Inhibition of the replication of different strains of ... - ACS Publications

compounds, satisfactory elemental analysis was obtained supporting greater than 95% purity. Electrospray mass ... Then a solution of 6a. 28 (224 mg, 0...
0 downloads 0 Views 687KB Size
Subscriber access provided by Rasmuson/BioSciences Library | University of Alaska Fairbanks

Article

Inhibition of the replication of different strains of chikungunya virus by 3-aryl-[1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones Asier Gomez-San Juan, Ana María Gamo, Leen Delang, Alfonso Pérez-Sánchez, Siti Naqiah Amrun, Rana Abdelnabi, Sofie Jacobs, Eva-María Priego, María-José Camarasa, Dirk Jochmans, Pieter LEYSSEN, Lisa F. P. Ng, Gilles Querat, Johan Neyts, and Maria-Jesus Perez-Perez ACS Infect. Dis., Just Accepted Manuscript • DOI: 10.1021/acsinfecdis.7b00219 • Publication Date (Web): 06 Feb 2018 Downloaded from http://pubs.acs.org on February 7, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Infectious Diseases is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Inhibition of the replication of different strains of chikungunya virus by 3-aryl-[1,2,3]triazolo [4,5-d]pyrimidin-7(6H)-ones

Asier Gómez-SanJuan, # Ana-María Gamo, # Leen Delang,† Alfonso Pérez-Sánchez,# Siti Naqiah Amrun,§ Rana Abdelnabi, † Sofie Jacobs, † Eva-María Priego,# María-José Camarasa,# Dirk Jochmans,† Pieter Leyssen,† Lisa F. P. Ng,§ Gilles Querat,◊ Johan Neyts,† and María-Jesús PérezPérez# * #



Instituto de Química Médica, IQM, CSIC, Juan de la Cierva 3, E-28006 Madrid, Spain

KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium.

§

Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, 04-06 Immunos, Singapore 138648, Singapore



UMR "Émergence des Pathologies Virales" (EPV: Aix-Marseille Univ – IRD 190 – Inserm 1207 – EHESP – IHU Méditerranée Infection), 27 Bd Jean Moulin, 13005 Marseille, France Corresponding author: [email protected]

1 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 51

The re-emergence of chikungunya virus (CHIKV) is a serious global health threat. CHIKV is an alphavirus that is transmitted to humans by Aedes mosquitoes; therefore, their wide distribution significantly contributes to the globalization of the disease. Unfortunately, no effective antiviral drugs are available. We have identified a series of 3-aryl-[1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones as selective inhibitors of CHIKV replication. New series of compounds have now been synthesized with the aim to improve their physicochemical properties and to potentiate the inhibitory activity against different CHIKV strains. Among these newly synthesized compounds modified at position 3 of the aryl ring, a tetrahydropyranyl and a N-t-butylpiperidine carboxamide derivatives have shown to elicit potent antiviral activity against different clinically relevant CHIKV isolates with EC50 values ranging from 0.30 to 4.5 µM in Vero cells, as well as anti-CHIKV activity in human skin fibroblasts (EC50 = 0.1 µM), a clinically relevant cell system for CHIKV infection.

KEY WORDS. Triazolopyrimidines; chikungunya virus; physicochemical properties.

2 ACS Paragon Plus Environment

Page 3 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

3 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 51

Chikungunya virus (CHIKV) is a positive single-stranded RNA enveloped virus belonging to the alphavirus genus and Togaviridae family. CHIKV is transmitted to humans through the bites of Aedes mosquitoes, particularly Ae. aegypti and Ae. albopictus and causes chikungunya fever (CHIKF) characterized by chronic and incapacitating arthralgia.1-3 Although mortality is low, symptoms can persist for months or years, and this has important social and economic consequences. In some cases, complications afflicting neonates and the elderly have been reported, and recent outbreaks are also associated with more severe forms of CHIKF involving neurological complications.4 Since 2005, CHIKV has been spreading worldwide resulting in epidemics in Africa, the Indian Ocean islands, Asia and more recently in the Americas.5 Since 2007, autochthonous chikungunya outbreaks in Italy and France have been documented, the latest one this year.6 There are three different CHIKV genotypes: West African, Asian and East/Central/South African (ECSA).7, 8 The ECSA genotype has been associated to epidemics in the Indian Ocean countries while the Asian genotype has been identified in the outbreaks in the Caribbean regions.9 Chikungunya is thus considered an important re-emerging global public health problem in countries where the distribution of the Aedes mosquito vectors continues to expand, particularly in the tropical and subtropical areas.3, 10-12 No vaccine or antiviral drug is available to fight against chikungunya infection.13 The use of chloroquine, the antimalarial drug, was evaluated in clinical trials during the outbreak in La Reunion.14 Despite the activity of the compound in cell culture, the benefits of this drug for CHIKF treatment were not clear. Traditional broad-spectrum antivirals such as ribavirin, mycophenolic acid or interferon have also been used but their effectiveness has not been confirmed.15 Thus, cell-based assays have been set up to identify compounds that may inhibit CHIKV replication.13, 16 Indeed, in the last three to four years, an increasing number of inhibitors have been described either from natural products such as flavonoids17 and diterpenes18 or synthetic compounds.19-22 Interestingly, some of the compounds reported to block infection affect host factors, such as berberine.23 4 ACS Paragon Plus Environment

Page 5 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Through a collaborative project, we identified the triazolopyrimidines, exemplified by compounds 1 and 2 (Figure 1), as selective inhibitors of CHIKV replication.24 Structure-activity relationship studies performed with a wider series of compounds24,

25

revealed that the [1,2,3]triazolo[4,5-

d]pyrimidin-7(6H)-one substituted with a methyl (1) or preferentially an ethyl (2) at position 5 was crucial for anti-CHIKV activity together with a meta-substituted aryl ring linked at position 3 of the triazole. As recently reported,25 the oximes 3 and 4 (Figure 1) have EC50 values in the very low µM range suggesting that the substituent at position 3 of the aryl moiety can be significantly enlarged without compromising the antiviral activity. Based on the anti-CHIKV activity of the isopropyl derivative 2, it was considered that enlarging the isopropyl substituent into a six membered ring to increase the fraction of sp3 atoms26 and further incorporating an heteroatom might maintain the antiviral activity and improve the physicochemical properties of the new series, as indicated by their theoretical octanol/water partition coefficient (cLogP), topological surface area (tPSA) and solubility values (see Table S1 in the Supporting Information), thus paving the way for compounds with a better profile towards potential “in vivo” testing.

RESULTS AND DISCUSSION Chemistry The synthesis of the first series of six-membered ring derivatives is shown in Scheme 1. Reaction of 3-azidophenol (5)27 with the mesylates 6a28, 6b29 or 629 in the presence of Cs2CO3 afforded the corresponding arylazides 7a-c in moderate yields. The modest yield of the substitution products is due to the competing elimination reaction of the mesylate, as reported for similar analogues.30 Attempts to minimize the elimination reaction by using different bases (K2CO3, Cs2CO3 or NaH), testing different temperatures (from rt to 80 ºC) or modifying the dilution did not meet with success. The best results were obtained using Cs2CO3 as the base and heating at 80 ºC for 2 h. Then, reaction of the arylazides 7a-c with cyanoacetamide in the presence of NaH afforded the 5-amino-1H-1,2,3triazole-4-carboxamides 8a-c in excellent yields. Finally, reaction of 8a and 8b with ethylpropionate, 5 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 51

or reaction of 8c with tert-butylpropionate in the presence of tBuOK in dioxane afforded the triazolopyrimidines 9a-c. Removal of the Boc protecting group in 9c by treatment with a 5% solution of TFA in DCM for 3 h at room temperature afforded quantitatively the free piperidyl derivative 9d. When these compounds (9a-d) were tested for anti-CHIKV activity in cell culture, compound 9b with a tetrahydropyranyl substituent, and compound 9c with a Boc-protected piperidyl showed very potent antiviral activity with EC50 values in the low µM range. The antiviral activity of the N-Boc derivative 9c was particularly attractive since Boc-removal and further acylation reaction on the piperidyl might lead to novel substitutions. Before that, and in an effort to further improve clogP and tPSA values (Table S1 in Suppl material), the piperidyl ring was replaced by a piperazinyl and a carbonyl group, instead of an oxygen, was introduced as the linker with the aryl ring. Thus, reaction of 3-azidobenzoic acid31 (10, Scheme 2) with tert-butyl piperazine-1-carboxylate, catalyzed by HCTU in the presence of DIPEA, in DMF:DCM at rt for 2h afforded the coupling product 11 in 77% yield. Further reaction of this azide with cyanoacetamide in the presence of NaH afforded the aminocarboxamide 12 that was treated with tert-butyl propionate under basic conditions and MW irradiation to obtain 13 (22% yield for the two steps). This compound was 8-fold less active than 9c (as it will be later discussed in the Biological Results section), so our efforts were turned back to the piperidyl derivatives. Thus compound 9c was used as a synthon to introduce novel substitutions at the N atom of the piperidine. Removal of the Boc protecting group by treatment with 5% TFA followed by reaction with different chloroformates in DCM in the presence of Et3N and DMAP (cat) at 0 ºC afforded compounds 14a-e (Scheme 3). In a similar way, a number of ureas were synthesized (compounds 15a-h, Scheme 3) by reaction of the free amine after Boc-removal with a variety of isocyanates, in DCM/DMF in the presence of Et3N or alternatively by reaction of the free amine with triphosgene and further reaction with a second amine. The conditions and yields obtained for each of these compounds are specified in Scheme 3. Amides and sulfonamides as substituents of the N of the piperazine were also synthesized (Scheme 4). Thus, Boc-removal in 9c followed by reaction with methyl 4-chloro-4-oxobutanoate 6 ACS Paragon Plus Environment

Page 7 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

afforded the N-acyl derivative 16 (30% yield). Alternatively, reaction of 9c with TFA followed by reaction of the free amine with nicotinic or isonicotinic acid catalyzed by HATU and DIPEA afforded compounds 17a and 17b in 47% and 50% yield, respectively. Finally, reaction of the free amine with 2-propanesulfonyl chloride afforded the sulfonamide 18 in 67% yield. Biological results Evaluation of anti-CHIKV activity and cytotoxicity The synthesized compounds were evaluated for their potential to inhibit the CHIKV-induced cytopathogenic effect (CPE) in Vero cells (Table 1). Chloroquine and our previously reported hits (compounds 1 and 2) were included as reference compounds. The antiviral activity is expressed as the 50% effective concentration (EC50) and the 90% effective concentration (EC90), indicating the concentration of compound required to inhibit the virus-induced CPE by 50 and 90%, respectively. The effect of the compounds on non-infected cells is expressed as CC50, which corresponds to the calculated concentration of compound that reduces the viability of compound-treated cells by 50% (as determined by microscopic observation). Compounds 9a-9d behaved very differently against CHIKV replication. While compounds 9b and 9c were very potent compounds with EC50 and EC90 values in the low µM range (Table 1), compounds 9a and 9d were inactive. In order to explain the lack of activity of the 9d compared to its oxygen counterpart 9b, it can be argued that under the physiological conditions the N atom of the free piperidyl might be charged (see Maps of electrostatic potential section) Such a positive charge may hamper the correct diffusion through membranes or imply a significant desolvation penalty to reach the target binding site. On the other hand, and in that concerning the tert-butyl carbamate 9c, it is interesting to mention that carbamates are present in many approved drugs, as recently reviewed32 and in particular tert-butyl carbamates of piperidine derivatives have been reported to accomplish drug-like properties.33 Compound 13 with a piperazine tert-butyl carbamate had an EC50 value of 27 µM, 8-fold less active than 9c, suggesting that the incorporation of the piperazine was detrimental for anti-CHIKV activity. On the other hand, other carbamates (14a-e) or urea (15a-h) derivatives of piperidine 7 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 51

showed significant anti-CHIKV activity with EC50 values between 1.5 to 14 µM, although in some cases this was accompanied with significant cytotoxicity as measured by microscopic scoring. Only compounds 15d and 15h were almost inactive against CHIKV replication. It is also interesting to mention that the incorporation of a pyridine at the distal site (compounds 17a and 17b, Table 1) led to poorly active or inactive compounds, further stressing that the incorporation of groups that may be protonated at physiological pH is detrimental for activity. In order to determine whether the cytotoxicity observed with carbamates and ureas was compound-dependent or class-dependent, for a few selected compounds with the best EC50 values (9c, 14a and 15e) additional experiments were performed to measure cytotoxicity in other cell lines (Vero E6, BHK, HeLa Rh or Huh7) either microscopically or by the MTS/PMS colorimetric method. The obtained data are shown in Table 2. Only compound 9c showed CC50 values between 50-120 µM, while compounds 14a and 15e were not toxic at concentrations of 220 µM in the tested cell lines, the only exception being the more sensitive Huh7 cells. Thus, from the new series of triazolopyrimidines here reported, compound 9b, with a tetrahydropyranyl substituent, and compounds 9c, 14a and 15e with a piperidine substituent functionalized as a carbamate (9c and 14a) or an urea (15e) represent potent and selective antiCHIKV inhibitors. Evaluation of anti-CHIKV activity against different clinical isolates Selected compounds from this series were evaluated for antiviral activity against several CHIKV clinical isolates (Table 3), including strains obtained from the outbreaks in Italy (Venturini 2008), Africa (Congo 2011), La Réunion Island (Opy 2006) and the Caribbean Americas (St Martin 2013 and EFS-1, Martinique 2013). It should be noted that all these compounds showed antiviral activity at the low µM or sub-µM range, well below their CC50 in Vero cells. In all cases the most potent inhibition was observed against the African Congo strain with EC50 values as low as 0.30 to 0.60 µM for compounds 9b, 14a and 15e. In addition these compounds showed lower EC50 values against St Martin strains than the reference compounds 1 and 2, so it can be concluded that the new derivatives have a wider profile against different CHIKV strains than previous series. 8 ACS Paragon Plus Environment

Page 9 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Evaluation of anti-CHIKV activity in human skin fibroblasts Next, the anti-CHIKV activity of some selected triazolopyrimidines was determined in human skin fibroblasts, which is a clinically more relevant system as the skin is the first organ supporting viral replication upon the bite of an infected mosquito.34, 35 BJ (CRL-2522) fibroblastic cells were infected with Gluc-tagged CHIKV-LR2006 OPY1 and the antiviral effect of selected compounds was determined. In this model, our reference compounds 1 and 2 had EC50 values of 1.10 ± 0.38 µM and 0.72 ± 0.56 µM, respectively. Interestingly compound 9b showed a more potent antiviral activity with an EC50 value of 0.12 ± 0.09 µM. Also compound 15e was significantly active in this assay with an EC50 of 0.55 ± 0.38 µM. It is relevant to mention that in all cases the EC50 values were observed to be in the low µM or sub-µM range while cell viability was not affected at concentrations two to three logs higher (CC50>100 µM). Compounds 9b and 15e were also tested against a different CHIKV strain (CHIKV-SGP11) in fibroblasts providing an EC50 value of 0.78 ± 0.07 µM and 0.20 ± 0.20 µM, respectively. Moreover, in this cellular system, significant antiviral activity was also found for 9b and 15e against the O´nyong-nyong virus (ONNV), the closest analogue to CHIKV among alphaviruses,7 with EC50 values of 0.66 ± 0.07 µΜ and 0.42 ± 0.07 µM, respectively. Evaluation against CHIKV strains resistant to compound 1. In order to determine if the new series of compounds behaved similarly to our initially reported compounds, some of the here described compounds were tested against a CHIKV strain that was selected under the selective pressure of compound 1. This strain is characterized by a single mutation in the nonstructural protein 1 (nsP1) gene (P34S).36 The CHIKV nsP1 is the main enzyme involved in the capping of the viral RNA. The importance of the P34S mutation for the resistance phenotype had been previously confirmed by reverse genetics.36 T-705 (favipiravir), a CHIKV inhibitor with a different mechanism of action,37 was included as a negative control. As shown in Table 4, all triazolopyrimidine compounds were less active against the CHIKV variant resistant against compound 1, albeit not to the same extent. Compound 9b behaved similarly to compound 2 (fold resistance= 54 and 64, respectively), whereas compounds 9c and 15e showed lower levels of cross9 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 51

resistance. The observed difference in fold resistance levels could be due to the difference in the selectivity indexes: compounds 1, 9c and 15e have lower selectivity indexes than compounds 2 and 9b; their toxicity might limit their resistance indexes. Maps of electrostatic potential In an attempt to explain the differences shown in the antiviral activity of these compounds, molecular electrostatic potential (MEP) maps were calculated for selected compounds (9b, 9c, 9d, and 15e). The geometry of the compounds was first optimized by means of the quantum-chemistry software Jaguar (implemented in the Schrödinger Suite), and MEP maps were calculated based on the optimized structures using DelPhi. Results are shown in Figure S1. As expected, MEP maps corresponding to the triazolopyrimidinone core and the aromatic ring, present in all the ligands, showed a common pattern. The mayor differences were observed at the distal part for compound 9d compared to the other three compounds, where a significant positive map was located around the protonated piperidine ring for compound 9d. This may partially account for the different behaviour in terms of antiviral activity between compound 9d and the other three compounds, On the other hand, the urea and carbamate derivatives, 9c and 15e respectively, both with a tert-butyl distal substituent, showed similar positive and negative MEPs. Microsomal stability studies and preliminary pharmacokinetics As indicated in the introduction, one of the objectives of the new series of triazolopyrimidines was to improve their physicochemical properties in order to identify a potential inhibitor for “in vivo” experiments. Among the newly synthesized derivatives, compounds 9b and 15e showed the best inhibition of CHIKV replication in the different systems assayed. Thus, in silico ADME predictions were performed for both compounds using QikProp program (Schrödinger Release 2015-4, Schrödinger, LLC, New York, NY, 2015). The obtained values for the minimum energy conformer of 9b and 15e (Table S2) in terms of Caco permeability (QPPCaco), binding to human albumin (QPlogKHsa) and percentage of human oral absorption indicated superior properties for 9b compared to 15e. Thus, compound 9b was selected for experimental determinations. Microsomal stability studies, in the absence of cofactors, indicated perfect stability while in the presence of 10 ACS Paragon Plus Environment

Page 11 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

cofactors the t1/2 was around 24 min and the clearance value was around 230 µL/min/mg. Data are shown in Table S3, together with reference compounds (diclofenac and imipramine). Interestingly, the values obtained for compound 9b were 3-fold better than those obtained with compound 2. By means of follow-up on these results, compound 9b was selected for a preliminary pharmacokinetics study in male BALB/c mice. The compound was administered as a single dose by intraperitoneal and subcutaneous route at 10 mg/kg body weight. The data obtained are compiled in Table S4. The Cmax (1168 and 806 ng/mL for intraperitoneal and subcutaneous dosing, respectively) were acceptable but the half-lives (0.34 and 0.47 h, respectively) were short, indicating that the compound is well absorbed and distributed, but that it is also easily metabolized and/or excreted.

CONCLUSIONS Based on the identification of 3-aryl-[1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones as selective antiCHIKV agents, exemplified by compounds 1 and 2, additional series of compounds were synthesized and evaluated, resulting in the identification of novel derivatives that inhibit CHIKV replication in the very low µM or sub-µM range. Among them, the tetrahydropyranyl derivative 9b or the N-tert-butyl piperidine carboxamide 15e have demonstrated to be potent and selective inhibitors of a variety of clinical CHIKV strains (Venturini, Congo, St Martin) with EC50 values ranging from 0.30 to 4.5 µM. Both compounds also showed potent antiviral activity against two CHIKV strains in human skin fibroblasts, which is a more relevant biological system since the skin is the primary infection site after the mosquito bite. Preliminary PK studies demonstrated that compound 9b has a reasonable Cmax although further improvement of the half-life is recommended before performing “in vivo” proof-of-concept experiments in an animal model of CHIKV infection. Compounds 9b, 9c and 15e showed a significant level of cross resistance when tested against a CHIKV strain selected under the pressure of the prototype triazolopyrimidine 1, the higher level of cross resistance was obtained for compound 9b that was also the less toxic.

11 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 51

METHODS Chemistry Procedures. Melting points were obtained on a Mettler Toledo M170 apparatus and are uncorrected. The elemental analysis was performed with a Heraus CHN-O-RAPID instrument. The elemental compositions of the compounds agreed to within ±0.4 of the calculated values. For all the tested compounds, satisfactory elemental analysis was obtained supporting greater than 95% purity. Electrospray mass spectra were measured on a quadrupole mass spectrometer equipped with an electrospray source (Hewlett-Packard, LC/MS HP 1100). 1H and 13C NMR spectra were recorded on a Varian INNOVA-300 operating at 300MHz (1H) and 75 MHz (13C), respectively, and a Varian INNOVA-400 operating at 400 MHz (1H) and 101 MHz (13C), respectively. Analytical TLC was performed on silica gel 60 F254 (Merck) precoated plates (0.2 mm). Spots were detected under UV light (254 nm) and/or charring with ninhydrin. Separations on silica gel were performed by preparative centrifugal circular thin-layer chromatography (CCTLC) on a ChromatotronR (Kiesegel 60 F254 gipshaltig (MercK)), with layer thicknesses of 1 and 2 mm and flow rates of 4 or 8 mL/min, respectively. Flash column chromatography was performed with silica gel 60 (230-400 mesh) (Merck). Microwave reactions were performed using the Biotage Initiator 2.0 single-mode cavity instrument from Biotage (Uppsala). Experiments were carried out in sealed microwave process vials utilizing the standard absorbance level (400 W maximum power). The temperature was measured with an IR sensor on the outside of the reaction vessel. 1-Azido-3-(cyclohexyloxy)benzene (7a). To a solution of 527 (85 mg, 0.63 mmol) in anhydrous DMF (1 mL) under argon atmosphere, Cs2CO3 (246 mg, 0.76 mmol) was added. The reaction mixture was stirred at rt for 5 min. Then a solution of 6a28 (224 mg, 0.8 mmol) in anhydrous DMF (1.2 mL) was added dropwise and the reaction was heated at 80 ºC for 2 h. After work-up, the residue was purified by CCTLC (hexane) to yield 30 mg (22%) of 7a as colourless oil. 1H NMR (400 MHz, CDCl3) δ 1.23-1.45 (m, 3H, Cy), 1.46-1.63 (m, 3H, Cy), 1.73-1.88 (m, 2H, Cy), 1.92-

12 ACS Paragon Plus Environment

Page 13 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

2.02 (m, 2H, Cy), 4.23 (tt, J = 8.9, 3.8 Hz, 1H, OCH), 6.55 (t, J = 2.2 Hz, 1H, Ar), 6.61 (ddd, J = 7.9, 2.1, 0.8 Hz, 1H, Ar), 6.68 (ddd, J = 8.3, 2.4, 0.8 Hz, 1H, Ar), 7.22 (t, J = 8.1 Hz, 1H, Ar). 4-(3-Azidophenoxy)tetrahydro-2H-pyran (7b). To a solution of 527 (188 mg, 1.39 mmol) in anhydrous DMF (3 mL) under argon atmosphere, Cs2CO3 (553 mg, 1.7 mmol) was added. The reaction mixture was stirred at rt for 5 min. Then a solution of 6b29 (250 mg, 1.39 mmol) in anhydrous DMF (4.2 mL) was added dropwise and the reaction was heated at 80 ºC for 2 h. After work-up, the residue was purified by flash chromatography (hexane/ethyl acetate, 8:2) to yield 140 mg (46%) of 7b as yellow oil. MS (ES, positive mode): m/z 637 (2M+H)+. 1H NMR (300 MHz, CDCl3) δ 1.69-1.88 (m, 2H, H-3’a(b), H-5’a(b)), 1.94-2.10 (m, 2H, H-3’b(a), H-5’b(a)), 3.47-3.66 (m, 2H, H-2’a(b), H-6’a(b)), 3.87-4.15 (m, 2H, H-2’b(a), H-6’b(a)), 4.48 (m, 1H, OCH), 6.56 (t, J = 2.2 Hz, 1H, Ar), 6.58-6.78 (m, 2H, Ar), 7.28 (t, J = 8.1 Hz, 1H, Ar). tert-Butyl 4-(3-azidophenoxy)piperidine-1-carboxylate (7c). To a solution of 527 (405 mg, 3 mmol) in anhydrous DMF (9 mL), Cs2CO3 (1.17 g) was added and the mixture was stirred for 5 minutes at rt. A solution of 6c29 (830 mg, 3 mmol) in anhydrous DMF (3 mL) was added dropwise. The reaction mixture was heated at 80 ºC for 2 h. Then a saturated solution of NH4Cl (10 mL) was added and the solution was extracted with ethyl acetate (3 x 10 mL). The organic phase was washed with NaOH 1M (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (hexane/ethyl acetate, 90:10) to yield 509 mg (55%) of 7c as yellow oil. MS (ES, positive mode) m/z: 638 (2M+H)+. 1H NMR (400 MHz, CDCl3): δ 1.47 (s, 9H, (CH3)3), 1.74 (m, 2H, CH2CH2N), 1.91 (m, 2H, CH2CH2N), 3.34 (m, 2H, CH2CH2N), 3.68 (m, 2H, CH2CH2N), 4.45 (m, 1H, CHO), 6.55 (pt, J = 2.2 Hz, 1H, Ar), 6.62-6.69 (m, 2H, Ar), 7.24 (pt, J = 8.1 Hz, 1H, Ar). General Procedure for the Synthesis of 5-Amino-1H-1,2,3-triazole-4-carboxamides (General Procedure A). To a solution of cyanoacetamide (1.1 mmol) in anhydrous DMF (3 mL) at 0 ºC, NaH (60% mineral oil, 1.8 mmol) was slowly added. After 30 min at this temperature, a solution of the corresponding azide (1.0 mmol) in anhydrous DMF (3 mL) was added. The reaction mixture was stirred 30 min at 0 ºC and additional 30 min at rt. The mixture was concentrated to dryness. The 13 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 51

crude obtained was dissolved in dichloromethane/methanol (9:1) (10 mL) and washed with H2O (3×10 mL). The combined aqueous phases were extracted with dichloromethane (3×10 mL). The combined organic phases were dried over anhydrous Na2SO4, filtered and evaporated. The residue was purified as indicated in each case 5-Amino-1-(3-(cyclohexyloxy)phenyl)-1H-1,2,3-triazole-4-carboxamide (8a). Following the general procedure A, to a solution of cyanoacetamide (13 mg, 0.15 mmol) in anhydrous DMF (1.0 mL) at 0 ºC, NaH (60% mineral oil, 10 mg, 0.25 mmol) was slowly added, followed by the addition of a solution of 7a (30 mg, 0.14 mmol) in anhydrous DMF (1.0 mL). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:2) to yield 31 mg (74%) of 8a as a white solid. Mp: 166-168 ºC. MS (ES, positive mode): m/z 302 (M+H)+. 1H NMR (400 MHz, DMSO-d6) δ 1.261.60 (m, 6H, Cy), 1.64-1.79 (m, 2H, Cy), 1.88-2.00 (m, 2H, Cy), 4.45 (m, 1H, OCH), 6.36 (br s, 2H, NH2), 7.04-7.15 (m, 3H, Ar), 7.22 (br s, 1H, CONH2a(b)), 7.48 (m, 1H, Ar), 7.61 (br s, 1H, CONH2b(a)). 5-Amino-1-(3-((tetrahydro-2H-pyran-4-yl)oxy)phenyl)-1H-1,2,3-triazole-4-carboxamide (8b). Following the general procedure A, to a solution of cyanoacetamide (49 mg, 0.58 mmol) in anhydrous DMF (1.3 mL) at 0 ºC, NaH (60% mineral oil, 38 mg, 0.93 mmol) was slowly added, followed by the addition of a solution of 7b (115 mg, 0.52 mmol) in anhydrous DMF (1.3 mL). After work-up, the solid was re-suspended in diethyl ether to yield 138 mg (87%) of 8b as a white solid. Mp: 171-173 ºC. MS (ES, positive mode): m/z 304 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 1.55-1.67 (m, 2H, H-3’a(b), H-5’a(b)), 1.97-2.02 (m, 2H, H-3’b(a), H-5’b(a)), 3.45-3.53 (m, 2H, H2’a(b), H-6’a(b)), 3.82-3.89 (m, 2H, H-2’b(a), H-6’b(a)), 4.69 (m, 1H, OCH), 6.37 (br s, 2H, NH2), 7.117.17 (m, 3H, Ar), 7.23 (br s, 1H, CONH2a(b)), 7.50 (t, J = 8.1 Hz, 1H, Ar), 7.62 (br s, 1H, CONH2b(a)). tert-Butyl

4-(3-(5-amino-4-carbamoyl-1H-1,2,3-triazol-1-yl)phenoxy)piperidine-1-

carboxylate (8c). To a solution of cyanoacetamide (110 mg, 1.30 mmol) in anhydrous DMF (3.25 mL) at 0 ºC NaH (60% mineral oil, 50 mg, 2.10 mmol) was slowly added. After 30 min at this temperature a solution of 7c (370 mg, 1.16 mmol) in anhydrous DMF (3 mL) was added. The 14 ACS Paragon Plus Environment

Page 15 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

reaction mixture was stirred 30 min at 0 ºC and for additional 30 min at rt. The mixture was concentrated to dryness. The residue was dissolved in DCM (10 mL) and washed with water (10 mL). The organic phase was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (dichloromethane/methanol, 95:5) to yield 460 mg (88%) of 8c as a white solid. MS (ES, positive mode): m/z 403 (M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 1.40 (s, 9H, (CH3)3), 1.55 (m, 2H, CH2CH2N), 1.93 (m, 2H, CH2CH2N), 3.19 (m, 2H, CH2CH2N), 3.66 (m, 2H, CH2CH2N), 4.67 (m, 1H, CHO), 6.37 (br s, 2H, NH2), 7.11-7.17 (m, 3H, Ar), 7.22 (br s, 1H, CONH2), 7.49 (t, J = 8.0 Hz, 1H, Ar), 7.62 (br s, 1H, CONH2). General Procedure for the Synthesis of [1,2,3]Triazolo[4,5-d]pyrimidin-7-ones (General Procedure B). A microwave vial was charged with the corresponding carboxamide (1.0 mmol), t

BuOK (1M in THF, 4.0 mmol) and ethyl propionate (4.0 mmol) in anhydrous dioxane (10 mL). The

reaction mixture was heated in a microwave reactor at 100 ºC for 1 h. The mixture was concentrated to dryness. The crude obtained was dissolved in dichloromethane (10 mL) and washed with a saturated solution of NaHCO3 (3×10 mL). The combined aqueous phases were extracted with dichloromethane (3×10 mL). The combined organic phases were dried over anhydrous Na2SO4, filtered and evaporated. The residue was purified as indicated in each case. 3-(3-(Cyclohexyloxy)phenyl)-5-ethyl-3,6-dihydro-7H-[1,2,3]triazolo[4,5-d]pyrimidin-7-one (9a). Following the general procedure B, a microwave vial was charged with 8a (30 mg, 0.10 mmol) t

BuOK (1M in THF, 0.4 mL, 0.4 mmol) and ethyl propionate (46 µL, 0.4 mmol) in anhydrous

dioxane (1.0 mL). After work-up, the residue was purified by flash chromatography (dichloromethane/methanol, 95:5) to yield 28 mg (83%) of 9a as a white solid. Mp: 262-264 ºC. MS (ES, positive mode): 340 (M+H)+. 1H NMR (400 MHz, DMSO-d6, δ) 1.25 (t, J = 7.5 Hz, 3H, CH2CH3), 1.28-1.62 (m, 6H, Cy), 1.67-1.82 (m, 2H, Cy), 1.89-2.07 (m, 2H, Cy), 2.72 (q, J = 7.5 Hz, CH2CH3), 4.42 (m, 1H, OCH), 7.09 (ddd, J = 8.2, 2.5 Hz, 1H, Ar), 7.52 (t, J = 8.1 Hz, 1H, Ar), 7.58 (m, 1H, Ar), 7.67 (t, J = 2.2 Hz, 1H, Ar), 12.72 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.1 (CH2CH3), 23.2 (Cy), 25.1 (Cy), 27.7 (CH2CH3), 31.2 (Cy), 74.9 (OCH), 108.8, 113.6, 116.5

15 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 51

(Ar), 129.0 (C-7a), 130.7, 136.4 (Ar), 148.6 (C-3a), 155.9, 157.9 (C-5, Ar), 164.6 (C-7). Anal calcd for (C18H21N5O2 0.5H2O): C, 62.05; H, 6.36; N, 20.10. Found: C, 62.54; H, 6.28; N, 19.84. 5-Ethyl-3-(3-((tetrahydro-2H-pyran-4-yl)oxy)phenyl)-3,6-dihydro-7H-[1,2,3]triazolo[4,5d]pyrimidin-7-one (9b). Following the general procedure B, a microwave vial was charged with 8b (100 mg, 0.33 mmol) tBuOK (1M in THF, 1.3 mL, 1.3 mmol) and ethyl propionate (160 µL, 1.3 mmol) in anhydrous dioxane (3.3 mL). After work-up, the residue was purified by flash chromatography (dichloromethane/methanol, 95:5) to yield 95 mg (85%) of 9b as a white solid. Mp: 237-239 ºC. MS (ES, positive mode): 342 (M+H)+. 1H NMR (400 MHz, DMSO-d6) δ 1.25 (t, J = 7.5 Hz, 3H, CH2CH3), 1.58-1.70 (m, 2H, H-3’a(b), H-5’a(b)), 1.99-2.08 (m, 2H, H-3’b(a), H-5’b(a)), 2.72 (q, J = 7.5 Hz, CH2CH3), 3.49 (ddd, J = 11.9, 9.5, 2.7 Hz, 2H, H-2’a(b), H-6’a(b)), 3.87 (dt, J = 11.7, 4.3 Hz, 2H, H-2’b(a), H-6’b(a)), 4.68 (m, 1H, OCH), 7.15 (ddd, J = 8.2, 2.5, 1.0 Hz, 1H, Ar), 7.54 (t, J = 8.1 Hz, 1H, Ar), 7.61 (ddd, J = 8.1, 1.9, 1.0 Hz, 1H, Ar), 7.70 (t, J = 2.2 Hz, 1H, Ar), 12.72 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.1 (CH2CH3), 27.7 (CH2CH3), 31.6 (C-3’, C5’), 64.5 (C-2’, C-6’), 71.9 (OCH), 109.1, 114.0, 116.5 (Ar), 128.9 (C-7a), 130.7, 136.4 (Ar), 148.6 (C-3a), 155.9, 157.5 (C-5, Ar), 164.6 (C-7). Anal calcd for (C17H19N5O3): C, 59.81; H, 5.61; N, 20.52. Found: C, 59.67; H, 5.64; N, 20.31. tert-Butyl

4-(3-(5-ethyl-7-oxo-triazolo[4,5-d]pyrimidin-3-yl)phenoxy)piperidine-1-

carboxylate (9c). A microwave vial was charged with 8c (360 mg, 0.9 mmol), tBuOK 1M in THF (3.6 mL, 3.6 mmol) and tert-butyl propionate (0.54 mL, 3.6 mmol) in anhydrous dioxane (9 mL). The reaction mixture was heated in a microwave reactor at 100 ºC for 1h. The mixture was concentrated to dryness. Water was added (15 mL) and the mixture was extracted with ethyl acetate (3 x 10 mL). The organic phase was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (dichloromethane/methanol, 95:5) to yield 296 mg (74%) of 9c as a white solid. Mp: 210-212 ºC. MS (ES, positive mode): m/z 441 (M+H)+, 881 (2M+H)+. 1H NMR (300 MHz, DMSO-d6): δ 1.25 (t, J = 7.5 Hz, 3H, CH2CH3), 1.40 (s, 9H, (CH3)3), 1.59 (m, 2H, CH2CH2N), 1.70 (m, 2H, CH2CH2N), 2.73 (q, J = 7.5 Hz, 2H, CH2CH3), 3.19 (m, 2H, CH2CH2N), 3.69 (m, 2H, CH2CH2N), 4.67 (m, 1H, OCH), 7.16 (m, 1H, Ar), 16 ACS Paragon Plus Environment

Page 17 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

7.55 (pt, J = 8.0 Hz, 1H, Ar), 7.60-7.64 (m, 1H, Ar), 7.69 (pt, J = 2.1 Hz, 1H, Ar), 12.70 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.7 (CH2CH3), 28.4 (CH2CH3), 28.7 (C(CH3)3), 30.9 (C3’, C-5’) 40.8 (C-2’, C-6’), 73.1 (C-4’), 79.3 (C(CH3)3), 110.0, 114.8, 117.1, 131.4 (Ar), 129.6 (C7a), 137.0 (CArN), 149.3 (C-3a), 154.6, 156.5, 158.2 (NCOO, C-5, CArO), 165.2 (C-7). Anal. Calc. for C22H28N6O4: C, 59.99; H, 6.41; N, 19.08. Found: C, 59.77; H, 6.41; N, 18.89. Trifluoroacetate

salt

of

5-ethyl-3-(3(piperidine-4-yloxy)phenyl)-3H-[1,2,3]triazolo[4,5-

d]pyrimidin-7(6H)-one (9d). A solution of 9c (35 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt for 2 h and concentrated to dryness. The residue obtained was treated with dichloromethane/hexane and concentrated to dryness. This procedure was repeated several times until a permanent weight was obtained (quantitative yield). A small portion was crystallized in MeOH/Et2O. Mp: 186-188 ºC. MS (ES, positive mode): m/z 341 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.24 (t, J = 7.5 Hz, 3H, CH2CH3), 1.80-1.95 (m, 2H, H-3’a(b), H-5’a(b)), 2.10-2.20 (m, 2H, H-3’b(a), H-5’b(a)), 2.73 (q, J = 7.5 Hz, 2H, CH2CH3), 3.11-.313 (m, 2H, H-2’a(b), H-6’a(b)), 3.273.30 (m, 2H, H-2’b(a), H-6’b(a)), 4.78 (m, 1H, H-4’), 7.20 (dd, J = 8.3, 2.5 Hz, 1H, Ar), 7.58 (pt, J = 8.1 Hz, 1H, Ar), 7.68 (m, 1H, Ar), 7.69 (pt, J = 2.2 Hz, 1H, Ar), 8.64 (br s, 2H, NH2), 12.76 (br s, 1H, NH).

13

C NMR (101 MHz, DMSO d6): δ 11.8 (CH2CH3), 27.8 (C-3’, C-5’), 28.4 (CH2CH3),

41.3 (C-2’, C-6’), 70.1 (C-4’), 110.2, 115.3, 117.0, 135.1 (Ar), 129.6 (C-7a), 137.1 (CArN), 149.3 (C-3a), 156.5, 157.8 (C-5, CArO), 165.3 (C-7). Anal. Calc. for C17H20N6O2·TFA·0.5H2O: C, 49.24; H, 4.79; N, 18.14. Found: C, 49.42; H, 4.93; N, 18.26. tert-Butyl 4-(3-azidobenzoyl)piperazine-1-carboxylate (11) A solution of tert-butyl piperazine1-carboxylate (0.14 g, 0.74 mmol), DIPEA (0.26 mL, 1.47 mmol) and HCTU (0.46 g, 0.88 mmol) in anhydrous dichloromethane (3 mL) was stirred at rt for 20 min. A solution of 3-azidobenzoic acid 1031 (0.12 g, 0.74 mmol) in anhydrous dichloromethane/DMF (9:1) (2 mL) was added and the reaction mixture was stirred at rt for 2 h. The reaction mixture was dissolved in dichloromethane (10 mL) and washed with a 5% aqueous solution of citric acid (5 mL), a saturated aqueous solution of NaHCO3 (5 mL) and brine (5 mL). The combined organic phase was dried on anhydrous Na2SO4, filtered and evaporated. The residue was purified by CCTLC (dichloromethane/methanol, 10:0.2) to 17 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 51

yield 0.19 g (77%) of 11 as yellow solid. Mp: 103-105 ºC. MS (ES, positive mode): m/z 332 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.39 (s, 9H, C(CH3)3), 3.26-3.39 (m, 6H, H-2’, H-6’, H3’a(b), H-5’a(b)), 3.55-3.57 (m, 2H, H-3’b(a), H-5’b(a)), 7.11 (t, J = 1.8 Hz, 1H, Ar), 7.22-7.16 (m, 2H, Ar), 7.46 (t, J = 7.8 Hz, 1H, Ar). tert-Butyl 4-(3-(5-amino-4-carbamoyl-1H-1,2,3-triazol-1-yl)benzoyl)piperazine-1-carboxylate (12). Following general procedure A, to a solution of cyanoacetamide (43 mg, 0.52 mmol) in anhydrous DMF (2.0 mL), NaH (60% mineral oil, 34 mg, 0.84 mmol) was slowly added at 0 ºC, followed by the addition of a solution of 11 (0.15 g, 0.47 mmol) in anhydrous DMF (1.5 mL). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:1) to yield 0.16 g (84%) of 12 as white solid. Mp: 125-127 ºC. MS (ES, positive mode): m/z 416 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.39 (s, 9H, C(CH3)3), 3.35-3.40 (m, 6H, H-2’, H-6’, H-3’a(b), H-5’a(b)), 3.57-3.59 (m, 2H, H-3’b(a), H-5’b(a)), 6.43 (s, 2H, NH2), 7.22 (s, 1H, CONH2a(b)), 7.54-7.56 (m, 1H, Ar), 7.59-7.60 (m, 1H, Ar), 7.61 (s, 1H, CONH2b(a)), 7.66-7.67 (m, 2H, Ar). tert-Butyl

4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3-

yl)benzoyl)piperazine-1-carboxylate (13). Following general procedure B, a microwave vial was charged with 12 (0.14 g, 0.035 mmol), tBuOK (1M in THF, 0.13 mL, 0.13 mmol) and tert-butyl propionate (19 µL, 0.13 mmol) in anhydrous dioxane (1.0 mL). After work-up, the residue was purified by CCTLC (hexane/ethyl acetate, 1:9) to yield 42 mg (26%) of 13 as white amorphous solid. MS (ES, positive mode): m/z 454 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.26 (t, J = 7.5 Hz, 3H, CH2CH3),1.41 (s, 9H, C(CH3)3), 2.74 (q, J = 7.5 Hz, 2H, CH2CH3), 3.40-3.43 (m, 6H, H-2’, H-6’, H-3’a(b), H-5’a(b)), 3.62-3.64 (m, 2H, H-3’b(a), H-5’b(a)), 7.60 (d, J = 7.7 Hz, 1H, Ar), 7.76 (t, J = 7.9 Hz, 1H, Ar), 8.08- 8.20 (m, 2H, Ar), 12.76 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.7 (CH2CH3), 28.3 (CH2CH3), 28.5 (C(CH3)3), 42.0, 43.5 (C-2’, C-6’), 41.9, 47.5 (C-3’, C-5’), 79.7 (C(CH3)3), 120.9, 123.4, 127.9, 130.6 (Ar), 129.4 (C-7a), 135.7 (CArCO), 137.5 (CArN), 149.3 (C-3a), 154.3, 156.3 (C-5, COC(CH3)3), 165.3, 168.3 (CArCON, C-7). Anal calcd for (C22H27N7O4): C, 58.27; H, 6.00; N, 21.62. Found: C, 58.19; H, 6.20; N, 21.22.

18 ACS Paragon Plus Environment

Page 19 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

General procedure for the synthesis of carbamates derived from 5-ethyl-3-(3-(piperidin-4yloxy)phenyl)-3,6-dihydro-7H-[1,2,3]triazolo[4,5-d]pyrimidin-7-one

(14a-e).

(General

procedure C). A solution of 9c (1.0 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt for 2 h. The mixture was concentrated to dryness. The residue obtained was treated with dichloromethane/hexane and concentrated to dryness. This procedure was repeated several times until a permanent weight was obtained. This obtained yellow solid was dissolved in anhydrous dichloromethane (1 mL) and triethylamine (3.0 mmol), DMAP (0.4 mmol) and the appropriate chloroformate (1.5 mmol) in anhydrous DCM (1mL) were added at 0 ºC. After 3 h of stirring at rt, the reaction was concentrated to dryness. A saturated solution of NaCl (10 mL) was added and the mixture was extracted with ethyl acetate (3×10 mL). The combined organic extracts were dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography to yield the corresponding carbamates 14a-e. Phenyl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxylate (14a). Following the general procedure C, a solution of 9c (44 mg, 0.1 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with triethylamine (42 µL, 0.3 mmol), DMAP (5 mg, 0.04 mmol) and phenylchloroformate (19 µL, 0.15 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:1) to yield 14 mg (30%) of 14a as a white solid. MS (ES, positive mode): m/z 461 (M+H)+. Mp > 225 ºC (decomp). 1H NMR (400 MHz, DMSO-d6): δ 1.23 (t, J = 7.5 Hz, 3H, CH2CH3), 1.71-1.73 (m, 2H, H-3’a(b), H-5’a(b)), 2.05-2.08 (m, 2H, H-3’b(a), H-5’b(a)), 2.70 (q, J = 7.5 Hz, 2H, CH2CH3), 3.263.48 (m, 2H, H-2’a(b), H-6’a(b)), 3.75-3.89 (m, 2H, H-2’b(a), H-6’b(a)), 4.75 (tt, J = 7.8, 3.6 Hz, 1H, H4’), 7.10-7.12 (m, 2H, Ar), 7.15-7.21 (m, 2H, Ar), 7.34-7.38 (m, 2H, Ar), 7.54 (pt, J = 8.1 Hz, 1H, Ar), 7.61 (m, 1H, Ar), 7.70 (pt, J = 2.2 Hz, 1H, Ar), 12.71 (br s, 1H, NH).

13

C NMR (101 MHz,

DMSO d6): δ 11.2 (CH2CH3), 27.9 (CH2CH3), 40.2 (C-2’, C-6’), 72.1 (C-4’), 109.3, 114.2, 116.5, 122.0, 125.2, 129.3, 130.8 (Ar), 129.0 (C-7a), 136.5 (CArN), 148.8 (C-3a), 150.5, 151.2, 153.0, 157.5 (NCOO, C-5, CArO), 164.8 (C-7). Anal. Calc. for C24H24N6O4: C, 62.60; H, 5.25; N, 18.25. Found: C, 62.24, H, 5.28; N, 17.88. 19 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 51

Benzyl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxylate (14b). Following the general procedure C, a solution of 9c (36 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with triethylamine (40 µL, 0.28 mmol), DMAP (5 mg, 0.04 mmol) and benzylchloroformate (22 µL, 0.15 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:1) to yield 23 mg (60%) of 14b as a white solid. Mp: 115-117 ºC. MS (ES, positive mode): m/z 475 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.23 (t, J = 7.5 Hz, 3H, CH2CH3), 1.57-1.65 (m, 2H, H3’a(b), H-5’a(b)), 1.95-2.01 (m, 2H, H-3’b(a), H-5’b(a)), 2.71 (q, J = 7.5 Hz, 2H, CH2CH3), 3.24-3.29 (m, 2H, H-2’a(b), H-6’a(b)), 3.71-3.77 (m, 2H, H-2’b(a), H-6’b(a)), 4.68 (m, 1H, H-4’), 5.07 (s, 2H, OCH2), 7.15 (d, J = 8.1 Hz, 1H, Ar), 7.28-7.38 (m, 5H, Ar), 7.55 (pt, J = 8.2 Hz, 1H, Ar), 7.60 (d, J = 8.1 Hz, 1H, Ar), 7.68 (pt, J = 2.2 Hz, 1H, Ar), 12.70 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.7 (CH2CH3), 28.4 (CH2CH3), 41.5 (C-2’, C-6’), 66.9 (OCH2), 72.9 (C-4’), 110.0, 114.9, 117.2, 128.2, 128.5, 129.6, 131.5 (Ar), 129.1 (C-7a ), 137.1, 137.7 (CArC, CArN), 149.3 (C-3a), 155.1, 156.5, 158.2 (NCOO, C-5, CArO), 165.3 (C-7). Anal. Calc. for C25H26N6O4·H2O: C, 60.97; H, 5.73; N, 17.06. Found: C, 61.38; H, 5.52; N, 16.92. Allyl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxylate (14c). Following the general procedure C, a solution of 9c (35 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with triethylamine (40 µL, 0.28 mmol), DMAP (5 mg, 0.04 mmol) and allylchloroformate (16 µL, 0.15 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:1) to yield 28 mg (83%) of 14c as a white solid. Mp: 182-184 ºC. MS (ES, positive mode): m/z 425 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.22 (t, J = 7.5 Hz, 3H, CH2CH3), 1.56-1.63 (m, 2H, H3’a(b), H-5’a(b)),1.93-2.00 (m, 2H, H-3’b(a), H-5’b(a)), 2.69 (q, J = 7.5 Hz, 2H, CH2CH3), 3.20-3.27 (m, 2H, H-2’a(b), H-6’a(b)), 3.68-3.74 (m, 2H, H-2’b(a), H-6’b(a)), 4.50 (dt, J = 5.2, 1.6 Hz, 2H, OCH2), 4.68 (m, 1H, H-4’), 5.16 (dq, J = 10.5, 1.5 Hz, 1H, CH=CH2(cis)), 5.25 (dq, J = 17.2, 1.7 Hz, 1H, CH=CH2(trans)), 5.90 (ddt, J = 17.3, 10.5, 5.2 Hz, 1H, CH=CH2), 7.13 (dd, J = 8.2, 2.5 Hz, 1H, Ar), 7.52 (pt, J = 8.1 Hz, 1H, Ar), 7.59 (dd, J = 8.0, 1.9 Hz, 1H, Ar), 7.67 (pt, J = 2.1 Hz, 1H, Ar), 12.70 20 ACS Paragon Plus Environment

Page 21 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

(br s, 1H, NH). 13C NMR (101 MHz, DMSO d6): δ 11.5 (CH2CH3), 28.2 (CH2CH3), 30.6 (C-3’, C5’) 41.1 (C-2’, C-6’), 65.6 (OCH2), 72.6 (C-4’), 109.7, 114.6, 116.9, 117.4, 131.2 (Ar, CH=CH2), 129.4 (C-7a), 133.9 (CH=CH2), 136.8 (CArN), 149.1 (C-3a), 154.6, 156.3, 157.9 (NCOO, C-5, CArO), 165.0 (C-7). Anal. Calc. for C21H24N6O4·H2O: C, 57.00; H, 5.92; N, 18.99. Found: C, 56.71; H, 6.20; N, 18.77. Isobutyl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxylate (14d). Following the general procedure C, a solution of 9c (42 mg, 0.09 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with triethylamine (40 µL, 0.28 mmol), DMAP (5 mg, 0.04 mmol) and isobutylchloroformate (19 µL, 0.15 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:1) to yield 8 mg (20%) of 14d as a white solid. Mp: 110-112 ºC. MS (ES, positive mode): m/z 441 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 0.87 (d, J = 6.7 Hz, 6H, CH(CH3)3), 1.23 (t, J = 7.5 Hz, 3H, CH2CH3), 1.55-1.64 (m, 2H, H-3’a(b), H-5’a(b)), 1.85 (hept, J = 6.7 Hz, 1H, CH(CH3)3), 1.902.00 (m, 2H, H-3’b(a), H-5’b(a)), 2.71 (q, J = 7.5 Hz, 2H, CH2CH3), 3.22-3.24 (m, 2H, H-2’a(b), H6’a(b)), 3.69-3.73 (m, 2H, H-2’b(a), H-6’b(a)), 3.78 (d, J = 6.6 Hz, 2H, OCH2), 4.68 (m, 1H, H-4’), 7.14 (dd, J = 8.3, 2.5 Hz, 1H, Ar), 7.53 (pt, J = 8.1 Hz, 1H, Ar), 7.61 (d, J = 8.1 Hz, 1H, Ar), 7.68 (pt, J = 2.2 Hz, 1H, Ar), 12.70 (br s, 1H, NH).

13

C NMR (101 MHz, DMSO d6): δ 11.8 (CH2CH3), 19.5

(CH(CH3)2), 28.2, 28.5 (CH2CH3, CH(CH3)2), 41.4 (C-2’, C-6’), 71.4 (OCH2), 73.0 (C-4’), 109.9, 114.8, 117.1, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN), 149.4 (C-3a), 155.3, 156.8, 158.2 (NCOO, C5, CArO), 165.4 (C-7). Anal. Calc. for C22H28N6O4·H2O: C, 57.63; H, 6.60; N, 18.33. Found: C, 57.95; H, 6.24; N, 17.97. Tetrahydro-2H-pyran-4-yl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5d]pyrimidin-3-yl)phenoxy)piperidine-1-carboxylate (14e). Following the general procedure C, a solution of 9c (36 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with triethylamine (40 µL, 0.28 mmol), DMAP (5 mg, 0.04 mmol) and tetrahydro-2H-pyran-4-yl chloroformate38 (25 mg, 0.15 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:1) to yield 8 mg (18%) of 14e as a white solid. 21 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 51

Mp: 174-176 ºC. MS (ES, positive mode): m/z 469 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.25 (t, J = 7.8 Hz, 3H, CH2CH3), 1.49-1.65 (m, 4H, H-3’a(b), H-5’a(b), H-3’’a(b), H-5’’a(b)), 1.82-1.88 (m, 2H, H-3’’b(a), H-5’’b(a)), 1.97-2.02 (m, 2H, H-3’b(a), H-5’b(a)), 2.73 (q, J = 7.5 Hz, 2H, CH2CH3), 3.27 (m, 2H, H-2’a(b), H-6’a(b)), 3.46 (ddd, J = 11.6, 8.6, 3.1 Hz, 2H, H-2’’a(b), H-6’’a(b)), 3.67-3.85 (m, 4H, H-2’b(a), H-6’b(a), H-2’’b(a), H-6’’b(a)), 4.68-4.77 (m, 2H, H-4’, H-4’’), 7.16 (dd, J = 8.3, 2.5 Hz, 1H, Ar), 7.56 (pt, J = 8.1 Hz, 1H, Ar), 7.63 (d, J = 8.0 Hz, 1H, Ar), 7.70 (pt, J = 2.2 Hz, 1H, Ar), 12.73 (br s, 1H, NH). 13C NMR (101 MHz, DMSO d6): δ 11.1 (CH2CH3), 27.7 (CH2CH3), 31.8 (C-3’’, C5’’), 40.7 (C-2’, C-6’), 64.4 (C-2’’, C-6’’), 69.4 (C-4’’), 72.3 (C-4’), 109.3, 114.2, 116.5, 130.8 (Ar), 128.9 (C-7a), 136.4 (CArN), 148.7 (C-3a), 153.9, 155.8, 157.5 (NCOO, C-5, CArO), 164.6 (C-7). Anal. Calc. for C23H28N6O5·H2O: C, 56.78; H, 6.22; N, 17.27. Found: C, 57.06; H, 6.19; N, 17.18. General procedures (D and E) for the synthesis of ureas derived from 5-ethyl-3-(3(piperidin-4-yloxy)phenyl)-3,6-dihydro-7H-[1,2,3]triazolo[4,5-d]pyrimidin-7-one

(15a-h).

General Procedure D. A solution of 9c (1.0 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt for 2 h and concentrated to dryness. The residue obtained was treated with dichloromethane/hexane and concentrated to dryness. This procedure was repeated several times until a permanent weight was obtained. A solution containing this yellow solid, the corresponding amine (1.0 mmol) and DIPEA (2.2 mmol) in anhydrous dichloromethane (3.5 mL) or dichloromethane/DMF (9:1, 3.5 mL) was treated a solution of triphosgene (0.37 mmol) in dichloromethane (0.5 mL). The reaction mixture was stirred at rt for 30 min. A saturated aqueous solution of NaHCO3 (5 mL) was added and the mixture was extracted with dichloromethane (10 mL). The organic phase was washed with brine (5 mL), dried on anhydrous Na2SO4, filtered and evaporated. The residue was purified as indicated in each case. General Procedure E. A solution of 9c (1.0 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt for 2 h and concentrated to dryness. The residue obtained was treated with dichloromethane/hexane and concentrated to dryness. This procedure was repeated several times until a permanent weight was obtained. A solution containing this yellow solid and Et3N (2.1 mmol) in anhydrous dichloromethane (10 mL) or dichloromethane/DMF (9:1, 10 mL) was stirred at 0 ºC 22 ACS Paragon Plus Environment

Page 23 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

for 30 min. The corresponding isocyanate (1.5 mmol) was added at 0 ºC and the reaction mixture was stirred at rt for 1 h. A saturated aqueous solution of NH4Cl (5 mL) was added and the mixture was extracted with dichloromethane (2×10 mL). The combined organic phase was dried on anhydrous Na2SO4, filtered and evaporated. The residue was purified as indicated in each case. 4-(3-(5-Ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3-yl)phenoxy)-Nphenylpiperidine-1-carboxamide (15a). Following the general procedure D, a solution of 9c (18 mg, 0.04 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with aniline (4 µL, 0.04 mmol), DIPEA (16 µL, 0.09 mmol) and triphosgene (4.50 mg, 0.02 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 10 mg (54%) of 15a as a white solid. Mp: 259-261 ºC. MS (ES, positive mode): m/z 460 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.24 (t, J = 7.5 Hz, 3H, CH2CH3), 1.59-1.68 (m, 2H, H-3’a(b), H-5’a(b)), 1.99-2.05 (m, 2H, H-3’b(a), H-5’b(a)), 2.72 (q, J = 7.5 Hz, 2H, CH2CH3), 3.24-3.33 (m, 2H, H-2’a(b), H-6’a(b)), 3.81-3.87 (m, 2H, H-2’b(a), H-6’b(a)), 4.70 (m, 1H, H-4’), 6.91 (t, J = 7.3 Hz, 1H, Ar), 7.157.23 (m, 3H, Ar), 7.42-7.45 (m, 2H, Ar), 7.55 (t, J = 8.1 Hz, 1H, Ar), 7.61 (m, 1H, Ar), 7.70 (s, 1H, Ar), 8.55 (br s, 1H, ArNHCO), 12.72 (br s, 1H, NH).

13

C NMR (101 MHz, DMSO-d6): δ 11.8

(CH2CH3), 28.4 (CH2CH3), 31.1 (C-3’, C-5’), 41.8 (C-2’, C-6’), 73.4 (C-4’), 109.9, 114.8, 117.2, 120.3, 122.3, 128.9, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN), 141.2 (CArN) 149.3 (C-3a), 155.6, 156.5, 158.2 (ArNHCO, C-5, CArO), 165.2 (C-7). Anal calcd for (C24H25N7O3. 0.5H2O): C, 61.53; H, 5.59; N, 20.93. Found: C, 61.44; H, 5.82; N, 20.88. 4-(3-(5-Ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3-yl)phenoxy)-N-(4methoxyphenyl)piperidine-1-carboxamide (15b). Following the general procedure E, a solution of 9c (37 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with 4-methoxyphenyl isocyanate (17 µL, 0.13 mmol) and Et3N (25 µL, 018 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 30 mg (73%) of 15b as a white solid. Mp: 229-231 ºC. MS (ES, positive mode): m/z 490 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.23 (t, J = 7.5 Hz, 3H, CH2CH3), 1.57-1.65 (m, 2H, H-3’a(b), H-5’a(b)), 1.98-2.02 (m, 2H, H-3’b(a), H-5’b(a)), 2.70 (q, J = 7.5 Hz, 2H, CH2CH3), 3.20-3.26 (m, 2H, H-2’a(b), 23 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 51

H-6’a(b)), 3.66 (s, 3H, OCH3), 3.78-3.84 (m, 2H, H-2’b(a), H-6’b(a)), 4.68 (m, 1H, H-4’), 6.78 (d, J = 9.2 Hz, 2H, Ar), 7.14 (d, J = 8.3 Hz, 1H, Ar), 7.30 (d, J = 8.3 Hz, 2H, Ar), 7.53 (d, J = 8.1 Hz, 1H, Ar), 7.59 (d, J = 8.1 Hz, 1H, Ar), 7.69 (s, 1H, Ar), 8.37 (br s, 1H, ArNHCO), 12.70 (br s, 1H, NH). 13

C NMR (101 MHz, DMSO-d6): δ 11.8 (CH2CH3), 28.4 (CH2CH3), 31.1 (C-3’, C-5’), 41.8 (C-2’,

C-6’), 55.8 (OCH3), 73.4 (C-4’), 109.9, 114.1, 114.8, 117.2, 122.3, 131.4 (Ar), 129.6 (C-7a), 134.2 (CArN), 137.1 (CArN), 149.3 (C-3a), 155.1, 155.8, 156.6 (ArNHCO, C-5, CArO), 158.3 (CArO), 165.2 (C-7). Anal calcd for (C25H27N7O4·0.5H2O): C, 60.23; H, 5.66; N, 19.67. Found: C, 59.91; H, 5.57; N, 19.29. N-Benzyl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxamide (15c). Following the general procedure E, a solution of 9c (36 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with benzyl isocyanate (15 µL, 0.12 mmol) and Et3N (24 µL, 017 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 28 mg (71%) of 15c as a white solid. Mp: 217-219 ºC. MS (ES, positive mode): m/z 474 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.22 (t, J = 7.5 Hz, 3H, CH2CH3), 1.49-1.57 (m, 2H, H-3’a(b), H-5’a(b)), 1.94-1.97 (m, 2H, H-3’b(a), H-5’b(a)), 2.69 (q, J = 7.5 Hz, 2H, CH2CH3), 3.09-3.16 (m, 2H, H-2’a(b), H-6’a(b)), 3.70-3.76 (m, 2H, H-2’b(a), H-6’b(a)), 4.21 (d. J = 5.6 Hz, 2H, CH2Ph), 4.64 (m, 1H, H-4’), 7.10-7.29 (m, 7H, Ar, BnNHCO), 7.52 (t, J = 8.1 Hz, 1H, Ar), 7.58 (s, 1H, Ar), 7.67 (m, 1H, Ar), 12.70 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.8 (CH2CH3), 28.4 (CH2CH3), 31.1 (C-3’, C-5’), 41.6 (C-2’, C-6’), 44.2 (CH2Ph), 73.5 (C-4’), 109.8, 114.8, 117.2, 127.1, 127.6, 128.8, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN), 141.2 (CArCH2) 149.3 (C-3a), 156.5, 157.9, 158.2 (BnNHCO, C-5, CArO), 165.2 (C-7). Anal calcd for (C25H27N7O3·2H2O): C, 58.93; H, 6.13; N, 19.24. Found: C, 58.62; H, 6.00; N, 18.87. N-Ethyl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxamide (15d). Following the general procedure E, a solution of 9c (72 mg, 0.16 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with ethyl isocyanate (20 µL, 0.24 mmol) and Et3N (48 µL, 0.34 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 16 mg (25%) of 15d as a white 24 ACS Paragon Plus Environment

Page 25 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

solid. Mp: > 255 ºC (decomp.). MS (ES, positive mode): m/z 412 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 0.99 (t, J = 7.1 Hz, 3H, NHCH2CH3), 1.23 (t, J = 7.5 Hz, 3H, CH2CH3), 1.47-1.56 (m, 2H, H-3’a(b), H-5’a(b)), 1.91-1.96 (m, 2H, H-3’b(a), H-5’b(a)), 2.71 (q, J = 7.5 Hz, 2H, CH2CH3), 2.963.10 (m, 4H, H-2’a(b), H-6’a(b), NHCH2CH3), 3.65-3.71 (m, 2H, H-2’b(a), H-6’b(a)), 4.63 (m, 1H, H-4’), 6.49 (t, J = 5.4 Hz, 1H, EtNHCO), 7.13 (m, 1H, Ar), 7.53 (t, J = 8.1 Hz, 1H, Ar), 7.59 (m, 1H, Ar), 7.67 (s, 1H, Ar), 12.71 (br s, 1H, NH).

13

C NMR (101 MHz, DMSO-d6): δ 11.8 (CH2CH3), 16.3

(NHCH2CH3), 28.4 (CH2CH3), 31.0 (C-3’, C-5’), 35.6 (NHCH2CH3), 41.5 (C-2’, C-6’), 73.6 (C-4’), 109.8, 114.7, 117.2, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN) 149.3 (C-3a), 156.5, 157.9, 158.3 (EtNHCO, C-5, CArO), 165.2 (C-7). Anal calcd for (C20H25N7O3·H2O): C, 55.93; H, 6.34; N, 22.83. Found: C, 56.37; H, 6.10; N, 22.48. N-(tert-Butyl)-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxamide (15e) Following the general procedure E, a solution of 9c (39 mg, 0.09 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with tert-butyl isocyanate (15 µL, 0.13 mmol) and Et3N (25 µL, 018 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 30 mg (78%) of 15e as a white solid. Mp: 223-225 ºC . MS (ES, positive mode): m/z 440 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.20-1.24 (m, 3H, CH2CH3), 1.22 (s, 9H, C(CH3)3), 1.46-1.55 (m, 2H, H-3’a(b), H5’a(b)), 1.89-1.94 (m, 2H, H-3’b(a), H-5’b(a)), 2.69 (q, J = 7.5 Hz, 2H, CH2CH3), 2.99-3.05 (m, 2H, H2’a(b), H-6’a(b)), 3.64-3.67 (m, 2H, H-2’b(a), H-6’b(a)), 4.60 (m, 1H, H-4’), 5.80 (s, 1H, tBuNHCO), 7.11 (m, 1H, Ar), 7.51 (t, J = 8.1 Hz, 1H, Ar), 7.56 (m, 1H, Ar), 7.66 (s, 1H, Ar), 12.70 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.8 (CH2CH3), 28.4 (CH2CH3), 29.9 (C(CH3)3), 31.1 (C3’, C-5’), 41.8 (C-2’, C-6’), 50.6 (C(CH3)3), 73.7 (C-4’), 109.8, 114.7, 117.4, 131.4 (Ar), 129.6 (C7a), 137.1 (CArN) 149.3 (C-3a), 156.5, 157.6, 158.3 (tBuNHCO, C-5, CArO), 165.2 (C-7). Anal calcd for (C22H29N7O3): C, 60.12; H, 6.65; N, 22.31. Found: C, 59.89; H, 6.61; N, 21.91. N-Cyclohexyl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3yl)phenoxy)piperidine-1-carboxamide (15f) Following the general procedure D, a solution of 9c (42 mg, 0.09 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction 25 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 51

with cyclohexylamine (11 µL, 0.09 mmol), DIPEA (36 µL, 0.21 mmol) and triphosgene (10 mg, 0.03 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 10 mg (24%) of 15f as a white solid. Mp: 245-247 ºC. MS (ES, positive mode): m/z 466 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.01-1.20 (m, 5H, Cy), 1.23 (t, J = 7.5 Hz, 3H, CH2CH3), 1.48-1.57 (m, 3H, Cy, H-3’a(b), H-5’a(b)), 1.64-1.74 (m, 4H, Cy), 1.90-1.97 (m, 2H, H3’b(a), H-5’b(a)), 2.71 (q, J = 7.5 Hz, 2H, CH2CH3), 3.06-3.10 (m, 2H, H-2’a(b), H-6’a(b)), 3.36 (m, 1H, Cy), 3.67-3.72 (m, 2H, H-2’b(a), H-6’b(a)), 4.62 (m, 1H, H-4’), 6.19 (d, J = 2.2 Hz, 1H, CyNHCO), 7.12 (m, 1H, Ar), 7.53 (t, J = 8.5 Hz, 1H, Ar), 7.60 (m, 1H, Ar), 7.67 (s, 1H, Ar), 12.70 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.8 (CH2CH3), 25.8 (Cy), 26.1 (Cy), 28.4 (CH2CH3), 31.0 (C-3’, C-5’), 33.8 (Cy), 41.6 (C-2’, C-6’), 49.9 (Cy), 73.6 (C-4’), 109.8, 114.7, 117.1, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN), 149.3 (C-3a), 156.5, 157.7, 158.2 (CyNHCO, C-5, CArO), 165.2 (C-7). Anal calcd for (C24H31N7O3·2H2O): C, 57.47; H, 7.03; N, 19.55. Found: C, 57.39; H, 6.67; N, 19.21. N-((1S,3R,5S)-Adamantan-1-yl-4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5d]pyrimidin-3-yl)phenoxy)piperidine-1-carboxamide (15g) Following the general procedure E, a solution of 9c (37 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with adamantyl isocyanate (22 mg, 0.13 mmol) and Et3N (25 µL, 018 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 26 mg (61%) of 15g as a white solid. Mp: 228-230 ºC. MS (ES, positive mode): m/z 518 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.22 (t, J = 7.5 Hz, 3H, CH2CH3), 1.46-1.60 (m, 9H, H-Ad, H-3’a(b), H-5’a(b)), 1.88-1.97 (m, 13H, H-Ad, H-3’b(a), H-5’b(a)), 2.69 (q, J = 7.5 Hz, 2H, CH2CH3), 2.98-3.04 (m, 2H, H-2’a(b), H-6’a(b)), 3.63-3.66 (m, 2H, H-2’b(a), H-6’b(a)), 4.59 (m, 1H, H-4’), 5.68 (br s, 1H, NH-Ad), 7.11 (m, 1H, Ar), 7.51 (t, J = 8.1 Hz, 1H, Ar), 7.58 (m, 1H, Ar), 7.66 (s, 1H, Ar), 12.70 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.8 (CH2CH3), 28.4 (CH2CH3), 29.7 (C-Ad), 31.0 (C-3’, C-5’), 41.9 (C-2’, C-6’), 42.3 (C-Ad), 51.0 (C-Ad), 73.7 (C-4’), 109.8, 114.7, 117.1, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN) 149.3 (C-3a), 155.6, 157.2, 158.3 (AdNHCO, C-5, CArO), 165.3 (C-7). Anal calcd for (C28H35N7O3·0.5H2O): C, 63.86; H, 6.89; N, 18.62. Found: C, 64.22; H, 6.86; N, 18.19. 26 ACS Paragon Plus Environment

Page 27 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

5-Ethyl-3-(3-((1-(morpholine-4-carbonyl)piperidin-4-yl)oxy)phenyl)-3,6-dihydro-7H[1,2,3]triazolo[4,5-d]pyrimidin-7-one (15h). Following the general procedure D, a solution of 9c (40 mg, 0.09 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with morpholine (8 µL, 0.09 mmol), DIPEA (35 µL, 0.20 mmol) and triphosgene (10 mg, 0.03 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 12 mg (26%) of 15h as a white solid. Mp: 238-240 ºC. MS (ES, positive mode): m/z 454 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.22 (t, J = 7.5 Hz, 3H, CH2CH3), 1.57-1.64 (m, 2H, H3’a(b), H-5’a(b)), 1.94-2.04 (m, 2H, H-3’b(a), H-5’b(a)), 2.69 (q, J = 7.5 Hz, 2H, CH2CH3), 3.01-3.07 (m, 2H, H-2’a(b), H-6’a(b)), 3.09-3.12 (m, 4H, H-3’’, H-5’’), 3.42-3.48 (m, 2H, H-2’b(a), H-6’b(a)), 3.523.54 (m, 4H, H-2’’, H-6’’), 4.64 (m, 1H, H-4’), 7.12 (m, 1H, Ar), 7.52 (t, J = 8.1 Hz, 1H, Ar), 7.59 (m, 1H, Ar), 7.67 (s, 1H, Ar), 12.70 (br s, 1H, NH).

13

C NMR (101 MHz, DMSO-d6): δ 11.8

(CH2CH3), 28.5 (CH2CH3), 30.9 (C-3’, C-5’), 44.1 (C-2’, C-6’), 47.7 (C-3’’, C-5’’), 66.6 (C-2’’, C6’’), 73.4 (C-4’), 109.9, 114.7, 117.1, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN) 149.4 (C-3a), 156.7, 158.2 (C-5, CArO), 163.7, 165.4 (NCON, C-7). Anal calcd for (C22H27N7O4·H2O): C, 56.04; H, 6.20; N, 20.79. Found: C, 56.10; H, 6.01; N, 20.46. Methyl

4-(4-(3-(5-ethyl-7-oxo-6,7-dihydro-3H-[1,2,3]triazolo[4,5-d]pyrimidin-3-

yl)phenoxy)piperidin-1-yl)-4-oxobutanoate (16). Following the general procedure C, a solution of 9c (36 mg, 0.08 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt, followed by reaction with triethylamine (40 µL, 0.28 mmol), DMAP (5 mg, 0.04 mmol) and methyl 4-chloro-4oxobutanoate (40 µL, 0.3 mmol). After work-up, the residue was purified by CCTLC (dichloromethane/methanol, 10:1) to yield 14 mg (30%) of 16 as a white solid. MS (ES, positive mode): m/z 455 (M+H)+. Mp: 86-88 ºC. 1H NMR (400 MHz, DMSO-d6): δ 1.25 (t, J = 7.5 Hz, 3H, CH2CH3), 1.55 (m, 1H, H-3’/H-5’), 1.67 (m, 1H, H-3’/H-5’), 1.94-2.06 (m, 2H, H-3’/H-5’), 2.432.48 (m, 2H, CH2COO), 2.62 (m, 2H, CH2CON), 2.73 (q, J = 7,5 Hz, 2H, CH2CH3), 3.23-3.38 (m, 2H, H-2’/H-6’), 3.57 (s, 3H, CH3O), 3.76 (m, 1H, H-2’/H-6’), 3.88 (m, 1H, H-2’/H-6’), 4.74 (tt, J = 7.9, 3.7 Hz, 1H, H-4’), 7.17 (dd, J = 8.3, 1.1 Hz, 1H, Ar), 7.56 (pt, J = 8.1 Hz, 1H, Ar), 7.63 (dd, J = 8.2, 1.2 Hz, 1H, Ar), 7.71 (pt, J = 2.2 Hz, 1H, Ar).

13

C NMR (101 MHz, DMSO d6): δ 11.2 27

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 51

(CH2CH3), 27.3 (CH2CH3), 27.8 (CH2CON), 28.6, 28.8 (C-3’, C-5’), 30.8 (CH2COO), 41.9 (C-2’, C6’), 51.3 (CH3O), 72.4 (C-4’), 109.3, 114.2, 116.5, 130.8 (Ar), 129.0 (C-7a), 136.4 (CArN), 148.7 (C3a), 156.0, 157.5 (C-5, CArO), 164.7 (C-7), 169.2 (CH2CON), 173.1 (CH2COO). Anal. Calc. for C22H26N6O5·2H2O: C, 53.87; H, 6.16; N, 17.13. Found: C, 54.29; H, 5.90; N, 16.85. 5-Ethyl-3-(3-((1-nicotinoylpiperidin-4-yl)oxy)phenyl)-3,6-dihydro-7H-[1,2,3]triazolo[4,5d]pyrimidin-7-one (17a). A solution of 9c (54 mg, 0.12 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt for 2 h and then concentrated to dryness. The residue obtained was treated with dichloromethane/hexane and evaporated. This procedure was repeated several times to obtain a yellow solid, which was used without further purification. A solution of this yellow solid, DIPEA (43 µL, 0.25 mmol), PyBOP (96 mg, 0.18 mmol) and nicotinic acid (15 mg, 0.12 mmol) in anhydrous dichloromethane/DMF (9:1, 1.0 mL) was stirred at rt for 2 h. The reaction mixture was diluted with dichloromethane (10 mL) and washed with an aqueous solution of citric acid (0.1M), with a saturated aqueous solution of NaHCO3 (10 mL)3 solution and brine (10 mL). The combined organic phases were dried on anhydrous Na2SO4, filtered and evaporated. The residue was purified by CCTLC (dichloromethane/methanol/Et3N 10:0.2:0.2) to yield 15 mg (28%) of 17a as a white amorphous solid. MS (ES, positive mode): m/z 446 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.25 (t, J = 7.5 Hz, 3H, CH2CH3), 1.74-1.76 (m, 2H, H-3’a(b), H-5’a(b)), 2.04-2.08 (m, 2H, H-3’b(a), H5’b(a)), 2.73 (q, J = 7.5 Hz, 2H, CH2CH3), 3.34-3.38 (m, 2H, H-2’/H-6’), 3.53 (m, 1H, H-2’/H-6’), 4.02 (m, 1H, H-2’/H-6’), 4.80 (m, 1H, H-4’), 7.19 (m, 1H, Ar), 7.49 (dd, J = 7.8, 4.9 Hz, 1H, Ar), 7.57 (t, J = 8.1 Hz, 1H, Ar), 7.64 (m, 1H, Ar), 7.72 (pt, J = 2.1 Hz, 1H, Ar), 7.87 (dt, J = 7.9, 1.9 Hz, 1H, Ar), 8.64-8.66 (m, 2H, Ar), 12.72 (br s, 1H, NH).

13

C NMR (101 MHz, DMSO-d6): δ 11.6

(CH2CH3), 28.2 (CH2CH3), 39.2, 44.8 (C-2’, C-6’), 72.7 (C-4’), 109.9, 114.7, 116.9, 124.0, 131.2, 135.1, 147.9, 150.8 (Ar), 129.4 (C-7a), 132.4 (CArCO), 136.9 (CArN), 149.1 (C-3a), 156.3, 157.9 (C5, CArO), 165.1 (C-7), 167.3 (CArCO). Anal calcd for (C23H23N7O3. 0.5H2O): C, 60.78; H, 5.32; N, 21.57. Found: C, 60.57; H, 5.46; N, 21.38. 5-Ethyl-3-(3-((1-isonicotinoylpiperidin-4-yl)oxy)phenyl)-3,6-dihydro-7H-[1,2,3]triazolo[4,5d]pyrimidin-7-one (17b). A solution of 9c (53 mg, 0.12 mmol) in TFA (5% in dichloromethane) (2 28 ACS Paragon Plus Environment

Page 29 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

mL) was stirred at rt for 2 h and then concentrated to dryness. The residue obtained was treated with dichloromethane/hexane and evaporated. This procedure was repeated several times to obtain a yellow solid, which was used without further purification. A solution of this yellow solid, DIPEA (42 µL, 0.24 mmol), HATU (69 mg, 0.18 mmol) and isonicotinic acid (15 mg, 0.12 mmol) in anhydrous DMF (1.0 mL) was stirred at rt for 2 h. The reaction mixture was diluted with dichloromethane (10 mL) and washed with an aqueous solution of citric acid (0.1M), with a saturated aqueous solution of NaHCO3 (10 mL)3 solution and brine (10 mL). The combined organic phases were dried on anhydrous Na2SO4, filtered and evaporated. The residue was purified by CCTLC (dichloromethane/methanol/Et3N 10:0.2:0.2) to yield 37 mg (50%) of 17b as a white amorphous solid. MS (ES, positive mode): m/z 446 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.25 (t, J = 7.5 Hz, 3H, CH2CH3), 1.69-1.79 (m, 2H, H-3’/H-5’), 1.99 (m, 1H, H-3’/H-5’), 2.12 (m, 1H, H-3’/H-5’), 2.73 (q, J = 7.5 Hz, 2H, CH2CH3), 3.28 (m, 1H, H-2’/H-6’), 3.46-3.53 (m, 2H, H-2’/H6’), 4.01 (m, 1H, H-2’/H-6’), 4.80 (m, 1H, H-4’), 7.19 (m, 1H, Ar), 7.42-7.44 (m, 2H, Ar), 7.57 (t, J = 8.1 Hz, 1H, Ar), 7.64 (m, 1H, Ar), 7.71 (pt, J = 2.1 Hz, 1H, Ar), , 8.67-8.68 (m, 2H, Ar), 12.72 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.6 (CH2CH3), 28.2 (CH2CH3), 38.9, 44.4 (C-2’, C6’), 72.7 (C-4’), 109.9, 114.7, 116.9, 121.5, 131.2, 150.5 (Ar), 129.4 (C-7a), 136.9 (CArN), 144.1 (CArCO), 149.1 (C-3a), 156.3, 157.9 (C-5, CArO), 165.0 (C-7), 167.2 (CArCO). Anal calcd for (C23H23N7O3. 0.5H2O): C, 60.78; H, 5.32; N, 21.57. Found: C, 60.63; H, 5.47; N, 21.31. 5-Ethyl-3-(3-((1-(isopropylsulfonyl)piperidin-4-yl)oxy)phenyl)-3,6-dihydro-7H[1,2,3]triazolo[4,5-d]pyrimidin-7-one (18) A solution of 9c (30 mg, 0.07 mmol) in TFA (5% in dichloromethane) (2 mL) was stirred at rt for 2 h. The mixture was concentrated to dryness. The residue obtained was treatment with dichloromethane/hexane and was concentrated to dryness. This procedure was repeated several times to obtain a yellow solid, which was used without further purification. A solution of this yellow solid and Et3N (20 µL, 0.14 mmol) in anhydrous dichloromethane/DMF (9:1, 1.0 mL) was stirred at 0 ºC for 30 min. 2-Propanesulfonyl chloride (11 µL, 0.10 mmol) was added at 0 ºC and the reaction mixture was stirred at rt for 1 h. A saturated aqueous solution of NH4Cl (5 mL) was added and the mixture was extracted with dichloromethane 29 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 51

(2×5 mL). The combined organic phase was dried on anhydrous Na2SO4, filtered and evaporated. The residue was purified by CCTLC (dichloromethane/methanol, 10:0.5) to yield 20 mg (67%) of 18 as a white solid. Mp: 245-247 ºC. MS (ES, positive mode): m/z 447 (M+H)+. 1H NMR (400 MHz, DMSO-d6): δ 1.20-1.25 (m, 9H, CH2CH3, CH(CH3)2), 1.65-1.73 (m, 2H, H-3’a(b), H-5’a(b)), 1.98-2.05 (m, 2H, H-3’b(a), H-5’b(a)), 2.71 (q, J = 7.5 Hz, 2H, CH2CH3), 3.19-3.25 (m, 2H, H-2’a(b), H-6’a(b)), 3.33 (m, 1H, CH(CH3)2), 3.47-3.53 (m, 2H, H-2’b(a), H-6’b(a)), 4.68 (m, 1H, H-4’), 7.15 (m, 1H, Ar), 7.54 (t, J = 8.1 Hz, 1H, Ar), 7.61 (m, 1H, Ar), 7.68 (s, 1H, Ar), 12.71 (br s, 1H, NH). 13C NMR (101 MHz, DMSO-d6): δ 11.8 (CH2CH3), 17.1 (CH(CH3)2), 28.4 (CH2CH3), 31.3 (C-3’, C-5’), 43.5 (C-2’, C-6’), 52.5 (CH(CH3)2), 72.4 (C-4’), 110.0, 114.9, 117.1, 131.4 (Ar), 129.6 (C-7a), 137.1 (CArN), 149.3 (C-3a), 156.5, 158.0 (C-5, CArO), 165.2 (C-7). Anal calcd for (C20H26N6O4S): C, 53.80; H, 5.87; N, 18.82; S, 7.18. Found: C, 53.50; H, 5.93; N, 18.38; S, 7.11.

Virus strains and cells. Chikungunya virus (CHIKV) Indian Ocean strain 899 (Genbank FJ959103.1) was generously provided by Prof. C. Drosten (University of Bonn, Germany). CHIKV La Reunion Island strain LR2006_OPY1 (Genbank DQ443544.2) and the clinical isolates Venturini (Italy 2008), Congo 95 (2011), St Martin (2013) and Martinique EFS-1 were used in EPV in Marseille and are freely disposable from the European Virus Archive (https://www.european-virus-archive.com/). All viruses were propagated in African green monkey kidney cells [Vero cells (ATCC CCL-81) or Vero E6 (ATCC CRL-1586)]. CHIKV isolates from Singapore (SGP11) and Reunion Island (LR2006 OPY1), and O’nyong’nyong virus (ONNV) infectious clones were tagged with Gaussia luciferase (Gluc) from marine copepod Gaussia princeps at a site in between the non-structural and structural genes. Viral stocks of infectious clones were produced in Vero E6 cells, tittered by plaque assays and stored at 80 °C. Vero cells were maintained in cell growth medium composed of minimum essential medium (MEM Rega-3, Gibco, Belgium) supplemented with 10% Foetal Bovine Serum (FBS, Integro, The 30 ACS Paragon Plus Environment

Page 31 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Netherlands), 1% L-glutamine (Gibco), and 1% sodium bicarbonate (Gibco). The antiviral assays were performed in the same medium but supplemented with 2% (instead of 10%) FBS. The E6 subclone of Vero cells were maintained in Eagles MEM (Gibco) supplemented with antibiotics, 1% glutamine, 1% non essential amino-acids (Gibco) and 7.5 % FCS. Plating prior to infection and antiviral assays were done in the same medium but with reduced serum concentration (2.5%). CRL2522 human skin fibroblasts were maintained in Dulbecco’s Modified Eagle Medium (HyClone), with 10% FBS (HyClone) and 1% penicillin/streptomycin (Gibco). Antiviral assays with CRL-2522 cells were performed in serum-free DMEM. All cell cultures were maintained at 37 °C in an atmosphere of 5% CO2 and 95-99% humidity. Antiviral assays. CPE-inhibition assay. Vero cells were seeded in 96-well tissue culture plates (Becton Dickinson, Aalst, Belgium) at a density of 2.5 × 104 cells/well in 100 µl 2% FBS medium and were allowed to adhere overnight. Next, a compound dilution series was prepared in the medium on top of the cells (100 µl) after which the cultures were infected with 0.01 MOI of CHIKV 899 inoculum or 0.01 MOI of a triazolopyrimidine-resistant CHIKV variant in 100 µl 2% FBS medium. The starting concentration was 100 µg/ml for all compounds tested. Each assay was performed in 3-fold in the same test and assays were repeated three times independently to assess for inter-experiment variability. On day 5 post-infection (p.i.), the plates were processed using the MTS/PMS method as described by the manufacturer (Promega, The Netherlands). The 50% effective concentration (EC50), which is defined as the compound concentration that is required to inhibit virus-induced cytopathic effect by 50%, was determined using logarithmic interpolation. The fold resistance value is defined as the ratio of the EC50 value of the resistant CHIKV strain and the EC50 value of the wild-type CHIKV strain (=EC50 resistant CHIKV/EC50 WT CHIKV). Virus yield assay. The antiviral activity of the best compounds was validated in an assay for CHIKV OPY 1, Venturini, Congo, Martinique EFS-1 and St. Martin, as previously described.25 Antiviral activity assay with CRL-2522. The assay was performed as previously described.22 CRL2522 cells were seeded at a density of 5 x 103 cells/well in 96-well microtiter plates (Corning). After 31 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 51

overnight incubation, cells were infected with the Gluc-tagged viruses at MOI 1. Virus overlay was removed and cells were treated with serial dilutions of compounds (ranging from 0.001 to 500 µM) prepared in medium in triplicates. After 24 h, 50 µl of conditioned medium was removed and transferred to a new plate, followed by the addition of 50 µl of 20 µM Xenolight RediJect coelenterazine h (Perkin Elmer) diluted in PBS with 5 mM NaCl. Luminescent signals were immediately read by the GloMax Multi-Detection System (Promega) at 1 s integration time. Cytotoxicity assays Cytotoxicity assay in Vero A, Vero E6, BHK, HeLa RH and Huh-7 cells: The cytotoxic and cytostatic effect of the compounds was evaluated in uninfected cells by means of microscopic scoring or by the MTS/PMS method. Vero cells were seeded in 96-well tissue culture plates (Becton Dickinson, Aalst, Belgium) at a density of 2.5 × 104 cells/well in 100 µl 2% FBS medium and were allowed to adhere overnight. Next, a compound dilution series was prepared in the medium on top of the cells. Following 5 days of incubation, the plates were checked microscopically for alterations of the cell or monolayer morphology. After microscopic inspection, the plates were processed using the MTS/PMS method as described by the manufacturer (Promega, The Netherlands). The 50% cytostatic/cytotoxic concentration (CC50; i.e. the concentration of compound that reduces cell viability by 50%) was calculated using logarithmic interpolation. Cytotoxicity in Vero E6 cells. Cytotoxicities were assayed by measuring the cell viabilities in the same culture settings along antiviral activities in parallel P96 wells plates. Two fold serial dilutions, starting 100 µM or 300 µ M, in duplicates or triplicates, were added in 25 µl to 100 µl of Vero E6 cells that have been plated the day before. At day two, the medium was removed and replaced by 70 µl of fresh medium containing 10 µl of CellTiter blue reagent (Promega) and incubated for 90 min at 37 °C. Cell viabilities were measured as resorufin fluorescence readings on a plate reader (Tecan Infinite M200 Pro). Cytotoxicity in CRL-2522 cells. Cell viability assay in CRL-2522 cells were previously described.22 Briefly, cells were seeded on 96-well microtiter plates (Corning) before being treated with serial dilutions of compounds (ranging from 0.001 to 500 µM) prepared in medium. After 24 h, 32 ACS Paragon Plus Environment

Page 33 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

cell viability assays using CellTiter-Glo Reagent (Promega) were performed according to the manufacturer’s protocol. Luminescent signals were detected by the GloMax Multi-Detection System (Promega) after 1 h of incubation. Maps of electrostatic potential The geometries of compounds 9b, 9c 9d and 15e were optimized using Jaguar 39 and the B3LYP/631G** basis set. Electrostatic potentials were computed solving the linearized Poisson-Boltzmann equation by a finite difference method as implemented in DelPhi software (v.4). 40, 41 Cubic grids of resolution 1.0 Å were centred on each molecule, leaving a separation of 10 Å between any solute atom and the borders of the box, and the atomic charges were distributed onto the grid points. ESP charges and AMBER radii were used. The boundary between the solvent [treated as a high-dielectric medium (€=80)], and the ligands [considered a low-dielectric medium (€=2)] was defined by the solvent-accessible surfaces using a probe radius of 1.4 Å. Microsomal stability studies. These studies have been performed by Anthem Biosciences according to their protocols using diclofenac and imipramine as reference compounds. Pharmacokinetic studies. These studies have been performed by Anthem Biosciences according to their protocols, using a single dose at 10 mg/kg body weight by subcutaneous or intraperitoneal administration to male Balb/c mice. In Silico ADME Calculations. Calculations were computed with the Schrödinger Molecular Modeling Suite (Schrödinger Release 2015-4, Schrödinger, LLC, New York, NY, 2015). The 3D structures of 9b and 15e were generated using Maestro (version 10.4,) and energy minimizations were carried out using Macromodel (version 9.9). Local minimum energy structures of each compound were used as input for ADME studies with QikProp.

Supporting Information

33 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 34 of 51

The following Supporting information accompanies this article: Figure S1: MEP maps of compounds 9b, 9c, 9d and 15e. Table S1: predicted clogP, tPSA and solubility values for the proposed compounds. Table S2: In silico rapid ADME predictions performed for compound 9b and 15e using QikProp program (Schrödinger) and range of recommended values for oral drugs. Table S3: In vitro microsomal stability in mouse liver microsomes. Table S4. Mean PK parameters after single dose intraperitoneal and subcutaneous administration of 9b in male BALB/c mice at 10 mg/kg body weight. This material is available free of charge via the Internet at http://pubs.acs.org

Corresponding Author Information: María-Jesús Pérez-Pérez Instituto de Química Médica (CSIC) Juan de la Cierva 3, 28006 Madrid (Spain) Phone: 34 91 2587516 Fax: 34 91 5644853 e-mail: [email protected] ORCID: 0000-0003-1336-7760

Author Contributions: AGSJ, AMG, AP have performed the synthesis of the compounds, supervised by MJPP; EMP has performed the theoretical calculations; MJC and MJPP have designed the synthesis; LD, SJ, RA, SAM, LFPN and GQ performed the in vitro infection assays and cytotoxicity studies. DJ and PL supervised the antiviral assays. MJPP, JN, LD, GQ have written the paper. Acknowledgments. We thank Caroline Collard and Nick Verstraeten for their excellent technical assistance in the acquisition of the antiviral data. This work has been supported by grants from MINECO/FEDER SAF2015-64629-C2-1-R and by European Union FP7 Program under SILVER grant agreement n° 260644.

34 ACS Paragon Plus Environment

Page 35 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Abbreviations: CC50: 50% cytotoxic concentration or concentration that reduces the overall metabolic activity of uninfected cells to 50%; CHIKF: chikungunya fever; CHIKV: chikungunya virus; cLogP: theoretical octanol/water partition coefficient; CPE : cytopathic effect; EC50 : 50% effective concentration; EC90 : 90% effective concentration; ECSA: East/Central/South African genotypes;

MTS/PMS:

3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-

sulfophenyl)-2H-tetrazolium) in the presence of phenazine methosulfate; ONNV: O´nyong-nyong; PK: pharmacokinetics; QPPCaco: calculated Caco permeability; QPlogKHsa: calculated binding to human albumin; tPSA: topological surface area;

REFERENCES

1.

Couderc, T., and Lecuit, M. (2015) Chikungunya virus pathogenesis: From bedside to bench, Antiviral Res. 121, 120-131, 10.1016/j.antiviral.2015.07.002.

2.

Thiberville, S. D., Moyen, N., Dupuis-Maguiraga, L., Nougairede, A., Gould, E. A., Roques, P., and de Lamballerie, X. (2013) Chikungunya fever: Epidemiology, clinical syndrome, pathogenesis and therapy, Antiviral Res. 99, 345-370, 10.1016/j.antiviral.2013.06.009

3.

Burt, F. J., Chen, W., Miner, J. J., Lenschow, D. J., Merits, A., Schnettler, E., Kohl, A., Rudd, P. A., Taylor, A., Herrero, L. J., Zaid, A., Ng, L. F. P., and Mahalingam, S. (2017) Chikungunya virus: an update on the biology and pathogenesis of this emerging pathogen, Lancet Infect. Dis. 17, e107-e117, 10.1016/s1473-3099(16)30385-1.

4.

Agarwal, A., Vibha, D., Srivastava, A. K., Shukla, G., and Prasad, K. (2017) Guillain-Barre syndrome complicating chikungunya virus infection, J. Neurovirol. 23, 504-507, 10.1007/s13365-017-0516-1.

5.

Nasci, R. S. (2014) Movement of Chikungunya Virus into the Western Hemisphere, Emerging Infect. Dis. 20, 1394-1395, 10.3201/eid2008.140333.

35 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

6.

Page 36 of 51

Calba, C., Guerbois-Galla, M., Franke, F., Jeannin, C., Auzet-Caillaud, M., Grard, G., Pigaglio, L., Decoppet, A., Weicherding, J., Savaill, M. C., Munoz-Riviero, M., Chaud, P., Cadiou, B., Ramalli, L., Fournier, P., Noël, H., De Lamballerie, X., Paty, M. C., and LeparcGoffart, I. (2017) Preliminary report of an autochthonous chikungunya outbreak in France, July to September 2017, Eurosurveillance 22, 5-10, 10.2807/1560-7917.ES.2017.22.39.1700647.

7.

Powers, A. M., Brault, A. C., Tesh, R. B., and Weaver, S. C. (2000) Re-emergence of chikungunya and O’nyong-nyong viruses: evidence for distinct geographical lineages and distant evolutionary relationships, J. Gen. Virol. 81, 471-479, 10.1099/0022-1317-81-2-471

8.

Weaver, S. C., and Forrester, N. L. (2015) Chikungunya: Evolutionary history and recent epidemic spread, Antiviral Res. 120, 32-39, 10.1016/j.antiviral.2015.04.016.

9.

Leparc-Goffart, I., Nougairede, A., Cassadou, S., Prat, C., and de Lamballerie, X. (2014) Chikungunya in the Americas, The Lancet 383, 514, 10.1016/S0140-6736(14)60185-9

10.

Burt, F. J., Rolph, M. S., Rulli, N. E., Mahalingam, S., and Heise, M. T. (2012) Chikungunya: A re-emerging virus, Lancet 379, 662-671, 10.1016/s0140-6736(11)60281-x.

11.

Powers, A. M. (2015) Risks to the Americas associated with the continued expansion of chikungunya virus, J. Gen. Virol. 96, 1-5, 10.1099/vir.0.070136-0.

12.

Rougeron, V., Sam, I. C., Caron, M., Nkoghe, D., Leroy, E., and Roques, P. (2015) Chikungunya, a paradigm of neglected tropical disease that emerged to be a new health global risk, J. Clin. Virol. 64, 144-152, 10.1016/j.jcv.2014.08.032.

13.

Abdelnabi, R., Neyts, J., and Delang, L. (2017) Chikungunya virus infections: time to act, time to treat, Curr. Opin. Virol. 24, 25-30, 10.1016/j.coviro.2017.03.016.

14.

De Lamballerie, X., Boisson, V., Reynier, J. C., Enault, S., Charrel, R. N., Flahault, A., Roques, P., and Grand, R. L. (2008) On chikungunya acute infection and chloroquine treatment, Vector-Borne Zoonotic Dis. 8, 837-839, 10.1089/vbz.2008.0049.

15.

Abdelnabi, R., Neyts, J., and Delang, L. (2015) Towards antivirals against chikungunya virus, Antiviral Res. 121, 59-68, 10.1016/j.antiviral.2015.06.017. 36 ACS Paragon Plus Environment

Page 37 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

16.

da Silva-Júnior, E. F., Leoncini, G. O., Rodrigues, É. E. S., Aquino, T. M., and AraújoJúnior, J. X. (2017) The medicinal chemistry of Chikungunya virus, Bioorg. Med. Chem. 25, 4219-4244, 10.1016/j.bmc.2017.06.049.

17.

Lani, R., Hassandarvish, P., Shu, M.-H., Phoon, W. H., Chu, J. J. H., Higgs, S., Vanlandingham, D., Abu Bakar, S., and Zandi, K. (2016) Antiviral activity of selected flavonoids against Chikungunya virus, Antiviral Res. 133, 50-61, 10.1016/j.antiviral.2016.07.009.

18.

Nothias-Scaglia, L. F., Retailleau, P., Paolini, J., Pannecouque, C., Neyts, J., Dumontet, V., Roussi, F., Leyssen, P., Costa, J., and Litaudon, M. (2014) Jatrophane diterpenes as inhibitors of chikungunya virus replication: Structure-activity relationship and discovery of a potent lead, J. Nat. Prod. 77, 1505-1512, 10.1021/np500271u.

19.

Ching, K. C., Kam, Y. W., Merits, A., Ng, L. F. P., and Chai, C. L. L. (2015) Trisubstituted thieno[3,2-b]pyrrole 5-carboxamides as potent inhibitors of alphaviruses, J. Med. Chem. 58, 9196-9213, 10.1021/acs.jmedchem.5b01047.

20.

Mishra, P., Kumar, A., Mamidi, P., Kumar, S., Basantray, I., Saswat, T., Das, I., Nayak, T. K., Chattopadhyay, S., Subudhi, B. B., and Chattopadhyay, S. (2016) Inhibition of Chikungunya Virus Replication by 1-[(2-Methylbenzimidazol-1-yl) Methyl]-2-Oxo-Indolin3-ylidene] Amino] Thiourea(MBZM-N-IBT), Sci. Rep. 6, 20122, 10.1038/srep20122

21.

Ching, K. C., Tran, T. N. Q., Amrun, S. N., Kam, Y. W., Ng, L. F. P., and Chai, C. L. L. (2017) Structural Optimizations of Thieno[3,2-b]pyrrole Derivatives for the Development of Metabolically Stable Inhibitors of Chikungunya Virus, J. Med. Chem. 60, 3165-3186, 10.1021/acs.jmedchem.7b00180.

22.

Ehteshami, M., Tao, S., Zandi, K., Hsiao, H. M., Jiang, Y., Hammond, E., Amblard, F., Russell, O. O., Merits, A., and Schinazi, R. F. (2017) Characterization of β-D-N4hydroxycytidine as a novel inhibitor of Chikungunya virus, Antimicrob. Agents Chemother. 61, e02395-16, 10.1128/AAC.02395-16.

37 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

23.

Page 38 of 51

Varghese, F. S., Thaa, B., Amrun, S. N., Simarmata, D., Rausalu, K., Nyman, T. A., Merits, A., McInerney, G. M., Ng, L. F. P., and Ahola, T. (2016) The Antiviral Alkaloid Berberine Reduces Chikungunya Virus-Induced Mitogen-Activated Protein Kinase Signaling, J. Virol. 90, 9743-9757, 10.1128/jvi.01382-16.

24.

Gigante, A., Canela, M. D., Delang, L., Priego, E. M., Camarasa, M. J., Querat, G., Neyts, J., Leyssen, P., and Pérez-Pérez, M. J. (2014) Identification of 1,2,3 Triazolo 4,5-d pyrimidin7(6H)-ones as Novel Inhibitors of Chikungunya Virus Replication, J. Med. Chem. 57, 40004008, 10.1021/jm401844c.

25.

Gigante, A., Gómez-SanJuan, A., Delang, L., Li, C., Bueno, O., Gamo, A. M., Priego, E. M., Camarasa, M. J., Jochmans, D., Leyssen, P., Decroly, E., Coutard, B., Querat, G., Neyts, J., and Pérez-Pérez, M. J. (2017) Antiviral activity of [1,2,3]triazolo[4,5-d]pyrimidin-7(6H)ones against chikungunya virus targeting the viral capping nsP1, Antiviral Res. 144, 216-222, 10.1016/j.antiviral.2017.06.003.

26.

Lovering, F., Bikker, J., and Humblet, C. (2009) Escape from Flatland: Increasing Saturation as an Approach to Improving Clinical Success, J. Med. Chem. 52, 6752-6756, 10.1021/jm901241e.

27.

Demeter, O., Fodor, E. A., Kállay, M., Mező, G., Németh, K., Szabó, P. T., and Kele, P. (2016) A Double-Clicking Bis-Azide Fluorogenic Dye for Bioorthogonal Self-Labeling Peptide Tags, Chem. Eur. J. 22, 6382-6388, 10.1002/chem.201504939.

28.

Albrecht, S., Defoin, A., and Tarnus, C. (2006) Simple preparation of O-substituted hydroxylamines from alcohols, Synthesis, 1635-1638, 10.1055/s-2006-926440.

29.

Zhang, D., Zhang, X., Ai, J., Zhai, Y., Liang, Z., Wang, Y., Chen, Y., Li, C., Zhao, F., Jiang, H., Geng, M., Luo, C., and Liu, H. (2013) Synthesis and biological evaluation of 2-amino-5aryl-3-benzylthiopyridine scaffold based potent c-Met inhibitors, Bioorg. Med. Chem. 21, 6804-6820, 10.1016/j.bmc.2013.07.032.

38 ACS Paragon Plus Environment

Page 39 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

30.

Bénard, C., Mohammad, R., Saraswat, N., Shan, R., Maiti, S. N., Wuts, P. G. M., Stier, M., Lints, T., Bradow, J., and Schwarz, J. B. (2008) Convenient Preparation of Optically Pure 3‐Aryloxy‐pyrrolidines, Synth. Commun. 38, 517-524, 10.1080/00397910701796691.

31.

Song, D., Park, Y., Yoon, J., Aman, W., Hah, J.-M., and Ryu, J.-S. (2014) Click approach to the discovery of 1,2,3-triazolylsalicylamides as potent Aurora kinase inhibitors, Bioorg. Med. Chem. 22, 4855-4866, 10.1016/j.bmc.2014.06.047.

32.

Ghosh, A. K., and Brindisi, M. (2015) Organic Carbamates in Drug Design and Medicinal Chemistry, J. Med. Chem. 58, 2895-2940, 10.1021/jm501371s.

33.

Jeon, M.-K., Lee, K. M., Kim, I. H., Jang, Y. K., Kang, S. K., Lee, J. M., Jung, K.-Y., Kumar, J. A., Rhee, S. D., Jung, W. H., Song, J. S., Bae, M. A., Kim, K. R., and Ahn, J. H. (2014) Synthesis and biological evaluation of thienopyrimidine derivatives as GPR119 agonists, Bioorg. Med. Chem. Lett. 24, 4281-4285, 10.1016/j.bmcl.2014.07.020.

34.

Briant, L., Desprès, P., Choumet, V., and Missé, D. (2014) Role of skin immune cells on the host susceptibility to mosquito-borne viruses, Virology 464-465, 26-32, 10.1016/j.virol.2014.06.023.

35.

Lum, F. M., and Ng, L. F. P. (2015) Cellular and molecular mechanisms of chikungunya pathogenesis, Antiviral Res. 120, 165-174, 10.1016/j.antiviral.2015.06.009.

36.

Delang, L., Li, C., Tas, A., Quérat, G., Albulescu, I. C., De Burghgraeve, T., Guerrero, N. A. S., Gigante, A., Piorkowski, G., Decroly, E., Jochmans, D., Canard, B., Snijder, E. J., PérezPérez, M. J., van Hemert, M. J., Coutard, B., Leyssen, P., and Neyts, J. (2016) The viral capping enzyme nsP1: a novel target for the inhibition of chikungunya virus infection, Sci. Rep. 6, 31819, 10.1038/srep31819

37.

Delang, L., Segura Guerrero, N., Tas, A., Quérat, G., Pastorino, B., Froeyen, M., Dallmeier, K., Jochmans, D., Herdewijn, P., Bello, F., Snijder, E. J., de Lamballerie, X., Martina, B., Neyts, J., van Hemert, M. J., and Leyssen, P. (2014) Mutations in the chikungunya virus nonstructural proteins cause resistance to favipiravir (T-705), a broad-spectrum antiviral, J. Antimicrob. Chemother. 69, 2770-2784, 10.1093/jac/dku209. 39 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

38.

Page 40 of 51

Blatt, L. M., Pan, L., Seiwert, S., Andrews, S. W., Martin, P., Schumacher, A., Beigelman, L., Liu, J., Condroski, K., Jiang, Y., Kaus, R., Kennedy, A., Kercher, T., Lyon, M., and Wang, B. (2008) Preparation of macrocyclic acylsulfonamides, N-hydroxy-, N-alkoxy- and N-aryloxyamides as inhibitors of HCV replication, p 315 pp., InterMune, Inc., USA; Array BioPharma, Inc. WO2008137779A2.

39.

Bochevarov, A. D., Harder, E., Hughes, T. F., Greenwood, J. R., Braden, D. A., Philipp, D. M., Rinaldo, D., Halls, M. D., Zhang, J., and Friesner, R. A. (2013) Jaguar: A highperformance quantum chemistry software program with strengths in life and materials sciences, Int. J. Quantum Chem. 113, 2110-2142, 10.1002/qua.24481.

40.

Rocchia, W., Sridharan, S., Nicholls, A., Alexov, E., Chiabrera, A., and Honig, B. (2002) Rapid grid-based construction of the molecular surface and the use of induced surface charge to calculate reaction field energies: Applications to the molecular systems and geometric objects, J. Comput. Chem. 23, 128-137, 10.1002/jcc.1161.

41.

Rocchia, W., Alexov, E., and Honig, B. (2001) Extending the applicability of the nonlinear Poisson-Boltzmann equation: Multiple dielectric constants and multivalent ions, J. Phys. Chem. B 105, 6507-6514, 10.1021/jp010454y.

40 ACS Paragon Plus Environment

Page 41 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Table 1. Antiviral evaluation of the triazolopyrimidines against CHIKV strain 899 in Vero cells.a Compound

EC50 (µM)b

EC90 (µM)c

CC50 (µM)d

Selectivity index

1

19 ± 2e

38 ± 16 e

116 ± 60

6.1

2

2.6 ± 1.0 e

8.8 ± 6.0 e

167 ± 94

64

9a

>49

>49

>49

naf

9b

1.2 ± 0.009

3.4

84 ± 19

70

9c

3.2 ± 1.0

8.2 ± 1.5

59 ± 23

18

9d

236

>241

241

1.0

13

27 ± 11

>82

82 ± 3

3.0

14a

1.5 ± 0.3

3.1 ± 1.3

>220

>147

14b

2.5 ± 0.3

>8.2

8.2 ± 0.2

3.3

14c

2.8 ± 0.2

>20

20 ± 6

7.1

14d

4.2 ± 0.4

>8.3

8.3 ± 0.5

2.0

14e

2.9 ± 0.7

19

45 ± 15

16

15a

7.4 ± 4.1

>13

13 ± 0.9

1.8

15b

14 ± 2.2

>23

>220

>16

15c

7.3 ± 1.5

>106

106

15

15d

129 ± 79

>243

>243

>1.9

41 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

a

Page 42 of 51

15e

8.9 ± 6.1

11 ± 4.1

201 ± 26

23

15f

5.1 ± 0.2

35 ± 13

135 ± 41

26

15g

2.0 ± 0.3

>46

46 ± 2.8

23

15h

66 ± 28

>221

>221

>3.4

16

26 ± 9.7

89

>220

>8.5

17a

>224

>224

>224

naf

17b

34

>129

129

3.8

18

11 ± 3.4

>22

22

2.0

Chloroquine

11 ± 7

21 ± 18

89 ± 28

8.1

All data are mean values +- standard deviation for at least three independent experiments

b

50% effective concentration or concentration required to protect 50% of the cells against the cytopathic effect of the virus. c

90% effective concentration or concentration required to protect 90% of the cells against the cytopathic effect of the virus. d

50% cytotoxic concentration or concentration that reduces the viability of uninfected cells to 50%. Measured by microscopic scoring e

Data as reported in24

f

na: not applicable

42 ACS Paragon Plus Environment

Page 43 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Table 2 Determination of cytotoxicity of selected compounds in different cell lines CC50 (µM)a

Cell type

a

CC50 (µM)b

9c

14a

15e

9c

14a

15e

Vero A

74 ± 0.9

>220

>220

59 ± 23

>220

201 ± 26

Vero E6

84 ± 1.8

>220

>220

60 ± 2

>220

>220

BHK

115 ± 6

>220

>220

57 ± 23

>220

>220

HeLa Rh

75 ± 9

>220

>220

72 ± 9

>220

>220

Huh7

60 ± 0.3

78 ± 5

152 ± 9

48 ± 2

91 ± 2

137 ± 23

Cytotoxicity determined by MTS

b

Cytotoxicity determined microscopically

43 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 44 of 51

Table 3. Antiviral evaluation of selected triazolopyrimidines against different laboratory strains and clinical isolates of CHIKV in Vero cells.

Comp

CC50

CHIKV

CHIKV

CHIKV

CHIKV

CHIKV St

Congo

Venturini

OPy

St Martin

Martin EFS

EC50 (µM)

EC50 (µM)

EC50 (µM)

EC50 (µM)

EC50 (µM)

(µM)a 1

>300

6.35 ± 0.05b

26 ± 2b

28

23.9 ± 0.5b

55.3

2

>300

0.75 ± 0.35

1.43 ± 0.01b

2.6 ± 0.5

2.9 ± 0.05 b

13 ± 6

b

a

9b

>100

0.3 ± 0.0

1.65 ± 0.5

5.2

2 ± 0.3

4.5 ± 0.5

9c

>100

1 ± 0.1

2.6 ± 1.1

11.5

NDd

8.7 ± 1.7

14a

>300b

0.35 ± 0.5

1.2 ± 0.2

3.4 ± 1.2

1.2 ± 0.5

3.7 ± 1.1

15e

>300

1.0 ± 0.4

2.9 ± 0.4

5.4 ± 2.4

3 ± 2.5

4.4 ± 3

50% cytotoxic concentration or concentration that reduces the overall metabolic activity of

uninfected cells to 50%. Measured by CellTiter blue read-out. b

Reported in ref. 24

c

Poorly soluble.

d

ND: Not determined

44 ACS Paragon Plus Environment

Page 45 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Table 4. In vitro anti-CHIKV activity of triazolopyrimidine compounds against WT and against a resistant CHIKV strain selected under the pressure of the prototype triazolopyrimidine 1.

EC50 (µM)a

a

WT CHIKV

Resistant CHIKV strain to 1

T-705

29 ± 21

12 ± 6

1

19 ± 2

>116b

2

2.6 ± 1

>167b

9b

1.2 ± 0.01

65 ± 40

9c

3.2 ± 1.0

35 ± 29

15e

8.9 ± 6.1

23 ± 14

Data shown are average values ± SD of at least two independent experiments.

b

Similar data were reported in ref 36

45 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 46 of 51

Figure 1

Previously identified [1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones as potent inhibitors of CHIKV replication

46 ACS Paragon Plus Environment

Page 47 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Scheme 1. Synthesis of [1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones with a six-membered ring as substituent at the aryl ring.a

N3

N3 X +

a

X

MsO

OH 5

O

6a X = CH2 6b X = O 6c X = NBoc

7a X = CH2 7b X = O 7c X = NBoc

b

O

O N

HN

H 2N

N N

N

X

c

H2N

d

a

N N

X O

O 9a 9b 9c 9d

N

X = CH2 X=O X = NBoc X = NH

8a X = CH2 8b X = O 8c X = NBoc

Reagents and conditions: (a) Cs2CO3, DMF, 80 ºC, 2 h (7a, 22% yield; 7b, 46% yield; 7c, 55%

yield); (b) cyanoacetamide, NaH 60%, DMF, 0 ºC to rt, 1 h (8a, 74% yield; 8b, 87% yield; 8c, 88% yield) (c) For 9a and 9b: EtCO2Et, tBuOK, dioxane, MW, 100 ºC, 1 h (9a, 83% yield; 9b, 85% yield); for 9c: EtCO2tBu, tBuOK, dioxane, MW, 100 ºC, 1 h (9c, 74%) ; (d) TFA, DCM, rt, 2h, (9d, quantitative yield).

47 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 48 of 51

Scheme 2. Synthesis of the N-Boc piperazine derivative.a

a

Reagents and conditions: (a) tert-butyl piperazine-1-carboxylate, HCTU, DIPEA, DMF:DCM, rt

2h (11, 77% yield); (b) cyanoacetamide, NaH 60%, DMF anh., 0 ºC to rt, 1 h (12, 84% yield); (c) EtCOOtBu, tBuOK, dioxane, MW, 100 ºC, 60 min (13, 26% yield)

48 ACS Paragon Plus Environment

Page 49 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Scheme 3. Synthesis of analogues of carbamates and ureas related to compound 9c

O

O N

HN

N

N

N

a

O N

Boc

N

N

N

HN

N

N

O

R O

O

9c

14a R = Ph 14b R = CH2Ph 14c R = CH2CH=CH2 14d R = CH2CH(CH3)2

b or c

14e R =

O

O N

HN

O N

N

N

N

R H N

H N

O 15a-h

a

R=

b

OCH3

H N

H N

H N

H N c

d

H N

O N

e

a

f

g

h

Reagents and conditions: (a) (i) TFA 5% in DCM, rt, 2 h (quant.); (ii) ROCOCl, Et3N, DMAP, DCM, 0º C

to rt (14a, 30% yield; 14b, 60% yield; 14c, 83% yield; 14d, 20% yield; 14e, 18% yield); (b) for 15a, 15f, 15h: (i) TFA 5% in DCM, rt, 2 h (quant.); (ii) triphosgene, RNH2, DCM/DMF, DIPEA, rt, 30 min. (15a, 54% yield; 15f, 24% yield; 15h, 26% yield); (c) for 15b, 15c, 15d, 15e, 15g: (i) TFA 5% in DCM, rt, 2 h (quant.); (ii) RNCO, DCM/DMF, Et3N, 0 ºC to rt (15b, 73% yield; 15c, 71% yield; 15d, 25% yield; 15e, 78% yield; 15g, 61% yield)

49 ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 50 of 51

Scheme 4. Synthesis of other acyl and sulfonamide derivatives related to compound 9c.a

O

O N

HN N

N

HN

N

Boc

N

a

N

O

OCH3

N N

N

N

O

O

O

16

9c b O c

O

N

HN

N N

N

O N N

X Y

O O O S N

N

HN

N

N

17a X=N; Y=CH 17b X=CH; Y=N

O 18

a

Reagents and conditions: (a) (i) TFA 5% in DCM, rt, 2 h (quant.); (ii) methyl 4-chloro-4-oxobutanoate,

Et3N, DMAP, DCM, 0º C to rt (16, 30% yield); (b) (i) TFA 5% in DCM, rt, 2 h (quant.); (ii) nicotinic acid or isonicitinic acid, PyBOP, DIPEA, DMF:DCM, rt, 2 h (17a, 28% yield; 17b, 50% yield); (c) (i) TFA 5% in DCM, rt, 2 h (quant.); (ii) 2-propanesulfonyl chloride, Et3N, DCM/DMF, 0º C to rt, 1h (18, 67% yield)

50 ACS Paragon Plus Environment

Page 51 of 51 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

For Table of Contents Use Only

51 ACS Paragon Plus Environment