European Journal of Medicinal Chemistry - 中国医药化工网...

12
Research paper Design and synthesis of novel xanthine derivatives as potent and selective A 2B adenosine receptor antagonists for the treatment of chronic inammatory airway diseases Sujay Basu * , Dinesh A. Barawkar, Vidya Ramdas, Meena Patel, Yogesh Waman, Anil Panmand, Santosh Kumar, Sachin Thorat, Minakshi Naykodi, Arnab Goswami, B. Srinivasa Reddy, Vandna Prasad, Sandhya Chaturvedi, Azfar Quraishi, Suraj Menon, Shalini Paliwal, Abhay Kulkarni, Vikas Karande, Indraneel Ghosh, Syed Mustafa, Siddhartha De, Vaibhav Jain, Ena Ray Banerjee, Sreekanth R. Rouduri, Venkata P. Palle, Anita Chugh, Kasim A. Mookhtiar ** Advinus Therapeutics Ltd., Drug Discovery Facility, Quantum Towers, Plot-9, Phase-I, Rajiv Gandhi Infotech Park, Hinjawadi, Pune 411057, India article info Article history: Received 15 November 2016 Received in revised form 1 April 2017 Accepted 7 April 2017 Available online 12 April 2017 Keywords: Adenosine cAMP Bioavailability Propargyl Heteroaryl Human liver microsomes abstract Adenosine induces bronchial hyperresponsiveness and inammation in asthmatics through activation of A 2B adenosine receptor (A 2B AdoR). Selective antagonists have been shown to attenuate airway reactivity and improve inammatory conditions in pre-clinical studies. Hence, the identication of novel, potent and selective A 2B AdoR antagonist may be benecial for the potential treatment of asthma and Chronic Obstructive Pulmonary Disease (COPD). Towards this effort, we explored several prop-2-ynylated C8-aryl or heteroaryl substitutions on xanthine chemotype and found that 1-prop-2-ynyl-1H-pyrazol-4-yl moiety was better tolerated at the C8 position. Compound 59, exhibited binding afnity (K i ) of 62 nM but was non-selective for A 2B AdoR over other AdoRs. Incorporation of substituted phenyl on the terminal acetylene increased the binding afnity (K i ) signicantly to <10 nM. Various substitutions on terminal phenyl group and different alkyl substitutions on N-1 and N-3 were explored to improve the potency, selectivity for A 2B AdoR and the solubility. In general, compounds with meta-substituted phenyl provided better selectivity for A 2B AdoR compared to that of para-substituted analogs. Substitutions such as basic amines like pyrrolidine, piperidine, piperazine or cycloalkyls with polar group were tried on terminal acetylene, keeping in mind the poor solubility of xanthine analogs in general. However, these sub- stitutions led to a decrease in afnity compared to compound 59. Subsequent SAR optimization resulted in identication of compound 46 with high human A 2B AdoR afnity (K i ¼ 13 nM), selectivity against other AdoR subtypes and with good pharmacokinetic properties. It was found to be a potent functional A 2B AdoR antagonist with a K i of 8 nM in cAMP assay in hA 2B -HEK293 cells and an IC 50 of 107 nM in IL6 assay in NIH-3T3 cells. Docking study was performed to rationalize the observed afnity data. Structure- activity relationship (SAR) studies also led to identication of compound 36 as a potent A 2B AdoR antagonist with K i of 1.8 nM in cAMP assay and good aqueous solubility of 529 mM at neutral pH. Compound 46 was further tested for in vivo efcacy and found to be efcacious in ovalbumin-induced allergic asthma model in mice. © 2017 Elsevier Masson SAS. All rights reserved. 1. Introduction Adenosine plays numerous important physiological roles via a family of four G-protein coupled receptors (GPCR), namely A 1 ,A 2A , A 2B and A 3 adenosine receptors (AdoRs) [1].A 1 and A 2A AdoRs are stimulated by low nanomolar adenosine concentrations, while * Corresponding author. ** Corresponding author. E-mail addresses: [email protected] (S. Basu), kasim.mookhtiar@advinus. com (K.A. Mookhtiar). Contents lists available at ScienceDirect European Journal of Medicinal Chemistry journal homepage: http://www.elsevier.com/locate/ejmech http://dx.doi.org/10.1016/j.ejmech.2017.04.014 0223-5234/© 2017 Elsevier Masson SAS. All rights reserved. European Journal of Medicinal Chemistry 134 (2017) 218e229

Transcript of European Journal of Medicinal Chemistry - 中国医药化工网...

Page 1: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

lable at ScienceDirect

European Journal of Medicinal Chemistry 134 (2017) 218e229

Contents lists avai

European Journal of Medicinal Chemistry

journal homepage: http: / /www.elsevier .com/locate/ejmech

Research paper

Design and synthesis of novel xanthine derivatives as potent andselective A2B adenosine receptor antagonists for the treatment ofchronic inflammatory airway diseases

Sujay Basu*, Dinesh A. Barawkar, Vidya Ramdas, Meena Patel, Yogesh Waman,Anil Panmand, Santosh Kumar, Sachin Thorat, Minakshi Naykodi, Arnab Goswami,B. Srinivasa Reddy, Vandna Prasad, Sandhya Chaturvedi, Azfar Quraishi, Suraj Menon,Shalini Paliwal, Abhay Kulkarni, Vikas Karande, Indraneel Ghosh, Syed Mustafa,Siddhartha De, Vaibhav Jain, Ena Ray Banerjee, Sreekanth R. Rouduri, Venkata P. Palle,Anita Chugh, Kasim A. Mookhtiar**

Advinus Therapeutics Ltd., Drug Discovery Facility, Quantum Towers, Plot-9, Phase-I, Rajiv Gandhi Infotech Park, Hinjawadi, Pune 411 057, India

a r t i c l e i n f o

Article history:Received 15 November 2016Received in revised form1 April 2017Accepted 7 April 2017Available online 12 April 2017

Keywords:AdenosinecAMPBioavailabilityPropargylHeteroarylHuman liver microsomes

* Corresponding author.** Corresponding author.

E-mail addresses: [email protected] (S. Basucom (K.A. Mookhtiar).

http://dx.doi.org/10.1016/j.ejmech.2017.04.0140223-5234/© 2017 Elsevier Masson SAS. All rights re

a b s t r a c t

Adenosine induces bronchial hyperresponsiveness and inflammation in asthmatics through activation ofA2B adenosine receptor (A2BAdoR). Selective antagonists have been shown to attenuate airway reactivityand improve inflammatory conditions in pre-clinical studies. Hence, the identification of novel, potentand selective A2BAdoR antagonist may be beneficial for the potential treatment of asthma and ChronicObstructive Pulmonary Disease (COPD). Towards this effort, we explored several prop-2-ynylated C8-arylor heteroaryl substitutions on xanthine chemotype and found that 1-prop-2-ynyl-1H-pyrazol-4-ylmoiety was better tolerated at the C8 position. Compound 59, exhibited binding affinity (Ki) of 62 nMbut was non-selective for A2BAdoR over other AdoRs. Incorporation of substituted phenyl on the terminalacetylene increased the binding affinity (Ki) significantly to <10 nM. Various substitutions on terminalphenyl group and different alkyl substitutions on N-1 and N-3 were explored to improve the potency,selectivity for A2BAdoR and the solubility. In general, compounds with meta-substituted phenyl providedbetter selectivity for A2BAdoR compared to that of para-substituted analogs. Substitutions such as basicamines like pyrrolidine, piperidine, piperazine or cycloalkyls with polar group were tried on terminalacetylene, keeping in mind the poor solubility of xanthine analogs in general. However, these sub-stitutions led to a decrease in affinity compared to compound 59. Subsequent SAR optimization resultedin identification of compound 46 with high human A2BAdoR affinity (Ki ¼ 13 nM), selectivity againstother AdoR subtypes and with good pharmacokinetic properties. It was found to be a potent functionalA2BAdoR antagonist with a Ki of 8 nM in cAMP assay in hA2B-HEK293 cells and an IC50 of 107 nM in IL6assay in NIH-3T3 cells. Docking study was performed to rationalize the observed affinity data. Structure-activity relationship (SAR) studies also led to identification of compound 36 as a potent A2BAdoRantagonist with Ki of 1.8 nM in cAMP assay and good aqueous solubility of 529 mM at neutral pH.Compound 46 was further tested for in vivo efficacy and found to be efficacious in ovalbumin-inducedallergic asthma model in mice.

© 2017 Elsevier Masson SAS. All rights reserved.

), kasim.mookhtiar@advinus.

served.

1. Introduction

Adenosine plays numerous important physiological roles via afamily of four G-protein coupled receptors (GPCR), namely A1, A2A,A2B and A3 adenosine receptors (AdoRs) [1]. A1 and A2AAdoRs arestimulated by low nanomolar adenosine concentrations, while

Page 2: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229 219

micromolar concentrations of adenosine are required for the acti-vation of A2B and A3AdoRs [2]. The low affinity AdoRs (A2B and A3)are activated only under stressful conditions wherein adenosinelevels are elevated to micromolar concentrations. Increased levelsof adenosine have been reported under pathophysiological condi-tions, such as hypoxic or ischemic conditions, after massive celldeath, or as a consequence of inflammatory processes [3,4]. Rodentand human A2BAdoRs share 86e87% amino acid sequence homol-ogy [5]. The A2BAdoRs show a ubiquitous distribution, with highestlevels observed in the cecum, colon, bladder, mast cells, hemato-poietic cells, and lung, while lower levels are detected in otherorgans, such as brain and liver [6].

Apart from the differences in affinity to adenosine, the receptorsalso differ in their signaling mechanism. A1 and A3AdoRs mediateinhibition of adenylate cyclase through Gi coupled pathway,whereas A2B and A2A stimulate adenylate cyclase activity via Gscoupling and result in increased intracellular cAMP levels [5]. Inaddition, coupling of A2A and A2BAdoRs to phospholipase C via Gq,resulting in mobilization of intracellular calcium and directcoupling to calcium channels (stimulation of calcium influx), havealso been reported [7e9].

Adenosine has been shown to cause mast cell degranulation[10], vasodilation [11], chloride secretion in epithelial cells [12,13],growth inhibition of smooth muscle cells [14], and stimulation ofglucose production in rat hepatocytes [15] and stimulate VEGFsecretion [16] via activation of A2BAdoR. A2BAdoR antagonists,therefore, can potentially be used as therapeutic agents for thetreatment of asthma and chronic obstructive pulmonary disease[4,17e19] type II diabetes [15], cystic fibrosis [12], inflammatorybowel disease [13] and cancer [20].

Adenosine induces bronchoconstriction in asthmatic patientsbut not in normal people [21,22]. In addition, adenosine levels areelevated in the bronchoalveolar lavage fluid and exhaled breathcondensates of asthmatic patients [23,24] and adenosine deami-nase deficient mice show evidence of bronchioli remodelling andemphysema-like lesions [25,26]. The mechanism of this adenosine-mediated damage lies with the activation of A2BAdoR on humanmast cells, which in turn leads to mast cell degranulation andrelease of inflammatory cytokines IL-4, IL-8 and IL-13 [27]. A2BAdoRactivation also increases the expression and release of IL-6 and TNF-a in bronchial epithelial cells [28] and this indicates that A2BAdoRplay a key role in the pathogenesis of inflammatory lung disease.There is also evidence that A2BAdoR is the predominant AdoRexpressed in bronchial smooth muscle cells, and its activation in-creases the expression and release of IL-6 and monocytic chemo-tactic peptide-1 (MCP-1) [29] and is implicated in airwayremodeling. In human lung fibroblasts, activation of A2BAdoR in-duces the release of IL-6 and differentiation of fibroblasts to myo-fibroblasts [30].

On account of all the evidence that implicates A2BAdoR activa-tion in inflammatory lung diseases such as asthma, COPD andfibrosis, a lot of research has been carried out with the goal ofidentifying potent and selective A2BAdoR antagonists to provide anovel approach to the management and treatment of asthma andCOPD.

The alkylxanthines theophylline 1 and enprofylline 2 (Fig. 1), areweak, non-selective AdoR antagonists used therapeutically for thetreatment of asthma [31,32]. The bronchodilating effect of 1 and 2has been attributed to a selective antagonism of A2BAdoR, whichprompted several groups to design and test a large number ofxanthine derivatives in the search for new, more potent and se-lective A2B AdoR antagonists. During the past decade, severalstructure-activity relationship studies of A2BAdoR antagonists havebeen published [33e37]. Though several potent and selectiveA2BAdoR antagonists like MRS-1754 (3), MRE2029-F20 (4) have

been reported, these compounds were highly lipophilic, possessingvery low solubility and poor oral bioavailability [38,39].

A potent and selective A2BAdoR antagonist CVT-6883 (5) [36,40]entered into clinics and later discontinued from Phase I clinicaltrials. It has shown inhibition of airway inflammation and airwayreactivity induced by allergen or AMP in mouse model of allergicasthma [41]. In addition, compound 5 attenuated pulmonaryinflammation, fibrosis, and alveolar airspace enlargement andreduced elevations of pro-inflammatory cytokines and chemokines,as well as mediators of fibrosis [42].

In our earlier communication we had reported a new series ofxanthine derivatives [43]. The objective of present study was toidentify and develop novel, potent and selective A2BAdoR antago-nists combined with good aqueous solubility and improving in vivohalf life. As we were interested in synthesizing and testing novelxanthine derivatives, first we explored different propargylated aryland heteroaryl groups at C8 position of xanthine and identified 8-(1-prop-2-ynyl-1H-pyrazol-4-yl)-xanthine as a potential core forSAR studies. Herein, we discuss the structure-activity relationship(SAR) studies carried out on 8-(1-prop-2-ynyl-1H-pyrazol-4-yl)-xanthine towards identification of novel, potent and selective A2B

AdoR antagonists with good oral bioavailability.

2. Results and discussion

2.1. Chemistry

The key intermediates 8-phenyl-, 8-isoxazolyl-, 8-(1H-pyrazol-3-yl)- and 8-(1H-pyrazol-4-yl)-1,3-dipropyl xanthines were pre-pared by classical method starting from 1,3-dipropyl-5,6-diaminouracil 6 (Scheme 1) [36]. The diamino uracil 6 wascoupled with phenyl carboxylic acid 7a, isoxazole carboxylic acid7b, pyrazole-3- or pyrazole-4-carboxylic acids 7c or 7d, using N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (EDCI)in methanol at room temperature to obtain corresponding car-boxamide derivatives 8a-d. Subsequent ring closure of these de-rivatives was performed by refluxing 8a-d with 10% sodiumhydroxide in methanol to afford 8-substituted xanthine derivatives9a-d. The N-7 position of 9a-d was protected with 2-(trime-thylsilyl)ethoxymethylchloride (SEM-Cl) using potassium carbon-ate in dimethylformamide to furnish 10a-dwhich on debenzylationusing hydrogen and 10% Pd/C provided the key intermediates 11a-cwith “O” and 11d with “N” as a handle for further derivatization[36]. These compounds were alkylated with various substituted (3-bromo-prop-1-ynyl)-benzene derivatives using potassium car-bonate in acetone at reflux temperature to provide the silyl pro-tected xanthine derivatives which on silyl deprotectionwith 2NHClfurnished the final compounds 12e40 in good yields (28e99%)Scheme 1.

In a similar fashion, various N-1, N-3 differentially substitutedxanthine derivatives were prepared by starting from 1-ethyl-3-propyl-5,6-diaminouracil 41a or 1H-3-propyl-5,6-diaminouracil41b [44]. The key intermediates, 3-ethyl-1-propyl-8-(1H-pyrazol-4-yl)-xanthine 42a or 1-propyl-8-(1H-pyrazol-4-yl)-xanthine 42bwere prepared (Scheme 2) by following the synthetic sequence asdescribed in Scheme 1. The alkylation of 42a or b with variousphenyl substituted propargyl bromides using standard alkylationconditions followed by N-7 silyl deprotection with 2N HCl fur-nished 43e56 in good yields (11e85%).

Propargyl and 2-butyne substituted 8-(1H-pyrazol-4-yl)-xanthine derivatives 59 and 60 were prepared (Scheme 3) byalkylation of intermediate 11d with corresponding bromo com-pounds using potassium carbonate in acetone at reflux condition,followed by N-7 deprotection of the resulting silyl protected com-pounds 57 and 58 with 2N HCl in ethanol at reflux condition.

Page 3: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

N

N N

NH

O

ON

N

CF3

NH

OO

CN

N

N N

NH

O

O

N

N N

NH

O

O

NH

N N

NH

O

O

N

N N

NH

O

O

NN

ONH

OO

O

1 Theophylline 2 Enprofylline 3 MRS-1754

4 MRE2029F20 5 CVT-6883

Fig. 1. Representative structures of xanthine based A2BAdoR antagonists.

12-16 17-21

22-23 24-40

Ar/Het = -Phenyl, Isoxazole, Pyrazole

a b

c d

6 7a-d 8a-d 9a-d

10a-d 11a-d

e, f

12-40

Scheme 1. Synthesis of compounds 12e40 a. aReagents and conditions: (a) EDCI, MeOH, r.t.; (b) 10% NaOH, MeOH, reflux; (c) SEM-Cl, K2CO3, DMF, r.t.; (d H2, 10% Pd/C, MeOH:DCM(2:1), r.t.; (e) K2CO3, acetone, 50 �C; (f) 2N HCl, ethanol, 80 �C.

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229220

Compounds substituted with basic amino moiety on terminalacetylene 61e66 were prepared (Scheme 4) by treating the 1,3-dipropyl-8-(1-prop-2-ynyl-1H-pyrazol-4-yl)-xanthine 57 withformaldehyde solution and acyclic or heterocyclic amines in pres-ence of CuI in dimethylsulfoxide at room temperature followed byN-7 silyl deprotection. The compounds, 8-{1-[3-(1-Hydroxy-

cycloalkyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-xanthine derivatives,67 and 68 were also obtained in a similar fashion. The reaction ofintermediate 57 with cyclopentanone or cyclohexanone in thepresence of lithiumhexamethyl disilazide in tetrahydrofuran at lowtemperatures followed by N-7 silyl deprotection furnished thecompounds 67 and 68, Scheme 4.

Page 4: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

41a, R1=Et41b, R1=H

42a, R1=Et42b, R1=SEM

43-49, R1=Et50-56, R1=H

Scheme 1

a, b

Scheme 2. Synthesis of compounds 43e49 and 50e56 a. aReagents and conditions: (a)K2CO3, acetone, 50 �C; (b) 2N HCl, ethanol, 80 �C.

a b

11d 57, R2=H58, R2=Me

R2=H; Me 59, R2=H60, R2=Me

Scheme 3. Synthesis of compounds 59e60 a. aReagents and conditions: (a) K2CO3, acetone, 50 �C; (b) 2N HCl, ethanol, 80 �C.

i) a or bii) c

57 61-68

Scheme 4. Synthesis of compounds 61e68 a. aReagents and conditions: (a) 37% HCHOsolution, amine, CuI, DMSO, r.t.; (b) LHMDS, cyclopentanone or cyclohexanone,THF, �78 to 20 �C; (c) 2N HCl, ethanol, 80 �C.

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229 221

2.2. Biological activity

As we were interested in synthesizing and testing novel de-rivatives of xanthinewith propargylated aryl and heteroaryl groupsat C8 position, we first explored different aryl and heteroarylgroups at the C8 position of xanthine. Herein, we discuss thestructure-activity relationship (SAR) of compounds 12e25 with

phenyl, isoxazolyl, 3-pyrazolyl and 1H-4-pyrazolyl substitution atthe C8 position (Table 1). Compounds 12e16 having phenyl, pyridyl(structure not shown), and compounds 22, 23 having 3-pyrazolylsubstitutions at C8 position were found to be inactive or showedlow binding affinity for A2BAdoR, indicating that phenyl, pyridyland 3-pyzolyl are not suitable at C8 of xanthine. These compoundsalso showed poor solubility. When isoxazolyl was placed at C8 ofxanthine, compounds 17 and 18 with small substitution like m-Fand p-F on terminal phenyl ring showed moderate binding affinityof 55 and 15 nM, respectively, for A2BAdoR. Compounds 19e21withlarger substituents such as trifluoroalkyl or trifluoroalkoxy substi-tution on terminal phenyl ring were inactive or showed low affinityfor A2BAdoR. 4-Pyrazolyl, between C8 of xanthine and m-F and p-F(24 and 25) substituted terminal phenyl ring had high binding af-finity of 6.7 and 5.9 nM, respectively, for A2BAdoR (Table 1). Basedon the binding affinity of compounds 12e25 for A2BAdoR, it wasconcluded that 4-pyrazolyl ring is the best structural requirementgroup at the C8 position. Hence, 8-(1-propargyl-4-pyrazolyl)-1,3-disubstituted xanthine derivative was selected for further SARstudies to identify a potent, selective and soluble A2BAdoRantagonist.

The binding selectivity of 8-(1-propargyl-4-pyrazolyl)-xanthineanalogs 24 and 25 over other AdoR subtypes A1AdoR and A2AAdoR

is shown in Table 2. These compounds were not very selective forA2BAdoR. Therefore, to increase the selectivity, we explored varioussubstitutions on the terminal phenyl ring, including electrondonating groups such as methyl and methoxy, electron with-drawing groups such as trifluoromethyl, trifluoromethyloxy, andcyano and polar carboxylic acids and hydroxyl groups with theintention to improve solubility. The data for these compounds istabulated in Table 2. The binding affinity of compounds 24e40clearly indicates that substituted phenyl groups are well toleratedon the acetylene moiety, majority of compounds showing highbinding affinity Ki of <10 nM for A2BAdoR, irrespective of the natureof substitution (e.g. electron donating or withdrawing group), andirrespective of the position of substitution (ortho, meta or para onphenyl ring), as outlined in Table 2. Compound 26, with orthosubstitution of trifluoromethyl, was found to be relatively lesspotent (Ki ¼ 29 nM), and in general, not much difference wasobserved between meta and para substitutions in terms of bindingaffinity for A2BAdoR. However, majority of compounds with parasubstitution showed modest selectivity over A2AAdoR with low orno selectivity over A1AdoR (except compounds 32 and 37). Com-pounds 25, 28, 30, and 40 exhibited similar binding affinity for A2Band A1AdoR. Majority of compounds with meta substitutions wereselective for A2BAdoR over both A1 and A2AAdoRs with Majority of

Page 5: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

Table 1Binding Affinity of Xanthine Derivatives with C8 variation for A2BAdoR.

Compd R hA2B Ki (nM)a

12 m-F >1 mM13 p-F NRb

14 p-CF3 NRb

15 m-OCF3 NRb

16 p-OCF3 >600

17 m-F 5518 p-F 1519 m-CF3 >60020 p-CF3 NR21 m-OCF3 >600

22 m-CF3 NR23 p-OCF3 NR

24 m-F 6.725 p-F 5.9

NR-no response.a Binding affinity for hA2BAdoRwas determined by using HEK-A2B cells with [3H]-MRS-1754 as the radioligand, All data points were evaluated in triplicates. Each compound

was evaluated at 8 concentrations with each data point in triplicates for Ki determination.b These compounds showed poor solubility.

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229222

the compounds exhibited highly selective over A3AdoR (�30% in-hibition at 1 mM) except for compound 38. Among these com-pounds, 29, 36, and 39were identified as the best compounds withhigh affinity and selectivity for A2BAdoR. It is interesting to notethat polar substitution like carboxylic acid and ester groups 36e40were well tolerated in terms of potency for A2BAdoR and selectivityover other AdoRs. Also, compound 36 with m-carboxylic acidsubstitution endowed with better aqueous solubility of 529 mM atpH 7.4.

To further improve the selectivity over other AdoR subtypes, wehave explored different substitutions at N-3 position [44]. Table 3shows follow-up SAR where the 4-pyrazolyl at C8 and propyl atN-1 side chain was fixed and the N-3 side chain was varied withethyl (43e49) or H (50e56). Unexpectedly, compounds 48, 49 and56 having carboxylic acid substitution showed lower binding af-finity for A2BAdoR compared to the corresponding 1,3-dipropylderivative 36 and 40. Consistent with dipropyl derivatives, evenin the case of 1-propyl-3-ethyl-xanthine derivatives, all parasubstituted compounds 44, 45, 47, 51, 53 and 55 exhibited modestselectivity over A2AAdoR and low selectivity over A1AdoR, whereasall the meta substituted compounds 46, 52, and 54 had betterselectivity over both the receptors. An exception was observed form-fluoro substituted compounds 43 and 50. Poor selectivityobserved in the case of compounds 43 and 50 could be due to the

presence of a smaller group like fluoro. All these compounds(43e56) retained high selectivity over A3AdoR (�30% inhibition at1 mM).

Compared to meta trifluoromethoxy phenyl substituteddipropyl xanthine derivative 29, corresponding 1-propyl-3-ethyl-xanthine derivative 46 had improved selectivity as shown inTable 3. N1-monopropylated xanthine derivatives 52 and 54 hadbetter binding affinity as well as selectivity than corresponding 1,3-dipropyl or 1-propyl-3-ethyl xanthine derivatives. However thesecompounds suffered from poor solubility issues.

In parallel, we pay attention towards replacement of highlylipophilic terminal aryl ring with the intension to achieve bettersolubility for the insoluble xanthine derivatives in combinationwith potency and selectivity. We replaced terminal aryl ring withsmall lipophilic group like H or methyl and explored polar sub-stitutions, e.g., alcohol or basic functions on the terminal acetylenemoiety as in compounds 61e68. First, we evaluated 8-(1-prop-2-ynyl-1H-pyrazol-4-yl)-1,3-dipropyl xanthine 59 and 8-(1-But-2-ynyl-1H-pyrazol-4-yl)-1,3-dipropyl xanthine 60, where terminalphenyl was replaced by “H” and methyl respectively for bindingaffinity and selectivity for A2BAdoR. These compounds had modestaffinity of Ki ¼ 62 and 23 nM, respectively for A2BAdoR but werepoorly selective (Table 4). This indicated clear need of optimizationof terminal substituent for potency as well as selectivity.

Page 6: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

Table 2Binding affinity of xanthine derivatives with different substitution on terminal phenyl for A2B, A1, A2A and A3AdoRs.

Compd R Ki (nM)hA2B

a% Inhibition at 0.1 mM % Inhibition at 1 mM

hA1a hA2A

a hA3a

24 m-F 6.7 87 64 025 p-F 5.9 105 66 1926 o-CF3 29 52 29 2527 m-CF3 4.9 43 64 3228 p-CF3 1.5 84 30 1229 m-OCF3 3.6 39 27 630 p-OCF3 5.5 92 51 3731 m-CN 3.7 52 84 032 p-CN 14 14 29 1433 m-CH3 5 57 55 934 p-CH3 6.5 70 33 035 m-OCH3 6.3 57 74 2136 m-CO2H 5.3 20 32 037 p-CO2H 17.6 19 44 2538 o-CO2Me 12 37 41 4339 m-CO2Et 7.8 29 29 040 p-CO2Et 3.5 80 19 27

a Binding affinities were determined by usingmembrane preparations fromHEK-293 cells overexpressing the relevant human AdoR isoform. All data points were evaluatedin triplicates. Each compound was evaluated at 8 concentrations with each data point in triplicates for Ki determination [45].

Table 3Binding affinity of N-1, N-3 differentially substituted xanthine derivatives for A2B, A1, A2A and A3AdoRs.

Compd R Ki (nM)hA2B

a% Inhibition at 0.1 mM % Inhibition at 1 mM

hA1a hA2A

a hA3a

43 m-F 7.4 73 58 2344 p-F 7.3 92 56 3345 p-CF3 12 66 21 1446 m-OCF3 13 17 37 047 p-OCF3 9.9 84 49 3248 m-CO2H 190 NT NT NT49 p-CO2H 130 NT NT NT

50 m-F 3 53 60 051 p-F 1.6 77 55 052 m-CF3 2.2 26 39 053 p-CF3 1.3 89 39 1554 m-OCF3 2.8 14 33 055 p-OCF3 1.6 88 46 1756 m-CO2H 48 NT NT NT

NT-not tested.a Binding affinities were determined by usingmembrane preparations fromHEK-293 cells overexpressing the relevant human AdoR isoform. All data points were evaluated

in triplicates. Each compound was evaluated at 8 concentrations with each data point in triplicates for Ki determination [45].

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229 223

Compounds 61e65 with substitutions like diethylamine, pyrroli-dine, piperidine, morpholine and N-methyl piperazine showed low

affinity for A2BAdoR and were non-selective over other AdoR sub-types (Table 4). However, compound 66 substituted with 4-(3-

Page 7: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

Table 4Binding affinity of xanthine derivatives with different substitution on acetylene for A2B, A1, A2A and A3AdoRs.

Compd R Ki (nM)hA2B

a% Inhibition at 0.1 mM % Inhibition at 1 mM

hA1a hA2A

a hA3a

59 H 62 56 69 3260 -CH3 23 42 60 NT61 200 71 59 69

62 120 66 67 59

63 140 64 74 73

64 330 28 39 51

65 83 55 73 79

66 12 69 10 31

67 79 NT NT NT

68 100 NT NT NT

NT-not tested.a Binding affinities were determined by using membrane preparations from HEK-293 cells overexpressing the relevant human AdoR isoform. All data points were evaluated

in triplicates. Each compound was evaluated at 8 concentrations with each data point in triplicates for Ki determination [45].

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229224

trifluoromethyl-phenyl)-piperazine showed high affinity(Ki ¼ 12 nM) for A2BAdoR, although it had only modest selectivityover A1AdoR. Compounds 67 and 68 with tertiary alcohol functionalso were of low affinity towards A2BAdoR.

In terms of affinity and selectivity, clear SAR was demonstratedwith focused optimization of 8-(1-prop-2-ynyl-1H-pyrazol-4-yl)-1,3-dipropyl xanthine analogs. Compoundswith substituted phenylon terminal propyne group showed high affinity compared to othernon-aryl substitutions. Low or no selectivity over A1 and A2A AdoRs,as observed with most of the para-substituted compounds andcompounds with smaller groups like fluoro (irrespective of sub-stitution at N-3 position), could be attributed to the presence ofrigid propargyl group between C8-pyrazolyl ring and terminalphenyl ring, which makes the right hand side of these compoundsclose to linear structures in these cases. Bringing a bulkier substi-tution at the meta position on the phenyl ring improved theselectivity for A2BAdoR over other AdoR subtypes.

Among all compounds, compounds 29, 36 and 46 were identi-fied as the best in terms of binding affinity, selectivity and hencewere taken forward for detailed profiling. Functional potency of

these compounds was determined in hA2BHEK-293 cells usingcAMP assay and in NIH-3T3 cells using IL-6 assay. Compounds 29,36 and 46 were found to be potent A2BAdoR antagonists andinhibited 50-N-ethylcarboxamidoadenosine (NECA)-induced in-crease in cAMP with Ki of 8.8, 1.8 and 8 nM, respectively, in hA2B-HEK-293 cells. The compounds 29, 36 and 46 also inhibited NECAinduced IL-6 release in NIH-3T3 cells with IC50 of 90, 2000 and107 nM, respectively.

2.3. Drug metabolism and pharmacokinetic studies

Based on affinity, potency and selectivity compound 29, 36 and46 were progressed for drug metabolism and pharmacokinetics(DMPK) evaluation and summarized in Table 5 [43]. All the threecompounds were metabolically stable in mouse, rat and humanliver microsomes and improved solubility. When dosed orally tomice at 10 mg/kg, compound 46 showed better pharmacokineticscompared to compound 29, with Cmax of 13 mM, systemic exposureAUC(0-t) of 60 mM h, t1/2 of 6 h and oral bioavailability of 76%,Compound 36 displayed poor systemic exposure with high

Page 8: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

Table 5Oral pharmacokinetic profile of compounds 29, 36 and 46 in C57BL/6J mice at 10mg/kg.a

Compd Cmax (mM) AUC(0-t) (mM.h) t1/2 (h) %F Solubility (mM)b

29 6.6 67 5 42 <636 1.9 2.2 1.8 10 52946 13 60 6.1 76 21

a PO formulation for 29 and 46: DMAC (10%), CrEL (10%), PEG300 (10%), MQwater(qs), pH ~9; for 36: DMSO(10%), CrEL (10%), PEG300 (10%), MQ water (qs), pH ~7.

b Aqueous solubility at pH 7.4.

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229 225

clearance in mice when dosed orally at 10 mg/kg dose, and wasfound to be a substrate for Pgp (B-A/A-B ¼ 12). Furthermore,compound 46 did not show any CYP liability (IC50 > 10 mM), cyto-toxicity inMDCK and HepG2 cells (IC50> 100 mM), or any significanthERG inhibition (IC50 > 25 mM).

2.4. Molecular docking analysis

As compound 46 showed good binding affinity, selectivity andacceptable pharmacokinetic properties, we were interested to un-derstand the binding mode of this compound (46) in conjunctionwith reference compound 5. Molecular docking analysis revealedthat both the compounds had similar binding mode with goodoverlay and forming same intermolecular interactions with theA2BAdoR binding pocket residues formed by trasnsmembrane re-gion TM3, TM5, TM6 and TM7 and extracellular loop ECL2 (Fig. 2a).Moreover, both the compounds showed comparable docking score(Gold Chemscore DG was �26.03 and �26.88 for compounds 5 and46, respectively). It was identified that compound 46 is an extendedanalog of 5, where acetylene linker of the former is mimicking thenorthern phenyl ring of the later (Fig. 2a). Both the compoundsxanthine ring showed hydrogen bonding with a key residue

Fig. 2. (a) Overlay of docking poses of the reference compound 5 (magenta) and the synthesi(cyan cartoon representation). (b) 2D interaction diagram of compound 46 (hydrophobic rereferences to colour in this figure legend, the reader is referred to the web version of this

Asn254 (TM6) and <pi>�<pi>; interaction with Phe173 (ECL2). N-1 ethyl and N-3 propyl of the compounds accommodate well in thehydrophobic pocket. Northern phenyl ring of the compoundsformed hydrophobic interaction with the ECL2 residue Leu172,whereas CF3 group exposed in the bulk solvent. 2D interaction di-agram of compound 46 is illustrated in Fig. 2b. This analysis dem-onstrates the important molecular recognition interactionsrequired for the A2BAdoR activity.

2.5. In vivo efficacy study

Compound 46, when evaluated for in vivo efficacy (Fig. 3),showed reduction of increased inflammatory cells in ovalbumin-induced allergy model in mice, and significantly inhibited NECA-induced airway hyper-responsiveness [43].

3. Conclusion

A focused SAR study on 8-(1-prop-2-ynyl-1H-pyrazol-4-yl)-xanthine derivatives led to the identification of novel, potent andselective A2BAdoR antagonists. Compound 46, 3-ethyl-1-propyl-8-{1-[3-(3-trifluoromethoxy-phenyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-xanthine emerged as the best compound in terms of affinity andselectivity over other subtypes for A2BAdoR, solubility and PK. SARstudy also led to identification of potent A2BAdoR antagonist 36with good aqueous solubility. Compound 46 showed high bindingaffinity (Ki¼ 13 nM) and great selectivity over other AdoR subtypes.It was also potent with Ki of 8 nM in cAMP assay and IC50 107 nM inthe IL-6 assay, and showed good PK profile. Furthermore, com-pound 46 showed significant in vivo efficacy in the ovalbumin-induced allergic asthma model in mice. Further developmentaround this series is in progress andwill be reported in a due courseof time.

zed novel active compound 46 (yellow) in the homology modeled A2BAdoR binding sitesidues are shown in green color and polar residues in pink). (For interpretation of thearticle.)

Page 9: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229226

4. Experimental section

4.1. General

Commercial chemicals and solvents were of reagent grade andwere used without further purification. Anhydrous solvents wereused without further drying. The following abbreviations are usedfor reagent and solvents: DCM-dichloromethane; DMF-dimethylformamide; DMSO-dimethyl sulfoxide; EtOAc-ethyl acetate; EtOH-Ethanol; andMeOH-methanol. Globe chemie silica gel (100e200 or230e400 mesh) was used for column chromatography. Analtechthin layer chromatography plates (20� 20 cm, 2000 mm)were usedfor preparative thin layer chromatography. Proton NMR (1H NMR)spectra were recorded on a Varian 400 spectrometer (400 MHz).Solutions were typically prepared in either deuterated dimethylsulfoxide (DMSO-d6), deuterated methanol (CD3OD) or deuteratedchloroform (CDCl3). Chemical shifts are reported in d units (partsper million) downfield from tetramethylsilane and are assigned assinglet (s), doublet (d), doublet of doublet (dd), triplet (t), quartet(q) and multiplets (m), and broad (br). Coupling constants (J) arereported in Hertz (Hz). Mass spectra (MS) were recorded on Agilent6110. HPLC were recorded on Agilent RRLC using Eclipse XOB-C18(250 � 4.6) mm Su column and 0.05% fumaric acid (aq.) andacetonitrile as mobile phase with flow rate of 1 ml/min at 30 �C forrun time of 17 min and the HPLC purity is � 95% unless otherwisestated.

4.2. Synthesis

Preparation of 1,3-Dipropyl-8-{1-[3-(3-trifluoromethoxy-phenyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-3,7-dihydro-purine-2,6-dione (29).

Preparation of 3-(3-Trifluoromethoxy-phenyl)-prop-2-yn-1-ol: A

Fig. 3. Effect of Compound 46 on A) NECA-induced airway hyper-responsiveness and B) totpresented as mean ± SEM. *significantly different compared to Vehicle treated group.

mixture of propargyl alcohol (1.0 g, 18 mmol), 1-Iodo-3-trifluoromethoxy-benzene (5.14 g, 18 mmol), copper iodide(0.342 g,1.8mmol), dichlorobis (triphenylphosphine) palladium (II)(0.632 g, 0.9 mmol), diethylamine (30 ml) was degassed for 10min.and stirred for 20 h at 25-25 �C. Excess of diethyl amine wasdistilled off under vacuum. The residue was diluted with water(50 ml) and extracted with ethyl acetate (3 � 50 ml). The organiclayer was washed with brine solution and dried over Na2SO4. Thesolvent was evaporated and the crude product was purified bycolumn chromatography (10% Ethyl acetate in hexane) to obtainpure 3-(3-Trifluoromethoxy-phenyl)-prop-2-yn-1-ol (3.8 g, 100%).1HNMR (CDCl3): d 1.71 (t, J ¼ 6 Hz, 1H); 4.52 (d, J ¼ 6 Hz, 2H); 7.20(d, J ¼ 7.2 Hz, 1H); 7.28e7.39 (m, 3H).

Preparation of 1-(3-Bromo-prop-1-ynyl)-3-trifluoromethoxy-ben-zene: 3-(3-Trifluoromethoxy-phenyl)-prop-2-yn-1-ol (3.9 g,18mmol) was taken in DCM (50ml). The reactionmixturewas thencooled to -5-0 �C. Tribromo phosphine (1 ml, 11 mmol) was addedslowly at -50 �C and stirred at the same temperature for 3 h. Thereaction mixture was quenched with saturated NaHCO3 solutionand the organic layer was separated. The aqueous layer wasextracted with DCM (2 � 40 ml). The combined organic layer waswashed with saturated brine solution, dried over Na2SO4, andevaporated under vacuum to get 1-(3-Bromo-prop-1-ynyl)-3-trifluoromethoxy-benzene (2.97 g, 60%). 1HNMR (CDCl3): d 4.12 (s,2H); 7.20 (d, J ¼ 6.8 Hz, 1H); 7.30 (s, 1H); 7.34e7.39 (m, 2H); MS (m/z) (Mþþ1): 279.3.

Preparation of 1, 3-Dipropyl-8-{1-[3-(3-trifluoromethoxy-phenyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-7-(2-trimethylsilanyl-ethoxymethyl)-3, 7-dihydro-purine-2, 6-dione: A mixture of 1, 3-Dipropyl-8-(1H-pyrazol-4-yl)-7-(2-trimethylsilanyl-ethoxymethyl)-3,7-dihydro-purine-2,6-dione 11d36 (1.0 g, 2.3 mmol), 1-(3-Bromo-prop-1-ynyl)-3-trifluoromethoxy-benzene (0.829 g, 3.0 mmol), K2CO3(0.731mg, 5.3mmol) and acetone (50ml) were refluxed at 80 �C for5 h. The reaction mixture was cooled to room temperature and the

al cell count in BALf (bronchoalveolar lavage fluid) in Ovalbumin treated mice. Data is

Page 10: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229 227

solid K2CO3 was filtered off and evaporated the organic solventunder vacuum. The residue obtained was purified by columnchromatography (2%methanol in dichloromethane) to get pure 1,3-Dipropyl-8-{1-[3-(3-trifluoromethoxy-phenyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-7-(2-trimethylsilanyl-ethoxymethyl)-3,7-dihydro-purine-2,6-dione (1.5 g, 100%). 1HNMR (DMSO-d6): d �0.01 (s, 9H);0.94e1.03 (m, 8H); 1.68e1.73 (m, 2H); 1.80e1.87 (m, 2H); 3.83 (t,J ¼ 8 Hz, 2H); 4.01 (t, J ¼ 7.6 Hz, 2H); 4.13 (t, J ¼ 7.2 Hz, 2H); 5.26 (s,2H); 5.81 (s, 2H); 7.35e7.42 (m, 4H); 8.19 (s, 1H); 8.40 (s, 1H); MS(m/z) (Mþþ1): 631.4.

Preparation of 1,3-Dipropyl-8-{1-[3-(3-trifluoromethoxy-phenyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-3,7-dihydro-purine-2,6-dione: 1,3-Dipropyl-8-{1-[3-(3-trifluoromethoxy-phenyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-7-(2-trimethylsilanyl-ethoxymethyl)-3,7-dihydro-purine-2,6-dione (1.4 g, 2.2 mmol) in EtOH (40 mL) was treatedwith 2N HCl (20 mL) and heated at 80 �C for 2 h. The reactionmixture was concentrated in vacuo, and the residue was trituratedwith diethyl ether to afford 1,3-Dipropyl-8-{1-[3-(3-trifluoromethoxy-phenyl)-prop-2-ynyl]-1H-pyrazol-4-yl}-3,7-dihydro-purine-2,6-dione (0.915 g, 80%) as a white solid. M.P.199e200 �C; 1HNMR (DMSO-d6): d 0.85e0.91 (m, 6H); 1.54e1.60(m, 2H); 1.69e1.75 (m, 2H); 3.85 (t, J ¼ 7.2 Hz, 2H); 3.98 (t,J ¼ 7.2 Hz, 2H); 5.42 (s, 2H); 7.45e7.50 (m, 2H); 7.54e7.56 (m, 2H);8.12 (s, 1H); 8.53 (s, 1H); 13.59 (s, 1H); 13C NMR (DMSO-d6,100 MHZ): d 153.8, 150.6, 148.4, 148.2, 145.0, 138.3, 131.0, 130.9,129.7, 124.0, 123.4, 122.1, 118.7, 112.7, 106.3, 85.3, 83.2, 44.3, 42.1,41.7, 20.8, 11.2, 11.0, MS (m/z) (Mþþ1): 501.2; HPLC purity 100%.

4.2.1. Radioligand binding for adenosine receptors A1, A2A, A2B andA3

Human adenosine receptor (A1, A2A, A2B and A3) cDNA wasstably transfected into HEK-293 cells (referred to as HEK-A1, HEK-A2A, HEK-A2B, HEK-A3 cells). The HEK-293 cell was obtained fromATCC. The cells monolayer was washed with PBS once and har-vested in a buffer containing 150mMNaCl,1mMEDTA, 50mMTris,pH 7.4 (10mMEDTA,10mMHEPES, pH 7.4 for HEK-A3) at 1500 rpmfor 5 min at room temperature. The cell pellet was incubated insonication buffer containing 1 mM EDTA, 5 mM Tris, pH 7.4 (1 mMEDTA,10mMHEPES, pH-7.4 for HEK-A3) for 10min at 4 �C followedby sonication on ice for 6 min. The lysate was centrifuged at 1000xgfor 10 min at 4 �C and the pellet was discarded. The supernatantwas centrifuged at 49,000xg for 45 min at 4 �C. The resultantprotein pellet was resuspended in sonication buffer supplementedwith 1 U/ml adenosine deaminase (ADA, Roche) and incubated for30min at room temperature with constant mixing. The proteinwaswashed twice with same buffer at 49,000xg for 45 min at 4 �C andthe final protein was stored in 50 mM Tris, pH 7.4 supplementedwith 1 U/mL ADA and 10% sucrose (1 mM EDTA, 5 mM Tris, pH 7.4, 1U/ml ADA and 10% sucrose for HEK-A3). The protein concentrationwas estimated by Bradford assay and aliquots were storedat �80 �C.

The binding affinity and selectivity of test compounds wasdetermined using radioligand binding assays. DPCPX (8-Cyclopentyl-1,3-dipropylxanthine, for A1AdoR), Preladenant [2-(2-furanyl)-7-(2-(4-(4-(2-methoxyethoxy)phenyl)-1-piperazinyl)ethyl)-7H-pyrazolo (4,3-e) (1,2,4)triazolo (1,5-c)pyrimidine-5-amine, for A2AAdoR], 3-Ethyl-3,9-dihydro-1-propyl-8-[1-[[3-(tri-fluoromethyl)phenyl]methyl]-1H-pyrazol-4-yl]-1H-purine-2,6-dione (CVT-6883, A2BAdoR) and 3-Ethyl-5-benzyl-2-methyl-4-phenylethynyl-6-phenyl-1,4-(±)-dihydropyridine-3,5-dicarboxylate (MRS-119, A3AdoR) were used as internal standards.Competition radioligand binding assays were started by mixing1 nM [3H]-DPCPX (A1), 1 nM [2e3H]-4-(2-[7-amino-2-{2-furyl}{1,2,4}triazolo{2,3-a}{1,3,5,}triazin-5-yl amino]ethyl)phenol ([3H]ZM241385) (A2A), 35 1.6 nM radiolabelled N-(4-Cyanophenyl)-2-

[4-(2,3,6,7-tetrahydro-2,6-dioxo-1,3-dipropyl-1H-purin-8-yl)phe-noxy]acetamide ([3H]-MRS-1754) (A2B)36 or 2 nM [3H]-HEMADO(A3) with various concentrations of test compounds and therespective membranes in assay buffer containing 50 mM Tris pH7.4,1mMEDTA (A1), 50mMTris, pH 7.4,10mMMgCl2,1mMEDTA,1 U/ml ADA (A2A, A3) or 50 mM Tris pH 6.5, 5 mM MgCl2, 1 mMEDTA (A2B) supplemented with 1 U/ml ADA. The assays wereincubated at room temperature for 90 min with gentle agitation,stopped by filtration using a Harvester (Molecular Devices), andwashed four times with ice-cold 50 mM Tris (pH 7.4). Nonspecificbinding was determined in the presence of 100 mM NECA. The af-finities of compounds (i.e., Ki values) were calculated usingGraphPad software.

cAMP Assays: The functional activity of compounds was deter-mined by evaluating the changes in cAMP levels post agonisttreatment in HEK-293 cells over-expressing human A2BAdoR. A 24 hculture of cells was treated with 1 U/mL of ADA in DMEM supple-mented with 10% FBS for 90 min at 37 �C and 5% CO2. Cells wereharvested using cell dissociation buffer (Sigma), washed withincomplete DMEM and incubatedwith increasing concentrations oftest compound for 15min in DMEM supplementedwith 1U/ml ADAat room temperature. The cAMP levels were induced using A2BAdoRagonist NECA for 15 min. The levels of cAMP were detected andestimated as per manufacturer's instruction using the Cisbio HTRFcAMP kit. The FRET between d2-labeled cAMP and cryptate labeledanti-cAMP antibody was read in HTRF mode in a Flex Station III(Molecular Devices) using an excitation maximum of 313 nm andemission maxima of 620 nm and 665 nm. Data was analyzed usingGraphPad Prism software to determine IC50 and Ki values.

4.2.2. Molecular modeling4.2.2.1. Homology model development. In the absence of anyexperimentally (X-ray or NMR) determined three dimensional (3d)structure of human A2BAdoR, homology model of this receptor wasbuilt using MOE program [46] by implementing the standard pro-tocol. On the basis of lowest E value, good Z score and highestpercentage identity (61%) with the query protein, PDB ID: 4EIY (X-ray crystal structure of A2AAdoR with small molecule antagonistZM241385) [47] was found to be the most suitable template forbuilding the homology model of human A2BAdoR. Among theseveral other crystal structures of A2AAdoR in the Protein Data Bank(PDB), this structure was particularly selected due to its high res-olution and better overall B-factor. Initially, 100 models weregenerated and the best model was selected on the basis of Gener-alized Born/Volume Integral (GB/VI) score, Ca RMSD with thetemplate protein, contact energy and packing score. The best modelwas checked for its stereochemistry using Ramachandran plot.Model refinement and minimization was done using Amber99(ReField) force field. The template ligand ZM241385 coordinateswere transferred in the homology modeled structure of A2BAdoR inorder to define the binding site.

4.2.2.2. Molecular docking. Ligands were built in MOE, hydrogenswere added, partial charges were assigned and minimized usingMMF94� force field. Molecular docking was performed by GOLD5.2.2. (Genetic Optimization for Ligand Docking) program fromCambridge Crystallographic Data Centre (CCDC), UK [48] usingstandard protocol as mentioned elsewhere. The binding site ofmodeled A2BAdoR for the purpose of docking was defined byselecting the bound ligand (ZM241385). All the atoms within 6 Årange from the centre of bound ligand were selected in order tocover all the residues of active site. Subsequently, all the H-bonddonors/acceptors were forced to be treated as solvent accessible. Amaximum of 10 docking GA runs per molecule were performed,allowing all the poses to generate and sorted by using ChemScore

Page 11: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229228

fitness scoring function. All other parameters were kept at theirdefault values for the docking. Most plausible docking poses for thecompounds were selected by visual inspection on the basis ofbinding orientation, key interactions and Gold Chemscore DG.

Acknowledgements

Authors are grateful to the senior management of the Advi-nusfor its support and encouragement. Analytical departments areacknowledged for their help during this work. We thank Dr. AnupRanade for managing intellectual property - Advinus publicationno. ADV-A-040.

Appendix A. Supplementary data

The contents of supporting information include the following:(1) experimental procedures and characterization (3) 1H NMR/13CNMR spectra of selected compounds.

Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.ejmech.2017.04.014.

References

[1] V. Ralevic, G. Burnstock, Receptors for purines and pyrimidines, Pharmacol.Rev. 50 (1998) 413e492.

[2] C.E. Muller, B. Stein, Adenosine receptor antagonists: structures and potentialtherapeutic applications, Curr. Pharm. Des. 2 (1996) 501e530.

[3] J. Linden, Molecular approach to adenosine receptors: receptor mediatedmechanisms of tissue protection, Annu. Rev. Pharmacol. Toxicol. 41 (2001)775e787.

[4] J.R. Fozard, J.P. Hannon, Adenosine receptor ligands: potential as therapeuticagents in asthma and COPD, Pulm. Pharmacol. Ther. 12 (1999) 111e114.

[5] I. Feoktistov, I. Biaggioni, Adenosine A2B receptors, Pharmacol. Rev. 49 (1997)381e402.

[6] P.G. Baraldi, R. Romagnoli, D. Preti, F. Fruttarolo, M.D. Carrion, M.A. Tabizi,Ligands for A2B adenosine receptor subtype, Curr. Med. Chem. 13 (2006)3467e3482.

[7] I. Feoktistov, J.J. Murray, I. Biaggioni, Positive modulation of intracellular Ca2þ

levels by adenosine A2B receptors, prostacyclin, and prostaglandin E1 via acholera toxin-sensitive mechanism in human erythroleukemia cells, Mol.Pharmacol. 45 (1994) 1160e1167.

[8] D.J. Mogul, M.E. Adams, A.P. Fox, Differential activation of adenosine receptorsdecreases N-Type but potentiates P-Type Ca2þ current in hippocampal CA3neurons, Neuron 10 (1993) 327e334.

[9] J.A. Auchampach, G.H. Caughey, J. Linden, A2B and not A3 adenosine receptorsmediate degranulation of canine BR mastocytoma cells, Drug Dev. Res. 38(1996) 147.

[10] J.A. Auchampach, X. Jin, T.C. Wan, G.H. Caughey, J. Linden, Canine mast celladenosine receptors: cloning and expression of the A3 receptor and evidencethat degranulation is mediated by the A2B receptor, Mol. Pharmacol. 52 (1997)846e860.

[11] M.F. Gurden, J. Cates, F. Ellis, B. Evans, M. Foster, E. Hornby, I. Kennedy,D.P. Martin, P. Strong, C.Y. Vardey, A. Wheeldon, Functional characterizationof three adenosine receptor types, Br. J. Pharmacol. 109 (1993) 693e698.

[12] J.P. Clancey, F.E. Ruiz, E.J. Sorscher, Adenosine and its nucleotides activatewild-type and R117H CFTR through an A2B receptor-coupled pathway, Am. J.Physiol. 276 (1999) C361eC369.

[13] G.R. Strohmeier, S.M. Reppert, W.I. Lencer, J.L. Madaa, The A2B adenosine re-ceptor mediates camp responses to adenosine receptor agonists in humanintestinal epithelia, J. Biol. Chem. 270 (1995) 2387e2394.

[14] R.K. Dubey, D.G. Gillespie, Z. Mi, E.K. Jackson, Adenosine inhibits growth ofhuman aortic smooth muscle cells via A2B receptors, Hypertension 31 (1998)516e521.

[15] H. Harada, O. Asano, Y. Hoshino, S. Yoshikawa, M. Matsukura, Y. Kabasawa,J. Nijima, Y. Kotake, N. Watanabe, T. Kawata, T. Inoue, T. Horizoe, N. Yasuda,H. Minami, K. Nagata, M. Murakami, J. Nagaoka, S. Kobayashi, I. Tanaka, S. Abe,2-Alkynyl-8-Aryl-9-Methyladenines as novel adenosine receptor antagonists:their synthesis and structure-activity relationships toward hepatic glucoseproduction induced via agonism of the A2B receptor, J. Med. Chem. 44 (2001)170e179.

[16] T.H. Adair, R. Cotton, M.R. McMullan, W. Li, J.W. Gu, Adenosine increasesplasma levels of VEGF in humans (Abstract), FASEB J. 15 (2001) A277.

[17] D. Marx, C.I. Ezeamuzie, K. Nieber, I. Szenlenyi, Therapy of bronchial asthmawith adenosine receptor agonists or antagonists, Drug News Perspect. 14(2001) 89e100.

[18] I. Feoktistov, R. Palosa, S.T. Holgate, I. Biaggioni, Adenosine A2B receptors: anovel therapeutic target in asthma, Trends Pharmacol. Sci. 19 (1998)148e153.

[19] J. Ham, D.A. Rees, The adenosine A2B receptor: its role in inflammation,endocrine, Metabolic Immune Disord. e Drug Targets 8 (2008) 244e254.

[20] K.A. Jacobson, C. Hoffman, F. Cattabeni, M.P. Abbracchio, Adenosine-inducedcell death: evidence for receptor-mediated signaling, Apoptosis 4 (1999)197e211.

[21] M.J. Cushley, A.E. Tattersfield, S.T. Holgate, Inhaled adenosine and guanosineon airway resistance in normal and asthmatic subjects, Brit. J. Clin. Pharmacol.15 (1983) 161e165.

[22] M.J. Cushley, A.E. Tattersfield, S.T. Holgate, Adenosine-induced bronchocon-striction in asthma. Antagonism by inhaled theophylline, Am. Rev. Respir. Dis.129 (1984) 380e384.

[23] A.G. Driver, C.A. Kukoly, C.S. Ali, S.J. Mustafa, Adenosine in bronchoalveolarlavage fluid in asthma, Am. Rev. Respir. Dis. 148 (1993) 91e97.

[24] E. Huszar, G. Vass, E. Vizi, Z. Csoma, E. Barat, G. Molnar Vilagos, I. Herjavecz,I. Horvath, Adenosine in exhaled breath condensate in healthy volunteers andin patients with asthma, Eur. Respir. J. 20 (2002) 1393e1398.

[25] J.L. Chunn, J.G. Molina, T. Mi, Y. Xia, R.E. Kellems, M.R. Blackburn, Adenosine-dependent pulmonary fibrosis in adenosine deaminase-deficient mice,J. Immunol. 175 (2005) 1937e1946.

[26] B. Ma, M.R. Blackburn, C.G. Lee, R.J. Homer, W. Lui, R.A. Flavell, L. Boyden,R.P. Lifton, C.X. Sun, H.W. Young, J.A. Elias, Adenosine metabolism and murinestrain-specific IL-4-induced inflammation, emphysema, and fibrosis, J. Clin.Invest. 116 (2006) 1274e1283.

[27] S. Ryzhov, A.E. Goldstein, A. Matafonov, D. Zeng, I. Biaggioni, I. Feoktistov,Adenosine-activated mast cells induce IgE Synthesis by B Lymphocytes: ana2B-mediated process involving Th2 cytokines IL-4 and IL-13 with implica-tions for asthma, J. Immunol. 172 (2004) 7726e7733.

[28] H. Zhong, Y. Wu, L. Belardinelli, D. Zeng, A2B adenosine receptors induce IL-19from bronchial epithelial cells resulting in TNF-alpha increase, Am. J. Respir.Cell Mol. Biol. 35 (2006) 587e592.

[29] H. Zhong, L. Belardinelli, T. Maa, I. Feoktistov, I. Biaggioni, D. Zeng, A2Badenosine receptors increase cytokine release by bronchial smooth musclecells, Am. J. Respir. Cell Mol. Biol. 30 (2004) 118e125.

[30] H. Zhong, L. Belardinelli, T. Maa, D. Zeng, Synergy between A2B adenosinereceptors and hypoxia in activating human lung fibroblasts, Am. J. Respir. CellMol. Biol. 32 (2005) 2e8.

[31] G.E. D'Alonzo, Theophylline revisited, Allergy Asthma. Proc. 17 (1996)335e339.

[32] K.A. Jacobson, A.P. Ijzerman, J. Linden, 1,3-Dialkylxanthine derivatives havinghigh potency as antagonists at human A2B adenosine receptors, Drug Dev. Res.47 (1999) 45e53.

[33] M.W. Beukers, I. Meurs, A.P. Ijzerman, Structure-affinity relationships ofadenosine A2B receptor ligands, Med. Res. Rev. 26 (2006) 667e698.

[34] B. Cacciari, G. Pastorin, C. Bolcato, G. Spalluto, M. Bacilieri, S. Moro, A2Badenosine receptor antagonists: recent developments, Mini-Rev. Med. Chem.5 (2005) 1053e1060.

[35] P.G. Baraldi, M.A. Tabrizi, D. Preti, A. Bovero, R. Romagnoli, F. Fruttarolo,N.A. Zaid, A.R. Moorman, K. Varani, S. Gessi, S. Merighi, P.A. Borea, Design,synthesis, and biological evaluation of new 8-heterocyclic xanthine de-rivatives as highly potent and selective human A2B adenosine receptor an-tagonists, J. Med. Chem. 47 (2004) 1434e1447.

[36] R. Kalla, E. Elzein, T. Perry, X. Li, V. Palle, V. Varkhedkar, A. Gimbel, T. Maa,D. Zeng, J. Zablocki, Novel 1,3-Disubstituted-8-(1-Benzyl-1H-Pyrazol-4-yl)xanthines: high affinity and selective A2B adenosine receptor antagonists,J. Med. Chem. 49 (2006) 3682e3692.

[37] A. Stefanachi, J.M. Brea, M.I. Cadavid, N.B. Centeno, C. Esteve, M.I. Loza,A. Martinez, R. Nieto, E. Ravi~na, F. Sanz, V. Segarra, E. Sotelo, B. Vidal,A. Carotti, 1-, 3- and 8-substituted-9-deazaxanthines as potent and selectiveantagonists at the human A2B adenosine receptor, Biorg. Med. Chem. 16(2008) 2852e2869.

[38] Y.-C. Kim, X-d. Ji, N. Melman, J. Linden, K.A. Jacobson, Anilide derivatives of an8-phenylxanthine carboxylic congener are highly potent and selective an-tagonists at human A2B adenosine receptors, J. Med. Chem. 43 (2000)1165e1172.

[39] X. Ji, Y.C. Kim, D.G. Ahern, J. Linden, K.A. Jacobson, [3H]-MRS 1754, a selectiveantagonist radioligand for A2B adenosine receptors, Biochem. Pharmacol. 61(2001) 657e663.

[40] E. Elzein, R.V. Kalla, X. Li, T. Perry, A. Gimbel, D. Zeng, D. Lustig, K. Leung,J. Zablocki, Discovery of a novel A2B adenosine receptor antagonist as a clinicalcandidate for chronic inflammatory airway diseases, J. Med. Chem. 51 (2008)2267e2278.

[41] S.J. Mustafa, A. Nadeem, M. Fan, H. Zhong, L. Belardinelli, D. Zeng, Effect of aspecific and selective A2B adenosine receptor antagonist on adenosine agonistAMP and allergen-induced airway responsiveness and cellular influx in amouse model of asthma, J. Pharmacol. Exp. Ther. 320 (2007) 1246e1251.

[42] C. Sun, H. Zhong, A. Mohsenin, E. Morschl, J.L. Chunn, J.G. Molina,L. Belardinelli, D. Zeng, M.R. Blackburn, Role of A2B adenosine receptorsignaling in adenosine-dependent pulmonary inflammation and injury, J. Clin.Invest. 116 (2006) 2173e2182.

[43] S. Basu, A.D. Barawkar, V. Ramdas, Y. Waman, M. Patel, A. Panmand, S. Kumar,S. Thorat, R. Bonagiri, D. Jadhav, P. Mukhopadhyay, V. Prasad, B.S. Reddy,A. Goswami, S. Chaturvedi, S. Menon, A. Quraishi, I. Ghosh, S. Dusange,S. Paliwal, A. Kulkarni, V. Karande, R. Thakre, G. Bedse, S. Rouduri, J. Gundu,V.P. Palle, A. Chugh, K.A. Mookhtiar, A2B adenosine receptor antagonists:design, synthesis and biological evaluation of novel xanthine derivatives, Eur.

Page 12: European Journal of Medicinal Chemistry - 中国医药化工网 ...wxjs.chinayyhg.com/upload/Files/20170512124818436/2… ·  · 2017-05-12... Abhay Kulkarni, Vikas Karande, Indraneel

S. Basu et al. / European Journal of Medicinal Chemistry 134 (2017) 218e229 229

J. Med. Chem. 127 (2017) 986e996.[44] R. Kalla, E. Elzein, T. Perry, X. Li, A. Gimbel, M. Yang, D. Zeng, J. Zablocki, Se-

lective, high affinity A2B adenosine receptor antagonists: N-1 mono-substituted 8-(Pyrazol-4-yl)Xanthines, Bioorg. Med. Chem. Lett. 18 (2008)1397e1401.

[45] Binding affinity for hA2BAdoR was determined using [3H]-MRS-1754 asligand; Binding affinity for hA1AdoR was determined using [3H]-DPCPX asligand; Binding affinity for hA2AAdoR was determined using [3H]-ZM-241385as ligand; and Binding affinity for hA3AdoR was determined using [3H]-HEMADO as ligand.

[46] 08, Molecular Operating Environment (MOE), Chemical Computing GroupInc., 1010 Sherbooke St. West, Suite #910, Montreal, QC, Canada, 2013. H3A2R7, 2017.

[47] W. Liu, E. Chun, A.A. Thompson, P. Chubukov, F. Xu, V. Katritch, G.W. Han,C.B. Roth, L.H. Heitman, A.P. IJzerman, V. Cherezov, R.C. Stevens, Structuralbasis for allosteric regulation of GPCRs by sodium ions, Science 337 (2012)232e236.

[48] G. Jones, P. Willett, R.C. Glen, A.R. Leach, R. Taylor, Development and valida-tion of a genetic algorithm for flexible docking, J. Mol. Biol. 267 (1997)727e748.