpine and diisopropylfluorophosphate (DFP).19, 26, 27 In vitro, microglia cultures from mice lacking EP2 have shown enhanced amyloid-β phagocytosis and are less sensitive to amyloid-β induced neurotoxicity.28 Although these results strongly support the notion that pharmacological inhibition of EP2 should be explored in preclinical and clinical setting, the currently available EP2 antagonists have sub-optimal drug-like features such as low in vivo metabolic stability, in vivo plasma half-life and aqueous solubility for chronic dosing and proof-of-concept testing in preclinical AD models.

Initially Pfizer,29 our laboratory30, 31 and recently Amgen32 identified four distinct classes of EP2 antagonists (Figure 1A). Only the Amgen compounds seems to be useful for chronic dosing, but the derivatives developed in Pfizer and in our laboratory displayed too low liver metabolic stability, plasma half-life, or brain permeability to be useful in chronic CNS disease models such as Alzheimer’s disease. We initially identified a cinnamic amide compound TG4–155 (Figure 1A) from high-throughput screening as a potent EP2 antagonist.30 Structure activity relationship study of this compound resulted in the first generation brain-permeable lead compound 1 (TG6-10-1, Figure 1B), which displays brain-to-plasma ratio of 1.7, but only 10-fold selectivity for EP2 over DP1, low aqueous solubility (27 μM) and moderate plasma half-life (1.7 h).33 Thus, developing a selective, aqueous-soluble compound with enhanced plasma half-life while maintaining potency and brain permeability is the main goal of our project. In the present study, we report further lead-optimization studies that led to the synthesis of several second-generation analogs and show that improvements are made in terms of selectivity, aqueous solubility and in vivo plasma half-life. Moreover, we developed a compound 20o.HCl (named TG11–77.HCl), which is water-soluble (2.52 mM), yet crosses the blood-brain-barrier with brain-to-plasma ratio of 0.4. Furthermore, several novel compounds in this class, including 20o.HCl exhibited anti-inflammatory activity in EP2-expressing microglia-derived BV2 cell cultures in vitro. Therefore, compound 20o.HCl and other derivatives in this class are now suitable candidates for dosing into - disease models such as AD.

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0049.jpg

A. First generation EP2 antagonist hits reported in the literature. B) Lead optimization pathway identifies a brain-permeable EP2 antagonist candidate 20o.HCl (TG11–77.HCl) from previous lead EP2 antagonist 1 (TG6-10-1)

RESULTS AND DISCUSSION

Lead optimization studies to improve DMPK properties.

Previous SAR studies around compound 1 (TG6-10-1, Figure 1B) indicated that the acrylamide moiety is not essential for EP2 bioactivity. Thus, we developed second-generation analogs with an amide linker for SAR study. Compound 2a and 3 (Figure 1B) are examples of the earlier leads. These two compounds displayed high EP2 potency and selectivity and improved aqueous-solubility. However, compound 2a displayed poor DMPK properties, for example < 2 minutes half-life in mouse liver microsomes.34 Although compound 3 displayed excellent stability in mouse liver microsomes (> 60 min) and in vivo plasma half-life in mice (10.5 h), it showed very poor brain-to-plasma ratio of 0.02.35 Therefore, it was not useful for proof-of concept studies in CNS disease models. The overall goal was to develop a compound with requisite brain penetration and plasma half-life. In the past, we have made several derivatives replacing the right-side ring (morpholine in 2a, tetrazole in 3) and learned that this ring region can be modified without substantial loss of potency at EP2 receptor. We also made limited derivatives for example, polar imidazole ring replacing the indole on the left-side ring to increase the aqueous solubility.34 We have not made modifications on the middle phenyl ring for SAR study in the past. Therefore, we expanded our lead optimization studies with structural modification in these areas as detailed below.

Chemical synthesis

Synthesis of novel derivatives.

To explore the middle phenyl ring for structural modification and SAR studies, novel compounds 2b-c have been synthesized in a single step (Scheme 1) using commercially available precursors (1b and 1c) by following the synthetic method reported for 2a.34 However, several other precursors (e.g. 6a-g, 10a-c, 13a-c) needed for synthesis of novel derivatives were not commercially available, therefore we have synthesized here as shown in Scheme 2. Commercially available boronic acid derivatives (4a-g) have been coupled to 6-bromonicotinic acid (5) in the presence of tetrakis(triphenylphosphine)palladium (0) catalyst and a base to provide intermediates 6a-g, which were then coupled to 2-(2-methyl-1H-indol-3-yl)ethan-1-amine (7) in the presence of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide.hydrochloride (EDCI.HCl) and (dimethylamino)pyridine (DMAP) to provide final compounds 8a-g. Analogously, boronic acid derivatives 4a-c on palladium catalyzed coupling with 5-bromopicolinic acid (9) resulted in intermediate derivatives 10a-c, which were further coupled to 7 to provide compounds 11a-c. Moreover, the boronic acid 4c was coupled to 2-chloropyrimidine-5-carboxylic acid (12a) or 4-bromo-3-fluorobenzoic acid (12b) or 4-bromo-benzoic acid (12c) in the presence of palladium (II) catalyst and a base to result in intermediates 13a-c, which were similarly coupled with amine 7 to furnish final compounds 14a-c.

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0055.jpg
Synthesis of middle ring modified derivatives 2b and 2ca

aReagents and conditions: (a) 7 (see structure in Scheme 2), EDCI.HCl, DMAP, DMF: CH2CI2 (1:1), RT, 12 h. The product yields are shown in the parenthesis

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0056.jpg
Synthesis of middle ring modified EP2 antagonist derivativesa

aReagents and conditions: (a) Pd(PPh3)4, Na2CO3 (1M), THF (or) toluene, 100 °C, N2; (b) Pd(dppf)CI2, dioxane:H20 (6:1), Na2CO3, 100 °C; (c) EDCI.HCl, DMAP, DMF: CH2CI2 (1:1), rt. Yield for each reaction product is shown in the parenthesis. Yields are isolated yields but are not optimized.

Furthermore, we synthesized a number of derivatives with ‘NH’ as a linking moiety between the middle ring and the last ring of the molecule. As shown in Scheme 3, the amine 7 was coupled to 2-aminopyrimidine-5-carboxylic acid (15) in presence of coupling reagent EDCI.HCl to provide intermediate amine 16, which was further coupled to 2-bromopyridines 17a-o in the presence of tris(dibenzylideneacetone)dipalladium (0) and a base to provide final products 20a-o. Analogously, intermediate 16 was coupled to commercially available 4-bromopyridine (18) and 3-bromopyridines 19a-b to provide 20p and 20q-r respectively as final products in good yields.

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0057.jpg
Synthesis of middle and right side ring modified EP2 antagonist derivativesa

aReagents and conditions: (a) DMAP, EDCI.HCl, DMF, rt, 24 h. (b) 17 or 18 or 19, Pd2(dba)3, Cs2CO3, xantphos, dioxane, 100 °C, 12–18 h; Yield for each reaction product is shown in the parenthesis.

Maintaining the middle ring as pyrimidine, and the last ring as 4,6-dimethyl pyridine (see next section for SAR discussion), we further synthesized novel compounds with substitutions on the left side indole ring. As shown in Scheme 4, ethyl-2-aminopyrimidine-5-carboxylate (21) on reaction with 17o in presence of palladium (II) acetate gave intermediate 22, which on hydrolysis afforded acid precursor 23. Requisite amines 7a-e (SI Figure 1) were synthesized using the known methods,36, 37 and they were individually coupled with 23 in the presence of DMAP and EDCI.HCl at 50 °C to furnish the final products 24a-e (Scheme 4). Similarly, compound 24f was synthesized using acid 23 and 2-trifluroindole-3-ethanolamine (7f),38 which was intern prepared form 2-vinyl-pyrrolidin-2-one (SI Figure 1). Moreover, the compound 2-(2-methylpyrazolo[1,5-a]pyridin-3-yl)ethan-1-amine (7g) has been synthesized starting from amino-pyridinium iodide (SI Figure 1).39 Compound 7g, and the commercially available 2-amino-1-(2-methyl-1H-indol-3-yl)ethan-1-one (7h) and 2-amino-1-(2-methyl-1H-indol-3-yl)ethan-1-ol (7i) were coupled to the acid precursor 23 in the presence of EDCI.HCl coupling reagents to provide final products 24g-i, respectively in moderate yields. All the intermediates and final products were characterized by complementary methods of NMR and LCMS. The compounds having purity > 95% were subjected to testing in bioassays as described below.

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0058.jpg
Synthesis of left side indole and ethylene linker modified analogsa

aReagents and conditions: (a) Pd(OAc)2, BINAP, Cs2CO3, dioxane, 100 °C, 48 h, 40%; (b) LiOH, THF:H2O, (7:3), 12 h, 60 °C; (c) 7a-7i, EDCI.HCl, DMAP, DMF, 50 °C, 24–48 h; Yield for each reaction product Is shown in the parenthesis. Yields are isolated but not optimized.

SAR analysis of novel derivatives

Incorporation of nitrogen into the structure of small molecules often enhances aqueous solubility, intestinal permeability and other DMPK properties.40 Therefore, we envisioned hitherto unexplored middle phenyl ring (see ring assignments in Figure 1B) for structural modification with nitrogen heterocyclic rings for SAR studies. Recently, several groups have analyzed the physicochemical properties of CNS candidates and drugs to propose a multi-parameter optimization (MPO) approach that can be used to design novel chemical agents with CNS desirable properties.4143 Based on this MPO approach, Wager et al. proposed six physicochemical properties (MW, cLogP, cLogD, HBD, TPSA and pKa, Table 1) that cumulatively guide CNS activity and permeability of molecules.43 Our previous lead compounds 2a and 3 attained desirable MPO score (>4 out of 6 maximum), however, compound 3 was found to be a CNS impermeable compound with in vivo brain-to-plasma ratio 0.02, whereas compound 2a was too short lived in vivo to determine the brain permeability. We also determined that 3 is not a substrate of efflux pumps, but it displayed low cell permeability Paap (A-B) of 0.41 × 10−6 cm/sec in MDR1 expressed MDCK- cell monolayers with efflux ratio 0.3 (Table 7). Examination of its properties (Table 1) indicate that further optimization of the total polar surface area and hydrogen bond donor (HBD) properties might improve CNS permeability.44 Moreover, compound 3 has a tetrazole ring, which is known to act as a surrogate for carboxylic acid, which could limit a passive brain permeability.45 Therefore, we used compound 2a as a starting point for the current lead optimization studies.

Table 1.

CNS desirable properties suggested by MPO approach and calculated biophysical properties of selected EP2 antagonists including earlier leads 2a, 3and current lead 20o.HCla

PropertyProperty range for CNS drugs and candidates2a38c8g14a20o.HCl
MW/FW305–360363346385415386436
cLogP2.3–3.33.252.644.023.863.493.58
cLogD (pH 7.4)1.7–2.23.251.044.023.863.493.57
pKa8.40.06−0.652.282.830.594.5
HBD1232223
PSA (Å2)44.8–51.257.3696.467.076.279.995
MPO scoredesired MPO score ≥ 44.75.03.83.74.33.6
aChemAxon software was used to calculate the values for compounds 2a, 3and 20olisted in Table 1

Table 7.

BBB-permeability, mouse and human liver microsomal stability, key pharmacokinetic properties for selected compoundsa

EntryMLMt 1/2(min)HLM t 1/2(min)In vivo plasma t 1/2 (h)aBrain- to- plasma
ratiob
Permeability across MDR1-MDCK cell linecEfflux ratio (B-A / A-B)MPO scored (Desired score for CNS permeability ≥ 4)
110111.7 (ip/po, 10 mg/kg dose)1.7B-A: 27.1 × 10−6 cm/s
A-B: 25.4 × 10−6 cm/s
1.14.7
3>60>60> 6h (po, 10mg/kg)0.02B-A: 0.12 × 10−6 cm/s
A-B: 0.41 × 10−6 cm/s
0.35.0
8c2.9ND<1h (ip, 10mg/kg)0.5B-A: 51.0 × 10−6 cm/s
A-B: 19.8 × 10−6 cm/s
2.63.8
8g2.9ND< 1h (ip, 10mg/kg)NDB-A: 55.3 × 10−6 cm/s
A-B: 20.6 × 10−6 cm/s
2.73.7
14a6.5ND< 1h (ip, 10mg/kg)0.16B-A: 71.1 × 10−6 cm/s
A-B: 4.54 × 10−6 cm/s
164.3
20o1787.71.1 h (IP, 10 mg/kg) & 2.4 h (po, 50 mg/kg)0.4B-A: 21.5 × 10−6 cm/s
A-B: 10.2 × 10−6 cm/s
2.13.6
In the presence of a pgp-inhibitor (verapamil)
20oB-A: 22.2 × 10−6 cm/s
A-B: 13.2 × 10−6 cm/s
1.7
aMLM = mouse liver microsomes; HLM = human liver microsomes. In liver microsomal stability tests, 1 μM compound test compound was incubated with 0.5 mg/mL liver microsomes for compounds 1and 3. However, other compounds in the table were incubated with 0.125 mg/mL liver microsomal concentration. The plasma half-life is estimated based on the 3-time point B/P ratio studies conducted with single injection of 10 mg/kg dose to male mice. A full-scale pharmacokinetic study with 8-time points was done for selected compounds (1, 3, 20o).
bBrain-to-plasma are derived from peak concentrations observed at 0.5 h after injection in to mice.
cBBB potential was determined using MDR1-expressed cell monolayers. All these studies are conducted at CRO laboratories following industry standard procedures.
dSee Table 1 for the physicochemical properties used to calculate the MPO score.

We previously reported compound 2a with good EP2 potency and selectivity which showed moderate aqueous solubility.34 It has a morpholine ring on the right, phenyl ring in the middle, and 3-indole moiety on the left side (Figure 1B). To learn whether the middle phenyl ring can be replaced with other rings, we synthesized and tested compound 2b with a nicotinic acid ring. Compound 2b displayed 4-fold less EP2 potency than 2a, but it showed 3-fold increased aqueous solubility reinforcing the notion that nitrogen in the ring will enhance the aqueous solubility.40 Keeping the nitrogen (nicotinic) in the middle ring, we substituted right side morpholine ring with piperidine ring (2c). This resulted in 6.4-fold loss of potency in comparison to 2a. However, replacement of morpholine with a phenyl ring on the right side resulted in compound 8a, which interestingly displayed nearly equal EP2 potency to 2a, but low aqueous solubility (Table 2). Incorporation of a methoxy group either at 3-position (8b), or methoxy or hydroxy at 2-position (8c-8d) on the phenyl ring improved the potency by 2-fold (~10 nM, Table 2) in comparison to 2a. Whereas, fluorine (8e) at 2-position reduced the potency by 2-fold in comparison to 2a, and 4-fold in comparison to 2-methoxy derivatives 8c. Interestingly, incorporation of 4-acetamido group (8f), or two methoxy groups at 3- and 5-positions (8g) further enhanced the potency with KB values 4.4 and 2.9 nM respectively. To see if changing the location of nitrogen atom in the middle ring from meta-to-ortho (see o-m positional assignments in Scheme 2), we synthesized derivatives 11a-c. Compound 11a was 4-fold less potent than equivalent derivative 8b. Similarly, 11b was 11-fold less potent than equivalent derivative 8g, and compound 11c was 3-fold less potent than equivalent compound 8f. These observations suggest that nitrogen atom is preferred at m-position than at o-position of the middle ring (meaning nicotinic ring is more favorable than picolinic ring) for optimal potency. Furthermore, we synthesized a pyrimidine derivative 14a and it showed 2.9-fold less potency than the corresponding nicotinic ring compound 8c, but it has the same potency as initial lead 2a in the current study. Nonetheless, nicotinic middle ring maintains high EP2 Schild potency (< 50 nM), except for 2b-c. It is important to emphasize here that derivatives 8a-g and 14a displayed enhanced solubility in simulated gastric fluid at pH 2.0, whereas compounds 11a-c have not showed solubility enhancement in the simulated gastric fluid (Table 2), suggesting nicotinic ring derivatives will have better aqueous solubility than picolinic ring derivatives. It is worth to indicate that the nitrogen in the middle ring is not absolutely essential for EP2 potency, because compound with phenyl ring, 14c has also showed high EP2 potency (cf. 8c), but a fluorinesubstitution on the middle phenyl ring drastically reduced the potency by 25-fold (cf. 14b vs. 14c or 8c) hinting that additional substitutions on the middle ring might not be tolerated for potency.

Table 2.

Middle ring modified EP2 antagonists. Potency and aqueous solubilitya

aThe potency of the compounds is presented in the form of Schild KB values, which are calculated using the formula log (dr-1) = log XB −log KB, where dr (dose ratio) = fold shift in EC50 of PGE2 by the antagonist compound, XB is antagonist concentration 1 μM. KB value indicates a concentration required to produce a 2-fold rightward shift of PGE2 EC50. KB values are average of 2 measurements run in duplicate.
bThe solubility of the compounds is measured in PBS buffer (pH 7.4) with 1% DMSO by nephelometry.46
cThe numbers are derived by measuring solubility in simulated gastric fluid (SGF) at pH 2.0 with 1% DMSO by nephelometry.

Although the requisite EP2 potency is obtained for several analogs shown in Table 2, only compound 8c has displayed good selectivity (>100-fold) against other receptors (see Table 6 and next section for selectivity discussion). Moreover, when representative compounds (8c, 8d, 8g, 14a) from the Table 2 were tested for stability in mouse liver microsomes, they displayed very short half-life (Table 5). Therefore, we envisioned synthesizing additional novel derivatives by keeping the middle ring as pyrimidine and modifying the third ring (Scheme 3). The other key difference between these novel set of derivatives shown in Table 3 in comparison to the ones shown in Table 2 is the NH functional group, which links the third ring and the middle pyrimidine ring. We envisioned that the NH group should enhance the molecular flexibility and potentially allow us to make anionic salts. In the process, we first synthesized and tested 2-pyridyl derivative 20a. Interestingly, this compound displayed high EP2 potency with KB = 10.7 nM. Addition of methyl group (20b) or a fluorine (20c) at 4-position maintained high potency (KB = < 10 nM). Moving the methyl group from 4- to 6- position also maintained a high potency (cf. 20e vs. 20b), but similarly moving the fluorine atom to 6- position recued the potency by 16-fold (cf. 20f vs. 20c). A methoxy group at 6- position also maintained high EP2 potency (20h), whereas cyano-group reduced the potency by 23-fold (20g). The acetyl group on pyridine ring whether it is at 6-,5- or 4- position (20j, 20l and 20k) reduced the potency by 4-, 20-and 12-fold respectively in comparison to 20b. A compound with bulkier group 3-hydroxybutane at 6- position showed 4-fold less potency than 20b. Similarly, tert-butyl group at 4- position reduced the potency by 5-fold (cf. 20d vs. 20b), or 6-position reduced by 13-fold (cf. 20n vs. 20e). A hydroxy group (20i) at 6-position eliminated the potency of the molecule (KB > 1000 nM) (Table 3). Incorporation of two methyl groups as in 20o did not have additive impact on the potency (cf. 20o vs. 20b or 20e). We also tested the 4-pyridyl derivative 20p, which showed complete loss of potency, whereas the 3-pyridyl derivative 20q regained the potency and shown nearly same potency as 20a. Addition of two methyl groups on 3-pyridyl ring (20r) similar to 20o indicated 10-fold loss of potency, thus we have not subjected 3-pyridyl ring for further modification.

Table 3.

Middle ring and right side ring modified EP2 antagonists. Potency and aqueous solubilitya

aSchild KB values are calculated similarly as indicated at Table 1 legend. KB values are average of 2 measurements run in duplicate. ND = not determined
bThe solubility of the compounds is measured in PBS buffer (pH 7.4) with 1% DMSO by nephelometry.46

Table 5.

Mouse liver microsomal (MLM) stability of selected novel EP2 antagonistsa

Compd.MLM T1/2 (minutes)
3a<2
8c2.9
8d13
8g2.9
14a6.5
20b8
20c17
20e12
20f96
20h16
20o17
24a17
24c26
24e15
24i56
aThe compounds at 1μM were incubated in 0.125 mg/mL liver microsomes that are activated with addition of NADPH (final concentration 1 mM) and quenched the reaction at 0, 5, 15, 30, 60 and 120 min using ice-cold acetonitrile. The % compound remaining was measured using LC-MS/MS from which half-life was calculated. The work was done at CRO laboratories.

Table 6.

EP2 Potency, selectivity index (S.I), and cytotoxicity (CC50) of selected novel EP2 antagonistsa

EntryEP2 KB (nM)DP1 KB (μM)S.I. (DP1/EP2)EP4 KB (μM)S.I (EP4/EP2)IP KB (μM)S.I (IP/EP2)CC50 (μM)
8a23.21.042NDNDNDNDND
8b9.50.995NDNDNDNDND
8c103.030022.822803.4735021
8e48.40.816NDNDNDNDND
8g2.91.24103.9113500.7325>50
11a48.61.735NDNDNDNDND
11b33.60.515NDNDNDNDND
14a29.66.630015.351721.5730>50
20a10.7>10>9006.7630>10>900>50
20c6.43.2500>10>1500>10>1500>50
20e6.12.25360>10>1500>10>1500>50
20h7.8>10>12806.7860>10>1280>50
20o9.77.327505.3550>10>1000>50
20q8.31.77213>10>1200>10>1200>50
24a13.09.06923.1240>10770>50
24b28.38.2290>10>350>10>350>50
24c20.08.24103.8193>10>500ND
aEP2 KB values are from average of 2 independent experiments run duplicate, except for 20o. EP4, DP1 and IP KB values are from 2 independent experiments run in duplicate. KB values for EP2 are in the nanomolar scale, whereas KB values for other receptors are shown in micromolar scale. CC50 (concentration required to kill 50% C6glioma cells) values are from 1–2 experiments run in triplicate using internal standard doxorubucin, which showed CC50 = 0.9 μM.

We then synthesized a new set of derivatives (Scheme 4) by modifying left side indole moiety and the two carbon linker, keeping the middle ring as pyrimidine and the right side ring as 4,6-dimethyl pyridyl ring as constant. As shown in Table 4, a fluoro, chloro, methoxy, difluoro or dichloro derivatives 24a-e all retained high EP2 potency, whereas changing indole ring to pyrazolo-pyridine ring as in 24g reduced the potency by 32-fold in comparison to 20o. Moreover, substituting the methyl group at second position of indole with a trifluoromethyl group (24f) also reduced the potency by 8-fold. Modification to the linker with ketone group next to indole (24h) eliminated the potency, where as a hydroxy group as in 24i recued the potency 23-fold in comparison to 20o suggesting unsubstituted ethylene linker is optimal for high EP2 potency within the scaffold. Overall, the SAR study on the leads indicated that a number of compounds exhibit high EP2 potency.

Table 4.

Modifications at indole ring and linker regions of the scaffold. Potency and aqueous solubilitya

Entry
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0038.jpg
EP2 KB (nM)Aqueous Solubilityb (μM)Aqueous solubility of corresponding HCl salt in SGFc (μM)Water solubility of corresponding HCl saltd (mM)
20o
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0039.jpg
9.7150>2502.52
24a
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0040.jpg
13.05ND1.16
24b
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0041.jpg
28.320NDND
24c
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0042.jpg
20ND>1001.73
24d
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0043.jpg
25.1NDND2.38
24e
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0044.jpg
8.820NDND
24f
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0045.jpg
6645ND0.7
24g
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0046.jpg
25470NDND
24h
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0047.jpg
>1000NDNDND
24i
An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-t0048.jpg
18357NDND
aSchild KB values are calculated similarly as indicated at Table 1 legend. KB values are average of 2 measurements run in duplicate, except compound 20ofor which the values are average of 6 repeats
bThe solubility of the compounds is measured in PBS buffer (pH 7.4) with 1% DMSO by nephelometry.46
cThe solubility of compounds measured in simulated gastric fluid at pH 2.0 by nephelometry. Compound 24dis a milky-white solution in DMSO and 24 h is an inactive compound. Therefore, their solubility is not determined by nephelometry, however, 24d.HClis highly water soluble.
dWater solubility is determined by 24 h shake flask thermodynamic solubility method in neat water. Briefly, the compounds were dissolved in water at 2 mg/mL and shaken at 2200 rpm for 24 h. Then, the solutions were filtered through 0.2 μ syringe filters and analyzed the area under the curve (AUC) using HPLC, along with standard DMSO solution of each compound. ND = not determined.

Determination of aqueous solubility of EP2 compounds

Aqueous solubility is an important property that plays crucial role in success of a drug candidate.47 We tested several compounds for aqueous solubility by using two different methods. First, we used a kinetic assay in which we dissolved the compounds in DMSO then made serial dilutions with phosphate buffered solution (PBS) at pH 7.4, maintaining 1% DMSO in the solutions. Solubility was determined by nephelometry.46 This assay works on the principle that when visible light is passed through a solution, part of the incident radiant energy will be scattered. The intensity of the scattered light is a function of the concentration of the dispersed phase. The nephelometric assay is used to determine either the point at which a solute begins to precipitate out of a true solution to form a suspension or the concentration at which a suspension becomes a solution when diluted further. The scattered light will remain at a constant intensity until precipitation occurs, at which point it will increase sharply. Several compounds shown in Table 2 and and33 displayed moderate solubility (< 100 μM) in PBS at pH 7.4. However, interestingly, several of those compounds including 8a-c, 8e-g, 14a, 20o, 24c have shown >100 μM aqueous solubility when determined in simulated gastric fluid (pH 2.0), suggesting that the intrinsic basic nitrogen can be explored to create salts that further increase solubility and facilitate the formulation. On this notion, compounds 20o, 24a, c, d and 24f have been converted to hydrochloride salts by stirring them in dichloromethane using dilute hydrochloride solution.

Second, to ask whether the hydrochloride salt compounds (20o, 24a, c d and 24f) are soluble in water, we used a thermodynamic assay (shake flask method)48 where the compounds are shaken (2 mg/mL, at 2200 rpm) in neat water and the soluble fraction was measured against standard DMSO solutions by HPLC or LC-MS/MS quantitation methods. As shown in Table 4, the hydrochloride salt compounds displayed millimolar level aqueous solubility in water. It is worth mentioning that the salt forms did not show increased solubility in PBS buffer at pH 7.4 by nephelometry method in comparison to their neutral compounds. This is likely due to a drop in pH in neat water, in comparison to a buffer. Indeed, the water pH was reduced from 6.4 to 3.5 when 20o.HCl was dissolved at 2 mg/mL. This observation is in line with our findings that the neutral compounds have higher solubility in simulated gastric fluid (pH 2.0) than in PBS at pH 7.4. Overall, the hydrochloride salts of compounds 20o, 24a, 24c and 24d showed 2.52, 1.16, 1.73 and 2.38 mM solubility in neat water (Table 4).

Selectivity assessment of novel EP2 antagonists

The structural identity among the prostanoid receptor family is rather low. EP1, EP2, EP3 and EP4 share only 20–30% structural homology.49 In contrast, EP2 is more homologous to DP1 (44%) and IP receptors (40%).49 In terms of cellular signaling, EP2, EP4, DP1 and IP induce cAMP mediated cell signaling, whereas EP1 promotes Ca2+ mediated signaling and EP3 inhibits cAMP mediated signaling. Functionally, EP2 and DP1 receptors seem to promote inflammation in a variety of disease conditions, whereas EP4 seems to act as pro and anti-inflammatory, and the IP receptor seems to have cardioprotective role.15 Thus, we determined the selectivity of several potent EP2 antagonists (KB < 50 nM for EP2) against DP1 first, and if any compound showed >100-fold selectivity to DP1, then it was tested against EP4 and IP receptors. As shown in Table 6, compounds 8a-b, 8e, 11a-b showed < 100-fold selectivity against the DP1 receptor, thus they were not tested against EP4 and IP receptors. The compounds 8c, 14a, 20a, 20c, 20e, 20h, 20o, and 24a-c displayed excellent selectivity against DP1, and subsequently against EP4 and IP receptors. However, compounds 8g and 24c displayed < 200-fold selectivity against at least one of the three (DP1, EP4, and IP) receptors suggesting selectivity will depend on the individual structure of the molecule within the scaffold. It is worth to note that we tested several compounds in the class for cytotoxicity in the C6-glioma cell line. As exemplified in Table 6, selected compounds including 20o displayed no cytotoxicity until 50 μM indicating > 2100-fold in vitro therapeutic index (CC50/EP2 KB), except compound 8c which showed CC50 21 μM with a therapeutic index of 2100-fold.

DMPK properties of novel EP2 antagonists

We tested several derivatives in pooled liver microsomal fractions to determine the half-life and intrinsic clearance by the mouse liver, and to project in vivo pharmacokinetics. The starting compound for the current study, 2a was metabolized quickly in liver microsomes (Table 5). To understand the potential metabolites, we incubated compound 2a in mouse liver microsomes and investigated metabolites after 5 minutes. As shown in Figure 2, the major metabolite is the amide bond cleaved product D (MW 190 Da), which can be formed via intermediate A that would be further cleaved to generate fragments B/C (MW 158 Da) and D. The compound D can also be generated via a vinylamine F (MW 362 Da) intermediate, formation of which can be envisioned via hydroxylation at the CH2 unit (E) next to 3-indole ring. Therefore, we synthesized compounds blocking the CH2 site with a hydroxy group (e.g. 24i). The half-life of 24i in liver microsomes increased by 3-fold (t1/2, 57 min) compared to its equivalent 20o, which showed 17 minutes of half-life (Table 5). However, the hydroxylated derivatives displayed reduced EP2 potency compared with the equivalent compounds. (cf. 24i displayed about 19-fold less potency than 20o, Table 4). In a similar experiment, the tetrazole compound 3 did not produce any fragments even after 20 minutes of incubation in liver microsomes, suggesting the metabolic hydroxylation or cleavage also depends on the right side moiety (Figure 2). The mouse liver microsomal half-life data presented in Table 5 also indicate that a fluorine atom in place of metabolically prone methyl group enhances the stability. For example, a fluoro- derivative 20c has 2-fold higher half-life in comparison to 20b, like wise 20f has 8-fold higher half-life than 20e. Regrettably, 20c is unstable in vivo, and 20f displayed less potency to EP2 receptor (Table 3), therefore, these compounds are not promoted for further studies.

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0050.jpg

Metabolites identified from the lead EP2 antagonists in mouse liver microsomes.Compound 2a was incubated for 5 min and metabolic fragments (A-F) were detected using LC-MS/MS. Unlike compound 2a, compound 3 has not shown any metabolic fragments even after 20 min of incubation in mouse liver microsomes.

We estimated the CNS drug-like properties for several of these compounds before synthesis using the CNS-MPO tool.43, 50 MPO scores indicated that several EP2 antagonists in the class as exemplified in Table 7, display a desired MPO score of ≥ 4, but several others do not achieve the desired score > 4 derived using their physicochemical properties. Compounds ≥ 4 MPO score should have desirable CNS activity and permeability features. However, the compounds 3 while it displayed > 5 score does not have good permeability properties and does not get in to the brain (Table 7). A corollary to this, the current lead compound 20o.HCl showed < 4 MPO score, yet crosses the blood-brain-barrier with in vivo brain-to-plasma ratio 0.4. Likewise, compounds 8c, 8g and 14a behaved similarly. Overall, we have seen a positive correlation between MPO score to in vivo brain to plasma ratio. Nonetheless, the MPO score does not quantitatively predict the brain permeability. We tested several compounds for permeability across MDR1 expressed MDCK cell line to investigate the potential of blood brain barrier (BBB) permeability within the class. As shown in Table 7, we found many of these compounds have good passive permeability from the apical-to-basolateral (A-B) side as well as the basolateral-to-apical side (> 0.6 ×10−6 cm/s) in comparison to compound 3. and their efflux ratio is < 3, except for compound 14a which displayed ER ratio 16 indicating it may be a substrate for efflux pumps (Table 7). On the other hand, the compound 20o found to show good permeability and showed similar efflux ratio in the presence and absence of an efflux pump inhibitor verapamil, confirming that it is not the substrate of efflux pumps. (Table 7). Moreover, compound 20o showed 0.4 % plasma protein unbound fraction in mouse plasma proteins, but we were not able to determine its fraction unbound in the relevant brain compartment (i.e., interstitial fluid) due lack of efficient method to extract cerebrospinal fluid (CSF) from mouse brain.

To determine in vivo brain-to-plasma ratio in mice, we tested several compounds whose mouse liver microsomal stability was greater than 15 min, by administering a dose of 10 mg/kg via intraperitoneal injection. Analysis of the compound concentration in the plasma and the brain tissues at 0.5, 2, and 4 h time points suggested that most of these compounds, exemplified as in Table 7 for 8c, 8g & 14a, will have a plasma half-life below 1 h, except compound 20o. A subsequent pharmacokinetic analysis on 20o.HCl with concentration analysis at 8-time points after a single i.p. injection (10 mg/kg) (Figure 3) indicated that it has terminal plasma half-life 1.1 h with clearance rate of 124.1 mL/min/kg and brainto-plasma ratio of 0.4. Additional studies with oral gavage dosing (50 mg/kg, B.I.D. dosing 8h apart) indicated that the plasma half-life could be extended to 2.4 h for 20o.HCl.

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0051.jpg

Pharmacokinetic parameters of 20o.HCl Compound 20o.HCl was administered in to male C57BL/6 mice via intraperitoneal injection at single dose of 10 mg/kg in the vehicle of 5% NMP, 5% Solutol-HS15 and 90% water. Concentrations in plasma and brain are determined by LC-MS/MS and plotted against time.

Competitive mode of EP2 antagonism by 20o.HCl and derivatives.

To determine whether the novel derivatives in this class exhibit competitive antagonism of EP2, we tested several compounds in a concentration-response manner against PGE2 concentration effect on EP2 receptors. The Schild KB indicates the antagonist concentration required for a two-fold rightward shift in the PGE2 concentration-response curve. Schild KB values are derived by the equation log (dr − 1) = log XB – log KB, where dr = dose ratio, i.e. the fold shift in agonist EC50 caused by the antagonist, XB is antagonist concentration. As illustrated in Figure 4B, a linear regression of log (dr-1) on log XB with slope of unity characterizes a competitive antagonism. A smaller KB value indicates a higher inhibitory potency. As shown in Figure 4A and andC,C, compound 20o (TG11–77 neutral and the hydrochloride salt form) induced a concentration-dependent, parallel rightward shift in the PGE2 concentration-response curve. Schild regression analyses (Figure 4B) is consistent with a competitive mechanism of antagonism on EP2 with average Schild KB 9.7 nM and average slope value 1.05. The average KB value is used throughout in this manuscript for discussion and comparisons. Moreover, several other compounds synthesized in this class displayed a concentration-dependent rightward shift of PGE2 EC50 and with slope of unity (SI Figure 2). Thus, the mechanism is competitive in general for the class of EP2 antagonists presented in this study. However, compound 20o did not show a concentration-dependent inhibition of DP1 receptor, indicating it is selective for EP2 over DP1 with selective index (DP1 KB/EP2 KB) of 750-fold. Moreover, this compound also showed high selectivity against EP4 and IP receptors with selective index of 550-fold and >1000-fold respectively (Table 6).

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0052.jpg

Competitive antagonism of EP2 receptor by 20o. A: Compound 20o (TG11–77 neutral) inhibited PGE2-induced human EP2 receptor activation in a concentration dependent manner. B. Schild regression analysis is performed to determine the modality of antagonism by this compound. C. Hydrochloride salt of 20o (20o.HCl) similarly inhibited PGE2-induced human EP2 receptor activation in a concentration-dependent manner. Schild KB values for neutral compound and the hydrochloride salt along with their slope values are shown in inset of Figure 4B. D. Concentration-response test of 20o.HCl on DP1 receptors indicates it does not significantly inhibit DP1 receptor activation by agonist BW245C. Data were normalized as percentage of maximum response; points represent mean ± SEM (n = 3).

EP2 antagonists display anti-inflammatory properties in a novel microglia cell line expressing human EP2.

The EP2 receptor acts as an immunomodulator with exacerbating role in chronic neurodegenerative disease such as epilepsy and Alzheimer’s disease. EP2 receptors also play an exacerbating role in chronic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease (colitis) and endometriosis.5153 To determine whether the new EP2 antagonists are anti-inflammatory, we tested several of these novel EP2 antagonists including 20o (TG11–77) for anti-inflammatory activity in vitro. A routine isolation of microglia from mouse brain proved to be low throughput and these primary cells behave variably depending on the animal. Thus, we created a hEP2-BV2 cell line, a mouse microglia cell line overexpressing human EP2 receptors.54. Upon activation of this cell line with 100 ng/mL lipopolysaccharide (LPS), mRNA levels of several proinflammatory genes including COX-2, IL-6 and IL-1β were induced. An EP2 specific agonist, ONO-AE1-259-01, at 30 nM further exacerbated the induction of these inflammatory genes. Gratifyingly, 20o blunted the upregulation of inflammatory genes COX-2, IL-1β, and IL-6 in a concentration dependent manner (Figure 5). We found that EP2 activation in hEP2-BV2 cell line decreased mRNA levels of TNF-α consistent with the result we found with primary microglia.55 Interestingly, these downregulated TNF-α levels are also reversed by the EP2 antagonist, reassuring that this compound is working by interacting with the EP2 receptor. In this experiment, neither the EP2 agonist (ONO-AE1-259-01), nor the antagonist (20o) alone showed significant effects on the expression of mRNA levels of EP2 and iNOS (an oxidative stress causing enzyme) in this cell line. Overall, these data support the use of this EP2 antagonist as an anti-inflammatory agent in inflammatory- disease models.

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0053.jpg

Mean fold change in mRNA expression of cytokines in BV2-hEP2 cells upon treatment with 20o (TG11–77), ONO-AE1-259-01 (abbreviated as Ono in the diagram) and LPS treatment. 200,000 cells/well were grown overnight and treated with vehicle or 20o for 1 h, vehicle or Ono for 1 h and vehicle or LPS for 2 hrs. Analyte mRNAs were measured by qRT-PCR (see SI Table 1 for primers used for this experiment). For statistical analysis, ΔΔCT values were used as they were normally distributed whereas fold changes were not. ANOVA-with Holm-Sidak multiple comparisons test for post-hoc analysis was used. P values were considered significant at *<0.05. Experimental repeats n = 4.

To determine whether there is a correlation between the potency of EP2 antagonists in the cAMPproduction assay and their ability to inhibit inflammatory gene expression in the BV2-hEP2 cell line, we tested four selected EP2 antagonists (3, 20o, 26 (TG4–155)56 and 27 (TG8–237)35) with KB values ranging between 2 and 50 nM. The fold concentrations are calculated and presented in SI Table 2. As shown in Figure 6, we plotted the percent inhibition of inflammatory genes against log of multiple Schild KB values. The results show that the inhibition of cytokines was positively correlated to their respective Schild KB, suggesting the higher the potency of antagonists in the cAMP assay, the lower the concentration required to display maximum effect on the inflammatory genes (Figure 6).

An external file that holds a picture, illustration, etc.
Object name is nihms-1607948-f0054.jpg

Concentration-related inhibition of inflammatory mediator expression by several EP2 antagonists. BV2-hEP2 microglia were incubated with combinations of the EP2 antagonists (0.3 or 1 μM), the EP2 agonist Ono (30 nM) and LPS (100 ng/ml) for 2 hours and the mRNA levels of 6 inflammatory mediators were measured. The concentrations of each antagonist on the x-axis is expressed as fold Schild KB measured in the cAMP assay performed in C6 glioma cells overexpressing human EP2. The y-axis shows the % inhibition of the induction produced by 30 nM Ono, which is 10-fold higher than its EC50 in the cAMP assay. The data were fitted to a logistic equation. Data points are mean and SEM from 4–7 independent cultures with technical duplicates in BV2-hEP2 cells and 34 experimental repeats for cAMP assay in C6 glioma cells. Color coding: Orange represents data obtained with 0.3 μM of the antagonist and green represents data obtained with 1 μM.

CONCLUSIONS

In conclusion, we synthesized 45 novel EP2 antagonists with good potency and selectivity. With the lead optimization following the DMPK properties on these antagonists, a novel compound 20o.HCl (named as TG11–77.HCl) was identified with acceptable brain permeability and excellent water solubility. Several compounds in this class displayed selective, competitive antagonism of EP2 receptors and increased solubility in simulated gastric fluid at pH 2.0 compared to PBS at pH 7.4. The hydrochloride salts of several compounds showed good water solubility at lower pH. Functionally, members of this class reversed inflammatory gene modulation by EP2 receptor activation, and the potency was shown to positively correlate to the anti-inflammatory actions measured following gene expression in a novel microglia cell line. Taken together, we conclude that 20o.HCl should be very useful for dosing into rodent models of CNS diseases.

EXPERIMENTAL SECTION

General experimental procedures:

Proton NMR spectra were recorded in solvent in DMSO-d6/CDCl3 on Varian and Inova-400 (400 MHz). Thin layer chromatography was performed on pre-coated, aluminum-backed plates (silica gel 60 F254, 0.25 mm thickness) from EM Science and was visualized by UV lamp, PMA solution and ninhydrin. Chemicals and drugs: PGE2, BW245C, iloprost, and rolipram were purchased from Cayman Chemical. LPS was purchased form Sigma-Aldrich. ONO-AE1-259-01 was generously provided by ONO Pharmaceuticals (Osaka, Japan). Column chromatography was performed with silica gel cartridges on Teledyne-ISCO instrument. Agilent LC-MS was used to determine the mass and purity of the products. LC-MS conditions: Mobile phase A: methanol (0.1% acetic acid); mobile phase B: water (0.1% acetic acid); column: ZORBAX Eclipse XDB C18 5μM, 4.6 × 150 mm. Gradient B 80% at 0 min, linearly decreased to 5% by 7 min, and then linear increase to 40% by 12 min; UV wavelength = 254 nm; flow rate = 1 mL/min. Furthermore, purity of several key compounds is determined by Water’s HPLC instrument. HPLC Conditions: Mobile phase A: water (0.1% trifluoroacetic acid); mobile phase B: acetonitrile (0.1% trifluoroacetic acid); column: XBridge C18 5μM, 4.6 × 150 mm; gradient: 10% B at 0 min, increased linearly to 90% by 10 min, then decreased to 10% by 12 min; UV wavelength = 230 nm; flow rate = 1 mL/min. Compounds with >95% purity by HPLC were tested in cellular bioassays and DMPK properties. Compounds 7a57, 7b58, 7e59 were reported in the literature and the characterization data for these derivatives was in good agreement with the literature data. The compound 2-(2-(trifluoromethyl)-1H-indol-3-yl)ethan-1-amine (7f) was synthesized as reported before60, 61 and 2-(2-methylpyrazolo[1,5-a]pyridin-3-yl)ethan-1-amine (7g) was synthesized following the literature procedure.62 2-Amino-1-(2-methyl-1H-indol-3-yl)ethan-1-one (7h), and 2-amino-1-(2-methyl-1H-indol-3-yl)ethan-1-ol (7i) were commercially available.

Procedure for the synthesis of 2b and 2c:

To a solution of commercially available acid 1b or 1c (0.4 mmol, 1 equiv.) and 7 (70 mg, 0.4 mmol, 1 equiv.) in mixture of dichloromethane and N,N-dimethylformamide (3 mL, 5:1) was added DMAP (catalytic amount, 2 mg) followed by EDCI.HCl (114 mg, 0.59 mmol, 1.3 equiv.) and the reaction mixture was stirred at room temperature for 10 h. Organic solvent was evaporated and reaction mixture was added a saturated solution of ammonium chloride (5 mL) and extracted with ethyl acetate (3 × 10 mL). Organic layer was separated and washed with saturated solution of sodium bicarbonate (5 mL) followed by brine solution (5 mL), dried over sodium sulfate and concentrated to dryness. The crude material was purified on silica gel chromatography using 60–70% ethyl acetate in hexanes to get the required product 2b or 2c (Scheme 1).

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-6-morpholinonicotinamide (2b):

1H NMR (400 MHz, DMSO-d6): δ 10.69 (s, 1H), 8.59 (d, J = 2.0 Hz, 1H), 8.40 (t, J = 5.6 Hz, 1H), 7.95 (dd, J = 8.9, 2.4 Hz, 1H), 7.44 (d, J = 7.6 Hz, 1H), 7.20 (d, J = 7.7 Hz, 1H), 6.97 – 6.85 (m, 2H), 6.81 (d, J = 9.0 Hz, 1H), 3.70 – 3.63 (m, 4H), 3.58 – 3.47 (m, 4H), 3.37 (q, J = 7.1 Hz, 2H), 2.84 (t, J = 7.4 Hz, 2H), 2.27 (s, 3H); LCMS (ESI): >97% purity. MS m/z, 365 [M + H]+; HPLC purity: 99.7%.

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-6-(piperidin-1-yl)nicotinamide (2c):

1H NMR (400 MHz, CDCl3): δ 8.36 (d, J = 2.3 Hz, 1H), 8.08 (s, 1H), 7.74 (dd, J = 9.0, 2.5 Hz, 1H), 7.51 (d, J = 7.5 Hz, 1H), 7.28 – 7.22 (m, 1H), 7.11 – 7.08 (m, 2H), 6.54 (d, J = 9.0 Hz, 1H), 6.02 (t, J = 5.6 Hz, 1H), 3.67 (q, J = 6.4 Hz, 2H), 3.57 (d, J = 5.0 Hz, 4H), 2.99 (t, J = 6.6 Hz, 2H), 2.32 (s, 3H), 1.67− 1.56 (m, 6H); LCMS (ESI): >97% purity. MS m/z 363 [M + H]+; HPLC purity: 98.9%.

General procedure for the synthesis of 6a-g:

A solution of boronic acid (4a-g) (4.1 mmol, 1 equiv.) and bromo-acid, 5 (4.1 mmol, 1 equiv.) in tetrahydrofuran or toluene and water (6:1) were loaded in to a sealed tube. To this solution, 1M Na2CO3 (8.2 mmol, 2 equiv.) was added and purged with nitrogen for 10 min. Then, Pd(PPh3)4 (0.2 mmol, 0.05 equiv.) catalyst was added to the reaction mixture, sealed and heated to 100 °C for 12 h. Reaction mixture was cooled to room temperature and solvent was evaporated under vacuum. The residue was washed with dichloromethane to remove organic impurities. Then, aqueous layer was acidified to pH 2 with concentrated HCl to result in white precipitate, which was filtered and dried under vacuum to provide the intermediates (6a, 6b63, 6c, 6d64, 6e65, 6f and 6g).

6-Phenylnicotinic acid (6a):

1H NMR (400 MHz, DMSO-d6): δ 13.36 (s, 1H), 9.24 – 9.03 (m, 1H), 8.35 – 8.31 (m, 1H), 8.17 (dd, J = 8.2, 1.3 Hz, 2H), 8.14 – 8.09 (m, 1H), 7.54 – 7.52 (m, 3H). LCMS (ESI): >95% purity; MS m/z, 198 [M - H]+.

6-(2-Methoxyphenyl)nicotinic acid (6c):

1H NMR (300 MHz, DMSO-d6): δ 12.52 (s, 1H), 9.14 (d, J = 2.2 Hz, 1H), 8.46 (dd, J = 8.4, 2.2 Hz, 1H), 8.11 (d, J = 8.3 Hz, 1H), 7.79 (dd, J = 7.7, 1.7 Hz, 1H), 7.59 – 7.47 (m, 1H), 7.23 (d, J = 8.2 Hz, 1H), 7.12 (dd, J = 11.5, 4.2 Hz, 1H), 3.87 (s, 3H). LCMS (ESI): >95% purity; MS m/z, 228 [M - H]+.

6-(4-Acetamidophenyl)nicotinic acid (6f):

1H NMR (300 MHz DMSO-d6): δ 13.32 (s, 1H), 10.17 (s, 1H), 9.08 (s, 1H), 8.26 (d, J = 6.5 Hz, 1H), 8.11 (d, J = 8.6 Hz, 2H), 8.02 (d, J = 8.3 Hz, 1H), 7.72 (d, J = 8.6 Hz, 2H), 2.06 (s, 3H). LCMS (ESI): >97% purity; MS m/z, 255 [M - H]+.

6-(3,5-Dimethoxyphenyl)nicotinic acid (6g):

1H NMR (300 MHz, DMSO-d6): δ 13.31 (bs, 1H), 9.11 (d, J = 2.2 Hz, 1H), 8.29 (dd, J = 8.3, 2.2 Hz, 1H), 8.12 (d, J = 8.4 Hz, 1H), 7.30 (d, J = 2.3 Hz, 2H), 6.61 (t, J = 2.2 Hz, 1H), 3.81 (s, 6H). LCMS (ESI): >95% purity; MS m/z, 258 [M - H]+.

General procedure for the synthesis of substituted 3-indole-ethylamines 7a-g:

To a solution of 25a-e (2 mmol, 1 equiv.) in acetonitrile (20 mL) was added cyclopropyl methyl ketone (4 mmol, 2 equiv.) and refluxed for 24 h. Then, reaction mixtures were cooled to room temperature. Solids precipitated were filtered to obtain corresponding hydrochloride salts of 7a-e. To a suspension of these salts in dichloromethane was added 50% ammonium hydroxide solution (1.2 equiv.) and stirred for 3 h at room temperature. Organic layer was extracted, dried over sodium sulfate and concentrated to dryness to get the amines 7a-e (see SI Figure 1). Corresponding references were provided in the general experimental section for reported compounds and data for 7c and 7d are shown below.

2-(5,7-Difluoro-2-methyl-1H-indol-3-yl)ethan-1-aminiumchloride (7c-HCl):

1H NMR (400 MHz, DMSO-d6): δ 11.48 (s, 1H), 8.15 (bs, 3H), 7.21 – 7.15 (m, 1H), 6.88 – 6.79 (m, 1H), 2.98 – 2.82 (m, 4H), 2.35 (s, 3H); LCMS (ESI): >95% purity. MS m/z, 211 [(M – HCl) + H]+.

2-(5,7-Dichloro-2-methyl-1H-indol-3-yl)ethan-1-aminiumchloride (7d-HCl):

1H NMR (400 MHz, DMSO-d6): δ 11.46 (s, 1H), 8.13 (bs, 3H) 7.58 (s, 1H), 7.14 (s, 1H), 3.02− 2.81 (m, 4H), 2.37 (s, 3H); LCMS (ESI): >95% purity. MS m/z, 243 [(M – HCl) + H] +.

General procedure for the synthesis of 8a-g:

To a solution of 6a-g (0.5 mmol, 1 equiv.) and 2-(2-methyl-1H-indol-3-yl)ethan-1-amine (7) (0.5 mmol, 1 equiv.) in N,N-dimethylformamide and dichloromethane (1:1) was added DMAP (catalytic amount) followed by EDCI.HCl (0.65 mmol, 1.3 equiv.) and the reaction mixture was stirred at room temperature for 10 h. Then, dichloromethane was evaporated and the crude reaction mixture was added a saturated solution of ammonium chloride (15 mL) and extracted with ethyl acetate (15 mL). Organic layer was separated and washed with saturated solution of sodium bicarbonate (15 mL) followed by brine solution (15 mL). Combined organic layer was dried over sodium sulfate, concentrated to dryness. The crude was purified on silica gel chromatography using 50–70% ethyl acetate in hexanes to get the required products (8a-g).

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-6-phenylnicotinamide (8a):

1H NMR (400 MHz, CDCl3): δ 8.85 (dt, J = 2.4, 0.8 Hz, 1H), 8.07 – 8.03 (m, 1H), 8.01 – 7.96 (m, 2H), 7.90 (s, 1H), 7.76 – 7.71 (m, 1H), 7.55 (d, J = 7.6 Hz, 1H), 7.51 – 7.41 (m, 3H), 7.31 (dd, J = 4.5, 4.0 Hz, 1H), 7.18 – 7.07 (m, 2H), 6.22 (t, J = 6.4 Hz, 1H), 3.76 (dd, J = 6.4, 3.4 Hz, 2H), 3.07 (t, J = 6.5 Hz, 2H), 2.40 (s, 3H); LCMS (ESI):. LCMS (ESI): >97% purity; MS m/z, 356 [M + H]+; HPLC purity: 99.4%.

6-(3-Methoxyphenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)nicotinamide (8b):

1H NMR (400 MHz, CDCl3): δ 8.85 (dd, J = 2.3, 0.7 Hz, 1H), 8.04 (dd, J = 8.3, 2.3 Hz, 1H), 7.92 (s, 1H), 7.74 (dd, J = 8.3, 0.8 Hz, 1H), 7.60 – 7.57 (m, 1H), 7.57 – 7.53 (m, 2H), 7.39 (t, J = 7.9 Hz, 1H), 7.32 – 7.28 (m, 1H), 7.18 – 7.07 (m, 2H), 7.00 (dd, J = 8.2, 2.6 Hz, 1H), 6.23 (t, J = 6.0 Hz, 1H), 3.89 (s, 3H), 3.75 (dd, J = 6.4, 3.6 Hz, 2H), 3.07 (t, J = 6.5 Hz, 2H), 2.39 (s, 3H); LCMS (ESI): LCMS (ESI): >97% purity; MS m/z, 386 [M + H]+; HPLC purity: 96.8%.

6-(2-Methoxyphenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)nicotinamide (8c):

1H NMR (400 MHz, DMSO-d6): δ 10.74 (s, 1H), 9.06 – 9.02 (m, 1H), 8.82 (t, J = 5.6 Hz, 1H), 8.19 – 8.15 (m, 1H), 7.96 – 7.92 (m, 1H), 7.81 – 7.76 (m, 1H), 7.52 – 7.40 (m, 2H), 7.25 – 7.15 (m, 2H), 7.11 – 7.05 (m, 1H), 7.01 – 6.89 (m, 2H), 3.85 (bs, 3H), 3.44 (dd, J = 7.4, 3.6 Hz, 2H), 2.91 (t, J = 7.7 Hz, 2H), 2.32 (s, 3H); LCMS (ESI): >99% purity; MS m/z, 386 [M + H]+.

6-(2-Hydroxyphenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)nicotinamide (8d):

1H NMR (400 MHz, DMSO-d6): δ 13.75 (s, 1H), 10.74 (s, 1H), 9.01 (d, J = 1.3 Hz, 1H), 8.89 (t, J = 5.6 Hz, 1H), 8.39 – 8.28 (m, 2H), 8.07 (d, J = 8.3 Hz, 1H), 7.48 (d, J = 7.5 Hz, 1H), 7.35 (t, J = 7.7 Hz, 1H), 7.23 (d, J = 8.1 Hz, 1H), 7.00 – 6.89 (m, 4H), 3.49 – 3.41 (m, 2H), 2.92 (t, J = 7.3 Hz, 2H), 2.32 (s, 3H); LCMS (ESI): LCMS (ESI): >97% purity; MS m/z, 372 [M + H]+; HPLC purity: 97.1%.

6-(2-Fluorophenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)nicotinamide (8e):

1H NMR (400 MHz, CDCl3): δ 8.89 (d, J = 1.6 Hz, 1H), 8.05 – 7.96 (m, 2H), 7.88 (s, 1H), 7.84 – 7.80 (m, 1H), 7.54 (d, J = 7.6 Hz, 1H), 7.44 – 7.37 (m, 1H), 7.31 – 7.25 (m, 2H), 7.19 – 7.07 (m, 3H), 6.21 (bs, 1H), 3.77 (q, J = 6.4 Hz, 2H), 3.07 (t, J = 6.5 Hz, 2H), 2.39 (s, 3H); LCMS (ESI): >99% purity; MS m/z, 374.0 [M + H]+.

6-(4-Acetamidophenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)nicotinamide (8f):

1H NMR (400 MHz, DMSO-d6): δ 9.03 (d, J = 1.6 Hz, 1H), 8.79 (t, J = 5.7 Hz, 1H), 8.22 (dd, J = 8.4, 2.3 Hz, 1H), 8.11 (d, J = 8.8 Hz, 2H), 8.01 (d, J = 8.4 Hz, 2H), 7.73 (d, J = 8.7 Hz, 2H), 7.48 (d, J = 7.7 Hz, 1H), 7.23 (d, J = 7.6 Hz, 1H), 7.01 – 6.89 (m, 2H), 3.49 – 3.38 (m, 2H), 2.96 – 2.86 (m, 2H), 2.32 (s, 3H), 2.08 (s, 3H); LCMS (ESI): LCMS (ESI): >95% purity; m/z, 413 [M + H]+; HPLC purity: 95.8%.

6-(3,5-Dimethoxyphenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)nicotinamide (8g):

1H NMR (400 MHz, CDCl3): δ 8.84 (s, 1H), 8.02 (dt, J = 8.3, 2.1 Hz, 1H), 7.92 (s, 1H), 7.70 (d, J = 8.3 Hz, 1H), 7.54 (d, J = 7.5 Hz, 1H), 7.30 (d, J = 7.9 Hz, 1H), 7.18 – 7.05 (m, 4H), 6.55 (q, J = 2.0 Hz, 1H), 6.23 (t, J = 4.8 Hz, 1H), 3.86 (d, J = 1.8 Hz, 6H), 3.77 (q, J = 6.3 Hz, 2H), 3.04 (t, J = 6.5 Hz, 2H), 2.39 (d, J = 1.5 Hz, 3H). LCMS (ESI): >99% purity; MS m/z, 416.2 [M + H]+.

General procedure for the synthesis of 10a-c:

A solution of boronic acid (4a-c) (4.1 mmol, 1 equiv.) and bromo-acid 9 (4.1 mmol, 1 equiv.) in tetrahydrofuran or toluene and water (6:1) were loaded in to a sealed tube. To this solution, 1M Na2CO3 (8.2 mmol, 2 equiv.) was added and purged with nitrogen for 10 min. Then, Pd(PPh3)4 (0.2 mmol, 0.05 equiv.) catalyst was added to the reaction mixture, sealed and heated to 100 °C for 12 h. Reaction mixture was cooled to room temperature and solvent was evaporated under vacuum. The residue was washed with dichloromethane to remove organic impurities. Then, aqueous layer was acidified to pH 2 with concentrated HCl to result in white precipitate, which was filtered and dried under vacuum to provide the intermediates 10a66, 10b66 and 10c). Often these compounds used for next reaction without purification.

5-(4-Acetamidophenyl)picolinic acid (10c):

1H NMR (400 MHz, DMSO-d6): δ 10.18 (s, 1H), 9.01 (s, 1H), 8.24 (d, J = 11 Hz, 1H), 8.08 (d, J = 11 Hz, 1H), 7.84 – 7.70 (m, 4 H), 2.08 (s, 3H). LCMS (ESI): >95% purity; MS m/z, 255 [M - H]+.

General procedure for the synthesis of 11a-c:

To a solution of 10a-c (0.5 mmol, 1 equiv.) and 2-(2-methyl-1H-indol-3-yl)ethan-1-amine (7) (0.5 mmol, 1 equiv.) in N,N-dimethylformamide and dichloromethane (1:1) was added DMAP (catalytic amount) followed by EDCI.HCl (0.65 mmol, 1.3 equiv.) and the reaction mixture was stirred at room temperature for 10 h. Then, dichloromethane was evaporated and the crude reaction mixture was added a saturated solution of ammonium chloride (15 mL) and extracted with ethyl acetate (15 mL). Organic layer was separated and washed with saturated solution of sodium bicarbonate (15 mL) followed by brine solution (15 mL). Combined organic layer was dried over sodium sulfate, concentrated to dryness. The crude was purified on silica gel chromatography using 50–70% ethyl acetate in hexanes to get the required products (11a-c).

5-(3-Methoxyphenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)picolinamide (11a):

1H NMR (400 MHz, CDCl3): δ 8.68 (d, J = 2.2 Hz, 1H), 8.25 (d, J = 8.1 Hz, 1H), 8.17 (t, J = 5.6 Hz, 1H), 8.04 (s, 1H), 8.00 – 7.95 (m, 1H), 7.57 (d, J = 7.4 Hz, 1H), 7.40 (t, J = 8.0 Hz, 1H), 7.28 (d, J = 7.7 Hz, 1H), 7.18 – 7.05 (m, 4H), 6.97 (dd, J = 8.3, 2.5 Hz, 1H), 3.86 (s, 3H), 3.72 – 3.75 (m, 2H), 3.04 (t, J = 6.9 Hz, 2H), 2.37 (s, 3H); LCMS (ESI): > 98% purity; MS m/z, 386 [M + H]+.

5-(3,5-Dimethoxyphenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)picolinamide (11b):

1H NMR (400 MHz, CDCl3): δ 8.69 (d, J = 6.8 Hz, 1H), 8.26 (d, J = 8.1 Hz, 1H), 8.16 (t, J = 5.9 Hz, 1H), 8.01 – 7.97 (m, 1H), 7.86 (s, 1H), 7.58 (d, J = 7.5 Hz, 1H), 7.31 – 7.27 (m, 1H), 7.16 – 7.06 (m, 2H), 6.71 (d, J = 2.2 Hz, 2H), 6.53 (t, J = 2.2 Hz, 1H), 3.86 (s, 6H), 3.74 (q, J = 6.8 Hz, 2H), 3.05 (t, J = 7.0 Hz, 2H), 2.39 (s, 3H); LCMS (ESI):LCMS (ESI): >97% purity; MS m/z, 416 [M + H]+; HPLC purity: 97.6%.

5-(4-Acetamidophenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)picolinamide (11c):

1H NMR (400 MHz, DMSO-d6): δ 10.69 (s, 1H), 10.14 (s, 1H), 8.87 (d, J = 2.1 Hz, 1H), 8.81 (t, J = 5.9 Hz, 1H), 8.20 (dd, J = 7.4, 3.1 Hz, 1H), 8.05 (d, J = 8.1 Hz, 1H), 7.66 – 7.78 (m, 4H), 7.58 – 7.43 (m, 1H), 7.20 (d, J = 7.7 Hz, 1H), 6.96 – 6.63 (m, 2H), 3.33 – 3.46 (m, 2H), 2.88 (t, J = 7.2 Hz, 2H), 2.29 (s, 3H), 2.04 (s, 3H); LCMS (ESI): LCMS (ESI): >97% purity; MS m/z, 413 [M + H]+.

General procedure for the synthesis 13a-c:

A solution of boronic acid, 4c (4.1 mmol, 1 equiv.) and with 12a-c (4.1 mmol, 1 equiv.) in dioxane and water (6:1) were loaded in to a sealed tube. To this solution, 1M Na2CO3 (8.2 mmol, 2 equiv.) was added and purged with nitrogen for 10 min. Then, Pd(dppf)Cl2 (0.2 mmol, 0.05 equiv.) catalyst was added to the reaction mixture, sealed and heated to 120 °C for 12 h. Reaction mixture was cooled to room temperature and solvent was evaporated under vacuum. The residue was washed with dichloromethane to remove organic impurities. Then, aqueous layer was acidified to pH 2 with concentrated HCl to result in white precipitate, which was filtered and dried under vacuum to provide the intermediates (13a,67, 68 13b6971 and 13c72).

General procedure for the synthesis of 14a-c:

To a solution of 13a-c (0.5 mmol, 1 equiv.) and 2-(2-methyl-1H-indol-3-yl)ethan-1-amine (7) (0.5 mmol, 1 equiv.) in N,N-dimethylformamide and dichloromethane (1:1) was added DMAP (catalytic amount) followed by EDCI.HCl (0.65 mmol, 1.3 equiv.) and the reaction mixture was stirred at room temperature for 10 h. Then, dichloromethane was evaporated and the crude reaction mixture was added a saturated solution of ammonium chloride (15 mL) and extracted with ethyl acetate (15 mL). Organic layer was separated and washed with saturated solution of sodium bicarbonate (15 mL) followed by brine solution (15 mL). Combined organic layer was dried over sodium sulfate, concentrated to dryness. The crude was purified on silica gel chromatography using 50–70% ethyl acetate in hexanes to get the required products (14a-c).

2-(2-Methoxyphenyl)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (14a):

1H NMR (400 MHz, CDCl3): δ 9.00 (s, 2H), 7.92 (s, 1H), 7.74 (dd, J = 7.6, 1.6 Hz, 1H), 7.53 (d, J = 7.4 Hz, 1H), 7.49 – 7.43 (m, 1H), 7.28 (d, J = 8.0 Hz, 1H), 7.17 – 6.99 (m, 4H), 6.25 (s, 1H), 3.86 (s, 3H), 3.75 (q, J = 6.4 Hz, 2H), 3.07 (t, J = 6.5 Hz, 2H), 2.37 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 387 [M + H]+.

2-Fluoro-2′-methoxy-N-(2-(2-methyl-1H-indol-3-yl)ethyl)-[1,1’-biphenyl]-4-carboxamide (14b):

1H NMR (400 MHz, CDCl3): δ 7.85 (s, 1H), 7.54 (d, J = 7.6 Hz, 1H), 7.44 −7.26 (m, 4H), 7.22 (d, J = 6.3 Hz, 2H), 7.17 – 7.06 (m, 2H), 7.05 – 6.95 (m, 2H), 6.16 (t, J = 5.3 Hz, 1H), 3.76 (s, 3H), 3.74 – 3.67 (m, 2H), 3.04 (t, J = 6.6 Hz, 2H), 2.38 (s, 3H); LCMS (ESI): >98% purity; MS m/z, 403 [M + H]+.

2′-Methoxy-N-(2-(2-methyl-1H-indol-3-yl)ethyl)-[1,1′-biphenyl]-4-carboxamide (14c):

1H NMR (400 MHz, DMSO-d6): δ 10.70 (s, 1H), 8.60 (t, J = 5.9 Hz, 1H), 7.87 – 7.79 (m, 2H), 7.56 – 7.49 (m, 2H), 7.46 (d, J = 7.5 Hz, 1H), 7.37 – 7.28 (m, 2H), 7.22 – 7.17 (m, 1H), 7.11 (dd, J = 5.5, 4.6 Hz, 1H), 7.05 – 6.98 (m, 1H), 6.98 – 6.86 (m, 2H), 3.75 (d, J = 1.9 Hz, 3H), 3.43 – 3.34 (m, 2H), 2.86 (t, J = 7.4 Hz, 2H), 2.29 (d, J = 1.9 Hz, 3H); LCMS (ESI): >97% purity; MS m/z, 385 [M + H]+; HPLC purity: 96%.

2-Amino-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (16):

To a solution of commercially available acid 15 (3 g, 21 mmol, 1 equiv.) in N,N-dimethylformamide (20 mL) was added DMAP (0.78 g, 6.3 mmol, 0.3 equiv.) followed by EDCI.HCl (5.35 g, 28 mmol, 1.3 equiv.) and stirred at room temperature for 10 minutes. Then, 2-(2-methyl-1H-indol-3-yl)ethan-1-amine (7) was added to the reaction mixture and stirred at room temperature for 24 h. Reaction mixture was added saturated solution of ammonium chloride (5 mL) and extracted with ethyl acetate (3 × 10 mL). Organic layer was separated and washed with saturated solution of sodium bicarbonate (5 mL) followed by brine solution. Combined organic layer was dried over sodium sulfate, concentrated to dryness. The crude material was purified on silica gel chromatography using 4–6% methanol in dichloromethane to get the required product 16 as solid (Yield: 59%). 1H NMR (400 MHz, DMSO-d6): δ 10.69 (s, 1H), 8.61 (s, 2H), 8.39 (t, J = 5.6 Hz, 1H), 7.42 (d, J = 7.7 Hz, 1H), 7.22 – 7.17 (m, 1H), 7.16 (s, 2H), 6.97 – 6.85 (m, 2H), 3.41 – 3.27 (m, 2H), 2.82 (t, J = 7.4 Hz, 2H), 2.27 (s, 3H); LCMS (ESI): > 98% purity. MS m/z, 296 [M + H]+.

General procedure for the synthesis of 20a-20r:

To a solution of 16 (0.5 mmol, 1 equiv.) and 2-bromopyridines (17a-o or 18 or 19a-b) (0.5 mmol, 1 equiv.) in dioxane was added Cs2CO3 (1.0 mmol, 2 equiv.). The solution was purged with nitrogen for 10 minutes. Then, Xantphos (0.05 mmol, 0.1 equiv.) was added followed by Pd2(dba)3 catalyst (0.05 mmol, 0.1 equiv.) and heated to 100 °C for 12–18 h. Reaction mixture was cooled to room temperature and added water (10 mL). Resultant solid was filtered and purified on silica gel chromatography using 3–5% methanol in dichloromethane to get the required products 20a-r.

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-2-(pyridin-2-ylamino)pyrimidine-5-carboxamide (20a):

1H NMR (400 MHz, DMSO-d6): δ 10.71 (s, 1H), 10.29 (s, 1H), 8.88 (s, 2H), 8.66 (t, J = 5.4 Hz, 1H), 8.29 (d, J = 4.9 Hz, 1H), 8.21 (d, J = 8.4 Hz, 1H), 7.79 – 7.72 (m, 1H), 7.44 (d, J = 7.6 Hz, 1H), 7.20 (d, J = 7.8 Hz, 1H), 7.06 – 6.99 (m, 1H), 6.97 – 6.86 (m, 2H), 3.35− 3.42 (m, 2H), 2.86 (t, J = 7.3 Hz, 2H), 2.28 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 373 [M + H]+; HPLC purity: 98.1%.

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-2-((4-methylpyridin-2-yl)amino)pyramidine-5-carboxamide (20b):

1H NMR (400 MHz, DMSO-d6): δ 10.74 (s, 1H), 10.22 (s, 1H), 8.92 – 8.89 (m, 2H), 8.67 (t, J = 5.7 Hz, 1H), 8.18 (d, J = 5.0 Hz, 1H), 8.08 (s, 1H), 7.47 (d, J = 7.6 Hz, 1H), 7.26 – 7.19 (m, 1H), 7.01 – 6.87 (m, 3H), 3.45 – 3.38 (m, 2H), 2.89 (t, J = 7.3 Hz, 2H), 2.34 (s, 3H), 2.32 (s, 3H); LCMS (ESI): > 95% purity; MS m/z, 387 [M + H]+.

2-((4-Fluoropyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20c):

1H NMR (400 MHz, DMSO-d6): δ 10.74 (s, 1H), 10.70 (s, 1H), 8.95 (s, 2H), 8.72 (t, J = 8 Hz 1H), 8.37 – 8.31 (m, 1H), 8.20 – 8.13 (m, 1H), 7.46 (d, J = 7.7 Hz, 1H), 7.23 (d, J = 8.0 Hz, 1H), 7.03 – 6.89 (m, 3H), 3.45− 3.38 (m, 2H), 2.89 (t, J = 6.7 Hz, 2H), 2.31 (d, J = 2.0 Hz, 3H); LCMS (ESI): >97% purity; MS m/z, 391 [M + H]+; HPLC purity: 98.1%.

2-((4-(tert-Butyl)pyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20d):

1H NMR (400 MHz, DMSO-d6): δ 10.71 (s, 1H), 10.23 (s, 1H), 8.89 (d, J = 2.8 Hz, 2H), 8.62 (t, J = 5.6 Hz, 1H), 8.27 – 8.30 (m, 1H), 8.21 – 8.17 (m, 1H), 7.43 (d, J = 7.6 Hz, 1H), 7.20 (dd, J = 7.8, 0.8 Hz, 1H), 7.05 (dd, J = 5.3, 1.8 Hz, 1H), 6.97 – 6.85 (m, 2H), 3.42 – 3.33 (m, 2H), 2.86 (t, J = 7.3 Hz, 2H), 2.28 (s, 3H), 1.29 – 1.24 (m, 9H); LCMS (ESI): >97% purity; MS m/z, 429 [M + H]+.

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-2-((6-methylpyridin-2-yl)amino)pyrimidine-5-carboxamide (20e):

1H NMR (400 MHz, DMSO-d6): δ 10.74 (s, 1H), 10.17 (s, 1H), 8.90 (d, J = 2.1 Hz, 2H), 8.67 (t, J = 5.3 Hz, 1H), 8.07 (d, J = 8.3 Hz, 1H), 7.68 (t, J = 7.8 Hz, 1H), 7.47 (d, J = 7.8 Hz, 1H), 7.23 (d, J = 7.7 Hz, 1H), 7.00 – 6.88 (m, 3H), 3.45− 3.38 (m, 2H), 2.89 (t, J = 7.4 Hz, 2H), 2.41 (s, 3H), 2.31 (s, 3H); LCMS (ESI): >98% purity; MS m/z, 387 [M + H]+.

2-((6-Fluoropyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20f):

1H NMR (400 MHz, DMSO-d6): δ 10.74 (s, 1H), 10.61 (s, 1H), 8.95 – 8.91 (m, 2H), 8.73 (t, J = 5.7 Hz, 1H), 8.22 – 8.17 (m, 1H), 7.97 (q, J = 8.4 Hz, 1H), 7.47 (d, J = 7.6 Hz, 1H), 7.25 – 7.20 (m, 1H), 7.01 – 6.89 (m, 2H), 6.77 (dd, J = 7.8, 2.4 Hz, 1H), 3.45 – 3.37 (m, 2H), 2.89 (t, J = 7.4 Hz, 2H), 2.31 (s, 3H); LCMS (ESI): >97% purity. MS m/z, 391 [M + H]+.

2-((6-Cyanopyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20g):

1H NMR (400 MHz, DMSO-d6): δ 10.89 (s, 1H), 10.74 (s, 1H), 8.95 (s, 2H), 8.74 (t, J = 4.8 Hz, 1H), 8.60 – 8.54 (m, 1H), 8.02 (t, J = 8.1 Hz, 1H), 7.71 – 7.61 (m, 1H), 7.46 (d, J = 7.6 Hz, 1H), 7.23 (d, J = 7.7 Hz, 1H), 7.00 – 6.89 (m, 2H), 3.42 (dd, J = 12.5, 6.2 Hz, 2H), 2.89 (t, J = 6.9 Hz, 2H), 2.31 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 398 [M + H]+; HPLC purity: 97%.

2-((6-Methoxypyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20h):

1H NMR (400 MHz, DMSO-d6) δ 10.74 (s, 1H), 10.07 (s, 1H), 8.91 (s, 2H), 8.68 (t, J = 5.7 Hz, 1H), 7.82 (d, J = 7.9 Hz, 1H), 7.70 (t, J = 7.9 Hz, 1H), 7.47 (d, J = 7.5 Hz, 1H), 7.23 (d, J = 7.6 Hz, 1H), 7.01 – 6.87 (m, 2H), 6.46 (d, J = 7.8 Hz, 1H), 3.85 (s, 3H), 3.45 – 3.37 (m, 2H), 2.89 (t, J = 7.1 Hz, 2H), 2.32 (s, 3H); LCMS (ESI): >96% purity; MS m/z, 403 [M + H]+.

2-((6-Hydroxypyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20i):

1H NMR (400 MHz, DMSO-d6): δ 12.01 (s, 1H), 10.74 (s, 2H), 8.95 (s, 2H), 8.74 (t, J = 5.5 Hz, 1H), 7.50 – 7.40 (m, 2H), 7.23 (d, J = 8.2 Hz, 1H), 7.01 – 6.87 (m, 2H), 6.41 (bs, 1H), 5.98 (d, J = 8.8 Hz, 1H), 3.46 – 3.38 (m, 2H), 2.89 (t, J = 7.4 Hz, 2H), 2.31 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 389 [M + H]+.

2-((6-Acetylpyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20j):

1H NMR (400 MHz, DMSO-d6): δ 10.71 (s, 1H), 10.49 (s, 1H), 8.91 (s, 2H), 8.73 – 8.63 (m, 1H), 8.41 (d, J = 9.2 Hz, 1H), 7.96 (t, J = 7.2 Hz, 1H), 7.58 (d, J = 7.5 Hz, 1H), 7.44 (d, J = 7.2 Hz, 1H), 7.20 (d, J = 8.5 Hz, 1H), 6.98 – 6.82 (m, 2H), 3.39 (dd, J = 13.5, 6.7 Hz, 2H), 2.86 (t, J = 7.4 Hz, 2H), 2.60 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 415 [M + H]+.

2-((4-Acetylpyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20k):

1H NMR (400 MHz, DMSO-d6): δ 10.71 (s, 1H), 10.61 (s, 1H), 8.92 (s, 2H), 8.71 – 8.60 (m, 2H), 8.49 (d, J = 5.1 Hz, 1H), 7.54 – 7.37 (m, 2H), 7.20 (d, J = 7.7 Hz, 1H), 6.92 (dd, J, 14.1, 7.1 Hz, 2H), 3.39 (dd, J = 13.9, 6.5 Hz, 2H), 2.86 (t, J = 7.5 Hz, 2H), 2.60 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 415 [M + H]+; HPLC purity: 96.2%.

2-((5-Acetylpyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20l):

1H NMR (400 MHz, DMSO-d6): δ 10.87 (s, 1H), 10.72 (s, 1H), 8.94 (s, 2H), 8.88 (d, J = 2.4 Hz, 1H), 8.72 (t, J = 5.7 Hz, 1H), 8.38 (d, J = 8.9 Hz, 1H), 8.27 (dd, J = 8.9, 2.3 Hz, 1H), 7.44 (d, J = 7.5 Hz, 1H), 7.20 (d, J = 7.5 Hz, 1H), 6.92 (dt, J = 14.6, 7.0 Hz, 2H), 3.39 (dd, J = 13.4, 6.6 Hz, 2H), 2.87 (t, J = 7.3 Hz, 2H), 2.54 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >98% purity; MS m/z, 415 [M + H]+.

2-((6-(2-Hydroxybutan-2-yl)pyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl) pyrimidine-5-carboxamide (20m):

1H NMR (400 MHz, DMSO-d6): δ 10.71 (s, 1H), 10.06 (s, 1H), 8.88 (s, 2H), 8.65 (t, J = 5.7 Hz, 1H), 8.03 (d, J = 8.2 Hz, 1H), 7.74 (t, J = 7.9 Hz, 1H), 7.43 (d, J = 7.6 Hz, 1H), 7.20 (dd, J = 7.7, 3.2 Hz, 2H), 6.91 (ddd, J = 14.7, 13.6, 6.2 Hz, 2H), 5.07 (s, 1H), 3.38 (dd, J = 13.5, 7.0 Hz, 2H), 2.86 (t, J = 7.3 Hz, 2H), 2.28 (s, 3H), 1.87 – 1.58 (m, 2H), 1.37 (s, 3H), 0.64 (t, J = 7.4 Hz, 3H); LCMS (ESI): >98% purity; MS m/z, 445 [M + H]+.

2-((6-(tert-Butyl)pyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20n):

1H NMR (400 MHz, DMSO-d6): δ 10.71 (s, 1H), 9.87 (s, 1H), 8.88 (d, J = 6.0 Hz, 2H), 8.64 (t, J = 5.6 Hz, 1H), 7.98 (d, J = 8.1 Hz, 1H), 7.68 (t, J = 7.9 Hz, 1H), 7.44 (d, J = 7.3 Hz, 1H), 7.20 (d, J = 7.3 Hz, 1H), 7.04 (d, J = 7.6 Hz, 1H), 7.02 – 6.80 (m, 2H), 3.43 – 3.34 (m, 2H), 2.86 (t, J = 7.2 Hz, 2H), 2.29 (s, 3H), 1.30 – 1.25 (m, 9H); LCMS (ESI): >97% purity; MS m/z, 429 [M + H]+.

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20o; TG11–77):

1H NMR (400 MHz, DMSO-d6): δ 10.74 (s, 1H), 10.08 (s, 1H), 8.90 (s, 2H), 8.65 (t, J = 5.5 Hz, 1H), 7.92 (s, 1H), 7.47 (d, J = 7.6 Hz, 1H), 7.23 (d, J = 7.8 Hz, 1H), 6.95 (dt, J = 20.4, 7.3 Hz, 2H), 6.76 (s, 1H), 3.45 – 3.37 (m, 2H), 2.89 (t, J = 7.3 Hz, 2H), 2.37 (s, 3H), 2.32 (s, 3H), 2.30 (s, 3H); LCMS (ESI): >95% purity. MS m/z, 401 [M + H]+; HPLC purity: 96.4%.

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide.Hydrochloride (20o-HCl; TG11–77-HCl):

To a solution of 20o (500 mg, 1.25 mmol, 1 equiv.) in dichloromethane (5 mL) was added 4M HCl in dioxane (0.62 mL, 2.5 mmol, 2 equiv.) at 0 °C and allowed to stir at room temperature for 12 h. The precipitated solid was filtered and washed with dichloromethane (5 mL) followed by ethyl acetate (5 mL) and dried to get the required salt, 20o.HCl (Yield: 86%). 1H NMR (400 MHz, DMSO-d6): δ 11.95 (s, 1H), 10.79 (s, 1H), 9.13 (s, 2H), 8.99 (t, J = 5.3 Hz, 1H), 7.61 (s, 1H), 7.46 (d, J = 7.7 Hz, 1H), 7.23 (d, J = 7.7 Hz, 1H), 7.18 (s, 1H), 7.00 – 6.88 (m, 2H), 3.66 (bs, 1H), 3.44 (q, J = 6.7 Hz, 2H), 2.91 (t, J = 7.3 Hz, 2H), 2.62 (s, 3H), 2.47 (s, 3H), 2.32 (s, 3H); LCMS (ESI): >97% purity. MS m/z, 401 [(M – HCl) + H]+; HPLC purity: 99%.

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-2-(pyridin-4-ylamino)pyrimidine-5-carboxamide (20p):

1H NMR (400 MHz, DMSO-d6): δ 10.78 (d, J = 2.3 Hz, 1H), 9.34 – 9.29 (m, 4H), 9.22 – 9.14 (m, 2H), 7.45 (d, J = 7.7 Hz, 1H), 7.20 (d, J = 7.9 Hz, 1H), 7.08 – 7.01 (m, 2H), 6.97 – 6.82 (m, 2H), 3.46 – 3.40 (m, 2H), 2.90 (t, J = 7.3 Hz, 2H), 2.29 (d, J = 2.0 Hz, 3H); LCMS (ESI): > 97% purity. MS m/z, 373 [M + H]+.

N-(2-(2-Methyl-1H-indol-3-yl)ethyl)-2-(pyridin-3-ylamino)pyrimidine-5-carboxamide (20q):

1H NMR (400 MHz, DMSO-d6): δ 10.70 (s, 1H), 10.26 (s, 1H), 8.91 (s, 1H), 8.88 (s, 2H), 8.62 (t, J = 5.5 Hz, 1H), 8.26 – 8.18 (m, 2H), 7.46 (d, J = 7.6 Hz, 1H), 7.35 (dd, J = 8.2, 4.8 Hz, 1H), 7.22 (d, J = 7.8 Hz, 1H), 7.00 – 6.87 (m, 2H), 3.48 – 3.35 (m, 2H), 2.89 (t, J = 7.3 Hz, 2H), 2.32 (s, 3H); LCMS (ESI): >97% purity. MS m/z, 373 [M + H]+; HPLC purity: 97.6%.

2-((2,6-Dimethylpyridin-3-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (20r):

1H NMR (400 MHz, DMSO-d6): δ 10.73 (s, 1H), 9.44 (s, 1H), 8.75 (s, 2H), 8.55 (t, J = 5.7 Hz, 1H), 7.64 (d, J = 8.1 Hz, 1H), 7.46 (d, J = 7.7 Hz, 1H), 7.21 (d, J = 7.6 Hz, 1H), 7.09 (d, J = 8.1 Hz, 1H), 7.00 – 6.88 (m, 2H), 3.43 – 3.35 (m, 2H), 2.87 (t, J = 7.3 Hz, 2H), 2.42 (s, 3H), 2.35 (s, 3H), 2.31 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 401[M + H]+; HPLC purity: 97.3%.

Ethyl 2-((4,6-dimethylpyridin-2-yl)amino)pyrimidine-5-carboxylate (22):

To a solution of 21 (250 mg, 1.6 mmol, 1 equiv.) and 17o (35 mg, 1.6 mmol, 1 equiv.) in dioxane (5 mL) were added Cs2CO3 (1.1 g, 3.26 mmol, 2 equiv.) followed by BINAP (100 g, 0.16 mmol, 0.1 equiv.). Reaction mixture was purged with nitrogen for 10 minutes and added Pd(OAc)2 (36 mg, 0.16 mmol, 0.1 equiv.) and heated to 100 °C for 48 h. Reaction mixture was cooled to room temperature and added water and filtered the solid, which was purified on column chromatography using 30–40% ethyl acetate in hexanes to get the required compound 22. 1H NMR (400 MHz, CDCl3): δ 11.75 (s, 1H), 9.12 (s, 2H), 8.62 (s, 1H), 6.84 (s, 1H), 4.50 – 4.28 (m, 2H), 2.70 (s, 3H), 2.54 (s, 3H), 1.40 (t, J = 7.1 Hz, 3H); LCMS (ESI): > 94% purity. MS m/z, 273 [M + H]+.

2-((4,6-Dimethylpyridin-2-yl)amino)pyrimidine-5-carboxylic acid (23):

To a solution of 22 (100 mg, 0.38 mmol, 1 equiv.) in tetrahydrofuran and water (7:3, 5 mL) was added LiOH.H2O (46 mg, 1.14 mmol, 3 equiv.) and heated to 60 °C for 12 h. Reaction mixture was brought to room temperature and acidified with 1N HCl and extracted with ethyl acetate (20 mL). Organic layer was concentrated to dryness to obtain the required acid 23. 1H NMR (400 MHz, DMSO-d6): δ 9.54 (s, 1H), 8.77 (s, 2H), 7.92 (s, 1H), 6.67 (s, 1H), 2.31 (s, 3H), 2.25 (s, 3H); LCMS (ESI): > 96% purity; MS m/z, 243 [M - H]+.

General procedure for the synthesis of compounds 24a-i:

To a solution of 23 (0.61 mmol, 1 equiv.) and compound 7a-i (0.61 mmol, 1 equiv.) in N,N-dimethylformamide (5 mL) was added DMAP (catalytic amount) followed by EDCI.HCl (0.78 mmol, 1.3 equiv.) and the reaction mixture was stirred at 50 °C for 24–48 h. Reaction mixture was brought to room temperature and added saturated solution of ammonium chloride (10 mL) and extracted with ethyl acetate (10 mL). Organic layer was separated and washed with saturated solution of sodium bicarbonate (10 mL) followed by brine solution (10 mL). Combined organic layer was dried over sodium sulfate, concentrated to dryness. The crude was purified on silica gel chromatography using 5–7% methanol in dichloromethane to get the required products (24a-i).

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-(5-fluoro-2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (24a):

1H NMR (400 MHz, DMSO-d6): δ 10.85 (s, 1H), 10.06 (s, 1H), 8.88 (s, 2H), 8.63 (t, J = 5.8 Hz, 1H), 7.91 (s, 1H), 7.22 – 7.17 (m, 2H), 6.83 – 6.74 (m, 2H), 3.43 – 3.35 (m, 2H), 2.85 (t, J = 7.3 Hz, 2H), 2.36 (s, 3H), 2.31 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >96% purity; MS m/z, 419 [M + H]+.

N-(2-(5-Chloro-2-methyl-1H-indol-3-yl)ethyl)-2-((4,6-dimethylpyridin-2-yl)amino)pyrimidine-5-carboxamide (24b):

1H NMR (400 MHz, DMSO-d6): δ 10.97 (s, 1H), 10.09 (s, 1H), 8.89 (s, 2H), 8.63 (t, J = 4.8 Hz, 1H), 7.92 (s, 1H), 7.48 (s, 1H), 7.23 (d, J = 8.5 Hz, 1 H) 7.08 – 6.88 (m, 1H), 6.76 (s, 1H), 3.44 – 3.37 (m, 2H), 2.91 – 2.82 (m, 2H), 2.36 (s, 3H), 2.31 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >95% purity; MS m/z, 435 [M + H]+.

N-(2-(5,7-Difluoro-2-methyl-1H-indol-3-yl)ethyl)-2-((4,6-dimethylpyridin-2-yl)amino)pyrimidine-5-carboxamide (24c):

1H NMR (400 MHz, DMSO-d6): δ 11.32 (s, 1H), 10.09 (s, 1H), 8.88 (s, 2H), 8.62 (t, J = 5.4 Hz, 1H), 7.91 (s, 1H), 7.11 (dd, J = 9.6, 1.8 Hz, 1H), 6.82 (t, J = 10.5 Hz, 1H), 6.76 (s, 1H), 3.44 – 3.36 (m, 2H), 2.86 (t, J = 7.0 Hz, 2H), 2.37 (s, 3H), 2.32 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 437 [M + H]+.

N-(2-(5,7-Dichloro-2-methyl-1H-indol-3-yl)ethyl)-2-((4,6-dimethylpyridin-2-yl)amino)pyrimidine-5-carboxamide (24d):

1H NMR (400 MHz, DMSO-d6): δ 11.29 (s, 1H), 10.04 (s, 1H), 8.82 (s, 2H), 8.56 (t, J = 6.1 Hz, 1H), 7.92 (s, 1H), 7.45 (s, 1H), 7.09 (s, 1H), 6.73 (s, 1H), 3.36 (dd, J = 12.0, 5.3 Hz, 2H), 2.85 – 2.81 (m, 2H), 2.69 (s, 3H), 2.33 (s, 3H), 2.26 (s, 3H); LCMS (ESI): >95% purity; MS m/z, 469 [M + H]+; HPLC purity: 95.5%.

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-(5-methoxy-2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (24e):

1H NMR (400 MHz, DMSO-d6): δ 10.57 (s, 1H), 10.07(s, 1H), 8.89 (s, 2H), 8.64 (t, J = 5.6 Hz, 1H), 7.91 (s, 1H), 7.11 (d, J = 8.6 Hz, 1H), 6.96 (d, J = 2.2 Hz, 1H), 6.76 (s, 1H), 6.61 (dd, J = 8.6, 2.3 Hz, 1H), 3.71 (s, 3H), 3.46 – 3.35 (m, 2H), 2.85 (t, J = 7.2 Hz, 2H), 2.36 (s, 3H), 2.29 (bs, 6H); LCMS (ESI): >96% purity; MS m/z, 431 [M + H]+.

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-(2-(trifluoromethyl)-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (24f):

1H NMR (400 MHz, DMSO-d6): δ 11.97 (s, 1H), 10.07 (s, 1H), 8.86 (bs, 2H), 8.71 (t, J = 5.7 Hz, 1H), 7.90 (s, 1H), 7.76 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.32 – 7.25 (m, 1H), 7.16 – 7.08 (m, 1H), 6.76 (s, 1H), 3.53 – 3.44 (m, 2H), 3.11 (t, J = 6.8 Hz, 2H), 2.36 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >97% purity; MS m/z, 455 [M + H]+.

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-(2-methylpyrazolo[1,5-a]pyridin-3-yl)ethyl)pyrimidine-5-carboxamide (24g):

1H NMR (400 MHz, DMSO-d6): δ 10.03 (s, 1H), 8.81 (s, 2H), 8.59 (t, J = 5.6 Hz, 1H), 8.45 (dd, J = 7.0, 0.8 Hz, 1H), 7.86 (s, 1H), 7.47 (d, J = 8.8 Hz, 1H), 7.13 – 6.97 (m, 1H), 6.77 – 6.64 (m, 2H), 3.42 – 3.37 (m, 2H), 2.87 (t, J = 6.1 Hz, 2H), 2.33 (s, 3H), 2.30 (s, 3H), 2.25 (s, 3H); LCMS (ESI): >96% purity; MS m/z, 402 [M + H]+.

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-(2-methyl-1H-indol-3-yl)-2-oxoethyl)pyrimidine-5-carboxamide (24h):

1H NMR (400 MHz, DMSO-d6): δ 11.97 (s, 1H), 10.10 (s, 1H), 8.97 (s, 2H), 8.82 (t, J = 6.6 Hz, 1H), 8.05 – 7.95 (m, 1H), 7.91 (s, 1H), 7.46 – 7.33 (m, 1H), 7.21 – 7.10 (m, 2H), 6.74 (s, 1H), 4.72 – 4.53 (m, 2H), 2.71 (s, 3H), 2.34 (s, 3H), 2.27 (s, 3H); LCMS (ESI): >98% purity; MS m/z, 415 [M + H]+.

2-((4,6-Dimethylpyridin-2-yl)amino)-N-(2-hydroxy-2-(2-methyl-1H-indol-3-yl)ethyl)pyrimidine-5-carboxamide (24i):

1H NMR (400 MHz, DMSO-d6): δ 10.77 (s, 1H), 10.06 (s, 1H), 8.63 (t, J = 8 Hz, 1H), 8.90 (s, 2H), 7.91 (s, 1H), 7.65 (d, J = 8 Hz, 1H), 7.26 – 7.20 (m, 1H), 7.00 – 6.88 (m, 2H), 6.76 (s, 1H), 5.21 (d, J = 3.3 Hz, 1H), 5.09 – 5.03 (t, J = 8.2 Hz, 1H), 3.66 – 3.44 (m, 2H), 2.36 (s, 3H), 2.35 (s, 3H), 2.29 (s, 3H); LCMS (ESI): >96% purity; MS m/z, 417 [M + H]+.

Cell Culture.

The rat C6 glioma (C6G) cells stably expressing human DP1, EP2, EP4, or IP receptors were created in the laboratory19, 30, 73 and grown in Dulbecco’s Modified Eagle Medium (DMEM) (Invitrogen) supplemented with 10% (v/v) fetal bovine serum (FBS) (Invitrogen), 100 U/mL penicillin, 100 μg/mL streptomycin (Invitrogen), and 0.5 μg/mL G418 (Invitrogen).

Cell-Based cAMP Assay.

Intracellular cAMP was measured with a cell-based homogeneous time-resolved fluorescence resonance energy transfer (TR-FRET) method (Cisbio Bioassays), as previously described.19, 30 The assay is based on generation of a strong FRET signal upon the interaction of two molecules, an anti-cAMP antibody coupled to a FRET donor (Cryptate) and cAMP coupled to a FRET acceptor (d2). Endogenous cAMP produced by cells competes with labeled cAMP for binding to the cAMP antibody and thus reduces the FRET signal. Cells stably expressing human DP1, EP2, EP4, or IP receptors were seeded into 384-well plates in 30 μL complete medium (4,000 cells/well) and grown overnight. The medium was carefully withdrawn and 10 μL Hanks’ Buffered Salt Solution (HBSS) (Hyclone) containing 20 μM rolipram was added into the wells to block phosphodiesterases. The cells were incubated at room temperature for 0.5–1 h and then treated with vehicle or test compound for 10 min before addition of increasing concentrations of appropriate agonist: BW245C for DP1, PGE2 for EP2 and EP4, or iloprost for IP. The cells were incubated at room temperature for 40 min, then lysed in 10 μL lysis buffer containing the FRET acceptor cAMP-d2 and 1 min later another 10 μL lysis buffer with anti-cAMP-Cryptate was added. After 60–90 min incubation at room temperature, the FRET signal was measured by an Envision 2103 Multilabel Plate Reader (PerkinElmer Life Sciences) with a laser excitation at 337 nm and dual emissions at 665 nm and 590 nm for d2 and Cryptate (50 μs delay), respectively. The FRET signal was expressed as: F665/F590 × 104.

Cytokine induction assay.

Stable BV2-hEP2 microglia cells were created in the lab54 and were grown overnight on poly-D-lysine coated 12 well plates at 200,000 cells per well in culture media. The cells were exposed to the test compounds 20o or others (0.3 μM or 1 μM) for 1 h, and EP2 selective agonist ONO-AE1-259-01 (30 nM) for an additional hour and subsequently LPS (100 ng/mL) for 2 h. All compounds were dissolved in DMSO and diluted in media just prior to cell treatment. Following incubation, media was removed from the wells and the cells were subjected to RNA extraction and purification using Trizol and the Zymo Research Quick-RNA miniprep kit according to the manufacturer’s protocol (Genesee Scientific). First-strand cDNA synthesis, qRT-PCR and analysis was performed using the primers. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a single internal control for relative quantification to determine whether EP2 activation modulates expression of inflammatory mediators in BV2-hEP2 microglia (Figure 5, ,6).6). PCR gene expression data are presented as the mean fold change of each gene of interest in the compound treated groups compared to vehicle.

Supplementary Material

Accepted SI information

ACKNOWLEDGEMENTS

This work was supported by NIH/NIA grant U01 AG052460 (T.G.), NINDS grants, R21 NS101167 (T.G.) R01 NS097776 (R.D.), and by ADDF grant 20131001 (T.G.). We also thank ONO Pharmaceutical Co (Osaka, Japan) for providing ONO-AE1-259-01.

ABBREVATIONS USED:

ADAlzheimer’s disease
CNScentral nervous system
PDParkinson’s disease
SEstatus epilepticus
TBItraumatic brain injury
COX-2cyclooxygenase-2
HTShigh-throughput screening
EDCI.HCl1-ethyl-3-(3-dimethylaminopropyl)carbodiimide.hydrochloride
DMAP4-(dimethylamino)pyridine
SARStructure activity relationship
MLMmouse liver microsomes
MPOmulti-parameter optimization
CSFCerebrospinal fluid

Footnotes

Supporting Information

The scanned NMR spectra of all the new compounds and HPLC spectra of key compounds. The Supporting Information is available free of charge on the ACS publications website. This material is available free of charge via the Internet at http://pubs.acs.org.

REFERENCES

1. Heneka MT; Carson MJ; El 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. [Europe PMC free article] [Abstract] [Google Scholar]
2. Hirsch EC; Vyas S; Hunot S Neuroinflammation in Parkinson’s Disease. Parkinsonism Relat Disord 2012, 18 Suppl 1, S210–212. [Abstract] [Google Scholar]
3. Minghetti L Role of Inflammation in Neurodegenerative Diseases. Curr. Opin. Neurol 2005, 18, 315–321. [Abstract] [Google Scholar]
4. Vezzani A; French J; Bartfai T; Baram TZ The Role of Inflammation in Epilepsy. Nat. Rev. Neurol 2011, 7, 31–40. [Europe PMC free article] [Abstract] [Google Scholar]
5. Niesman IR; Schilling JM; Shapiro LA; Kellerhals SE; Bonds JA; Kleschevnikov AM; Cui W; Voong A; Krajewski S; Ali SS; Roth DM; Patel HH; Patel PM; Head BP Traumatic Brain Injury Enhances Neuroinflammation and Lesion Volume in Caveolin Deficient Mice. J. Neuroinflammation 2014, 11, 39–52. [Europe PMC free article] [Abstract] [Google Scholar]
6. Vezzani A; Friedman A; Dingledine RJ The Role of Inflammation in Epileptogenesis. Neuropharmacology 2013, 69, 16–24. [Europe PMC free article] [Abstract] [Google Scholar]
7. Block ML; Zecca L; Hong JS Microglia-Mediated Neurotoxicity: Uncovering the Molecular Mechanisms. Nat. Rev. Neurosci 2007, 8, 57–69. [Abstract] [Google Scholar]
8. Vliet EAV; Aronica E; Vezzani A; Ravizza T Review: Neuroinflammatory Pathways as Treatment Targets and Biomarker Candidates in Epilepsy: Emerging Evidence from Preclinical and Clinical Studies. Neuropathol Appl Neurobiol 2018, 44, 91–111. [Abstract] [Google Scholar]
9. Krstic D; Knuesel I Deciphering the Mechanism Underlying Late-Onset Alzheimer Disease. Nat. Rev. Neurol 2013, 9, 25–34. [Abstract] [Google Scholar]
10. Ho L; Pieroni C; Winger D; Purohit DP; Aisen PS; Pasinetti GM Regional Distribution of Cyclooxygenase-2 in the Hippocampal Formation in Alzheimer’s Disease. J. Neurosci. Res 1999, 57, 295–303. [Abstract] [Google Scholar]
11. Hoozemans JJ; van Haastert ES; Veerhuis R; Arendt T; Scheper W; Eikelenboom P; Rozemuller AJ Maximal Cox-2 and Pprb Expression in Neurons Occurs During Early Braak Stages Prior to the Maximal Activation of Astrocytes and Microglia in Alzheimer’s Disease. J. Neuroinflammation 2005, 2, 27–31. [Europe PMC free article] [Abstract] [Google Scholar]
12. Pasinetti GM; Aisen PS Cyclooxygenase-2 Expression is Increased in Frontal Cortex of Alzheimer’s Disease Brain. Neuroscience 1998, 87, 319–324. [Abstract] [Google Scholar]
13. Aisen PS; Schafer KA; Grundman M; Pfeiffer E; Sano M; Davis KL; Farlow MR; Jin S; Thomas RG; Thal LJ; Alzheimer’s Disease Cooperative S Effects of Rofecoxib or Naproxen Vs Placebo on Alzheimer Disease Progression: A Randomized Controlled Trial. JAMA 2003, 289, 2819–2826. [Abstract] [Google Scholar]
14. Arehart E; Stitham J; Asselbergs FW; Douville K; MacKenzie T; Fetalvero KM; Gleim S; Kasza Z; Rao Y; Martel L; Segel S; Robb J; Kaplan A; Simons M; Powell RJ; Moore JH; Rimm EB; Martin KA; Hwa J Acceleration of Cardiovascular Disease by a Dysfunctional Prostacyclin Receptor Mutation: Potential Implications for Cyclooxygenase-2 Inhibition. Circ. Res 2008, 102, 986–993. [Europe PMC free article] [Abstract] [Google Scholar]
15. Egan KM; Lawson JA; Fries S; Koller B; Rader DJ; Smyth EM; Fitzgerald GA Cox-2-Derived Prostacyclin Confers Atheroprotection on Female Mice. Science 2004, 306, 1954–1957. [Abstract] [Google Scholar]
16. Grosser T; Yu Y; Fitzgerald GA Emotion Recollected in Tranquility: Lessons Learned from the Cox-2 Saga. Annu. Rev. Med 2010, 61, 17–33. [Abstract] [Google Scholar]
17. Ganesh T Prostanoid Receptor EP2 as a Therapeutic Target. J. Med. Chem 2014, 57, 4454–4465. [Europe PMC free article] [Abstract] [Google Scholar]
18. Jiang J; Dingledine R Prostaglandin Receptor EP2 in the Crosshairs of Anti-Inflammation, Anti-Cancer, and Neuroprotection. Trends Pharmacol. Sci 2013, 34, 413–423. [Europe PMC free article] [Abstract] [Google Scholar]
19. 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. [Europe PMC free article] [Abstract] [Google Scholar]
20. 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. [Europe PMC free article] [Abstract] [Google Scholar]
21. Liang X; Wang Q; Hand T; Wu L; Breyer RM; Montine TJ; Andreasson K Deletion of the Prostaglandin E2 EP2 Receptor Reduces Oxidative Damage and Amyloid Burden in a Model of Alzheimer’s Disease. J. Neurosci 2005, 25, 10180–10187. [Europe PMC free article] [Abstract] [Google Scholar]
22. 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, 2–11. [Europe PMC free article] [Abstract] [Google Scholar]
23. 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. [Europe PMC free article] [Abstract] [Google Scholar]
24. 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. [Europe PMC free article] [Abstract] [Google Scholar]
25. Montine TJ; Milatovic D; Gupta RC; Valyi-Nagy T; Morrow JD; Breyer RM Neuronal Oxidative Damage from Activated Innate Immunity is EP2 Receptor-Dependent. J. Neurochem 2002, 83, 463–470. [Abstract] [Google Scholar]
26. 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. [Europe PMC free article] [Abstract] [Google Scholar]
27. 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. [Europe PMC free article] [Abstract] [Google Scholar]
28. Shie FS; Breyer RM; Montine TJ Microglia Lacking E Prostanoid Receptor Subtype 2 Have Enhanced Abeta Phagocytosis yet Lack Abeta-Activated Neurotoxicity. Am. J. Pathol 2005, 166, 1163–1172. [Europe PMC free article] [Abstract] [Google Scholar]
29. Forselles KJA; 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. [Europe PMC free article] [Abstract] [Google Scholar]
30. 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. [Europe PMC free article] [Abstract] [Google Scholar]
31. 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. [Europe PMC free article] [Abstract] [Google Scholar]
32. 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. [Abstract] [Google Scholar]
33. Ganesh T; Jiang J; Yang MS; Dingledine R Lead Optimization Studies of Cinnamic Amide EP2 Antagonists. J. Med. Chem 2014, 57, 4173–4184. [Europe PMC free article] [Abstract] [Google Scholar]
34. Ganesh T; Jiang J; Dingledine R Development of Second Generation EP2 Antagonists with High Selectivity. Eur. J. Med. Chem 2014, 82, 521–535. [Europe PMC free article] [Abstract] [Google Scholar]
35. Ganesh T; Banik A; Dingledine R; Wang W; Amaradhi R Peripherally Restricted, Highly Potent, Selective, Aqueous-Soluble EP2 Antagonist with Anti-Inflammatory Properties. Mol. Pharm 2018, 15, 5809–5817. [Europe PMC free article] [Abstract] [Google Scholar]
36. Salikov RF; Belyy AY; Khusnutdinova NS; Vakhitova YV; Tomilov YV Synthesis and Cytotoxic Properties of Tryptamine Derivatives. Bioorg. Med. Chem. Lett 2015, 25, 3597–3600. [Abstract] [Google Scholar]
37. Salikov RF; Trainov KP; Levina AA; Belousova IK; Medvedev MG; Tomilov YV Synthesis of Branched Tryptamines via the Domino Cloke-Stevens/Grandberg Rearrangement. J. Org. Chem 2017, 82, 790–795. [Abstract] [Google Scholar]
38. Shmatova OI; Shevchenko NE; Nenajdenko VG Fischer Reaction with 2-Perfluoroalkylated Cyclic Imines ? An Efficient Route to 2-Perfluoroalkyl-Substituted Tryptamines and Their Derivatives and Homologues. Eur. J. Org. Chem 2015, 6479–6488. [Google Scholar]
39. Shultz MD; Cao X; Chen CH; Cho YS; Davis NR; Eckman J; Fan J; Fekete A; Firestone B; Flynn J; Green J; Growney JD; Holmqvist M; Hsu M; Jansson D; Jiang L; Kwon P; Liu G; Lombardo F; Lu Q; Majumdar D; Meta C; Perez L; Pu M; Ramsey T; Remiszewski S; Skolnik S; Traebert M; Urban L; Uttamsingh V; Wang P; Whitebread S; Whitehead L; Yan-Neale Y; Yao YM; Zhou L; Atadja P Optimization of the in vitro Cardiac Safety of Hydroxamate-Based Histone Deacetylase Inhibitors. J. Med. Chem 2011, 54, 4752–4772. [Abstract] [Google Scholar]
40. Pennington LD; Moustakas DT The Necessary Nitrogen Atom: A Versatile High-Impact Design Element for Multiparameter Optimization. J. Med. Chem 2017, 60, 3552–3579. [Abstract] [Google Scholar]
41. Hitchcock SA; Pennington LD Structure-Brain Exposure Relationships. J. Med. Chem 2006, 49, 7559–7583. [Abstract] [Google Scholar]
42. Rankovic Z CNS Drug Design: Balancing Physicochemical Properties for Optimal Brain Exposure. J. Med. Chem 2015, 58, 2584–2608. [Abstract] [Google Scholar]
43. Wager TT; Chandrasekaran RY; Hou X; Troutman MD; Verhoest PR; Villalobos A; Will Y Defining Desirable Central Nervous System Drug Space through the Alignment of Molecular Properties, in vitro ADME, and Safety Attributes. ACS Chem. Neurosci 2010, 1, 420434. [Europe PMC free article] [Abstract] [Google Scholar]
44. Desai PV; Raub TJ; Blanco M-J How Hydrogen Bonds Impact P-Glycoprotein Transport and Permeability. Bioorg. Med. Chem. Lett 2012, 22, 6540–6548. [Abstract] [Google Scholar]
45. Herr RJ 5-Substituted-1h-Tetrazoles as Carboxylic Acid Isosteres: Medicinal Chemistry and Synthetic Methods. Bioorg. Med. Chem 2002, 10, 3379–3393. [Abstract] [Google Scholar]
46. Bevan CD; Lloyd RS A High-Throughput Screening Method for the Determination of Aqueous Drug Solubility Using Laser Nephelometry in Microtiter Plates. Anal. Chem 2000, 72, 1781–1787. [Abstract] [Google Scholar]
47. Di L; Fish PV; Mano T Bridging Solubility between Drug Discovery and Development. Drug Discov. Today 2012, 17, 486–495. [Abstract] [Google Scholar]
48. Baka E; Comer JE; Takacs-Novak K Study of Equilibrium Solubility Measurement by Saturation Shake-Flask Method Using Hydrochlorothiazide as Model Compound. J. Pharm. Biomed. Anal 2008, 46, 335–341. [Abstract] [Google Scholar]
49. Sugimoto Y; Narumiya S Prostaglandin E Receptors. J. Biol. Chem 2007, 282, 11613–11617. [Abstract] [Google Scholar]
50. Wager TT; Hou X; Verhoest PR; Villalobos A Moving Beyond Rules: The Development of a Central Nervous System Multiparameter Optimization (CNS MPO) Approach to Enable Alignment of Druglike Properties. ACS Chem. Neurosci 2010, 1, 435–449. [Europe PMC free article] [Abstract] [Google Scholar]
51. 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, 44169–44178. [Europe PMC free article] [Abstract] [Google Scholar]
52. Sheibanie AF; Khayrullina T; Safadi FF; Ganea D Prostaglandin E2 Exacerbates Collagen-Induced Arthritis in Mice through the Inflammatory Interleukin-23/Interleukin-17 Axis. Arthritis Rheumatol 2007, 56, 2608–2619. [Abstract] [Google Scholar]
53. Sheibanie AF; Yen JH; Khayrullina T; Emig F; Zhang M; Tuma R; Ganea D The Proinflammatory Effect of Prostaglandin E2 in Experimental Inflammatory Bowel Disease is Mediated through the Il-23-->Il-17 Axis. J. Immunol 2007, 178, 8138–8147. [Abstract] [Google Scholar]
54. Rojas A; Banik A; Chen D; Flood K; Ganesh T; Dingledine R Novel Microglia Cell Line Expressing the Human EP2 Receptor. ACS Chem. Neurosci 2019, 10, 4280–4292. [Abstract] [Google Scholar]
55. Quan Y; Jiang J; Dingledine R EP2 Receptor Signaling Pathways Regulate Classical Activation of Microglia. J. Biol. Chem 2013, 288, 9293–9302. [Europe PMC free article] [Abstract] [Google Scholar]
56. 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. [Europe PMC free article] [Abstract] [Google Scholar]
57. Salikov RF; Trainov KP; Belousova IK; Belyy AY; Fatkullina US; Mulyukova RV; Zainullina LF; Vakhitova YV; Tomilov YV Branching Tryptamines as a Tool to Tune Their Antiproliferative Activity. Eur. J. Med. Chem 2018, 144, 211–217. [Abstract] [Google Scholar]
58. Baggett AW; Cournia Z; Han MS; Patargias G; Glass AC; Liu S-Y; Nolen BJ Structural Characterization and Computer-Aided Optimization of a Small-Molecule Inhibitor of the Arp2/3 Complex, a Key Regulator of the Actin Cytoskeleton. ChemMedChem 2012, 7, 1286–1294. [Europe PMC free article] [Abstract] [Google Scholar]
59. Liang X-W; Liu C; Zhang W; You S-L Asymmetric Fluorinative Dearomatization of Tryptamine Derivatives. ChemComm 2017, 53, 5531–5534. [Abstract] [Google Scholar]
60. Shevchenko NE; Balenkova ES; Röschenthaler G-V; Nenajdenko VG Practical Synthesis of A-Perfluoroalkyl Cyclic Imines and Amines. Synthesis 2010, 120–126. [Google Scholar]
61. Shmatova OI; Shevchenko NE; Nenajdenko VG Fischer Reaction with 2-Perfluoroalkylated Cyclic Imines ― an Efficient Route to 2-Perfluoroalkyl-Substituted Tryptamines and Their Derivatives and Homologues. Eur. J. Org. Chem 2015, 6479–6488. [Google Scholar]
62. Shultz MD; Cao X; Chen CH; Cho YS; Davis NR; Eckman J; Fan J; Fekete A; Firestone B; Flynn J; Green J; Growney JD; Holmqvist M; Hsu M; Jansson D; Jiang L; Kwon P; Liu G; Lombardo F; Lu Q; Majumdar D; Meta C; Perez L; Pu M; Ramsey T; Remiszewski S; Skolnik S; Traebert M; Urban L; Uttamsingh V; Wang P; Whitebread S; Whitehead L; Yan-Neale Y; Yao Y-M; Zhou L; Atadja P Optimization of the in vitro Cardiac Safety of Hydroxamate-Based Histone Deacetylase Inhibitors. J. Med. Chem 2011, 54, 4752–4772. [Abstract] [Google Scholar]
63. Sum P-E; How DB; Sabatini JJ; Xiang JS; Ipek M; Feyfant E Preparation of Glutamate Derivatives as Aggrecanase Inhibitors WO2007008994A2, 2007.
64. Blake TD; Hamper BC; Huang W; Kiefer JR; Moon JB; Neal BE; Olson KL; Pelc MJ; Schweitzer BA; Thorarensen A; Trujillo JI; Turner SR Preparation of Nicotinamide Derivatives as H-Pgds (Hematopoietic Prostaglandin D Synthase) Inhibitors US20080146569A1, 2008.
65. Westaway SM; Thompson M; Rami HK; Stemp G; Trouw LS; Mitchell DJ; Seal JT; Medhurst SJ; Lappin SC; Biggs J; Wright J; Arpino S; Jerman JC; Cryan JE; Holland V; Winborn KY; Coleman T; Stevens AJ; Davis JB; Gunthorpe MJ Design and Synthesis of 6-Phenylnicotinamide Derivatives as Antagonists of Trpv1. Bioorg. Med. Chem. Lett 2008, 18, 5609–5613. [Abstract] [Google Scholar]
66. Bentzien JM; Boyer SJ; Burke J; Eldrup AB; Guo X; Huber JD; Kirrane TM; Soleymanzadeh F; Swinamer AD Pyrrolidinyl and Piperidinyl Compounds Useful as Nhe-1 Inhibitors and Their Preparation and Pharmaceutical Compositions WO2010005783A1, 2010.
67. Fandrick DR; Reinhardt D; Desrosiers J-N; Sanyal S; Fandrick KR; Ma S; Grinberg N; Lee H; Song JJ; Senanayake CH General and Rapid Pyrimidine Condensation by Addressing the Rate Limiting Aromatization. Org. Lett 2014, 16, 2834–2837. [Abstract] [Google Scholar]
68. Juby PF; Hudyma TW; Brown M; Essery JM; Partyka RA Antiallergy Agents. 2. 2-Phenyl-5-(1H-tetrazol-5-yl)pyrimidin-4(3H)-ones. J. Med. Chem 1982, 25, 1145–1150. [Abstract] [Google Scholar]
69. Ricci P; Kramer K; Cambeiro XC; Larrosa I Arene-metal Π-complexation as a Traceless Reactivity Enhancer for C-H Arylation. J. Am. Chem. Soc 2013, 135, 13258–13261. [Abstract] [Google Scholar]
70. Senaweera S; Weaver JD Dual C-F, C-H Functionalization via Photocatalysis: Access to Multifluorinated Biaryls. J. Am. Chem. Soc 2016, 138, 2520–2523. [Europe PMC free article] [Abstract] [Google Scholar]
71. Shen Y; Wu X-X; Chen S; Xia Y; Liang Y-M Synthesis of Polyfluoroarene-Substituted Benzofuran Derivatives via Cooperative Pd/Cu Catalysis. ChemComm 2018, 54, 2256–2259. [Abstract] [Google Scholar]
72. Hansen AH; Sergeev E; Bolognini D; Sprenger RR; Ekberg JH; Ejsing CS; McKenzie CJ; Rexen Ulven E; Milligan G; Ulven T Discovery of a Potent Thiazolidine Free Fatty Acid Receptor 2 Agonist with Favorable Pharmacokinetic Properties. J. Med. Chem 2018, 61, 9534–9550. [Abstract] [Google Scholar]
73. 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. [Europe PMC free article] [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/73703323
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/73703323

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1021/acs.jmedchem.9b01218

Supporting
Mentioning
Contrasting
3
18
0

Article citations


Go to all (11) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

Alzheimer&apos;s Drug Discovery Foundation (1)

NIA NIH HHS (1)

NINDS NIH HHS (4)

National Institute of Neurological Disorders and Stroke (2)

National Institute on Aging (1)

Ono Pharmaceutical (1)