2018年3月28日水曜日

FBDD Approach Targetting IM=PDH 2016

 

 

Targeting Inosine-5′-monophosphate Dehydrogenase (IMPDH) from Mycobacterium tuberculosis




Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
§ MRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine and Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Rondebosch 7701, Cape Town, South Africa
Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, 30 Flemington Road, Parkville, Victoria 3052, Australia
J. Med. Chem., Article ASAP
DOI: 10.1021/acs.jmedchem.7b01622
Publication Date (Web): March 16, 2018
Copyright © 2018 American Chemical Society
*T.L.B. email: tom@cryst.bioc.cam.ac.uk., *D.B.A: phone, +61 3903 54794; e-mail, david.ascher@unimelb.edu.au., *C.A.: phone, +44 (0) 1223 336405; e-mail, ca26@cam.ac.uk.
ACS AuthorChoice - This is an open access article published under a Creative Commons Attribution (CC-BY) License, which permits unrestricted use, distribution and reproduction in any medium, provided the author and source are cited.

Abstract

Abstract Image
Tuberculosis (TB) remains a major cause of mortality worldwide, and improved treatments are needed to combat emergence of drug resistance. Inosine 5′-monophosphate dehydrogenase (IMPDH), a crucial enzyme required for de novo synthesis of guanine nucleotides, is an attractive TB drug target. Herein, we describe the identification of potent IMPDH inhibitors using fragment-based screening and structure-based design techniques. Screening of a fragment library for Mycobacterium thermoresistible (Mth) IMPDH ΔCBS inhibitors identified a low affinity phenylimidazole derivative. X-ray crystallography of the Mth IMPDH ΔCBS–IMP–inhibitor complex revealed that two molecules of the fragment were bound in the NAD binding pocket of IMPDH. Linking the two molecules of the fragment afforded compounds with more than 1000-fold improvement in IMPDH affinity over the initial fragment hit.

Introduction


Tuberculosis (TB) is a contagious infectious disease caused by Mycobacterium tuberculosis (Mtb), which can be transmitted through the air as droplets. The infection predominantly affects the lungs, but it can spread to other parts of the body, especially in patients with a suppressed immune system.
The World Health Organization (WHO) has estimated that nearly one-third of the world’s population is infected with Mtb, leading to 1.8 million TB deaths in 2015.(1) Although there has been a slow decline in new TB cases and TB-related deaths in recent years, the emergence and spread of multidrug resistant (MDR) and extensively drug resistant (XDR) strains of Mtb has increased the threat that this disease poses for global public health. According to the WHO, approximately 480,000 cases of MDR-TB emerged in 2015, and the cure rate of those patients was only 50%.(1)
Current TB treatments require combinations of four first-line drugs, isoniazid, rifampicin, ethambutol, pyrazinamide, and streptomycin, which must be taken for six months or longer.(2) Resistant strains are not susceptible to the standard drugs, and although MDR-TB is treatable using second-line drugs, such treatments have a number severe side effects.(3) Consequently, there is an urgent need for the development of novel and more effective drugs for the treatment of drug resistant TB.
Inosine-5′-monophosphate dehydrogenase (IMPDH, E.C. 1.1.1.205) has received considerable interest in recent years as an important target enzyme for immunosuppressive,(4) anticancer,(5,6) and antiviral drugs.(7) Most recently, IMPDH has emerged as a promising antimicrobial drug target.(8−11)
IMPDH catalyzes the first unique step in the de novo synthesis of guanine nucleotides, the oxidation of inosine 5′-monophosphate (IMP) to xanthosine 5′-monophosphate (XMP) with the concomitant reduction of the cofactor nicotinamide adenine dinucleotide (NAD+) to NADH (Figure 1).(12) XMP is then subsequently converted to guanosine 5′-monophosphate (GMP) by a GMP synthetase.(13)
figure
Figure 1. Purine nucleotide biosynthesis. The commonly occurring guanine nucleotide biosynthetic and salvage reactions are shown, as is the adenine nucleotide biosynthetic pathway. The IMPDH reaction is boxed. NK, nucleoside kinase; HPRT, hypoxanthine phosphoribosyl transferase; XPRT, xanthine phosphoribosyl transferase; GPRT, guanine phosphoribosyl transferase; GMPS, guanosine 5′-monophosphate synthetase; GMPR, guanosine 5′-monophosphate reductase; ADSS, adenylosuccinate synthetase; ADSL, adenylosuccinate lyase.
IMPDH has been deemed essential in every pathogen analyzed to date, including Mtb, Staphylococcus aureus, and Pseudomonas aeruginosa, which are three of the most serious bacterial threats.(14−16) However, this has been somewhat contradictory,(17) in comparing cell versus animal work. IMPDH is an ubiquitous enzyme present in several eukaryotes, bacteria, and protozoa.(18) The IMPDH reaction involves two chemical transformations. The first step of the IMPDH catalyzed reaction involves the attack of catalytic Cys on substrate IMP followed by hydride transfer to NAD+, forming the covalent enzyme intermediate E-XMP*. In the second step, E-XMP* is hydrolyzed to XMP.(11) The enzyme exists in two different conformations, an open form that accommodates both the substrate and cofactor during the first step and a closed form where the active site flap moves into the NAD+-binding site for the E-XMP* hydrolysis.(19)
In recent years, there has been considerable effort aimed at identifying small molecule inhibitors of IMPDH as potential antitubercular agents. X-ray crystal structures of a truncated form of the Mtb enzyme in complex with some of these compounds have been reported.(20−26)
In antibacterial drug discovery, and especially in TB drug discovery, high-throughput screening (HTS) typically identifies a number of leads that show high potency in vitro, but most did not show any translation to an in vivo effect. It is also inevitable that the HTS libraries represent only a small fraction of possible chemical space and so limit confidence in finding a good starting point for subsequent development. Phenotypic screens can potentially lead to the identification of a molecule that modifies a disease phenotype by acting on a previously undescribed target or by acting simultaneously on more than one target.(27) However, for many of these hits the relevant target or targets has not yet been identified, thus preventing further target-based optimization of the compounds.(28,29)
The previously reported IMPDH inhibitors, such as compounds 7759844 (1), MAD1, P41, VCC234718, and DDD00079282 (Figure 2), were identified by phenotypic screening or target based HTS of compound libraries.(21,23−25)
figure
Figure 2. Structures of previously reported IMPDH inhibitors (α Ki values against IMP). All compounds showed uncompetitive inhibition with respect to IMP.
We have sought to develop IMPDH inhibitors using a fragment-based approach. Fragment-based drug discovery (FBDD) is now established in both industry and academia as an alternative approach to high-throughput screening for the generation of hits or chemical tools for drug targets.(30)
We have previously reported the discovery of several series of novel and potent inhibitors using FBDD to target Mtb. Previously we have reported the fragment elaboration strategies that we have applied which have included fragment growing, merging, and linking. Although fragment linking is conceptually the most appealing strategy for fragment elaboration, in practice, this strategy can be challenging where the choice of the optimal fragment linker can be crucial.(31,32)
In the fragment-based approach, biophysical techniques are usually used to identify small chemical compounds (fragments) that bind with low affinity to the drug target. X-ray crystallography is then usually employed to establish the binding mode of the fragment and to facilitate the design of elaborated fragments. The availability of high-resolution X-ray crystal structures of a truncated form of the IMPDH,(21,24,25) in both the substrate-free and substrate/ligand-bound forms, makes this enzyme attractive for a fragment-based approach.
In this work, the discovery of a new class of potent nM inhibitors of IMPDH using a FBDD approach is reported. A library of 960 fragments was screened against Mth IMPDH ΔCBS using a biochemical assay. The fragment hits from this assay were examined using X-ray crystallography, and an X-ray crystal structure of one of the fragment complexes was solved to a resolution of 1.45 Å. Examination of the X-ray crystal structure suggested a strategy of fragment-linking for optimization of this fragment hit.

Results and Discussion


Fragment Screening
An in-house fragment library composed of 960 fragments was screened using a biochemical assay against Mth IMPDH ΔCBS. Mth IMPDH, which shares 85% sequence identity with Mtb IMPDH and is 100% identical in the active site,(24,25) was chosen for the fragment screening and structural studies because it gave higher protein expression yields and better diffracting crystals than the Mtb orthologue. IMPDH activity was monitored spectrophotometrically by measuring the formation of NADH at 340 nm. The biochemical assay was performed at a fragment concentration of 1 mM, and hits were retested in triplicate. Compound 1 (7759844) previously reported as IMPDH inhibitor was used as a positive control in assays (Table 1).(23)
Table 1. Structures and Activities for Compound 1 (7759844) and the Most Potent Fragment Hits Found in the Screen against Mth IMPDH ΔCBS

a
% Inhibition at 10 μM.
The screen resulted in 18 hits (1.9% hit rate), where a hit was defined as a compound that gave greater than 50% inhibition at a concentration of 1 mM. A complete list of fragment hits identified is included in Table 1. A number of common scaffolds were observed, in particular phenylimidazole (fragments 24), aminopyrazole (fragments 56), and naphthol (fragments 711), and the remaining compounds contained a substituted phenyl or a heterocyclic five membered ring (fragments 1219).
The IC50 values of six of the most active fragments were measured and the IC50 and ligand efficiency (LE) values of these fragments are summarized in Table 2. The fragment screen provided an array of hits with IC50 ranging from 325 μM to 674 μM and ligand efficiencies from 0.31 to 0.42. The inhibition constant [Ki] with respect to both substrates IMP and NAD+ was determined by assaying various concentrations of each inhibitor with five different concentrations of substrate and a fixed saturating concentration of the cosubstrate. The inhibition data for these fragments are summarized in Table 2. All compounds yielded an uncompetitive inhibition pattern with respect to NAD+ with Ki values ranging from 262 to 525 μM. Fragments 14, 16, and 19 yielded a mixed inhibition with respect to IMP with Ki values ranging from 126 to 398 μM, and compounds 1, 2, 11, and 18 yielded an uncompetitive inhibition with Ki values ranging from 361 to 609 μM.
Table 2. IC50, Ligand Efficiency, and Alpha Ki Values of Fragment Hits and Compound 1 (7759844) against Mth IMPDH ΔCBS
CompdIC50 (μM) (LE)aIMP αKi (μM)NAD αKi (μM)
10.77 ± 0.06 (0.40)0.86 ± 0.03 (UC)b0.55 ± 0.02 (UC)
2674 ± 53 (0.36)609 ± 3 (UC)512 ± 23 (UC)
11336 ± 6 (0.39)361 ± 53 (UC)310 ± 30 (UC)
14400 ± 7 (0.42)238 ± 6 (Mixed)262 ± 39 (UC)
16433 ± 60 (0.31)398 ± 81 (Mixed)525 ± 64 (UC)
18512 ± 37 (0.37)554 ± 70 (UC)452 ± 26 (UC)
19325 ± 9 (0.37)126 ± 34 (Mixed)355 ± 47 (UC)
a
Ligand efficiency was calculated using the equation LE = (1.37 × pIC50)/HA, where HA means heavy atom, i.e., a non-hydrogen atom.
b
UC: Uncompetitive inhibition.
Inhibition constants of compound 2 toward full-length Mtb IMPDH were also determined. Compound 2 inhibited full-length Mtb IMPDH enzyme with a Ki value of 572 ± 14 μM with IMP as the substrate and a Ki value of 534 ± 18 μM with NAD as the substrate, which are similar to the Ki values observed for the Mth IMPDH ΔCBS enzyme.
X-ray Structure of Compounds 1 (7759844) and 2
Compound 1 and the six fragment hits (2, 11, 14, 16, 18, and 19) were selected for structural characterization using X-ray crystallography by soaking into preformed crystals of Mth IMPDH ΔCBS as previously described.(20) After molecular replacement, clear electron density was observed in the 2F0Fc difference map (σ = 3.0) for IMP, and in addition density was observed for one molecule of compound 1 (Figure 3A) and two molecules of compound 2 (Figure 3B), which were partially occupying the NAD+ binding site. None of the other fragments (11, 14, 16, 18, and 19) showed any electron density in the X-ray crystal structures. Although the kinetic studies of compounds 1 and 2 suggested that these two compounds are uncompetitive with respect to NAD+, the binding mode of compounds 1 and 2 closely resembles that of other previously reported uncompetitive inhibitors of Mtb IMPDH.(21,24,25) It has been proposed that the uncompetitive mode of inhibition of IMPDH inhibitors with respect to NAD+ is consistent with their binding preferentially to the covalent IMPDH-XMP* intermediate after NADH has been released.(24,25,33)
figure
Figure 3. X-ray crystal structure of compounds 1 and 2 bound to Mth IMPDH ΔCBS. Ligand interactions are represented as dotted lines; hydrogen bonds are represented in red, polar interactions in orange, ionic interactions in yellow, and aromatic and π interactions in green dotted lines. Protein–ligand interactions were analyzed using Arpeggio. (a) Interactions made by 1 (green) in the X-ray crystal structure of the complex of IMPDH with IMP (orange). (b) Interactions made by 2 (yellow) in the X-ray crystal structure of the complex of IMPDH with IMP. (c) Structural alignment of the IMPDH crystal structures of 1 (green) and 2 (yellow).
The structure of Mth IMPDH ΔCBS with compound 1 showed that the inhibitor binds in the NAD pocket in a near identical manner to our recently described IMPDH inhibitors.(24,25) Compound 1 formed strong π interactions with the hypoxanthine group of IMP, P45′, Y471′, and polar interactions with G409, E442, P45′, and G470′. The electron density revealed two molecules of compound 2 within the NAD binding pocket of IMPDH. One molecule of compound 2 stacked with IMP, forming extensive π interactions with the hypoxanthine group of IMP. This fragment was further stabilized through polar interactions, hydrogen bonds, and π interactions to surrounding residues in the active site pocket, including A269, G318, and E442. The other molecule of compound 2 sits closer to the opening of the active site, making polar interactions with N273 and E442, and π interactions with H270 and Y471′.
A comparison of the structure of Mth IMPDH ΔCBS with compound 1 with the fragment 2 structure shows that the two molecules of 2 mimic the position of the larger inhibitor 1 (Figure 3C).
Fragment Elaboration
Fragment 2 was selected as the starting point for exploration because of the ease of synthetic modification and the availability of a X-ray crystal structure to guide the optimization. For chemical elaboration of 2, fragment linking as well as fragment growing were considered. As the two molecules of fragment 2 are found to bind in adjacent regions of the target protein, the fragment-linking approach was the more attractive option. However, before fragment linking, fragment 2 was further optimized with the aim of improving the binding affinity. The structures and inhibitory activities of these compounds against Mth IMPDH ΔCBS are summarized in Tables 3 and 4. The corresponding data for fragment 2 have also been included for comparative purposes.
Table 3. Structures and Activities for Fragment 2 and Compounds 2028 against Mth IMPDH ΔCBS

Table 4. Structures and Activities for Fragment 2 and Compounds 2935 against Mth IMPDH ΔCBS

a
Ligand efficiency was calculated using the equation LE = (1.37 × pIC50)/HA, where HA means heavy atom, i.e., a non-hydrogen atom.
b
ND: not determined. IC50 values were determined for compounds that showed >50% inhibition. c%Inhibition at 10 μM.
All compounds were evaluated at a concentration of 100 μM with Mth IMPDH ΔCBS.
Fragment Growing
The fragment-growing strategy involves using structure-based drug design to form additional interactions by growing out from the starting fragment. Fragment 2 was modified at the 2-position of the imidazole ring to explore the introduction of various aromatic rings linked by a thioacetamide (2022) to form π interactions with the hypoxanthine group of IMP (Figure S1). Such modifications gave compounds with improved Mth IMPDH ΔCBS inhibition. The phenyl and benzofuran derivatives (20 and 21) showed 13 and 31% inhibition, respectively. Mth IMPDH ΔCBS inhibition was shown to be sensitive to minor modifications of the phenyl substituent groups; for example, the 4-iodo substituted 22 showed 30% inhibition at 100 μM, whereas the nonsubstituted compound, 20, showed 13% inhibition at the same concentration. The effect of the removal of the 4-bromo group was investigated and compounds 2325 were synthesized. Removal of the bromo substituent in compounds 2022 (13–31% inhibition at 100 μM) was tolerated (2325, 3–46% inhibition at 100 μM). The importance of the aromatic amide linked by a thioacetamide was subsequently examined. Replacing the phenyl with isopropyl (26) resulted in complete loss of activity. Substitution of the thioacetamide by a thioacetic acid also led to a complete loss in activity (compounds 27 and 28).
Fragment Linking and SAR
Examination of the X-ray crystal structure of the previously reported inhibitor 1 when overlaid with fragment 2 revealed that the distance between the 4-position of the phenyl ring of fragment 2 and the 2-position of the imidazole ring represents the closest approach of the molecules (Figure S1). On the basis of this structural information, three different linkers were designed to connect the two copies of the fragment 2 at these positions (compounds 2931). Initially, a thioacetamide and urea linker moieties were examined. Compounds 29 and 30 showed 20% and 56% of Mth IMPDH ΔCBS inhibition, respectively, at 100 μM (Table 4). Interestingly, compound 30 showed a 12-fold improvement in Mth IMPDH ΔCBS inhibitory activity with an IC50 of 58 μM, compared to the fragment 2.
The lactate linker was then used, but all attempts to link 4-phenylimidazole with 4-(4-bromophenyl)-1H-imidazole were unsuccessful. We therefore decided to synthesize compound 31, which contains 1-methyl-4-phenyl-1H-imidazole and 4-(4-bromophenyl)-1H-imidazole linked with a lactate analogue, as in compound 1. Compound 31 (Table 4) showed markedly improved Mth IMPDH ΔCBS inhibition with a LE of 0.29 and IC50 of 0.52 μM, which is 1300-fold more potent compared to the fragment 2. Although compound 31 binds in the cofactor site, the mechanism of inhibition can vary depending on its relative affinities for the E·IMP and E-XMP* complexes.(8,11) Kinetic evaluation of compound 31 showed the mode of Mth IMPDH ΔCBS inhibition was uncompetitive with respect to both IMP and the NAD+ cofactor (see Figure S2, Supporting Information) with a Ki value of 0.30 ± 0.02 μM with IMP as the substrate and a Ki value of 0.20 ± 0.01 with NAD as the substrate.
Inhibition constants of compound 31 toward full-length Mtb IMPDH were also determined. Compound 31 inhibited full-length Mtb IMPDH enzyme with a Ki value of 0.61 ± 0.05 μM with IMP as the substrate and a Ki value of 0.39 ± 0.02 μM with NAD as the substrate. The inhibition constants were consistent with the data using the Mth IMPDH ΔCBS enzyme.
Removal of the bromo substituent in compound 29 to give compound 32 was well tolerated (Table 4). The importance of the imidazole group for the inhibitory activity against Mth IMPDH ΔCBS was confirmed by replacing of the 4-(4-bromophenyl)-1H-imidazole substituent of compound 31 with 4-(4-bromophenyl)oxazole (33) which resulted in a 4-fold loss of activity (Table 4). Replacing the 4-(4-bromophenyl)-1H-imidazole of 31 by a phenyl (34) resulted in complete loss of activity (Table 4). However, the 4-iodophenyl derivative 35 demonstrated slightly improved activity (IC50 = 0.47 μM, LE = 0.34) compared to compounds 31 (IC50 = 0.52 μM, LE = 0.29) and 1 (IC50 = 0.77 μM, LE = 0.40). It is noteworthy that LE of compound 35 was comparable to that of the original fragment hit 2 (LE = 0.36) and other reported IMPDH inhibitors.(34)
The importance of the 4-bromo substituent on the phenyl ring in compound 31 was also explored (compounds 3639, Table 5). Removal of the bromo substituent (36) resulted in a 5-fold loss in activity, whereas replacing this group with an iodine (39) or a morpholine ring (37) resulted in less than 1.3-fold loss in activity. The electronic nature of the substituent in this position had little effect on inhibitory activity. For example, an electron-donating methoxy (38) retained activity comparable to that of the bromine derivative (31). Among them, imidazoles 31, 3738, and 39 were found to be potent inhibitors of Mth IMPDH ΔCBS with IC50 values ranging from 520 to 690 nM.
Table 5. Structures and Activities for Compounds 31, 36–39, and (S)-31 against Mth IMPDH ΔCBS

a
Ligand efficiency was calculated using the equation LE = (1.37 × pIC50)/HA, where HA means heavy atom, i.e., a non-hydrogen atom.
The (S)-isomer of 31 was found to bind preferentially (Table 5), with the racemate 31 having approximately half the Mth IMPDH ΔCBS inhibition of the (S)-isomer 31. This observation accords with the results previously reported for other series of IMPDH inhibitors.(21,35)
X-ray Structure of Compound 31
From crystals soaked with compound 31, the 2F0Fc difference map (σ = 3.0) revealed strong density for the inhibitor. The structure of compound 31 (Figure 4A) showed that it bound in a nearly identical manner to compounds 1 and 2 in the NAD binding pocking (Figure 4B), stacking with IMP, and maintaining the interactions with H270, N273, E442, and P45 and Y471 from the neighboring subunit. Compound 31 made additional interactions in the binding pocket, including polar interactions with D267 and N297.
figure
Figure 4. X-ray crystal structure of compound 31 bound to Mth IMPDH ΔCBS. Protein–ligand interactions were analyzed using Arpeggio. (a) Interactions made by 31 in the X-ray crystal structure of the complex of IMPDH with IMP (orange). Ligand interactions are represented as dotted lines; hydrogen bonds are represented in red, polar interactions in orange, ionic interactions in yellow, and aromatic and π interactions in green dotted lines. (b) Structural alignment of the IMPDH crystal structures of 1 (lilac), 2 (yellow), and 31 (green).
Whole-Cell Activity against Mtb
The whole-cell activity of the most potent analogues 31, 3339, and (S)-31 in vitro was determined against Mtb H37Rv (see Table S1, Supporting Information). No significant inhibition of bacterial growth was detected for any of the compounds (MIC90 ≥ 50 μM) over the tested concentration range (0–100 μM). There are currently ongoing efforts to explain the lack of efficacy of the potent Mth IMPDH ΔCBS inhibitors described which could be caused by low membrane permeability, poor metabolic stability, and/or drug-efflux mechanisms.
Synthetic Chemistry
Compounds 2029 and 32 were synthesized from 4-phenyl-1H-imidazole-2-thiol or 4-(4-bromophenyl)-1H-imidazole-2-thiol according to the sequence described in Scheme 1. Thioacetic derivatives 27 and 28 were prepared by treatment of 4-phenyl-1H-imidazole-2-thiol or 4-(4-bromophenyl)-1H-imidazole-2-thiol with 2-chloroacetic acid in the presence of NaOH followed by neutralization with hydrochloric acid.
figure
Scheme 1. Synthesis of Thioacetamide and Thioacetic Derivatives 2029 and 32a
aReagent and conditions: (a) 2-chloroacetic acid, NaOH, EtOH, 80 °C, 69–72%; (b) isopropylamine, HATU, DIPEA, EtOAc, rt, 73%; (c) chloroacetyl chloride, TEA, DCM, rt, 89–99%; (d) NaOH, MeOH, H2O, 70 °C, 69–96%.

Thioacetamide 26 was synthesized by amide coupling between thioacetic derivative 28 and isopropylamine. Similarly, thioacetamides 2025, 29, and 32 were prepared by cross-linking 4-phenyl-1H-imidazole-2-thiol or 4-(4-bromophenyl)-1H-imidazole-2-thiol with α-chloroacetamides 4043, which were obtained by acylation of anilines with various substituents with chloroacetyl chloride.
The synthesis of urea 30 was achieved as shown in Scheme 2 by coupling amine 44 and N-(4-(1H-imidazol-4-yl)phenyl)-1H-imidazole-1-carboxamide, which was obtained from commercially available 4-(1H-imidazol-4-yl)aniline as a crude intermediated, in the presence of N,N-diisopropylethylamine. Amine 44 was synthesized from benzyl ((5-(4-bromophenyl)-1H-imidazol-2-yl)methyl)carbamate,(36) followed by the deprotection of the benzyloxycarbonyl group under acidic condition.
figure
Scheme 2. Synthesis of Urea 30a
aReagent and conditions: (a) CDI, THF, rt, quant.; (b) HCl (4 M), dioxane, 100 °C, quant.; (c) DIPEA, DMF, rt, 26%.

Imidazole derivatives 31, 3638, and 39 were prepared following the synthetic procedure outlined in Scheme 3. 2-Aminoimidazoles 5054 were synthesized according to a published microwave-assisted protocol.(37) In brief, 2-aminoimidazoles 5054 were prepared by reaction of the commercially available α-haloketones and N-acetylguanidine, followed by deacetylation (Scheme 3). Acid 59 was synthesized starting with imidazole 55, which was prepared by reaction of 2-bromo-4′-hydroxyacetophenone with formamide as reported previously.(38) The phenol 57 was synthesized by alkylation of imidazole 55, followed by deprotection of the methyl ether with BBr3. Substituted phenol 57 was converted to the ether 58 upon treatment with methyl 2-bromopropionate in the presence of Cs2CO3. Enantiomerically pure phenyl ethers were synthesized by using Mitsunobu reaction conditions with ethyl d-lactate (Scheme 4). After the hydrolysis of the ester group, the resulting carboxylic acid 59 was treated with thionyl chloride to give the acid chloride 60, which was reacted with 2-aminoimidazoles 5054 to afford imidazole derivatives 31, 3638, and 39.
figure
Scheme 3. Synthesis of 2-Acylaminoimidazole Derivatives 31, 36–38, and 39a
aReagent and conditions: (a) acetylguanidine, CH3CN, μW, 100 °C, 71–88%; (b) (1) 20% H2SO4, MeOH:H2O (1:1 v/v), μW, 100 °C; (2) aq Na2CO3, 79–95%; (c) formamide, 150 °C, 99%; (d) CH3I, Cs2CO3, DMF, rt, 88%; (e) BBr3 (1 M in DCM), −78 °C to rt, DCM, 96%; (f) Cs2CO3, methyl 2-bromopropionate, DMF, 60 °C, 92%; (g) NaOH, THF:H2O (2:1 v/v), 80 °C, 65%; (h) SOCl2, 80 °C, quant.; (i) TEA, DCM, 40 °C, 61–72%.

figure
Scheme 4. Synthesis of the S-Enantiomer of 31a
aReagent and conditions: (a) (1) Ethyl d-lactate, PPh3, DEAD, THF, rt; (2) NaOH, THF:H2O (2:1 v/v), rt, 56%; (b) SOCl2, 80 °C, quant.; (c) 50, TEA, DCM, 40 °C, 69%.

The syntheses of 2-acylaminooxazole 33 and amides 3435 were achieved as shown in Scheme 5. 2-Acylaminooxazole derivative 33 was obtained by coupling the acid chloride derivative 60 with 2-aminooxazole derivative 61, which was prepared by reaction of 2,4′-dibromoacetophenone with urea. Similarly, amides 3435 were prepared by coupling the corresponding anilines with the acid chloride derivative 60.
figure
Scheme 5. Synthesis of 2-Acylaminooxazole Derivative 33 and Amides 3435a
aReagent and conditions: (a) urea, CH3CN, 80 °C, 78%; (b) TEA, DCM, rt (for 34 and 35) or 40 °C (for 33), 64–76%.

Conclusions


FBDD has emerged as a robust approach to identify small molecules that bind to a wide range of therapeutic targets. Fragment elaboration strategies have resulted in the development of a number of compounds that have progressed into clinical trials. Within the area of TB drug discovery, a number of HTS and phenotypic screens have been performed during the past decade. Although HTS identified a number of leads that show high potency in vitro, the translation to an in vivo effect has proven challenging.
This study illustrates the successful application of a fragment-based approach followed by fragment optimization to obtain nanomolar affinity ligands of IMPDH. A library of 960 fragments were screened against Mth IMPDH ΔCBS, and from the screen the phenylimidazole fragment hit 2 (IC50 = 674 μM) was identified. Kinetic experiments showed that 2 was an uncompetitive inhibitor of Mth IMPDH ΔCBS with respect to NAD+ and IMP. Two molecules of the fragment 2 were shown to bind at the NAD binding site of the enzyme. The X-ray crystal structure also revealed that one molecule of fragment 2 makes π interactions with IMP and the other molecule sits closer to the opening of the active site, making polar interactions with N273 and E442 and π interactions with H270 and Y471′. This provides potential for further optimization of fragment 2. To explore better the possibilities given by fragment 2, fragment-linking and fragment-growing strategies were employed, resulting in low micromolar to nanomolar affinity compounds. Among them, compounds 31, 35, and 3739 were the most potent IMDPH inhibitors of the series described in this work with IC50 values between 0.47 and 0.69 μM, which represent >1000-fold improvement in Mth IMPDH ΔCBS potency over the initial fragment hit. Compound 31 was shown to bind at the NAD binding site of the enzyme, and the X-ray crystal structure also revealed that it makes π interactions with IMP, maintaining the interactions with H270, N273, E442, and P45 and Y471 from the neighboring subunit. Moreover, compound 31 made additional interactions in the binding pocket, including polar interactions with D267 and N297. A comparison of this structure with the fragment 2 structure shows that the two molecules of 2 mimic the position of the larger inhibitor 31. This is the first example of utilizing the fragment-based approach specifically to identify new potent inhibitors of IMPDH. Further structural optimization to improve the cellular activity of these analogues is ongoing with the aim of developing novel classes of anti-TB agents.

Experimental Section


1. Chemistry
1.1. General Experimental Methods
Solvents were distilled prior to use and dried by standard methods. Unless otherwise stated, 1H and 13C NMR spectra were obtained in CDCl3, MeOD, or DMSO-d6 solutions using either a Bruker 400 MHz AVANCE III HD Smart Probe, 400 MHz QNP cryoprobe, or 500 MHz DCH cryoprobe spectrometer. Chemical shifts (δ) are given in ppm relative to the residual solvent peak (CDCl3: 1H, δ = 7.26 ppm; 13C, δ = 77.16 ppm), and the coupling constants (J) are reported in hertz (Hz). Optical rotations were measured on a PerkinElmer Polarimeter 343 at 589 nm (Na D-line), and specific rotations are reported in 10–1 deg cm2 g–1. Microwave reactions were performed using a Biotage Initiator system under reaction conditions as indicated for each individual reaction.
Reactions were monitored by TLC and LCMS to determine consumption of starting materials. Flash column chromatography was performed using an Isolera Spektra One/Four purification system and the appropriately sized Biotage SNAP column containing KP-silica gel (50 μm). Solvents are reported as volume/volume eluent mixture where applicable.
High resolution mass spectra (HRMS) were recorded using a Waters LCT Premier Time of Flight (TOF) mass spectrometer or a Micromass Quadrapole-Time of Flight (Q-TOF) spectrometer.
Liquid chromatography mass spectrometry (LCMS) was carried out using an Ultra Performance Liquid Chromatographic system (UPLC) Waters Acquity H-class coupled to a Waters SQ Mass Spectrometer detector. Samples were detected using a Waters Acquity TUV detector at 2 wavelengths (254 and 280 nm). Samples were run using an Acquity UPLC HSS column and a flow rate of 0.8 mL/min. The eluent consisted of 0.1% formic acid in water (A) and acetonitrile (B); gradient, from 95% A to 5% A over a period of 4 or 7 min.
All final compounds had a purity greater than 95% by LCMS analysis.
General Method A: Synthesis of Thioacetamides 2025, 29, and 32
To a solution of the 2-chloroacetamide derivative (0.17 mmol) in MeOH (8 mL) was added 4-(4-bromophenyl)-1H-imidazole-2-thiol or 4-phenyl-1H-imidazole-2-thiol (0.17 mmol), followed by a solution of NaOH (0.67 mmol) in H2O (2.5 mL). The reaction mixture was stirred at 70 °C for 3 h. After cooling to rt, the solvents were removed in vacuo and the resulting residue was taken up in 30 mL of EtOAc and washed with H2O (15 mL). The aqueous layer was extracted with EtOAc (3 × 30 mL) and the combined organic layers were washed with brine and dried over MgSO4. Filtration and evaporation afforded crude products, which were purified as indicated below.
General Method B: Synthesis of Thioacetic Derivatives 27 and 28
A solution of 4-phenyl-1H-imidazole-2-thiol or 4-(4-bromophenyl)-1H-imidazole-2-thiol (2.80 mmol) and NaOH (5.0 mmol) in EtOH (5 mL) was reflux for 1 h. After cooling to rt, a solution of 2-chloroacetic acid (2.80 mmol) in EtOH (2 mL) was added. The reaction was stirred at reflux for an additional 3 h and then cooled to 0 °C. The reaction mixture was diluted with cold water (5 mL) and acidified with 1 M HCl. The precipitated product was collected by filtration and washed with DCM (2 × 2 mL).
General Method C: Synthesis of Compounds 31, 33, and 3639
A mixture of acid 59 (0.20 mmol) and SOCl2 (2 mL) was heated at 80 °C for 2 h. The solvent was removed under reduced pressure to give the acid chloride 60 as a white solid. The resulting solid was immediately dissolved in anhydrous DCM, and the resulting solution was added slowly dropwise at 0 °C to a solution of the corresponding substituted 2-aminoimidazoles (0.20 mmol) and TEA (0.80 mmol) in anhydrous DCM (5 mL). The reaction mixture was stirred at 40 °C for 36 h and then diluted with DCM (20 mL) and washed with saturated aqueous NaHCO3. The aqueous phase layer was then extracted with DCM (2 × 20 mL), and the combined organic layers were dried over MgSO4 and filtered, and the solvent was removed under reduced pressure to afford a yellow oil, which was purified by flash chromatography, eluting with the solvent system specified.
General Method D: Synthesis of Amides 3435
A mixture of acid 59 (0.20 mmol) and SOCl2 (2 mL) was heated at 80 °C for 2 h. The solvent was removed under reduced pressure to give 60 as a white solid. The resulting solid was immediately dissolved in anhydrous DCM, and the resulting solution was added slowly dropwise at 0 °C to a solution of the corresponding aniline (0.20 mmol) and triethylamine (0.80 mmol) in anhydrous DCM (5 mL). The reaction mixture was stirred at rt for 4 h and then diluted with DCM (20 mL) and washed with saturated aqueous NaHCO3. The aqueous phase layer was then extracted with DCM (2 × 20 mL), and the combined organic layers were dried over MgSO4 and filtered, and the solvent was removed under reduced pressure to afford a yellow oil, which was purified by flash chromatography, eluting with the solvent system specified.
General Method E: Synthesis of α-Chloroacetamides 4043
Et3N (4.77 mmol) followed by a solution of chloroacetyl chloride (4.77 mmol) in DCM (3 mL) were added to a stirred solution of the corresponding aniline (4.38 mmol) in DCM (5 mL) at rt. The reaction mixture was stirred at rt for 2–4 h. The reaction was then diluted with DCM (20 mL) and washed with saturated aqueous NaHCO3, 1 M HCl, and brine and dried over anhydrous MgSO4, and the solvent was removed under reduced pressure. Compound 43 was purified by flash chromatography eluting with the solvent system specified, although other analogues were used in subsequent reactions without further purification.
General Method F: Synthesis of Substituted N-(1H-Imidazol-2-yl)acetamides 4549
A mixture of the corresponding 2-bromoacetophenone derivative (0.38 mmol) and acetylguanidine (1.13 mmol) in anhydrous acetonitrile (3 mL) was heated at 100 °C using microwave irradiation for 15 min. The solvent was removed, and the residue was taken in H2O (3 mL) and filtered, and the solid was washed with H2O (2 mL × 2) and DCM (2 mL). The solid obtained was used in the next step without further purification.
General Method G: Synthesis of Substituted 2-Aminoimidazoles 5054
To a solution of the corresponding substituted N-(1H-imidazol-2-yl)acetamides (0.31 mmol) in a 1:1 v/v mixture of MeOH and H2O (2.4 mL) was added concentrated H2SO4 (0.6 mL), and the reaction mixture was heated at 100 °C under microwave irradiation for 15–30 min. The reaction mixture was concentrated, and the resulting residue was resuspended in H2O (5 mL), and a saturated aqueous Na2CO3 was added until pH 8. The product was extracted into EtOAc (3 × 40 mL). The combined organic fractions were dried over MgSO4, and the solvent was removed under reduced pressure. The resulting solid was used in the next reaction without further purification.
2-((4-(4-Bromophenyl)-1H-imidazol-2-yl)thio)-N-phenylacetamide (20)
Compound 40 (29 mg, 0.17 mmol) was reacted with 4-(4-bromophenyl)-1H-imidazole-2-thiol (43 mg, 0.17 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 20 (59 mg, 0.15 mmol, 89% yield) as a white solid. 1H NMR (400 MHz, MeOD) δ 7.62 (br s, 2H), 7.56–7.39 (m, 5H), 7.36–7.17 (m, 2H), 7.13–7.00 (m, 1H), 3.82 (s, 2H) ppm. 13C NMR (100 MHz, MeOD) δ 169.6, 141.3, 139.6, 132.8, 129.8, 129.8, 127.5, 125.5, 121.4, 121.2, 121.2, 116.7, 39.6 ppm. LCMS (ESI−) m/z 388.0 [M – H], retention time 2.20 min (100%). HRMS (ESI+): m/z calculated for C17H15BrN3OS [M + H]+: 388.0119. Found: 388.0121.
N-(Benzofuran-5-yl)-2-((4-(4-bromophenyl)-1H-imidazol-2-yl)thio)acetamide (21)
Compound 41 (36 mg, 0.17 mmol) was reacted with 4-(4-bromophenyl)-1H-imidazole-2-thiol (43 mg, 0.17 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 21 (70 mg, 0.16 mmol, 96% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 12.50 (br s, 1H), 10.35 (br s, 1H), 7.94 (dd, J = 10.6, 2.0 Hz, 2H), 7.73–7.64 (m, 2H), 7.52–7.44 (m, 4H), 7.34 (dd, J = 8.8, 2.2 Hz, 1H), 6.91 (dd, J = 2.2, 1.0 Hz, 1H), 3.99 (s, 2H) ppm. 13C NMR (100 MHz, DMSO-d6) δ 166.5, 150.8, 146.7, 140.2, 139.9, 134.3, 133.6, 131.4, 127.4, 126.2, 119.0, 116.9, 115.7, 111.6, 111.3, 107.0, 37.9. LCMS (ESI−) m/z 428.0 [M – H], retention time 2.22 min (100%). HRMS (ESI+): m/z calculated for C19H15BrN3O2S [M + H]+: 428.0068. Found: 428.0065.
2-((4-(4-Bromophenyl)-1H-imidazol-2-yl)thio)-N-(4-iodophenyl)acetamide (22)
Compound 42 (50 mg, 0.17 mmol) was reacted with 4-(4-bromophenyl)-1H-imidazole-2-thiol (43 mg, 0.17 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 22 (60 mg, 0.11 mmol, 69% yield) as a white solid. 1H NMR (500 MHz, MeOD) δ 7.63–7.58 (m, 4H), 7.56–7.45 (m, 3H), 7.38–7.31 (m, 2H), 3.81 (s, 2H) ppm.13C NMR (125 MHz, MeOD) δ 169.6, 139.6, 138.9, 132.7, 127.5, 127.1, 123.0, 116.6, 111.4, 88.1, 39.7 ppm. LCMS (ESI−) m/z 511.7 [M – H], retention time 2.30 min (100%). HRMS (ESI+): m/z calculated for C17H14BrIN3OS [M + H]+: 513.9086. Found: 513.9087.
N-Phenyl-2-((4-phenyl-1H-imidazol-2-yl)thio)acetamide (23)
Compound 40 (42 mg, 0.25 mmol) was reacted with 4-phenyl-1H-imidazole-2-thiol (44 mg, 0.25 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 23 (61 mg, 0.20 mmol, 82% yield) as a white solid. 1H NMR (500 MHz, MeOD) δ 7.71 (br s, 2H), 7.54–7.49 (m, 2H), 7.47 (br s, 1H), 7.35 (t, J = 7.4 Hz, 2H), 7.31–7.20 (m, 3H), 7.11–7.03 (m, 1H), 3.83 (s, 2H) ppm. 13C NMR (125 MHz, MeOD) δ 168.3, 142.7, 139.5, 138.5, 138.2, 128.4, 128.3, 126.7, 124.4, 124.1, 123.7, 119.8, 38.3 ppm. LCMS (ESI−) m/z 308.0 [M – H], retention time 1.82 min (95%). HRMS (ESI+): m/z calculated for C17H16N3OS [M + H]+: 310.1014. Found: 310.1004.
N-(Benzofuran-5-yl)-2-((4-phenyl-1H-imidazol-2-yl)thio)acetamide (24)
Compound 41 (36 mg, 0.17 mmol) was reacted with 4-phenyl-1H-imidazole-2-thiol (30 mg, 0.17 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 24 (42 mg, 0.12 mmol, 70% yield) as a white solid. 1H NMR (500 MHz, MeOD) δ 7.87 (d, J = 2.1 Hz, 1H), 7.72 (d, J = 2.2 Hz, 1H), 7.68 (d, J = 7.7 Hz, 2H), 7.46 (br s, 1H), 7.40 (d, J = 8.8 Hz, 1H), 7.37–7.29 (m, 3H), 7.23 (t, J = 7.4 Hz, 1H), 6.76 (dd, J = 2.2, 0.9 Hz, 1H), 3.84 (s, 2H) ppm. 13C NMR (125 MHz, MeOD) δ 169.6, 153.5, 147.4, 140.9, 134.7, 129.8, 129.1, 128.1, 125.8, 118.9, 114.2, 112.1, 107.7, 39.7 ppm. LCMS (ESI−) m/z 348.0 [M – H], retention time 1.81 min (100%). HRMS (ESI+): m/z calculated for C19H16N3O2S [M + H]+: 350.0963. Found: 350.0971.
N-(4-Iodophenyl)-2-((4-phenyl-1H-imidazol-2-yl)thio)acetamide (25)
Compound 42 (74 mg, 0.25 mmol) was reacted with 4-phenyl-1H-imidazole-2-thiol (44 mg, 0.25 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 25 (70 mg, 0.16 mmol, 70% yield) as a white solid. 1H NMR (500 MHz, MeOD) δ 7.66 (d, J = 7.7 Hz, 2H), 7.61–7.54 (m, 2H), 7.44 (br s, 1H), 7.38–7.31 (m, 4H), 7.23 (t, J = 6.7 Hz, 1H), 3.80 (s, 2H) ppm. 13C NMR (125 MHz, MeOD) δ 169.7, 140.8, 139.6, 138.9, 129.8, 128.2, 125.8, 123.0, 88.1, 39.8 ppm. LCMS (ESI−) m/z 433.9 [M – H], retention time 2.13 min (98%). HRMS (ESI+): m/z calculated for C17H15IN3OS [M + H]+: 435.9981. Found: 435.9989.
N-Isopropyl-2-((4-phenyl-1H-imidazol-2-yl)thio)acetamide (26)
A solution of propan-2-amine (22 μL, 0.25 mmol), 2-((4-phenyl-1H-imidazol-2-yl)thio)acetic acid (60 mg, 0.25 mmol), HATU (194 mg, 0.50 mmol), and DIPEA (90 μL, 0.50 mmol) in EtOAc (10 mL) was stirred for 5 h at rt. The reaction mixture was diluted with EtOAc (40 mL) and washed with saturated aqueous NaHCO3 (20 mL), water (20 mL), and brine (20 mL). The organic layer was dried over MgSO4 and filtered. The solvent was removed under reduced pressure to give an oil residue, which was purified by flash chromatography (5–100% v/v EtOAc in petroleum ether) to give the desired product as a white solid (50 mg, 0.18 mmol, 73% yield).1H NMR (500 MHz, MeOD) δ 7.37 (d, J = 7.8 Hz, 2H), 7.14 (s, 1H), 7.05 (t, J = 7.7 Hz, 2H), 6.93 (t, J = 7.4 Hz, 1H), 3.62 (dt, J = 13.1, 6.4 Hz, 1H), 3.00 (s, 2H), 0.78 (d, J = 6.6 Hz, 6H) ppm.13C NMR (125 MHz, MeOD) δ 170.3, 141.1, 140.8, 133.7, 129.7, 128.1, 125.7, 119.7, 43.0, 38.8, 22.4 ppm. LCMS (ESI−) m/z 273.9 [M – H], retention time 1.52 min (100%). HRMS (ESI+): m/z calculated for C14H18N3OS [M + H]+: 276.1171. Found: 276.1172.
2-((4-(4-Bromophenyl)-1H-imidazol-2-yl)thio)acetic Acid (27)
Following general procedure B, from 4-(4-bromophenyl)-1H-imidazole-2-thiol (250 mg, 0.98 mmol) was obtained 27 (220 mg, 0.70 mmol, 72% yield) as a white solid. 1H NMR (400 MHz, MeOD) δ 7.65–7.56 (m, 2H), 7.57–7.39 (m, 3H), 3.80 (s, 2H) ppm. 13C NMR (100 MHz, MeOD) δ 172.9, 141.9, 140.2, 132.9, 132.6, 127.5, 121.8, 118.9, 37.4 ppm. LCMS (ESI−) m/z 311.0 [M – H], retention time 1.59 min (98%). HRMS (ESI+): m/z calculated for C11H10BrN2O2S [M + H]+: 312.9646. Found: 312.9654.
2-((4-Phenyl-1H-imidazol-2-yl)thio)acetic Acid (28)
Following general procedure B, from 4-phenyl-1H-imidazole-2-thiol (500 mg, 2.80 mmol) was obtained 28 (450 mg, 1.92 mmol, 69% yield) as a white solid. 1H NMR (500 MHz, MeOD) δ 7.68 (d, J = 7.2 Hz, 2H), 7.59 (s, 1H), 7.41 (t, J = 7.7 Hz, 2H), 7.32 (t, J = 7.4 Hz, 1H), 3.86 (s, 2H) ppm. 13C NMR (125 MHz, MeOD) δ 172.7, 141.7, 139.6, 131.6, 130.0, 129.1, 126.0, 118.9, 37.5 ppm. LCMS (ESI+) m/z 235.1 [M + H]+, retention time 1.22 min (100%). HRMS (ESI+): m/z calculated for C11H11N2O2S [M + H]+: 235.0541. Found: 235.0536.
N-(4-(1H-Imidazol-4-yl)phenyl)-2-((4-(4-bromophenyl)-1H-imidazol-2-yl)thio)acetamide (29)
Compound 43 (40 mg, 0.17 mmol) was reacted with 4-(4-bromophenyl)-1H-imidazole-2-thiol (43 mg, 0.17 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 29 (51 mg, 0.12 mmol, 69% yield) as a white solid. 1H NMR (400 MHz, MeOD) δ 7.72 (d, J = 1.1 Hz, 1H), 7.66 (d, J = 8.5 Hz, 4H), 7.59–7.47 (m, 5H), 7.40 (br s, 1H), 3.86 (s, 2H) ppm. 13C NMR (125 MHz, MeOD) δ 169.4, 141.2, 138.4, 137.0, 132.8, 127.5, 126.2, 121.4, 116.7, 111.4, 39.7 ppm. LCMS (ESI−) m/z 451.9 [M – H], retention time 1.65 min (100%). HRMS (ESI+): m/z calculated for C20H17BrN5OS [M + H]+: 454.0337. Found: 454.0334.
1-(4-(1H-Imidazol-4-yl)phenyl)-3-((5-(4-bromophenyl)-1H-imidazol-2-yl)methyl)urea (30)
To a solution of 4-(1H-imidazol-4-yl)aniline (50 mg, 0.31 mmol) in THF (5 mL) was added 1,1-carbonyldiimidazole (76 mg, 0.47 mmol). The mixture was stirred at rt overnight, and then it was filtered. The filter was washed with THF (2 × 3 mL) and the resulting solid (30 mg, 0.12 mmol) was dissolved in DMF (3 mL), and 44 (42 mg, 0.12 mmol) and N,N-diisopropylethylamine (74 μL, 0.48 mmol) were added. The reaction mixture was stirred at rt for 14 h, and then it was poured into water, extracted with EtOAc, dried over Na2SO4, and concentrated. After cooling to 0 °C, a 1:5 v/v mixture of MeOH and DCM (12 mL) was added and the suspended solid was collected by filtration and dried at vacuum to yield 30 (35 mg, 0.08 mmol, 26% yield) as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 12.03 (br s, 2H), 8.64 (br s, 1H), 7.73–7.68 (m, 2H), 7.64–7.59 (m, 2H), 7.57 (d, J = 1.9 Hz, 1H), 7.52–7.47 (m, 2H), 7.47–7.39 (m, 2H), 7.39–7.32 (m, 2H), 6.58 (br s, 1H), 4.33 (d, J = 5.5 Hz, 2H) ppm. 13C NMR (125 MHz, DMSO-d6) δ 155.5, 146.9, 140.6, 138.9, 138.9, 135.9, 134.6, 131.7, 128.8, 126.6, 125.1, 125.0, 119.0, 118.4, 118.2, 113.7, 111.7, 37.6 ppm. LCMS (ESI−) m/z 437.0 [M – H], retention time 5.07 min (97%). HRMS (ESI+): m/z calculated for C20H18BrN6O [M + H]+: 437.0725. Found: 437.0725.
N-(4-(4-Bromophenyl)-1H-imidazol-2-yl)-2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanamide (31)
4-(4-Bromophenyl)-1H-imidazol-2-amine 50 (48 mg, 0.20 mmol) was reacted with the acid chloride 60 (53 mg, 0.20 mmol) and Et3N (112 μL, 0.80 mmol) according to general method C. The crude product was purified by flash chromatography (0–20% v/v MeOH in DCM) to give compound 31 as a white solid (64 mg, 0.14 mmol, 67% yield). 1H NMR (500 MHz, CDCl3) δ 10.79 (br s, 1H), 9.49 (br s, 1H), 7.69 (d, J = 8.7 Hz, 2H), 7.58–7.41 (m, 5H), 7.18–6.97 (m, 2H), 6.93 (d, J = 8.8 Hz, 2H), 4.86 (d, J = 6.8 Hz, 1H), 3.71 (s, 3H), 1.65 (d, J = 6.7 Hz, 3H) ppm. 13C NMR (125 MHz, CDCl3) δ 171.5, 155.2, 141.8, 141.2, 138.0, 137.2, 131.7, 129.1, 127.6, 126.3, 120.5, 115.8, 115.6, 115.3, 108.1, 74.6, 33.5, 18.5 ppm. LCMS (ESI+) m/z 465.9 [M + H]+, retention time 2.73 min (95%). HRMS (ESI+): m/z calculated for C22H21BrN5O2 [M + H]+: 466.0879. Found: 466.0874. ((S)-31): 69% yield. [α]D25 −40.2 (c 1.0, CHCl3). LCMS (ESI+) m/z 466.0 [M + H]+, retention time 2.70 min (95%). HRMS (ESI+): m/z calculated for C22H21BrN5O2 [M + H]+: 466.0879. Found: 466.0879.
N-(4-(1H-Imidazol-4-yl)phenyl)-2-((4-phenyl-1H-imidazol-2-yl)thio)acetamide (32)
Compound 43 (40 mg, 0.17 mmol) was reacted with 4-(4-bromophenyl)-1H-imidazole-2-thiol (30 mg, 0.17 mmol) according to general method A. Purification by flash chromatography (1–20% v/v MeOH in DCM) afforded 32 (41 mg, 0.11 mmol, 65% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.46 (br s, 2H), 10.42 (br s, 1H), 7.76 (br s, 2H), 7.68 (d, J = 8.2 Hz, 4H), 7.56 (d, J = 8.5 Hz, 2H), 7.52 (s, 1H), 7.34 (br s, 2H), 7.19 (br s, 1H), 3.99 (s, 2H) ppm. 13C NMR (125 MHz, DMSO-d6) δ 167.0, 141.7, 139.9, 137.7, 136.2, 134.6, 128.9, 126.7, 125.2, 124.6, 119.7, 115.4, 39.5 ppm. LCMS (ESI+) m/z 376.1 [M + H]+, retention time 0.88 min (100%). HRMS (ESI+): m/z calculated for C20H18N5OS [M + H]+: 376.1232. Found: 376.1241.
N-(4-(4-Bromophenyl)oxazol-2-yl)-2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanamide (33)
4-(4-Bromophenyl)oxazol-2-amine 61 (39 mg, 0.16 mmol) was reacted with the acid chloride 60 (43 mg, 0.16 mmol) and Et3N (90 μL, 0.64 mmol) according to general method C. The crude product was purified by flash chromatography (0–15% v/v MeOH in DCM) to give compound 33 as a white solid (49 mg, 0.10 mmol, 64% yield). 1H NMR (400 MHz, CDCl3) δ 9.13 (br s, 1H), 7.81–7.66 (m, 3H), 7.63–7.50 (m, 4H), 7.47 (s, 1H), 7.13 (d, J = 1.3 Hz, 1H), 7.03–6.94 (m, 2H), 4.90 (q, J = 6.6 Hz, 1H), 3.74 (s, 3H), 1.70 (d, J = 6.8 Hz, 3H) ppm. 13C NMR (100 MHz, CDCl3) δ 155.1, 141.7, 138.0, 131.9, 131.7, 130.3, 129.4, 129.3, 127.8, 127.0, 126.7, 126.4, 122.2, 116.0, 115.4, 75.1, 33.5, 18.4 ppm. LCMS (ESI+) m/z 468.9 [M + H]+, retention time 2.86 min (95%). HRMS (ESI+): m/z calculated for C22H20BrN4O3 [M + H]+: 467.0719. Found: 467.0711.
2-(4-(1-Methyl-1H-imidazol-4-yl)phenoxy)-N-phenylpropanamide (34)
Aniline (28 μL, 0.30 mmol) was reacted with the acid chloride 60 (53 mg, 0.20 mmol) and Et3N (112 μL, 0.80 mmol) according to general method D. The crude product was purified by flash chromatography (0–20% v/v MeOH in DCM) to give compound 34 as a white solid (48 mg, 0.14 mmol, 74% yield). 1H NMR (400 MHz, CDCl3) δ 8.25 (br s, 1H), 7.79–7.69 (m, 2H), 7.60–7.52 (m, 2H), 7.47 (d, J = 1.3 Hz, 1H), 7.35 (dd, J = 8.5, 7.3 Hz, 2H), 7.19–7.06 (m, 2H), 7.05–6.97 (m, 2H), 4.82 (q, J = 6.7 Hz, 1H), 3.73 (s, 3H), 1.69 (d, J = 6.8 Hz, 3H) ppm. 13C NMR (100 MHz, CDCl3) δ 170.3, 155.6, 141.8, 137.9, 137.0, 129.0, 129.0, 126.3, 124.7, 120.0, 116.0, 115.3, 75.6, 33.5, 18.7 ppm. LCMS (ESI+) m/z 322.2 [M + H]+, retention time 1.86 min (97%). HRMS (ESI+): m/z calculated for C19H20N3O2 [M + H]+: 322.1556. Found: 322.1552.
N-(4-Iodophenyl)-2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanamide (35)
4-Iodoaniline (65 mg, 0.30 mmol) was reacted with the acid chloride 60 (53 mg, 0.20 mmol) and Et3N (112 μL, 0.80 mmol) according to the general method D. The crude product was purified by flash chromatography (0–20% v/v MeOH in DCM) to give compound 35 as a pink solid (62 mg, 0.14 mmol, 68% yield). 1H NMR (400 MHz, CDCl3) δ 8.28 (br s, 1H), 7.77–7.69 (m, 2H), 7.69–7.57 (m, 2H), 7.47 (d, J = 1.4 Hz, 1H), 7.41–7.31 (m, 2H), 7.11 (d, J = 1.3 Hz, 1H), 7.03–6.91 (m, 2H), 4.80 (q, J = 6.7 Hz, 1H), 3.73 (s, 3H), 1.67 (d, J = 6.8 Hz, 3H) ppm. 13C NMR (100 MHz, CDCl3) δ 170.4, 155.5, 141.7, 137.9, 136.9, 129.0, 126.3, 126.0, 121.8, 116.0, 115.3, 87.9, 75.6, 33.5, 18.6 ppm. LCMS (ESI+) m/z 448.2 [M + H]+, retention time 2.47 min (96%). HRMS (ESI+): m/z calculated for C19H19IN3O2 [M + H]+: 448.0522. Found: 448.0517.
2-(4-(1-Methyl-1H-imidazol-4-yl)phenoxy)-N-(4-phenyl-1H-imidazol-2-yl)propanamide (36)
4-Phenyl-1H-imidazol-2-amine 51 (33 mg, 0.20 mmol) was reacted with the acid chloride 60 (53 mg, 0.20 mmol) and Et3N (112 μL, 0.80 mmol) according to general method C. The crude product was purified by flash chromatography (0–15% v/v MeOH in DCM) to give compound 36 as a brown solid (57 mg, 0.15 mmol, 72% yield). 1H NMR (400 MHz, CDCl3) δ 10.84 (br s, 1H), 9.51 (br s, 1H), 7.78–7.56 (m, 4H), 7.46 (d, J = 1.2 Hz, 1H), 7.39 (t, J = 7.7 Hz, 2H), 7.22–7.26 (m, 1H), 7.14 (s, 1H), 7.11 (d, J = 1.4 Hz, 1H), 7.02–6.89 (m, 2H), 4.89 (q, J = 6.8 Hz, 1H), 3.73 (s, 3H), 1.68 (d, J = 6.8 Hz, 3H) ppm. 13C NMR (125 MHz, CDCl3) δ 171.3, 155.2, 141.8, 137.9, 129.1, 128.7, 126.9, 126.3, 124.6, 115.9, 115.3, 107.8, 74.5, 33.5, 18.5 ppm. LCMS (ESI+) m/z 388.1 [M + H]+, retention time 4.44 min (100%). HRMS (ESI+): m/z calculated for C22H22N5O2 [M + H]+: 388.1773. Found: 388.1767.
2-(4-(1-Methyl-1H-imidazol-4-yl)phenoxy)-N-(4-(4-morpholinophenyl)-1H-imidazol-2-yl)propanamide (37)
4-(4-Morpholinophenyl)-1H-imidazol-2-amine 52 (40 mg, 0.16 mmol) was reacted with the acid chloride 60 (43 mg, 0.16 mmol) and Et3N (90 μL, 0.65 mmol) according to general method C. The crude product was purified by flash chromatography (3–15% v/v MeOH in DCM) to give compound 37 as a brown solid (51 mg, 0.11 mmol, 67% yield). 1H NMR (400 MHz, CDCl3) δ 10.74 (br s, 1H), 7.71 (d, J = 8.8 Hz, 2H), 7.54 (br s, 2H), 7.47 (d, J = 1.4 Hz, 1H), 7.11 (d, J = 1.4 Hz, 1H), 7.03–6.88 (m, 5H), 4.90 (q, J = 6.7 Hz, 1H), 4.06–3.83 (m, 4H), 3.73 (s, 3H), 3.27–3.01 (m, 4H), 1.69 (d, J = 6.8 Hz, 3H) ppm. 13C NMR (125 MHz, CDCl3) δ 171.5, 155.3, 150.3, 141.8, 141.0, 137.9, 137.8, 128.9, 126.3, 125.5, 115.8, 115.3, 74.4, 66.9, 49.3, 33.6, 18.7 ppm. LCMS (ESI+) m/z 473.2 [M + H]+, retention time 2.22 min (100%). HRMS (ESI+): m/z calculated for C26H29N6O3 [M + H]+: 473.2301. Found: 473.2297.
N-(4-(4-Methoxyphenyl)-1H-imidazol-2-yl)-2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanamide (38)
4-(4-Methoxyphenyl)-1H-imidazol-2-amine 53 (30 mg, 0.16 mmol) was reacted with the acid chloride 60 (43 mg, 0.16 mmol) and Et3N (90 μL, 0.65 mmol) according to general method C. The crude product was purified by flash chromatography (1–20% v/v MeOH in DCM) to give compound 38 as a yellow solid (45 mg, 0.11 mmol, 67% yield). 1H NMR (400 MHz, CDCl3) δ 10.73 (br s, 1H), 9.62 (br s, 1H), 7.78–7.65 (m, 2H), 7.57 (br s, 2H), 7.46 (d, J = 1.4 Hz, 1H), 7.10 (d, J = 1.3 Hz, 1H), 7.02 (s, 1H), 6.95 (dd, J = 10.5, 8.8 Hz, 4H), 4.88 (q, J = 6.8 Hz, 1H), 3.84 (s, 3H), 3.73 (s, 3H), 1.68 (d, J = 6.8 Hz, 3H) ppm. 13C NMR (125 MHz, CDCl3) δ 171.4, 158.7, 155.3, 141.8, 140.8, 137.9, 129.1, 126.3, 125.8, 115.8, 115.3, 114.2, 74.7, 55.3, 33.4, 18.4 ppm. LCMS (ESI+) m/z 418.3 [M + H]+, retention time 4.69 min (100%). HRMS (ESI+): m/z calculated for C23H24N5O3 [M + H]+: 418.1879. Found: 418.1875.
N-(4-(4-Iodophenyl)-1H-imidazol-2-yl)-2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanamide (39)
4-(4-Iodophenyl)-1H-imidazol-2-amine 54 (46 mg, 0.16 mmol) was reacted with the acid chloride 60 (43 mg, 0.16 mmol) and Et3N (90 μL, 0.65 mmol) according to general method C. The crude product was purified by flash chromatography (1–20% v/v MeOH in DCM) to give compound 39 as a yellow solid (50 mg, 0.10 mmol, 61% yield). 1H NMR (500 MHz, CDCl3) δ 10.75 (br s, 1H), 9.42 (br s, 1H), 7.69 (t, J = 8.3 Hz, 4H), 7.53–7.33 (m, 3H), 7.10 (d, J = 8.3 Hz, 2H), 6.95 (d, J = 8.9 Hz, 2H), 4.88 (q, J = 6.7 Hz, 1H), 3.71 (s, 3H), 1.66 (d, J = 6.7 Hz, 3H) ppm. 13C NMR (125 MHz, CDCl3) δ 171.5, 155.2, 141.7, 138.0, 137.7, 129.1, 126.5, 126.3, 115.8, 115.3, 108.1, 91.8, 74.5, 33.5, 18.5 ppm. LCMS (ESI+) m/z 514.2 [M + H]+, retention time 4.61 min (95%). HRMS (ESI+): m/z calculated for C22H21IN5O2 [M + H]+: 514.0740. Found: 514.0740.
2-Chloro-N-phenylacetamide (40)
Aniline (400 μL, 4.38 mmol) was reacted with chloroacetyl chloride (380 μL, 4.77 mmol) and Et3N (670 μL, 4.77 mmol) according to general method E, and used in subsequent reactions without further purification. Compound 40 was obtained (720 mg, 4.24 mmol, 96% yield) as a green solid and was used without further purification. 1H NMR (400 MHz, CDCl3) δ 8.27 (br s, 1H), 7.58 (d, J = 7.7 Hz, 2H), 7.39 (t, J = 7.9 Hz, 2H), 7.20 (t, J = 7.4 Hz, 1H), 4.22 (s, 2H) ppm. 13C NMR (100 MHz, CDCl3) δ 163.9, 136.8, 129.3, 125.4, 120.2, 43.0 ppm. LCMS (ESI+) m/z 170.1 [M + H]+, retention time 1.58 min (100%). HRMS (ESI+): m/z calculated for C8H9ClNO [M + H]+: 170.0373. Found: 170.0366. NMR data is in accordance with literature values.(39)
N-(Benzofuran-5-yl)-2-chloroacetamide (41)
Benzofuran-5-amine (140 mg, 1.05 mmol) was reacted with chloroacetyl chloride (93 μL, 1.16 mmol) and Et3N (162 μL, 1.16 mmol) according to general method E, and it was used in subsequent reactions without further purification. Compound 41 was obtained (219 mg, 1.04 mmol, 99% yield) as a green solid and was used without further purification. 1H NMR (400 MHz, CDCl3) δ 8.41 (br s, 1H), 7.94 (d, J = 2.2 Hz, 1H), 7.64 (d, J = 2.2 Hz, 1H), 7.47 (dd, J = 8.8, 0.8 Hz, 1H), 7.32 (dd, J = 8.8, 2.2 Hz, 1H), 6.76 (dd, J = 2.2, 0.9 Hz, 1H), 4.23 (s, 2H) ppm. 13C NMR (100 MHz, CDCl3) δ 163.9, 152.4, 146.1, 131.9, 127.9, 117.7, 113.3, 111.6, 106.8, 43.0 ppm. LCMS (ESI+) m/z 210.1 [M + H]+, retention time 1.71 min (100%). HRMS (ESI+): m/z calculated for C11H9ClNO2 [M + H]+: 210.0322. Found: 210.0319.
2-Chloro-N-(4-iodophenyl)acetamide (42)
4-Iodoaniline (500 mg, 2.28 mmol) was reacted with chloroacetyl chloride (200 μL, 2.51 mmol) and Et3N (350 μL, 2.51 mmol) according to general method E. The crude product was purified by flash chromatography (10–100% v/v EtOAc in petroleum ether) to give compound 42 as a brown solid (600 mg, 2.03 mmol, 89% yield). 1H NMR (400 MHz, CDCl3) δ 8.32–8.11 (br s, 1H), 7.69 (d, J = 8.8 Hz, 2H), 7.36 (d, J = 8.7 Hz, 2H), 4.20 (s, 2H) ppm. 13C NMR (100 MHz, CDCl3) δ 163.8, 138.1, 136.4, 121.8, 88.7, 42.9 ppm. LCMS (ESI−) m/z 293.7 [M – H], retention time 2.38 min (100%). HRMS (ESI−): m/z calculated for C8H6ClINO [M – H]: 293.9183. Found: 293.9187. NMR data is in accordance with literature values.(40)
N-(4-(1H-Imidazol-4-yl)phenyl)-2-chloroacetamide (43)
4-(1H-Imidazol-4-yl)aniline (200 mg, 1.25 mmol) was reacted with chloroacetyl chloride (109 μL, 1.38 mmol) and Et3N (192 μL, 1.38 mmol) according to general method E. The crude product was purified by flash chromatography (3–20% v/v MeOH in DCM) to give compound 43 as a yellow solid (264 mg, 1.12 mmol, 89% yield). 1H NMR (400 MHz, DMSO-d6) δ 10.43 (br s, 1H), 9.83 (br s, 1H), 7.98 (br s, 1H), 7.77–7.65 (m, 2H), 7.70–7.47 (m, 3H), 4.27 (s, 2H) ppm. LCMS (ESI+) m/z 236.2 [M + H]+, retention time 1.15 min (100%). HRMS (ESI+): m/z calculated for C11H10ClN3O [M + H]+: 236.0591. Found: 236.0590.
(5-(4-Bromophenyl)-1H-imidazol-2-yl)methanamine Hydrochloride (44)
4 M HCl (2 mL) was added slowly to a stirred solution of benzyl ((5-(4-bromophenyl)-1H-imidazol-2-yl)methyl)carbamate(36) (105 mg, 0.26 mmol) in dioxane (1 mL). The mixture was stirred at 100 °C for 4 h, and then the solvents were removed under reduced pressure to give the desired product as a white solid (86 mg, 0.26 mmol, 100% yield). 1H NMR (500 MHz, MeOD) δ 8.00 (s, 1H), 7.75–7.67 (m, 4H), 4.55 (s, 2H) ppm. 13C NMR (125 MHz, MeOD) δ 140.8, 136.0, 133.7, 128.4, 127.6, 124.7, 117.8, 111.4, 34.8 ppm. LCMS (ESI−) m/z 250.1 [M – H], retention time 2.13 min (100%). HRMS (ESI−): m/z calculated for C10H9BrN3 [M + H]+: 249.9980. Found: 249.9987.
N-(4-(4-Bromophenyl)-1H-imidazol-2-yl)acetamide (45)
Following general method F, from 2-bromo-1-(4-bromophenyl)ethanone (104 mg, 0.38 mmol) was obtained 45 (88 mg, 0.31 mmol, 84% yield) as a green solid. 1H NMR (500 MHz, DMSO-d6) δ 11.66 (br s, 1H), 11.20 (br s, 1H), 7.72–7.57 (m, 2H), 7.54–7.42 (m, 2H), 7.30 (d, J = 1.6 Hz, 1H), 2.05 (s, 3H) ppm. 13C NMR (125 MHz, DMSO-d6) δ 169.0, 141.9, 135.5, 134.4, 131.7, 126.4, 119.0, 110.4, 23.3 ppm. LCMS (ESI+) m/z 282.1 [M + H]+, retention time 2.13 min (100%). HRMS (ESI+): m/z calculated for C11H11BrN3O [M + H]+: 280.0085. Found: 280.0080. NMR data is in accordance with literature values.(37)
N-(4-Phenyl-1H-imidazol-2-yl)acetamide (46)
Following general method F, from 2-bromo-1-phenylethan-1-one (100 mg, 0.50 mmol) was obtained 46 (79 mg, 0.39 mmol, 78% yield) as a green solid. 1H NMR (400 MHz, DMSO-d6) δ 11.59 (br s, 1H), 11.24 (br s, 1H), 7.71 (d, J = 7.6 Hz, 2H), 7.32 (t, J = 7.6 Hz, 2H), 7.25 (br s, 1H), 7.16 (t, J = 7.4 Hz, 1H), 2.07 (s, 3H) ppm.13C NMR (100 MHz, DMSO-d6) δ 169.0, 141.7, 136.6, 128.9, 126.3, 124.4, 109.6, 23.3 ppm. LCMS (ESI−) m/z 200.0 [M – H], retention time 1.44 min (100%). HRMS (ESI+): m/z calculated for C11H12N3O [M + H]+: 202.0980. Found: 202.0977. NMR data is in accordance with literature values.(37)
N-(4-(4-Morpholinophenyl)-1H-imidazol-2-yl)acetamide (47)
Following general method F, from 2-bromo-1-(4-(piperidin-1-yl)phenyl)ethan-1-one (106 mg, 0.38 mmol) was obtained 47 (90 mg, 0.31 mmol, 83% yield) as a brown solid. 1H NMR (500 MHz, DMSO-d6) δ 11.46 (br s, 1H), 11.17 (br s, 1H), 7.55 (d, J = 8.2 Hz, 2H), 7.06 (br s, 1H), 6.89 (d, J = 8.5 Hz, 2H), 3.93–3.53 (m, 4H), 3.07 (t, J = 4.8 Hz, 4H), 2.04 (s, 3H) ppm.13C NMR (125 MHz, DMSO-d6) δ 168.8, 149.8, 141.4, 136.8, 125.2, 115.6, 113.5, 107.9, 66.6, 49.0, 23.2 ppm. LCMS (ESI−) m/z 285.1 [M – H], retention time 1.60 min (95%). HRMS (ESI+): m/z calculated for C15H19N4O2 [M + H]+: 287.1508. Found: 287.1505.
N-(4-(4-Methoxyphenyl)-1H-imidazol-2-yl)acetamide (48)
Following general method F, from 2-bromo-1-(4-methoxyphenyl)ethan-1-one (150 mg, 0.66 mmol) was obtained 48 (110 mg, 0.47 mmol, 71% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.52 (br s, 1H), 11.20 (br s, 1H), 7.62 (d, J = 8.1 Hz, 2H), 7.11 (s, 1H), 6.89 (d, J = 8.4 Hz, 2H), 3.75 (s, 3H), 2.06 (s, 3H) ppm. 13C NMR (100 MHz, DMSO-d6) δ 168.5, 157.8, 141.2, 136.1, 127.6, 125.3, 113.9, 107.9, 55.1, 22.9 ppm. LCMS (ESI−) m/z 230.1 [M – H], retention time 1.52 min (96%). HRMS (ESI+): m/z calculated for C12H14N3O2 [M + H]+: 232.1086. Found: 232.1082. NMR data is in accordance with literature values.(37)
N-(4-(4-Iodophenyl)-1H-imidazol-2-yl)acetamide (49)
Following general method F, from 2-bromo-1-(4-iodophenyl)ethan-1-one (214 mg, 0.66 mmol) was obtained 49 (190 mg, 0.58 mmol, 88% yield) as a green solid. 1H NMR (500 MHz, DMSO-d6) δ 11.66 (br s, 1H), 11.21 (br s, 1H), 7.64 (d, J = 8.1 Hz, 2H), 7.51 (d, J = 8.1 Hz, 2H), 7.30 (s, 1H), 2.05 (s, 3H) ppm. 13C NMR (125 MHz, DMSO-d6) δ 168.5, 141.4, 137.1, 135.1, 134.3, 126.2, 109.9, 90.9, 22.8. LCMS (ESI−) m/z 326.0 [M – H], retention time 2.43 min (100%). HRMS (ESI+): m/z calculated for C11H11IN3O [M + H]+: 327.9947. Found: 327.9945.
4-(4-Bromophenyl)-1H-imidazol-2-amine (50)
Following general method G, from N-(4-(4-bromophenyl)-1H-imidazol-2-yl)acetamide 45 (88 mg, 0.31 mmol) was obtained 50 (70 mg, 0.29 mmol, 95% yield) as a brown solid.1H NMR (500 MHz, DMSO-d6) δ 7.52 (d, J = 8.1 Hz, 2H), 7.42 (d, J = 8.6 Hz, 2H), 7.04 (s, 1H), 5.40 (br s, 2H) ppm. 13C NMR (125 MHz, DMSO-d6) δ 150.4, 134.0, 131.2, 126.0, 125.4, 117.5, 110.5 ppm. LCMS (ESI+) m/z 240.1 [M + H]+, retention time 1.50 min (97%). HRMS (ESI+): m/z calculated for C9H9BrN3 [M + H]+: 237.9980. Found: 237.9980. NMR data is in accordance with literature values.(37)
4-Phenyl-1H-imidazol-2-amine (51)
Following general method G, from N-(4-phenyl-1H-imidazol-2-yl)acetamide 46 (70 mg, 0.35 mmol) was obtained 51 (47 mg, 0.29 mmol, 84% yield) as a red solid. 1H NMR (400 MHz, MeOD) δ 7.62–7.46 (m, 2H), 7.31 (t, J = 7.7 Hz, 2H), 7.16 (td, J = 7.5, 1.2 Hz, 1H), 6.91 (s, 1H) ppm.13C NMR (125 MHz, MeOD) δ 150.3, 132.9, 128.4, 128.1, 125.6, 123.5, 111.2 ppm. LCMS (ESI+) m/z 160.1 [M + H]+, retention time 1.45 min (100%). HRMS (ESI+): m/z calculated for C9H10BrN3 [M + H]+: 160.0875. Found: 160.0873. NMR data is in accordance with literature values.(37)
4-(4-Morpholinophenyl)-1H-imidazol-2-amine (52)
Following general method G, from N-(4-(4-morpholinophenyl)-1H-imidazol-2-yl)acetamide 47 (85 mg, 0.30 mmol) was obtained 52 (60 mg, 0.24 mmol, 82% yield) as a brown solid. 1H NMR (400 MHz, MeOD) δ 7.47 (d, J = 8.8 Hz, 2H), 6.96 (d, J = 8.8 Hz, 2H), 6.78 (s, 1H), 3.91–3.64 (m, 4H), 3.19–3.02 (m, 4H) ppm. 13C NMR (125 MHz, MeOD) δ 149.9, 149.8, 129.3, 125.0, 124.5, 115.8, 113.0, 66.6, 49.4 ppm. LCMS (ESI+) m/z 245.1 [M + H]+, retention time 1.50 min (100%). HRMS (ESI+): m/z calculated for C13H17N4O [M + H]+: 245.1402. Found: 245.1390.
4-(4-Methoxyphenyl)-1H-imidazol-2-amine (53)
Following general method G, from N-(4-(4-methoxyphenyl)-1H-imidazol-2-yl)acetamide 48 (83 mg, 0.36 mmol) was obtained 53 (65 mg, 0.34 mmol, 95% yield) as a brown solid. 1H NMR (400 MHz, MeOD) δ 7.54–7.39 (m, 2H), 7.04–6.85 (m, 2H), 6.78 (s, 1H), 3.81 (s, 3H) ppm. 13C NMR (100 MHz, MeOD) δ 158.3, 150.0, 125.9, 125.0, 124.8, 113.6, 109.8, 54.3 ppm. LCMS (ESI+) m/z 190.2 [M + H]+, retention time 1.61 min (100%). HRMS (ESI+): m/z calculated for C10H12N3O [M + H]+: 190.0980. Found: 190.0979. NMR data is in accordance with literature values.(37)
4-(4-Iodophenyl)-1H-imidazol-2-amine (54)
Following general method G, from N-(4-(4-iodophenyl)-1H-imidazol-2-yl)acetamide 49 (150 mg, 0.46 mmol) was obtained 54 (103 mg, 0.36 mmol, 79% yield) as a red solid. 1H NMR (500 MHz, DMSO-d6) δ 7.58 (d, J = 8.3 Hz, 2H), 7.38 (d, J = 8.2 Hz, 2H), 7.03 (s, 1H), 5.35 (br s, 2H) ppm. 13C NMR (125 MHz, DMSO-d6) δ 150.8, 137.4, 134.6, 133.4, 126.0, 111.1, 90.2 ppm. LCMS (ESI+) m/z 286.1 [M + H]+, retention time 2.09 min (100%). HRMS (ESI+): m/z calculated for C9H9IN3 [M + H]+: 285.9841. Found: 285.9846.
4-(1H-Imidazol-4-yl)phenol (55)
2-Bromo-1-(4-hydroxyphenyl)ethan-1-one (1.0 g, 4.67 mmol) was dissolved in formamide (5 mL), and the reaction mixture was heated at 150 °C for 24 h. After cooling to rt, the resulting mixture was diluted with EtOAc (40 mL) and washed with saturated aqueous NaHCO3 (40 mL). The aqueous layer was extracted with EtOAc (2 × 40 mL), and the combined organic layers were dried over MgSO4, filtered, and concentrated to give 55 (0.75 g, 4.67 mmol, 99% yield) as a brown oil. 1H NMR (400 MHz, MeOD) δ 8.17 (br s, 1H), 7.68 (d, J = 1.2 Hz, 1H), 7.64–7.44 (m, 2H), 7.25 (d, J = 1.1 Hz, 1H), 6.82 (d, J = 8.6 Hz, 2H) ppm.13C NMR (100 MHz, MeOD) δ 156.3, 135.1, 125.8, 124.3, 115.1 ppm. LCMS (ESI+) m/z 161.2 [M + H]+, retention time 0.54 min (100%). HRMS (ESI+): m/z calculated for C9H9N2O [M + H]+: 161.0715. Found: 161.0720. NMR data is in accordance with literature values.(38)
4-(4-Methoxyphenyl)-1-methyl-1H-imidazole (56)
To a solution of 55 (2.77 g, 17 mmol) in DMF (20 mL) containing cesium carbonate (12.4 g, 38 mmol) at rt was added iodomethane (2.4 mL, 38 mmol). The reaction mixture was stirred at rt for 8 h. The reaction mixture was then cooled to 0 °C and quenched with H2O (70 mL) and extracted with EtOAc (3 × 60 mL). The combined organic layers were washed with brine (3 × 100 mL), dried over MgSO4, filtered, and concentrated to give 56 (2.8 g, 15 mmol, 88% yield) as a brown solid. 1H NMR (400 MHz, CDCl3) δ 7.71 (d, J = 8.8 Hz, 2H), 7.46 (d, J = 1.3 Hz, 1H), 7.10 (d, J = 1.4 Hz, 1H), 6.94 (d, J = 8.9 Hz, 2H), 3.85 (s, 3H), 3.73 (s, 3H) ppm. 13C NMR (100 MHz, CDCl3) δ 158.6, 142.4, 137.8, 127.2, 126.0, 114.8, 114.0, 55.3, 33.5. LCMS (ESI+) m/z 189.2 [M + H]+, retention time 1.38 min (100%). HRMS (ESI+): m/z calculated for C11H13N2O [M + H]+: 189.1028. Found: 189.1031. NMR data is in accordance with literature values.(41)
4-(1-Methyl-1H-imidazol-4-yl)phenol (57)
A solution of 4-(4-methoxyphenyl)-1-methyl-1H-imidazole 56 (1.21 g, 6.42 mmol) in anhydrous DCM (30 mL) at −78 °C was treated with BBr3 (16 mL, 16.05 mmol, 1 M in DCM). The reaction mixture was stirred at −78 °C for 10 min and 2 h at rt. The reaction mixture was then cooled to −78 °C and quenched with MeOH (4 mL). The solvents were then removed under reduced pressure, and the crude product was redissolved in EtOAc (50 mL) and washed with saturated NaHCO3 solution. The aqueous phase was extracted with EtOAc (2 × 50 mL), and the combined organic fractions were dried with anhydrous MgSO4 and filtered off, and the solvent was removed under vacuum to give 57 (1.30 g, 6.17 mmol, 96% yield) as a brown solid, which was used in the next step without further purification. 1H NMR (400 MHz, MeOD) δ 8.99–8.80 (m, 1H), 7.77 (d, J = 1.6 Hz, 1H), 7.55 (d, J = 8.7 Hz, 2H), 6.93 (d, J = 8.8 Hz, 2H), 3.99 (s, 3H) ppm. 13C NMR (100 MHz, MeOD) δ 159.1, 135.1, 134.6, 126.9, 117.3, 117.2, 115.8, 35.0 ppm. LCMS (ESI+) m/z 175.1 [M + H]+, retention time 1.09 min (100%). HRMS (ESI+): m/z calculated for C10H11N2O [M + H]+: 175.0871. Found: 175.0870.
Methyl 2-(4-(1-Methyl-1H-imidazol-4-yl)phenoxy)propanoate (58)
Cs2CO3 (3.46 g, 10.62 mmol) and methyl 2-bromopropanoate (1.29 g, 6.81 mmol) were added to a solution of phenol derivative 57 (1.12 g, 5.32 mmol) in anhydrous DMF (10 mL), and the mixture was stirred at rt for 1 h and at 60 °C for 5 h. After cooling to rt, the reaction was diluted with water (80 mL) and extracted with EtOAc (2 × 80 mL). The combined organic layers were washed with brine (3 × 100 mL), dried over MgSO4, filtered, and concentrated. The crude product was purified by flash chromatography (0–20% v/v MeOH in DCM) to give 58 (1.30 g, 4.99 mmol, 92% yield) as an orange oil. 1H NMR (400 MHz, CDCl3) δ 7.74–7.61 (m, 2H), 7.45 (d, J = 1.3 Hz, 1H), 7.08 (d, J = 1.4 Hz, 1H), 6.96–6.81 (m, 2H), 4.81 (q, J = 6.8 Hz, 1H), 3.77 (s, 3H), 3.71 (s, 3H), 1.64 (d, J = 6.8 Hz, 3H) ppm. 13C NMR (100 MHz, CDCl3) δ 172.8, 156.5, 142.1, 137.8, 128.1, 126.0, 115.3, 115.1, 72.7, 52.3, 33.5, 18.6 ppm. LCMS (ESI+) m/z 261.3 [M + H]+, retention time 1.50 min (100%). HRMS (ESI+): m/z calculated for C14H17N2O3 [M + H]+: 261.1239. Found: 261.1232.
2-(4-(1-Methyl-1H-imidazol-4-yl)phenoxy)propanoic Acid (59)
Methyl 2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanoate 58 (0.53 g, 2.04 mmol) was dissolved in a 2:1 v/v mixture of THF and H2O (7.5 mL), and then NaOH (0.16 g, 4.08 mmol) was added. The reaction mixture was heated at 80 °C for 40 min. After the reaction mixture was allowed to cool to rt, the solvents were removed. The mixture was then diluted with H2O (5 mL) and acidified to pH 7–8 using 1 N HCl. The mixture was cooled, and the resulting precipitate was collected by filtration, washed with water (2 × 3 mL) and petroleum ether (2 × 3 mL), and dried in vacuo to afford acid 59 (0.33 g, 1.33 mmol, 65% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 7.69–7.55 (m, 3H), 7.46 (d, J = 1.3 Hz, 1H), 6.84 (d, J = 8.8 Hz, 2H), 4.81 (q, J = 6.8 Hz, 1H), 3.66 (s, 3H), 1.49 (d, J = 6.7 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 173.3, 156.1, 140.4, 138.2, 127.8, 125.4, 115.8, 114.8, 71.7, 33.1, 18.4. LCMS (ESI+) m/z 247.2 [M + H]+, retention time 1.43 min (100%). HRMS (ESI+): m/z calculated for C13H15N2O3 [M + H]+: 247.1083. Found: 247.1080.
(S)-2-(4-(1-Methyl-1H-imidazol-4-yl)phenoxy)propanoic Acid ((S)-59)
To a solution of 57 (212 mg, 1.21 mmol) in anhydrous THF (5 mL) was added ethyl d-lactate (215 mg, 1.82 mmol). After the reaction mixture was cooled to 0 °C, PPh3 (478 mg, 1.82 mmol) and DEAD (286 μL, 1.82 mmol) were added, and the reaction mixture was stirred for 16 h at rt. The reaction mixture was poured into ice–water (40 mL) and extracted with DCM (3 × 50 mL). The combined organic layers were washed with brine, dried over anhydrous MgSO4, filtered, and concentrated. The crude product was purified by flash chromatography (0–15% v/v MeOH in DCM) to give ethyl (S)-2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanoate as an orange solid. LCMS (ESI+) m/z 275.3 [M + H]+, retention time 2.06 min (94%). Ethyl (S)-2-(4-(1-methyl-1H-imidazol-4-yl)phenoxy)propanoate (200 mg, 0.73 mmol) was dissolved in a 2:1 v/v mixture of THF and H2O (7.5 mL), and then NaOH (58 mg, 1.46 mmol) was added. The reaction mixture was stirred at rt for 3 h, and the solvents were removed. The residue was dissolved in EtOAc (40 mL) and then extracted with H2O (40 mL). The aqueous layer was acidified to pH 7–8 using 1 N HCl. The mixture was cooled, and the resulting precipitate was collected by filtration, washed with water (2 × 3 mL) and petroleum ether (2 × 3 mL), and dried in vacuo to afford acid (S)-59 (100 mg, 0.40 mmol, 56% yield) as a white solid. [α]D25 −51.6 (c 1.0, CHCl3). LCMS (ESI+) m/z 247.2 [M + H]+, retention time 1.43 min (100%). HRMS (ESI+): m/z calculated for C13H15N2O3 [M + H]+: 247.1083. Found: 247.1079.
4-(4-Bromophenyl)oxazol-2-amine (61)
A mixture of 2-bromo-1-(4-bromophenyl)ethanone (200 mg, 0.72 mmol) and urea (435 mg, 7.2 mmol) in anhydrous acetonitrile (5 mL) was heated at 80 °C for 18 h. The solvent was removed, and the residue was purified by flash chromatography (0–10% v/v MeOH in DCM) to afford 61 (134 mg, 0.56 mmol, 78% yield) as a yellow solid. 1H NMR (400 MHz, MeOD) δ 7.68 (s, 1H), 7.59–7.42 (m, 4H) ppm. 13C NMR (125 MHz, MeOD) δ 163.9, 139.8, 132.7, 132.1, 128.8, 127.9, 122.1 ppm. LCMS (ESI+) m/z 238.8 [M + H]+, retention time 1.95 min (100%). HRMS (ESI+): m/z calculated for C9H8BrN2O [M + H]+: 238.9820. Found: 238.9816.
2. Enzyme Assay
The activity of Mth IMPDH ΔCBS was determined using a plate reader by monitoring the production of NADH in absorbance at 340 nm and corrected for noncatalyzed chemical reactions in the absence of Mth IMPDH ΔCBS. All the measurements were done in the assay buffer (50 mM Tris HCl pH 8, 1 mM DTT, 1 mM EDTA, and 100 mM KCl) at 37 °C with 20 nM Mth IMPDH ΔCBS, 2.8 mM NAD+, and 1 mM IMP in a total of 150 μL volume in a 96 well plate-based format, and data were collected for 32 min. The reaction was initiated by the addition of the substrate, IMP, at a concentration of 1 mM. All reactions were performed in triplicate. Prior to reaction initiation, the compounds were preincubated in a buffer with enzyme for 5 min. The inhibitors were dissolved in DMSO-d6 and diluted to a final concentration of 1% v/v in experimental reactions.
IC50 values were calculated by plotting the percentage of inhibition against the logarithm of inhibitor concentration, and dose–response curves were fitted using Prism software (GraphPad).
The Ki value for NAD+ was determined at a constant saturating IMP concentration (1 mM) and five different concentrations of NAD+ (0.35, 0.70, 1.0, 1.4, and 2.8 mM) in the presence of increasing concentrations of inhibitor. The value of Ki for IMP was determined at fixed saturating concentration of NAD+ (2.8 mM) and different concentrations of IMP (0.12, 0.18, 0.25, 0.5, and 1 mM) and inhibitor.
The initial velocities at various inhibitor concentrations were determined based on the slope in the linear part of each reaction containing the inhibitor and the uninhibited reaction. To determine the inhibition constant (Ki values), the initial rate data versus substrate concentration at different inhibitor concentrations were fit using Prism software (GraphPad) to equations for uncompetitive or mixed inhibition. For each inhibitor concentration, the reciprocal of enzyme reaction velocity versus the reciprocal of the substrate concentration was plotted in a Lineweaver–Burk plot to determine the pattern of inhibition.
3. Protein Purification, Crystallization, and Data Collection of Mth IMPDH ΔCBS
Mth IMPDH was expressed, purified, and crystallized as previously described.(24,25) Briefly, hexahistidine tagged Mth IMPDH ΔCBS in pHat2 was expressed overnight in BL21 DE3 (NEB) cells at 18 °C by addition of 500 μM IPTG. Cells were lysed in 50 mM Hepes, pH 8.0, 500 mM NaCl, 5% glycerol, 10 mM β-mercaptoethanol, and 20 mM imidazole, and the recombinant protein purified using a Hi-Trap IMAC FF column (GE Healthcare) charged with nickel and an elution gradient of up to 300 mM imidazole. The hexahistidine tag was cleaved by TEV protease, and the purified Mth IMPDH ΔCBS was obtained by negative nickel gravity-flow purification(42) and size exclusion chromatography on a Superdex 200 gel filtration column equilibrated in 20 mM Hepes pH 8.0, 500 mM NaCl, 5% glycerol, and 1 mM TCEP step. The recombinant Mth IMPDH ΔCBS was then concentrated to 12.5 mg/mL for crystallization.
Mth IMPDH ΔCBS protein crystallized in 1 μL + 1 μL hanging drops with 100 mM sodium acetate, pH 5.5, 200 mM calcium chloride, and 8–14% isopropanol. Crystals were soaked overnight in drops of well solution + 5 mM IMP and either 5 mM Fragment 2 or Compound 1 or 1 mM Compound 31. Crystals were cryoprotected by passing through drops containing well solution + 25% glycerol and flash-frozen in liquid nitrogen. Data were collected from the crystals at Diamond Light Source beamline.
4. Structure Solution, Ligand Fitting, and Refinement
Data were processed using XDS(43) and Pointless (CCP4). To solve the structure, molecular replacement was performed with Phenix Phaser(44) using a previously solved IMP-bound Mth IMPDH ΔCBS structure as a probe (PDB IDs: 5J5R; 5K4X; 5K4Z). Refinement was performed using Phenix.refine and manually in Coot.(45) IMP and the inhibitors were sequentially fitted into the density using the LigandFit function of Phenix, and the structures were manually refined further using Coot. Information regarding the crystallographic statistics can be found in Table S2. Protein–ligand interactions were analyzed using Arpeggio(46) and CSM-Lig.(47,48) Compound properties were evaluated using pkCSM.(49) All figures made using Pymol (Schrodinger).
5. Drug Susceptibility Testing against Mtb
An Alamar Blue fluorescence-based broth microdilution assay was used to assess the minimum inhibitory concentration (MIC) of compounds against Mtb H37Rv, as described previously.(25,50) Briefly, Mtb H37Rv was grown in standard Middlebrook 7H9 broth (BD) supplemented with OADC (BD), 0.2% glycerol, and 0.05% Tween-80 to midexponential phase. Compounds dissolved in DMSO (1%) were tested in clear-bottomed, round-well 96-well microtiter plates at eight different concentrations using the standard anti-TB drugs, rifampin and isoniazid, as positive controls. An inoculum of ∼105 bacteria was added to each well, and the plates were incubated at 37 °C for 7 d. On day 7, 10 μL of Alamar Blue (Invitrogen) was added to each well, and plates were further incubated at 37 °C for 24 h. The fluorescence (excitation 544 nm; emission 590 nm) was measured in a FLUOstar OPTIMA plate reader (BMG LABTECH, Offenberg, Germany). Data were normalized to the minimum and maximum inhibition controls to generate a dose response curve (% inhibition) from which the MIC90 was determined.

Supporting Information


The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jmedchem.7b01622.
  • 1H, 13C, and 2D NMR spectra of all new compounds, supplementary Figures S1–S2, whole-cell activity of compounds against M. tuberculosis H37Rv (Table S1), and X-ray data collection and refinement statistics (Table S2) (PDF)
  • Molecular formula strings data file for all chemical structures mentioned in the manuscript (CSV)

Fragment-Based Approach to Targeting Inosine-5′-monophosphate Dehydrogenase (IMPDH) from Mycobacterium tuberculosis

Showing 1/2: jm7b01622_si_002.csv
ABC
1Compound_IDSMILESMth IMPDH IC50 (uM)
21COC1=CC=C(OC(C)C(NC2=CC=C(I)C=C2)=O)C=C10.77 ± 0.06
32BrC1=CC=C(C2=CNC=N2)C=C1674 ± 53
43C1(C2=CC=CC=C2)=CNC=N1
54ClC1=CC=C(C2=CNC=N2)C=C1
65BrC1=C(C2=CC=CC=C2)NN=C1N
76NC1=C(C2=CC=CC(Br)=C2)C=NN1
87OC1=C2C=CC=CC2=CC=C1
98OC1=CC=C2C=CC=CC2=C1
109O=C(O)C1=C(O)C=C2C(O)=CC=CC2=C1
1110OC1=CC=C2C=CC(OC)=CC2=C1
1211OC1=CC2=CC(O)=CC=C2C=C1336 ± 6
1312OC1=CC=C(N2C=CN=C2)C=C1
1413O=C(O)C1=CC=CC=C1N2C=CC=C2
1514O=C(C1=CC2=C(C=CS2)S1)O400 ± 7
1615N#CC1=CNC2=C1C=CC=C2
1716CC1=CC=C(C2=NC(C(O)=O)=CS2)C=C1433 ± 60
1817OCC1=CC=C2C(C=CS2)=C1
1918CC(NC1=CC(C)=CC=C1O)=O512 ± 37
2019OCC1=C(OC(C)(C)C2)C2=CC=C1325 ± 9
2120O=C(NC1=CC=CC=C1)CSC2=NC(C3=CC=C(Br)C=C3)=CN2
2221O=C(NC1=CC=C(OC=C2)C2=C1)CSC3=NC(C4=CC=C(Br)C=C4)=CN3
2322O=C(NC1=CC=C(I)C=C1)CSC2=NC(C3=CC=C(Br)C=C3)=CN2
2423O=C(NC1=CC=CC=C1)CSC2=NC(C3=CC=CC=C3)=CN2
2524O=C(NC1=CC=C(OC=C2)C2=C1)CSC3=NC(C4=CC=CC=C4)=CN3
2625O=C(NC1=CC=C(I)C=C1)CSC2=NC(C3=CC=CC=C3)=CN2
2726O=C(NC(C)C)CSC1=NC(C2=CC=CC=C2)=CN1
2827O=C(O)CSC1=NC(C2=CC=C(Br)C=C2)=CN1
2928O=C(O)CSC1=NC(C2=CC=CC=C2)=CN1
3029O=C(NC1=CC=C(C2=CNC=N2)C=C1)CSC3=NC(C4=CC=C(Br)C=C4)=CN3
3130O=C(NCC1=NC(C2=CC=C(Br)C=C2)=CN1)NC3=CC=C(C4=CNC=N4)C=C358 ± 8
3231CN1C=C(C2=CC=C(OC(C)C(NC3=NC(C4=CC=C(Br)C=C4)=CN3)=O)C=C2)N=C10.52 ± 0.004
3332O=C(NC1=CC=C(C2=CNC=N2)C=C1)CSC3=NC(C4=CC=CC=C4)=CN3
3433CN1C=C(C2=CC=C(OC(C)C(NC3=NC(C4=CC=C(Br)C=C4)=CO3)=O)C=C2)N=C12.21 ± 0.08
3534CN1C=C(C2=CC=C(OC(C)C(NC3=CC=CC=C3)=O)C=C2)N=C1
3635CN1C=C(C2=CC=C(OC(C)C(NC3=CC=C(I)C=C3)=O)C=C2)N=C10.47 ± 0.03
3736CN1C=C(C2=CC=C(OC(C)C(NC3=NC(C4=CC=CC=C4)=CN3)=O)C=C2)N=C12.76 ± 0.21
3837CN1C=C(C2=CC=C(OC(C)C(NC3=NC(C4=CC=C(N5CCOCC5)C=C4)=CN3)=O)C=C2)N=C10.52 ± 0.01
3938CN1C=C(C2=CC=C(OC(C)C(NC3=NC(C4=CC=C(OC)C=C4)=CN3)=O)C=C2)N=C10.64 ± 0.03
4039CN1C=C(C2=CC=C(OC(C)C(NC3=NC(C4=CC=C(I)C=C4)=CN3)=O)C=C2)N=C10.69 ± 0.06
4140O=C(CCl)NC1=CC=CC=C1
4241O=C(CCl)NC1=CC2=C(OC=C2)C=C1
4342O=C(CCl)NC1=CC=C(I)C=C1
4443O=C(CCl)NC1=CC=C(C2=CNC=N2)C=C1
4544BrC1=CC=C(C2=CN=C(CN)N2)C=C1.Cl
4645O=C(C)NC1=NC(C2=CC=C(Br)C=C2)=CN1
4746O=C(C)NC1=NC(C2=CC=CC=C2)=CN1
4847O=C(C)NC1=NC(C2=CC=C(N3CCOCC3)C=C2)=CN1
4948O=C(C)NC1=NC(C2=CC=C(OC)C=C2)=CN1
5049O=C(C)NC1=NC(C2=CC=C(I)C=C2)=CN1


#Author Present Address
(V.S.) H3D Drug Discovery and Development Centre, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, Cape Town, South Africa.
The authors declare no competing financial interest.
Notes
Additional data related to this publication is available at the University of Cambridge data repository: https://doi.org/10.17863/CAM.20512. Structures have been deposited in the Protein Data Bank (PDB codes 5OU1 for IMPDH:IMP:Compound 1; 5OU2 for IMPDH:IMP:Compound 2 and 5OU3 for IMPDH:IMP:Compound 31 complex structures). Authors will release the atomic coordinates and experimental data upon article publication.

Acknowledgments


This work was supported by the Bill and Melinda Gates Foundation (Hit-TB) and FP7 European Project MM4TB Grant no. 260872. D.B.A was supported by a C. J. Martin Research Fellowship from the National Health and Medical Research Council of Australia (APP1072476) and the Jack Brockhoff Foundation (JBF 4186, 2016). D.B.A. and T.L.B. were also funded by a Newton Fund RCUK-CONFAP Grant awarded by The Medical Research Council and Fundação de Amparo à Pesquisa do Estado de Minas Gerais (MR/M026302/1). V.M. and V.S. were also supported by grants from the HHMI (Senior International Research Scholars grant to V.M.), the South African Medical Research Council, and the National Research Foundation.
Abbreviations Used
TB
tuberculosis
Mycobacterium tuberculosis
Mtb
MDR
multidrug resistant
XDR
extensively drug resistant
IMPDH
inosine-5′-monophosphate dehydrogenase
IMP
inosine 5′-monophosphate
CBS
cystathionine β-synthase
XMP
xanthosine monophosphate
NAD+
nicotinamide adenine dinucleotide
Mth
Mycobacterium thermoresistible
MIC
minimal inhibitory concentration.
WHO
World Health Organization

References


This article references 50 other publications.
  1. 1.
    WHO. Tuberculosis: Fact sheet; World Health Organization 2016: Geneva, Switzerland. 2016.
  2. 2.
    Onyebujoh, P.; Zumla, A.; Ribeiro, I.; Rustomjee, R.; Mwaba, P.; Gomes, M.; Grange, J. M. Treatment of tuberculosis: present status and future prospects. Bull. World Health Organ. 2005, 83, 857865
  3. 3.
    Orenstein, E. W.; Basu, S.; Shah, N. S.; Andrews, J. R.; Friedland, G. H.; Moll, A. P.; Gandhi, N. R.; Galvani, A. P. Treatment outcomes among patients with multidrug-resistant tuberculosis: systematic review and meta-analysis. Lancet Infect. Dis. 2009, 9, 153161, DOI: 10.1016/S1473-3099(09)70041-6
  4. 4.
    Ratcliffe, A. J. Inosine 5′-monophosphate dehydrogenase inhibitors for the treatment of autoimmune diseases. Curr. Opin. Drug Discovery Dev. 2006, 9, 595605
  5. 5.
    Chen, L.; Pankiewicz, K. W. Recent development of IMP dehydrogenase inhibitors for the treatment of cancer. Curr. Opin. Drug Discovery Dev. 2007, 10, 403412
  6. 6.
    Olah, E.; Kokeny, S.; Papp, J.; Bozsik, A.; Keszei, M. Modulation of cancer pathways by inhibitors of guanylate metabolism. Adv. Enzyme Regul. 2006, 46, 176190, DOI: 10.1016/j.advenzreg.2006.01.002
  7. 7.
    Nair, V.; Shu, Q. Inosine monophosphate dehydrogenase as a probe in antiviral drug discovery. Antiviral Chem. Chemother. 2007, 18, 245258, DOI: 10.1177/095632020701800501
  8. 8.
    Shu, Q.; Nair, V. Inosine monophosphate dehydrogenase (IMPDH) as a target in drug discovery. Med. Res. Rev. 2008, 28, 219232, DOI: 10.1002/med.20104
  9. 9.
    Shah, C. P.; Kharkar, P. S. Inosine 5′-monophosphate dehydrogenase inhibitors as antimicrobial agents: recent progress and future perspectives. Future Med. Chem. 2015, 7, 14151429, DOI: 10.4155/fmc.15.72
  10. 10.
    Hedstrom, L.; Liechti, G.; Goldberg, J. B.; Gollapalli, D. R. The antibiotic potential of prokaryotic IMP dehydrogenase inhibitors. Curr. Med. Chem. 2011, 18, 19091918, DOI: 10.2174/092986711795590129
  11. 11.
    Hedstrom, L. IMP dehydrogenase: structure, mechanism, and inhibition. Chem. Rev. 2009, 109, 29032928, DOI: 10.1021/cr900021w
  12. 12.
    Jackson, R. C.; Weber, G.; Morris, H. P. IMP dehydrogenase, an enzyme linked with proliferation and malignancy. Nature 1975, 256, 331333, DOI: 10.1038/256331a0
  13. 13.
    Gilbert, H. J.; Lowe, C. R.; Drabble, W. T. Inosine 5′-monophosphate dehydrogenase of Escherichia coli. Purification by affinity chromatography, subunit structure and inhibition by guanosine 5′-monophosphate. Biochem. J. 1979, 183, 481494, DOI: 10.1042/bj1830481
  14. 14.
    Lee, S. A.; Gallagher, L. A.; Thongdee, M.; Staudinger, B. J.; Lippman, S.; Singh, P. K.; Manoil, C. General and condition-specific essential functions of Pseudomonas aeruginosa. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 51895194, DOI: 10.1073/pnas.1422186112
  15. 15.
    Valentino, M. D.; Foulston, L.; Sadaka, A.; Kos, V. N.; Villet, R. A.; Santa Maria, J., Jr.; Lazinski, D. W.; Camilli, A.; Walker, S.; Hooper, D. C.; Gilmore, M. S. Genes contributing to Staphylococcus aureus fitness in abscess- and infection-related ecologies. mBio 2014, 5, e0172914, DOI: 10.1128/mBio.01729-14
  16. 16.
    Sassetti, C. M.; Boyd, D. H.; Rubin, E. J. Genes required for mycobacterial growth defined by high density mutagenesis. Mol. Microbiol. 2003, 48, 7784, DOI: 10.1046/j.1365-2958.2003.03425.x
  17. 17.
    Hedstrom, L. The bare essentials of antibiotic target validation. ACS Infect. Dis. 2017, 3, 24, DOI: 10.1021/acsinfecdis.6b00185
  18. 18.
    Petrelli, R.; Vita, P.; Torquati, I.; Felczak, K.; Wilson, D. J.; Franchetti, P.; Cappellacci, L. Novel inhibitors of inosine monophosphate dehydrogenase in patent literature of the last decade. Recent Pat. Anti-Cancer Drug Discovery 2013, 8, 103125, DOI: 10.2174/1574892811308020001
  19. 19.
    Makowska-Grzyska, M.; Kim, Y.; Wu, R.; Wilton, R.; Gollapalli, D. R.; Wang, X. K.; Zhang, R.; Jedrzejczak, R.; Mack, J. C.; Maltseva, N.; Mulligan, R.; Binkowski, T. A.; Gornicki, P.; Kuhn, M. L.; Anderson, W. F.; Hedstrom, L.; Joachimiak, A. Bacillus anthracis inosine 5′-monophosphate dehydrogenase in action: the first bacterial series of structures of phosphate ion-, substrate-, and product-bound complexes. Biochemistry 2012, 51, 61486163, DOI: 10.1021/bi300511w
  20. 20.
    Usha, V.; Gurcha, S. S.; Lovering, A. L.; Lloyd, A. J.; Papaemmanouil, A.; Reynolds, R. C.; Besra, G. S. Identification of novel diphenyl urea inhibitors of Mt-GuaB2 active against. Microbiology 2011, 157, 290299, DOI: 10.1099/mic.0.042549-0
  21. 21.
    Makowska-Grzyska, M.; Kim, Y.; Gorla, S. K.; Wei, Y.; Mandapati, K.; Zhang, M.; Maltseva, N.; Modi, G.; Boshoff, H. I.; Gu, M.; Aldrich, C.; Cuny, G. D.; Hedstrom, L.; Joachimiak, A. Mycobacterium tuberculosis IMPDH in complexes with substrates, products and antitubercular compounds. PLoS One 2015, 10, e0138976 DOI: 10.1371/journal.pone.0138976
  22. 22.
    Chen, L.; Wilson, D. J.; Xu, Y.; Aldrich, C. C.; Felczak, K.; Sham, Y. Y.; Pankiewicz, K. W. Triazole-linked inhibitors of inosine monophosphate dehydrogenase from human and. J. Med. Chem. 2010, 53, 47684778, DOI: 10.1021/jm100424m
  23. 23.
    Usha, V.; Hobrath, J. V.; Gurcha, S. S.; Reynolds, R. C.; Besra, G. S. Identification of novel Mt-Guab2 inhibitor series active against M. tuberculosis. PLoS One 2012, 7, e33886 DOI: 10.1371/journal.pone.0033886
  24. 24.
    Park, Y.; Pacitto, A.; Bayliss, T.; Cleghorn, L. A.; Wang, Z.; Hartman, T.; Arora, K.; Ioerger, T. R.; Sacchettini, J.; Rizzi, M.; Donini, S.; Blundell, T. L.; Ascher, D. B.; Rhee, K. Y.; Breda, A.; Zhou, N.; Dartois, V.; Jonnala, S. R.; Via, L. E.; Mizrahi, V.; Epemolu, O.; Stojanovski, L.; Simeons, F. R.; Osuna-Cabello, M.; Ellis, L.; MacKenzie, C. J.; Smith, A. R.; Davis, S. H.; Murugesan, D.; Buchanan, K. I.; Turner, P. A.; Huggett, M.; Zuccotto, F.; Rebollo-Lopez, M. J.; Lafuente-Monasterio, M. J.; Sanz, O.; Santos Diaz, G.; Lelievre, J.; Ballell, L.; Selenski, C.; Axtman, M.; Ghidelli-Disse, S.; Pflaumer, H.; Boesche, M.; Drewes, G.; Freiberg, G.; Kurnick, M. D.; Srikumaran, M.; Kempf, D. J.; Green, S. R.; Ray, P. C.; Read, K. D.; Wyatt, P. G.; Barry Rd, C. E.; Boshoff, H. I. Essential but not vulnerable: indazole sulfonamides targeting inosine monophosphate dehydrogenase as potential leads against. ACS Infect. Dis. 2017, 3, 1823, DOI: 10.1021/acsinfecdis.6b00103
  25. 25.
    Singh, V.; Donini, S.; Pacitto, A.; Sala, C.; Hartkoorn, R. C.; Dhar, N.; Keri, G.; Ascher, D. B.; Mondesert, G.; Vocat, A.; Lupien, A.; Sommer, R.; Vermet, H.; Lagrange, S.; Buechler, J.; Warner, D. F.; McKinney, J. D.; Pato, J.; Cole, S. T.; Blundell, T. L.; Rizzi, M.; Mizrahi, V. The inosine monophosphate dehydrogenase, GuaB2, is a vulnerable new bactericidal drug target for tuberculosis. ACS Infect. Dis. 2017, 3, 517, DOI: 10.1021/acsinfecdis.6b00102
  26. 26.
    Cox, J. A.; Mugumbate, G.; Del Peral, L. V.; Jankute, M.; Abrahams, K. A.; Jervis, P.; Jackenkroll, S.; Perez, A.; Alemparte, C.; Esquivias, J.; Lelievre, J.; Ramon, F.; Barros, D.; Ballell, L.; Besra, G. S. Novel inhibitors of Mycobacterium tuberculosis GuaB2 identified by a target based high-throughput phenotypic screen. Sci. Rep. 2016, 6, 38986, DOI: 10.1038/srep38986
  27. 27.
    Moffat, J. G.; Vincent, F.; Lee, J. A.; Eder, J.; Prunotto, M. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat. Rev. Drug Discovery 2017, 16, 531543, DOI: 10.1038/nrd.2017.111
  28. 28.
    Koul, A.; Arnoult, E.; Lounis, N.; Guillemont, J.; Andries, K. The challenge of new drug discovery for tuberculosis. Nature 2011, 469, 483490, DOI: 10.1038/nature09657
  29. 29.
    Payne, D. J.; Gwynn, M. N.; Holmes, D. J.; Pompliano, D. L. Drugs for bad bugs: confronting the challenges of antibacterial discovery. Nat. Rev. Drug Discovery 2007, 6, 2940, DOI: 10.1038/nrd2201
  30. 30.
    Scott, D. E.; Coyne, A. G.; Hudson, S. A.; Abell, C. Fragment-based approaches in drug discovery and chemical biology. Biochemistry 2012, 51, 49905003, DOI: 10.1021/bi3005126
  31. 31.
    Mendes, V.; Blundell, T. L. Targeting tuberculosis using structure-guided fragment-based drug design. Drug Discovery Today 2017, 22, 546554, DOI: 10.1016/j.drudis.2016.10.003
  32. 32.
    Marchetti, C.; Chan, D. S.; Coyne, A. G.; Abell, C. Fragment-based approaches to TB drugs. Parasitology 2018, 145, 184195, DOI: 10.1017/S0031182016001876
  33. 33.
    Wei, Y.; Kuzmic, P.; Yu, R.; Modi, G.; Hedstrom, L. Inhibition of inosine-5′-monophosphate dehydrogenase from Bacillus anthracis: Mechanism revealed by pre-steady-state kinetics. Biochemistry 2016, 55, 52795288, DOI: 10.1021/acs.biochem.6b00265
  34. 34.
    Dunkern, T.; Prabhu, A.; Kharkar, P. S.; Goebel, H.; Rolser, E.; Burckhard-Boer, W.; Arumugam, P.; Makhija, M. T. Virtual and experimental high-throughput screening (HTS) in search of novel inosine 5′-monophosphate dehydrogenase II (IMPDH II) inhibitors. J. Comput.-Aided Mol. Des. 2012, 26, 12771292, DOI: 10.1007/s10822-012-9615-5
  35. 35.
    Gorla, S. K.; Kavitha, M.; Zhang, M.; Chin, J. E.; Liu, X.; Striepen, B.; Makowska-Grzyska, M.; Kim, Y.; Joachimiak, A.; Hedstrom, L.; Cuny, G. D. Optimization of benzoxazole-based inhibitors of Cryptosporidium parvum inosine 5′-monophosphate dehydrogenase. J. Med. Chem. 2013, 56, 40284043, DOI: 10.1021/jm400241j
  36. 36.
    Kitagawa, H.; Ozawa, T.; Takahata, S.; Iida, M.; Saito, J.; Yamada, M. Phenylimidazole derivatives of 4-pyridone as dual inhibitors of bacterial enoyl-acyl carrier protein reductases FabI and FabK. J. Med. Chem. 2007, 50, 47104720, DOI: 10.1021/jm0705354
  37. 37.
    Soh, C. H.; Chui, W. K.; Lam, Y. An efficient and expeditious synthesis of di- and monosubstituted 2-aminoimidazoles. J. Comb. Chem. 2008, 10, 118122, DOI: 10.1021/cc700143n
  38. 38.
    Kumar, S.; Jaller, D.; Patel, B.; LaLonde, J. M.; DuHadaway, J. B.; Malachowski, W. P.; Prendergast, G. C.; Muller, A. J. Structure based development of phenylimidazole-derived inhibitors of indoleamine 2,3-dioxygenase. J. Med. Chem. 2008, 51, 49684977, DOI: 10.1021/jm800512z
  39. 39.
    Jost, C.; Nitsche, C.; Scholz, T.; Roux, L.; Klein, C. D. Promiscuity and selectivity in covalent enzyme inhibition: a systematic study of electrophilic fragments. J. Med. Chem. 2014, 57, 75907599, DOI: 10.1021/jm5006918
  40. 40.
    Fortin, S.; Moreau, E.; Lacroix, J.; Cote, M. F.; Petitclerc, E.; Gaudreault, R. C. Synthesis, antiproliferative activity evaluation and structure-activity relationships of novel aromatic urea and amide analogues of N-phenyl-N’-(2-chloroethyl)ureas. Eur. J. Med. Chem. 2010, 45, 29282937, DOI: 10.1016/j.ejmech.2010.03.018
  41. 41.
    Bellina, F.; Guazzelli, N.; Lessi, M.; Manzini, C. Imidazole analogues of resveratrol: synthesis and cancer cell growth evaluation. Tetrahedron 2015, 71, 22982305, DOI: 10.1016/j.tet.2015.02.024
  42. 42.
    Ascher, D. B.; Cromer, B. A.; Morton, C. J.; Volitakis, I.; Cherny, R. A.; Albiston, A. L.; Chai, S. Y.; Parker, M. W. Regulation of insulin-regulated membrane aminopeptidase activity by its C-terminal domain. Biochemistry 2011, 50, 26112622, DOI: 10.1021/bi101893w
  43. 43.
    Kabsch, W. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 125132, DOI: 10.1107/S0907444909047337
  44. 44.
    Adams, P. D.; Afonine, P. V.; Bunkoczi, G.; Chen, V. B.; Davis, I. W.; Echols, N.; Headd, J. J.; Hung, L. W.; Kapral, G. J.; Grosse-Kunstleve, R. W.; McCoy, A. J.; Moriarty, N. W.; Oeffner, R.; Read, R. J.; Richardson, D. C.; Richardson, J. S.; Terwilliger, T. C.; Zwart, P. H. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 213221, DOI: 10.1107/S0907444909052925
  45. 45.
    Emsley, P.; Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2004, 60, 21262132, DOI: 10.1107/S0907444904019158
  46. 46.
    Jubb, H. C.; Higueruelo, A. P.; Ochoa-Montano, B.; Pitt, W. R.; Ascher, D. B.; Blundell, T. L. Arpeggio: A web server for calculating and visualising interatomic interactions in protein structures. J. Mol. Biol. 2017, 429, 365371, DOI: 10.1016/j.jmb.2016.12.004
  47. 47.
    Pires, D. E.; Blundell, T. L.; Ascher, D. B. mCSM-lig: quantifying the effects of mutations on protein-small molecule affinity in genetic disease and emergence of drug resistance. Sci. Rep. 2016, 6, 29575, DOI: 10.1038/srep29575
  48. 48.
    Pires, D. E.; Ascher, D. B. CSM-lig: a web server for assessing and comparing protein-small molecule affinities. Nucleic Acids Res. 2016, 44, W55761, DOI: 10.1093/nar/gkw390
  49. 49.
    Pires, D. E.; Blundell, T. L.; Ascher, D. B. pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J. Med. Chem. 2015, 58, 40664072, DOI: 10.1021/acs.jmedchem.5b00104
  50. 50.
    Singh, V.; Brecik, M.; Mukherjee, R.; Evans, J. C.; Svetlikova, Z.; Blasko, J.; Surade, S.; Blackburn, J.; Warner, D. F.; Mikusova, K.; Mizrahi, V. The complex mechanism of antimycobacterial action of 5-fluorouracil. Chem. Biol. 2015, 22, 6375, DOI: 10.1016/j.chembiol.2014.11.006


0 件のコメント:

コメントを投稿