Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Article

pubs.acs.org/jmc

Encoded Library Technology as a Source of Hits for the Discovery


and Lead Optimization of a Potent and Selective Class of Bactericidal
Direct Inhibitors of Mycobacterium tuberculosis InhA
Lourdes Encinas,‡ Heather O’Keefe,† Margarete Neu,§ Modesto J. Remuiñań ,‡ Amish M. Patel,†
Ana Guardia,‡ Christopher P. Davie,† Natalia Pérez-Macías,∥ Hongfang Yang,† Maire A. Convery,§
Jeff A. Messer,† Esther Pérez-Herrán,‡ Paolo A. Centrella,⊥ Daniel Á lvarez-Gómez,‡ Matthew A. Clark,⊥
Sophie Huss,‡ Gary K. O’Donovan,† Fátima Ortega-Muro,‡ William McDowell,# Pablo Castañeda,‡
Christopher C. Arico-Muendel,† Stane Pajk,∞ Joaquín Rullás,‡ Iñigo Angulo-Barturen,‡
Emilio Á lvarez-Ruíz,× Alfonso Mendoza-Losana,‡ Lluís Ballell Pages,‡ Julia Castro-Pichel,*,‡
and Ghotas Evindar*,†

ELT Boston, Platform Technology & Science, GlaxoSmithKline, Waltham, Massachusetts 02451, United States

Diseases of the Developing World, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres
Cantos, Madrid, Spain
§
Computational and Structural Chemistry, Platform Technology & Science, GlaxoSmithKline, Stevenage SG1 2NY, Hertfordshire,
U.K.

Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain

X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
#
Biological Reagent and Assay Development, Platform Technology & Science, GlaxoSmithKline, Stevenage SG1 2NY, Hertfordshire,
U.K.

Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
×
Centro de Investigación Básica, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain
*
S Supporting Information

ABSTRACT: Tuberculosis (TB) is one of the world’s oldest and


deadliest diseases, killing a person every 20 s. InhA, the enoyl-
ACP reductase from Mycobacterium tuberculosis, is the target of the
frontline antitubercular drug isoniazid (INH). Compounds that
directly target InhA and do not require activation by
mycobacterial catalase peroxidase KatG are promising candidates
for treating infections caused by INH resistant strains. The
application of the encoded library technology (ELT) to the
discovery of direct InhA inhibitors yielded compound 7 endowed
with good enzymatic potency but with low antitubercular potency.
This work reports the hit identification, the selected strategy for
potency optimization, the structure−activity relationships of a
hundred analogues synthesized, and the results of the in vivo
efficacy studies performed with the lead compound 65.

■ INTRODUCTION
Despite the existence of treatments for tuberculosis (TB), 8.7
compliance is monitored by healthcare workers and has been
successful when appropriately implemented (cure rate of >90%).
million people fell ill with TB and 1.4 million died from TB in Despite this, there is an urgent need for the development of
2011 (latest WHO report1). The increasing prevalence of newer, safer, and effective antitubercular drugs with new targets
multidrug resistant (MDR)2−7 and extremely drug resistant and novel modes of action (MoA).
(XDR)8,9 TB strains represents a threat to public health InhA is an NADH-dependent 2-trans enoyl-acyl carrier
worldwide. Resistance to TB drugs results primarily from protein (ACP) reductase of the type II fatty acid synthase
nonadherence by patients, incorrect drug prescription, or poor
quality drugs. The WHO sponsored implementation of DOTS10 Received: September 4, 2013
(directly observed treatment short course) in which treatment Published: January 22, 2014

© 2014 American Chemical Society 1276 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry Article

(FASII) pathway in Mycobacterium tuberculosis (Mtb).11,12 There


is a strong body of evidence indicating that it is the primary target
of the frontline antitubercular drug isoniazid (INH).13,14 Clinical
isolates and laboratory modified mycobacteria overexpressing
InhA show resistance to INH.14−16 The drug inhibits InhA
enzymatic activity, inducing an accumulation of saturated C24−
C26 fatty acids and blocking the production of longer molecules,
including mycolic acids. This inhibition correlates with Figure 1. Design of DNA-encoded library (DEL).
mycobacterial cell death. The essentiality of InhA has also been
demonstrated by the use of temperature sensitive mutants of
InhA in Mycobacterium smegmatis, where a shift to the diamino acid scaffolds in cycle 1, followed by elaboration with
nonpermissive temperature results in rapid lysis and cell death.12 over 800 amine-capping building blocks (carboxylic acids,
INH is a bactericidal drug and part of the first-line drug aldehydes, sulfonyl chlorides, and isocyates) in cycles 2 and 3
combination regimen for antitubercular therapy and is to generate a 16.1 million compound library. The details of the
specifically active against M. tuberculosis. INH is activated within library synthesis will be the subject of a different publication in
the mycobacterial cell by KatG. The activated form is thought to the near future.
react with NADH within the InhA active site to form an A cubic scatter plot in which each axis represents a cycle of
inhibitory adduct. diversity in the library was used to analyze and visualize the
In vitro activated INH also forms adducts with NAD(P) enriched library members for the selected chemotype (Figure
cofactors that bind to and inhibit InhA and other enzymes like 2).35 Individual points, corresponding to discrete small molecule
DHFR.17 The physiological relevance of these interactions in warheads in the library, are shown in pink and sized according to
vivo is clear in the case of InhA, while the potential role of other number of unique instances recorded by DNA sequencing. The
possible targets of INH in the antitubercular activity of the drug display is set to a minimum of two unique copies per warhead,
remains to be solidly proved. which should indicate significantly enriched binders under these
Resistance to INH, which is one of the hallmarks of MDR selection conditions. In this analysis, the feature of interest is
strains, has been associated with at least five different genes represented by a plane defined by the cycle 1 building block
(katG, inhA, ahpC, kasA, and ndh).18−20 While this finding still (BB1) (2S,4R)-4-aminopyrrolidine-2-carboxylic acid (1), which
remains to be clearly explained, 60−70% of resistant isolates are was attached to DNA through its carboxylic acid functional
directly linked to defects in the katG gene (often with group. Within the selected plane, there are multiple lines due to
compensatory mutations in other genes) and less commonly in preferred disynthon combinations of 1 with specific cycle 3
the inhA structural gene and upstream promoter region. building blocks (BB3). The most prominent line is the BB3 3-
With this background, we and others21−26 were interested in ethyl-1-methyl-1H-pyrazole-5-carboxylic acid (2) that is high-
the development of inhibitors targeted directly at InhA that do lighted in Figure 2. The selected disynthon combination of this
not require previous KatG activation. Specifically, it was our goal selected BB3 and the BB1 (2S,4R)-4-aminopyrrolidine-2-
to develop a new agent that was able to interact with InhA carboxylic acid (1) provides the core structure of the InhA
differently from the known NAD-INH complex, enabling a new feature selected from this library as a single stereoisomer. There
pharmacological profile when compared to INH that should are a number of BB2s selected within the line each represented by
result in activity against INH resistant clinical isolates. a single point; therefore, the BB2 is a variable component of the
In search of potent and selective InhA inhibitors, we utilized selected scaffold that provides additional SAR in the form of
encoded library technology (ELT), a proven novel and robust possible moieties tolerated by the site of interaction on the
hit/lead identification platform27−29 wherein a large collection of protein. This “selection” SAR is a valuable tool in compound
chemotypically diverse DNA-encoded small molecule libraries prioritization for off-DNA synthesis and can provide exquisite
are screened for affinity toward a desired protein target. The guidance in early chemotype lead optimization.
broad chemical diversity and modest target protein requirements In preparation for the off-DNA activity confirmation, three
are two of the major benefits of ELT for early drug discovery. exemplars within the selected disynthon line were chosen to
Successful ELT hit identification campaigns against diverse follow up. The synthetic strategy was designed to put together
targets have been previously reported.30−34 Herein we report the the pharmacophore earlier in the synthesis and add on the
discovery of a novel chemotype identified through ELT affinity variable components later (Scheme 1). This allowed for testing
selection against M. tuberculosis InhA and its subsequent lead of the intermediate in the activity assay and therefore additional
optimization.


SAR exploration. Synthesis was initiated with peptide coupling of
amine 1 to acid 2 that afforded methyl ester 3. Saponification of
RESULTS AND DISCUSSION the ester 3 followed by another amide formation with ethylamine
As described in earlier reports, affinity selection of InhA was gave the desired Boc-protected intermediate 4. TFA removal of
performed by immobilizing biotinylated protein on a streptavidin the Boc protecting group followed by acylation of the free amine
matrix in PhyNexus tips.27 Selections were conducted in parallel produced the desired final compounds (6, 7, and 8). In order to
against a panel of DNA-encoded libraries (DELs), representing a submit the corresponding ester and acids for testing, the desired
diversity of compounds, under conditions of protein alone, compounds were prepared through Boc removal of precursor 3
protein with NAD+, and protein with NADH. While enrichment followed by coupling with the appropriate carboxylic acid to give
of specific chemotypes was observed under all selection the corresponding methyl ester 9. After saponification of methyl
conditions, it was particularly evident for selections run in the ester 9 the corresponding acid 10 was accessible for testing as
presence of NADH cofactor. A putative hit from an aminoproline well.
scaffold (Figure 1) was chosen for further follow-up. The DNA- The prepared compounds were then assayed for their activity
encoded library (Figure 1) utilized 22 orthogonally protected against M. tuberculosis InhA as reported in Table 1. The first three
1277 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry Article

Figure 2. (a) Spotfire cube data view of InhA selection feature in the presence of NADH. (b) Selected chemotype: BB1, cycle 1 building block; BB3,
cycle 3 building block; RCO-, variable cycle 2 building block.

compounds (6, 7, and 8) synthesized off-DNA demonstrated Taking into account the moderate intrinsic clearance found in
inhibitory activity in the InhA assay. These data indicated some the hepatocyte assay, the potential metabolites of 7 were
potential for further modification of the R1 moiety with the 3- examined after incubation with hepatocytes and metabolite
substitued benzofuran 7 giving an IC50 potency of 34 nM. follow-up by MS/MS. Results from N-dealkylation of the
Preliminary SAR exploration around the methylamide was ethylamide and pyrazole unit in addition to the formation of
initiated with synthesis and testing of the corresponding methyl oxidation products were the main detected metabolites (Figure
ester and carboxylic acid. As demonstrated in Table 1, the S1 and Table S1).
inhibitor potency diminished to 215 nM for the methyl ester (9) The complex crystal structure showed that compound 7 binds
and to 1290 nM for the corresponding carboxylic acid (10). to InhA in the cleft normally occupied by substrate/product14,41
In order to better understand the properties of compound 7, (Figure 3). The pyrazole 2-nitrogen makes a hydrogen bonded
early activity and in vitro profiling of the hit structure were interaction to the 2′-hydroxyl group of NADH. In other InhA−
completed (Table 2) along with obtaining a complex crystal inhibitor bound structures an interaction with the NADH 2′-
structure with InhA. As a key issue in any target based program, hydroxyl is also present (see Figure 3B for the triclosan complex
we were able to ascertain the relationship between enzymatic structure14,21) typically as part of a hydrogen bond network
InhA inhibition and antitubercular MIC through the use of an involving Tyr158. In the current complex a network to Tyr158 is
InhAMTB overexpressor strain.36 The compound showed a not formed in part because of Tyr158 maintaining the position it
selective antibacterial activity against M. tuberculosis and was occupies in the apo-enzyme.14 The substrate binding loop is
found to be endowed with good potency against the target and ordered in the structure with helix α6 (residues 197−206)
modest, yet selective antitubercular activity. Furthermore, the closing the volume of the pocket. There are direct hydrogen
compound exhibited an encouraging balance among solubility, bond interactions between main chain atoms of residues in the
lipophilicity, and permeability. The physicochemical properties helix and atoms flanking the central proline of the ligand.
of a compound are fundamental in the drug discovery process, In order to expand the explored chemical space beyond the
mainly because of their influence on determining ADMET building blocks contained within the ELT library and also with
(absorption, distribution, metabolism, excretion, and toxicity) the idea of clarifying any possible bias introduced by the DNA
properties and the overall suitability of drug candidates. For the tags, we decided to introduce novel modifications in the three
oral route of administration the ‘ideal’ set of physicochemical positions of interest around the proline scaffold (Figure 4). The
properties is well established.37−40 The hit was metabolically main objectives of this initial exploration were to increase both
stable when exposed to both human and mice microsomal enzymatic activity and antitubercular potency. The 4-aminopro-
fractions and showed moderate intrinsic clearence in hepato- line central core was left unmodified, since we believed it
cytes. No measurable activity was found against the common provided a desirable scaffold for the appropriate disposition of
cytochrome P450 (CYP450) isoforms (1A2, 2C9, 2D6, 3A4) or the three different arms driving interactions within the InhA
in the hERG dofetilide binding assay (pIC50 < 4.3). Cytotoxicity active site. Chirality at the proline core proved to be essential for
evaluation in HepG2 showed TOX50 > 50 μM. activity (synthesis of 11’s enantiomer in Scheme S1). These
1278 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry Article

Scheme 1. Off-DNA Synthesis of InhA Selected Hitsa

a
Reagents and conditions: (i) N-[(dimethylamino)-1H-1,2,3-triazolo[4,5-b]pyridine-1-ylmethyene]-N-methylmethanaminium hexafluorophosphate
N-oxide (HATU, 1.2 equiv), N,N-diisopropylethylamine (DIPEA, 3.0 equiv), N,N-dimethylformamide (DMF); (ii) lithium hydroxide (LiOH, 3.0
equiv); (iii) HATU (1.2 equiv), DIPEA (3.0 equiv); ethylamine hydrochloride (2.0 equiv); (iv) 20% trifluoroacetic acid in dichloromethane; (v)
carboxylic acid (RCOOH, 1.2 equiv), HATU (1.2 equiv), DIPEA (3.0 equiv), DMF.

conclusions were supported by the evidence generated during This early SAR investigation showed how the replacement of
the ELT selection process, where 22 orthogonally protected the amide bond by an amine group (compound 13) resulted in a
diamino acid scaffolds were deselected for InhA interaction. complete loss of potency (synthesis in Scheme S3). N-alkylation
Preliminary SAR at P1, P2, and P3 Positions. The of the amide (compound 15) was not tolerated. A number of
molecule was divided into three different regions, and one novel replacements (Table 3) gave rise to compounds with excellent
modification at a time was introduced in each position. This enzymatic potency and activities in cellular assay much less
strategy allowed for the quick exploration and combination of the impressive or simply flat (see compounds 17, 26, 36, and 37). On
best chemical features in P1, P2, and P3 (Figure 4). the other hand, some compounds exhibited higher IC50 values
We first turned our attention to the introduction of new that cannot be easily related to the potency of antitubercular
diversity in P1. In order to confirm the importance of P1 in the activity (see compounds 32, 34, and 35). The fact that the
pharmacophore, this position was suppressed. Compound 12 context of the FAS II multienzyme complex is not truly
was still able to inhibit the enzyme with moderate activity (IC50 = represented in the enzymatic assay could be involved in this
disconnection.42 Among the active compounds, 11 was 1 order
0.12 μM), but no whole cell activity was found (synthesis in
of magnitude more potent in the whole cell assay than 7 and
Scheme S2). This evidence together with the realization that this
several times more potent than the rest and had the best
was the DNA-anchoring site for the ELT studies prompted us to physicochemical properties in terms of molecular weight,
explore a wider chemical diversity at this site (Table 3). A solubility, and lipophilicity (data not shown). This served as a
number of modifications were attempted: truncation of the side basis for further optimization at other positions.
chain to the primary amide, elongation and branching of the alkyl Transformations in P2 required the synthesis of the special
chain, aliphatic cycles, introduction of different heteroatoms and precursors 54 and 55 (Scheme 2).
amino acids in addition to exploration of a number of aromatic The effects of different heteroaromatic ring substitutions on
substitutions as depicted in compounds 25−48 where ring the P2 2,4-pyrazole ring were examined. The SAR exploration
changes, linker exploration, and variations of the substitution was restricted to a small number of analogues of the pyrazole
pattern in the aromatic rings were explored. Synthesis of the unit. Other P2 derivatives caused a significant or complete
starting material 10 depicted in Scheme 1 allowed for a facile decrease in enzymatic activity (the SAR is summarized in Table
access of P1 modifications. S2).
1279 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry Article

Table 1. Activity of the First Compounds Made, Off-DNA

Figure 3. Compound 7 bound to the active site of InhA. In (A),


compound 7 is shown in green ball-and-sticks, NADH is shown as thick
lines, and hydrogen bonds are shown as dashed lines. Helix α6 and
Tyr158 are also highlighted. In (B), the complex structure of triclosan
(PDB code 1P45) is shown in gold superposed on the 7 complex
structure. The different positions of helix α6 and Tyr 158 in the two
complexes are visible.

Synthesis of the starting material 5 as depicted in Scheme 1


allowed for a facile access to P3 modifications. Among the
heteroaromatic substituents investigated as possible P3 position
replacements, only the benzofuran and benzothiophene moieties
were associated with a significant in vitro antitubercular potency
(Table S3). This tight SAR was confirmed with additional studies
as shown further below. Figure 4. Three positions of the target compound structure.

Table 2. Complete Profile of 7: In Vitro InhA Inhibition, Antitubercular Activity, Cytotoxicity, and Physicochemical Properties
and in Vitro DMPK Profile

a
Young, R. J.; Green, D. V. S.; Luscombe, C. N.; Hill, A. P. Drug Discovery Today 2011, 16, 822−830. bCLND solubility values that are within 85% of
maximum possible concentration (as determined from DMSO stock concentration).

1280 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288


Journal of Medicinal Chemistry Article

Table 3. P1 Modifications: Structures and Potency of Compounds 11−48

Further Optimization of P1, P2, and P3 Positions. On was fixed as the most suitable group in terms of potency. A
the basis of the preliminary established SAR, a second round of in selected group of mainly nonpolar aliphatic substitutions was
depth exploration of the P2 and P3 positions was initiated. evaluated around P2 (Table 4). The compounds bearing at
Despite the obvious ester hydrolysis liabilities,43 the glycine at P1 position 4 cyclopropyl (58) and propyl (56) groups were found
1281 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry Article

Scheme 2. Synthesis of 54 and 55 Starting from 49a

a
Reagents and conditions: (i) ethylamine hydrochloride (1.2 equiv) or glycine methyl ester (1.1 equiv), HATU (1.2 equiv), DIPEA (3.0 equiv),
DMF; (ii) 20% trifluoroacetic acid in dichloromethane; (iii) carboxylic acid (benzofuran-3-carboxylic acid, 1.2 equiv), HATU (1.2 equiv), DIPEA
(3.0 equiv), DMF; (iv) piperidine/DMF 1:8 v/v, 0.6 M.

Table 4. SAR of the Pyrazole Derivatives: Structures and Potency of Compounds 56−62 Compared with 11

to be equipotent to ethyl (11) in MIC. In contrast, compound 106). With the aim of generating isosteric substitution patterns
57, with the larger isobutyl in the 4-pyrazole position, was found more suitable for further development, different P1 analogues
to be inactive. With regard to possible pyrazole replacement were prepared aimed at overcoming the blood instability issues
patterns, the 2-ethyl substitution, compound 59, showed similar associated with the ester group (Table S5).43 First, ester
IC50 values but 1 order of magnitude improvement in terms of variations were attempted with the aim of altering the rate of
MIC. The three-carbon chain propyl (60) was still active but the hydrolysis (entries 107−113). This approach was unsuccessful
polar 2-hydroxyethyl group (61) and the bulkier benzyl as described in Table S5. Other alternatives, such as replacements
substitution (62) were not tolerated. From this moment onward, with different amide combinations (primary, secondary or
the diethylpyrazole was selected as the P2 substitution pattern of tertiary amides; entries 114, 115, 116) or oxazole (entry 45),
choice. were used, but all led to significant drops in whole cell potency.
With regard to P3, the SAR exploration is summarized in Table Intriguingly, while low enzymatic IC50 values could be achieved,
S4. The synthetic route to prepare the starting material 63 that no compounds with MIC < 1 μM could be identified.
afforded compounds 103−107 is outlined in Scheme S4. These At this stage and given the improved activity of the
studies showed how nonpolar group substitutions at the 2- diethylpyrazole P2 substitution, we chose to re-examine P1
benzofuran position were tolerated. The fact that none of the (Table 5). The synthetic route to prepare the starting material 64
replacements tested at P3 showed any significant improvement that afforded compounds 65−70 is outlined in Scheme S5. A
confirmed how the ELT work around this position was able to selection of some of the best scaffolds in terms of potency,
effectively cover a significant amount of chemical space. The structural diversity, and physicochemical properties led to the
benzofuran at P3 was hence fixed ahead of further P1 synthesis of the new compounds 65−70. However, the desirable
optimization. synergistic nature of the SAR of the diethylpyrazole and P1
Unfortunately, the ester functionality of compound 11, which substituents previously observed with the glycine derivative 59
stood out with an MIC value of 0.5 μM, was deemed unsuitable was not transferable to the same extent to other P1 replacements.
because of the liabilities associated with enzymatic hydrolysis to All the compounds 65−70 showed InhA inhibition at low
generate the likely nonpermeable carboxylic acid product (entry concentrations combined with good activity against M. tuber-
1282 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry Article

Table 5. SAR of Diethylpyrazole Derivatives: Structures and Potency of Compounds 65−70

a
Young, R. J.; Green, D. V. S.; Luscombe, C. N.; Hill, A. P. Getting physical in drug discovery II: the impact of chromatographic hydrophobicity
measurements and aromaticity. Drug Discovery Today 2011, 16, 822−830. bLigand efficiency (LE): enzyme pIC50/heavy (non-hydrogen) atom
count. cMortenson, P. N.; Murray, C. W. J. Comput.-Aided Mol. Des. 2011, 25, 663−667.

Table 6. Complete Profile of Compound 65: In Vitro InhA Inhibition, Antitubercular Activity, and Physicochemical Properties
and Early in Vitro DMPK Profile

culosis with MIC values in the range of 0.2−1 μM. We have Profile of Compound 65. In order to complete its profile,
chosen to use the ligand lipophilicity efficiency index (LLE) compound 65 was tested for activity against intracellular bacteria,
designed by Leeson and Springthorpe. This useful index helps to showing an MIC value of 0.24 μM. This significant intracellular
rank compounds based on potency and lipophilicity along the activity (in THP-1 cells) was accompanied by a clean P450
lead optimization.44 We have also taken into account the LLEAT, profile (1A2, 2C9, 2C19, 2D6, and 3A4; IC50 > 50 μM in all
which combines lipophilicity, size, and potency and has been cases) and good stability values in mouse and human microsomal
designed to have the same target value of 0.3 kcal/mol and
fractions (Table 6).
dynamic range as LE.45−48 As an indication of druglike
properties, compound 65 showed the best ligand efficiency The bactericidal potential of compound 65 was assessed in
value of 0.37 kcal mol−1 atom−1, an LLEAT of 0.42. This was a killing rate experiments with linezolid and moxifloxacin as
significant improvement when compared to compound 7 (LE = bacteriostatic and bactericidal controls, respectively (Figure S2).
0.32, LLEAT = 0.36). The good potency values, combined with a The results show that compound 65 was able to reduce >2 log cfu
reasonable lipophilicity and the lower molecular weight, led us to counts after 1 week of incubation very close to the moxifloxacine
progress this lead molecule to further in vivo evaluation. behavior.
1283 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry


Article

Given its interest, the pharmacokinetic profile of 65 in CONCLUSIONS


C57BL/6J mice following oral administration was evaluated at 50 A class of potent and selective M. tuberculosis direct InhA
mg/kg po (1% methylcellulose) (Figure 5 and Table 7). inhibitors was identified by means of the application of DNA
encoded library technology. Compound 7 was considered a
suitable starting point for further medicinal chemistry
optimization. In order to systematically investigate the chemical
space around the proline core, one modification at a time was
introduced in the molecule around the three possible positions
with the initial goal of improving potency. This approach was
chemically flexible and was able to quickly provide SAR
information. After combination of the best features in each
position, P3 was found to be the least malleable part in terms of
SAR manipulations. The nature of the group at this position was
found to be key for potent enzymatic activity. The diethylpyr-
Figure 5. Peripheral blood levels of 65 after oral administration to azole at position P2 emerged as the most suitable moiety for
C57BL/6J mice (n = 3) at a dose of 50 mg/kg as suspension in 1% antitubercular potency optimizing. The SAR analysis around P1
methylcellulose. Individual values and mean values for each time point provided the ability to improve the physicochemical and ADME
are represented in the plot. properties. Feature combination resulted in the synthesis of the
optimized lead compound 65. Despite its good balance between
InhA inhibition, antitubercular potency, and pharmacokinetic
profile, compound 65 was shown to be inactive in vivo against a
Additionally, compound 65 was assayed by intravenous route murine TB acute infection model.
at 4 mg/kg and showed a high in vivo clearance (CL = 85.5 mL Overall we presented here the optimization of a new
min−1 kg−1) and a short half-life (t1/2 = 0.5 h) in mice (Table 7). antitubercular hit series showing good InhA enzymatic and
The oral PK profile was characterized by a Cmax value of 1.75 μg/ antitubercular potencies and adequate physicochemical proper-
mL at 0.83 h postdosing and an AUC(0−∞) of 3.59 μg·h/mL ties as reflected in attractive LE and LLE numbers. A clear
corresponding to 36% bioavailability (F). disconnection was found between the antitubercular in vitro and
In view of its balanced pharmacological, physicochemical, and in vivo profile of lead compound 65.


in vivo ADME profiles, compound 65 was progressed to both
tolerability and acute efficacy studies in a murine model of M. EXPERIMENTAL SECTION
tuberculosis infection.49 The aim of the preliminary safety study
was to estimate the maximal tolerable single oral dose level of 65 General Procedures. All commercially available reagents and
in female C57BL/6J mice. The MNLD (maximal nonlethal solvents were used without further purification. (2S,4R)-1-tert-Butyl 2-
methyl 4-aminopyrrolidine-1,2-dicarboxylate hydrochloride 1 was
dose) in mice treated with 65 (single oral dose formulated in 1% purchased from Apollo, Advanced Tech, and Tyger. 3-Ethyl-1-methyl-
methylcellulose by gavage followed by observation for 7 days) 1H-pyrazole-5-carboxylic acid 2 was purchased from Chembridge.
was higher than 1000 mg/kg. No adverse clinical events were (2S,4R)-4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-1-(tert-
found. The mean total exposure (AUC), obtained at 24 h, butoxycarbonyl)pyrrolidine-2-carboxylic acid 49 was purchased from
associated with a 1000 mg/kg dose was 73.69 μg·h/mL, and the Aldrich and Cheminpex. Benzofuran-3-carboxylic acid 79 was purchased
corresponding mean Cmax was 23 μg/mL. Subsequently, from Apollo, Bionet, and Maybridge. 1,3-Diethyl-1H-pyrazole-5-
compound 65 was in vivo tested to determine the therapeutic carboxylic acid 84 was purchased from Sinova SL. HATU was purchased
from Carbosynth. The rest of the reagents were obtained from Aldrich,
efficacy against a murine acute M. tuberculosis infection model Maybridge, Apollo, Panreac, Chembridge, Novabiochem, Enamine,
(Figure S3). No significant reduction of colony forming units Combiblocks, Artchem, and JW Pharmlab.
(cfu) at any dose tested was observed in the lungs of infected Reactions were monitored by thin layer chromatography (TLC)
mice. The positive control moxifloxacin (30 mg/kg body weight) using Merck 60 F254 silica gel glass backed plates. Elution was with
reduced the cfu by 3.48 log with respect to untreated mice. suitable solvent mixtures, and visualization was by UV light. Reactions
Despite an acceptable in vivo pharmacokinetic profile and have also been followed by mass spectrometry and reversed phase
good antitubercular potency (0.5 μM), compound 65 was found chromatography using a high resolution spectrometer Waters ZMD
2000 coupled with LC Agilent 1100 with DAD detection. The products
to be inactive in an acute TB infection model. This result
were purified by column chromatography or preparative HPLC.
underlines how antitubercular compounds with good in vitro Automated flash chromatography was performed on a Flash Biotage
activity and aceptable PK properties can still be devoid of in vivo Isolera SP4 system with peak detection at 254 nm, and preparative
activity. Further studies regarding possible explanations for this HPLC was performed in an Agilent 1100 and 1200 with peak detection
disconnection would need to be done. at 254 nm. All products were obtained as amorphous solids, and melting

Table 7. Pharmacokinetic Parameters of 65 after Intravenous Administration of 4 mg/kg a Single Dose (20% Encapsin/%5
DMSO) and Oral Administration of 50 mg/kg b Single Dose (1% Methylcellulose) to C57BL/6J Mice (n = 3)c
route Tmax b (h) Cmax b (μg/mL) AUC(0−∞) b (μg·h/mL) t1/2 a (h) Vd a (L/kg)) CL a (mL min−1 kg−1) F (%)
Compound 65
iv 0.47 (0.20) 3.68 (2.08) 85.51 (15.93)
po 0.83 (0.14) 1.75 (0.54) 3.59 (0.53) 35.65 (5.07)

a
t1/2, half-life; Vd, volume of distribution; CL, clearance. bTmax, time at maximum concentration; Cmax, maximum concentration; AUC(0−∞), area
under the curve for total exposure to infinite time. cValues are included as the mean and standard deviation (SD) of individual values.

1284 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288


Journal of Medicinal Chemistry Article

points were not measured. All NMR spectra were recorded on a Bruker 3-Ethyl-N-(5-(ethylcarbamoyl)pyrrolidin-3-yl)-1-methyl-1H-
DPX Avance 400 MHz instrument equipped with a QNP probe. pyrazole-5-carboxamide 2,2,2-Trifluoroacetate (5). Compound 5
Measurements were made at room temperature and at 80 °C and in an was synthesized from Boc-deprotection of (2S,4R)-tert-butyl 4-(3-ethyl-
appropriate deuterated solvent using residual hydrogenated solvent as 1-methyl-1H-pyrazole-5-carboxamido)-2-(ethylcarbamoyl)pyrrolidine-
standard (CDCl3, δ = 7.26 ppm and DMSO-d6, δ = 2.50 ppm); 1-carboxylate (4) using the protocol described in general method C.
conditions were indicated in each case. Chemical shifts are expressed in The reaction mixture was concentrated to dryness (coevaporated with
parts per million (ppm, δ units). Coupling constants (J) are in units of CH2Cl2 × 5, EtOAc × 1) to give the title compound (4.22 g, 128% yield)
hertz (Hz). Splitting patterns describe apparent multiplicities and are as an orange oil. The yield was >100% because the product contained
designated as s (singlet), d (doublet), t (triplet), q (quartet), dd (double EtOAc which could not be removed under reduced pressure. The crude
doublet), m (multiplet), br (broad). Analytical purity was ≥95% unless product was carried on to next step. MS (ESI) m/z [M + H]+ = 294; 1H
stated otherwise, as determined by 1H NMR and HPLC analyses. The NMR (400 MHz, DMSO-d6) δ 9.46 (br s, 1H), 8.78 (br s, 1H), 8.56 (d, J
purity of final compounds was checked using a Waters ZQ2000 coupled = 6.3 Hz, 1H), 8.50−8.47 (m, 1H), 6.65 (s, 1H), 4.51−4.43 (m, 1H),
with LC Waters 2795 and Waters 2996 PDA detector. All mass spectra 4.36−4.26 (m, 1H), 3.95 (s, 3H), 3.62−3.52 (m, 1H), 3.25−3.12 (m,
were performed by electrospray ionization (ESI). Representative 3H), 2.57−2.49 (m, 2H), 2.39−2.27 (m, 1H), 2.18−2.08 (m, 1H),
procedures and physical properties and characterization for main 1.19−1.13 (m, 3H), 1.05 (t, J = 7.25, 3H).
compounds are described. Characterization data for the rest of the N-((3R,5S)-1-(Benzofuran-3-carbonyl)-5-(ethylcarbamoyl)-
compounds are detailed in the Supporting Information. pyrrolidin-3-yl)-3-ethyl-1-methyl-1H-pyrazole-5-carboxamide
Synthesis of Compounds. General Method A. General (7). Compound 7 was synthesized from Boc-deprotection of (2S,4R)-
Procedure for HATU-Mediated Coupling. To the desired carboxylic tert-butyl 4-(3-ethyl-1-methyl-1H-pyrazole-5-carboxamido)-2-
acid (1.0 equiv) and HATU (1.2 equiv) in N,N-dimethylformamide was (ethylcarbamoyl)pyrrolidine-1-carboxylate (4) using the protocol
added Hunig’s base (3.0 equiv). To the reaction mixture was then added described in general method C followed by coupling with benzofuran-
the desired amine (1.0 equiv). The reaction mixture was allowed to stir 3-carboxylic acid using the procedure described in general method A.
at room temperature for 3−12 h. The reaction mixture was then diluted The reaction mixture was concentrated in vacuo, and the residue was
with EtOAc (100 mL), washed with 10% NH4Cl (2 × 100 mL), purified by preparative HPLC. The reaction afforded the title compound
saturated NaHCO3 (1 × 100 mL), saturated NaCl (1 × 100 mL), dried (40 mg, 29% yield). MS (ESI) m/z [M + H]+ = 437.9; 1H NMR (400
(MgSO4), and evaporated to dryness. The crude product was assessed MHz, DMSO-d6) δ 8.49 (s br, 2H), 7.98 (m, 2H), 7.64 (d, J = 7.6 Hz,
for purity and the desired mass by LCMS analysis. 1H), 7.34 (m, 2H), 6.61 (s, 1H), 4.59 (m, 2H), 4.11 (m, 1H), 3.99 (s,
General Method B. General Procedure for the Deprotection 3H), 3.67 (m, 3H), 3.08 (m, 2H), 2.24 (m, 1H), 2.08 (m, 1H), 1.12 (t, J
of Methyl Esters. To the desired methyl ester starting material (1.0 = 7.2, 3H), 1.01 (t, J = 6.8, 3H); 13C NMR (100 MHz, DMSO-d6, 25 °C)
equiv) in tetrahydrofuran (THF)/methanol (5:1) was added a solution δ ppm 13.7, 14.7, 20.6, 33.4, 34.6, 38.4, 48.6, 53.1, 59.3, 105.3, 111.4,
of lithium hydroxide (3.0 equiv). The reaction mixture was stirred 116.2, 122.3, 123.6, 125.2, 126.0, 135.3, 151.3, 153.9, 159.5, 161.8, 170.8.
overnight at room temperature. The reaction was analyzed by LCMS. [α]23.2D −34.71 (c 0.68, EtOH).
The reaction mixture was diluted with EtOAc, washed with 10% N-((3R,5S)-1-(Benzofuran-3-carbonyl)-5-carbamoylpyrroli-
KH2SO4, saturated NaCl, dried over MgSO4, and evaporated to dryness. din-3-yl)-1,3-diethyl-1H-pyrazole-5-carboxamide (65). The title
General Method C. General Procedure for Boc-Deprotection compound was synthesized from 64 using the protocol described in
of Carbamates. To the desired Boc-protected starting material (1.0 general method A. After preparative HPLC purification (XBridge-30, 35
equiv) in dichloromethane was added TFA (20% by volume).The mL/min, 10−100% acetonitrile in water, 10 nM NH4HCO3, basic, 20
reaction mixture was then stirred at room temperature for 2 h. min) the reaction afforded the title compound (423 mg, 77% yield). MS
Deprotection process was monitored by LCMS. The solvents were (ESI) m/z [M + H]+ = 424; 1H NMR (400 MHz, DMSO-d6, 80 °C) δ
evaporated in vacuo, and the product was azeotroped with 2 × CH2Cl2. 8.34 (br s, 1H), 8.28 (d, J = 6.6 Hz, 1H), 8.00 (d, J = 7.6 Hz, 1H), 7.62 (d,
General Method D. Removal of Fmoc-Protection Group or J = 8.3 Hz, 1H), 7.41−7.31 (m, 2H), 7.13−6.82 (br s, 2H), 6.60 (s, 1H),
Fmoc Deprotection. A solution of piperidine/dimethylformamide 4.73 (dd, J = 8.3, 4.9 Hz, 1H), 4−62−4.57 (m, 1H), 4.40−4.35 (m, 2H),
(1:8 v/v, 0.6 M) was added to the resin and shaken at room temperature 4.11−4.07 (m, 1H), 3.71 (dd, J = 10.8, 5.6 Hz, 1H), 2.58−2.53 (m, 2H),
for 1 h. The reaction was checked by LCMS and showed that the 2.34−2.22 (m, 2H), 1.27 (t, J = 7.1 Hz, 3H), 1.19 (t, J = 7.6 Hz, 3H); 13C
deprotection had been successful. The remaining piperidine and NMR (100 MHz, DMSO-d6, 25 °C) δ ppm 13.7, 15.8, 20.7, 34.5, 45.4,
dimethylformamide were removed by rotary evaporator heating at 80 48.6, 53.1, 59.1, 105.4, 111.4, 116.2, 122.2, 123.6, 125.2, 126.0, 134.7,
°C. This crude was dissolved in MeOH, and a white precipitated 151.4, 153.9, 159.5, 161.8, 173.2. [α]23.2D −46.53 (c 0.68, EtOH).
appeared. The solid was filtered off. The crude was added to a silica gel Materials and Methods. The human biological samples were
column and was eluted with CH2Cl2/MeOH 8%. sourced ethically, and their research use was in accord with the terms of
(2S,4R)-1-tert-Butyl 2-Methyl 4-(3-ethyl-1-methyl-1H-pyra- the informed consents. All animal studies were ethically reviewed and
zole-5-carboxamido)pyrrolidine-1,2-dicarboxylate (3). Com- carried out in accordance with European Directive 2010/63/EU and the
pound 3 was synthesized from the (2S,4R)-1-tert-butyl 2-methyl-4- GSK Policy on the Care, Welfare and Treatment of Animals.
aminopyrrolidine-1,2-dicarboxylate (1) and 3-ethyl-1-methyl-1H-pyr- Affinity Selection. Selections were done using streptavidin matrix
azole-5-carboxylic acid (2) using the same procedure as described in tips (PhyNexus) and biotinylated InhA protein (full length (270aa)).
general method A. The reaction produced a brown oil in 96% yield (1.57 Three rounds of selections were performed. Each tip in each round of
g) with a purity of 95%. No further purification was needed. MS (ESI) selection had 10 μg of protein loaded/attached to the matrix. The InhA
m/z [M + H]+ = 380.8. protein was chemically biotinylated using NHS-chromogenic biotin.
(2S,4R)-tert-Butyl 4-(3-Ethyl-1-methyl-1H-pyrazole-5-carbox- Biotinylated protein ran as a dimer (76 kDa) and retained activity.
amido)-2-(ethylcarbamoyl)pyrrolidine-1-carboxylate (4). Com- Before use, tips were prewashed in selection buffer (50 mM Tris (pH
pound 4 was synthesized from saponification of (2S,4R)-1-tert-butyl 2- 7.5), 150 mM NaCl, 0.1% Tween 20) and 1.0 mg/mL sheared salmon
methyl 4-(3-ethyl-1-methyl-1H-pyrazole-5-carboxamido)pyrrolidine- sperm DNA (sssDNA, Ambion) and 10 mM β-mercaptoethanol
1,2-dicarboxylate (3) using the same procedure as described in general (BME).
method B followed by coupling with ethylamine using the same For selection round 1, 10 μg of biotinylated InhA protein was
procedure as described in general method A. The reaction produced a immobilized on a previously prepared streptavidin matrix tip and the tip
brown oil in 92% yield (1.38 g) with a purity of 95%. The crude product was washed four times with selection buffer to remove excess protein.
was carried on to next step as is. MS (ESI) m/z [M + H]+ = 393.8; 1H The selection conditions included buffer with and without NADH.
NMR (400 MHz, DMSO-d6) δ 8.42 (br s, 1H), 7.91 (br s, 1H), 6.66 (s, When the experimental conditions included NADH, it was added to all
1H), 4.41 (m, 1H), 4.11 (m, 1H), 3.93 (s, 3H), 3.63 (m, 2H), 3.31 (m, relevant buffers, with the exception of elution buffer, to a final
1H), 3.26 (m, 2H), 3.09 (m, 2H), 2.14 (m, 1H), 2.01 (m, 1H), 1.32 (s, concentration of 133μM. Then 5 nmol of pyrrolidine library was diluted
9H), 1.14 (t, J = 7.2, 3H), 1.01 (t, J = 6.8, 3H). in 60 mL of selection buffer. The appropriate library samples were

1285 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288


Journal of Medicinal Chemistry Article

passed over the streptavidin matrix tip for 1 h at room temperature. The they were incubated at 37 °C without shaking for 6 days. A resazurin
tip was washed 8 times with selection buffer and 2 times with DNA free solution was prepared by dissolving one tablet of resazurin (resazurin
selection buffer. Binders were eluted by passing heated (80 °C) DNA tablets for milk testing; ref 330884Y′ VWR International Ltd.) in 30 mL
free selection buffer over the tip for 12 min. The eluted material was of sterile PBS (phosphate buffered saline). Of this solution, an amount
passed over a fresh streptavidin matrix tip for 10 min at room of 25 μL was added to each well. Fluorescence was measured
temperature to remove any denatured protein (postclear). The postclear (Spectramax M5 Molecular Devices, excitation 530 nm, emission 590
step was repeated a second time to the round 1 selection output (1 μL of nm) after 48 h to determine the MIC value.
the round 1 selection output was retained to be used for qPCR in order Pharmacokinetic Studies. For pharmacokinetic studies C57BL/6
to monitor the output from this round of selection). The remaining female mice of 18−20 g weight were used (n = 3 mice). Experimental
round 1 selection output was brought to 60 μL by adding the necessary compounds were administered by intravenous (iv) bolus at 4 mg/kg at a
volume of sssDNA and selection buffer. volume of 10 mL/kg to n = 3 mice and by oral gavage (po) at 50 mg/kg
For selection round 2, the above selection procedure was repeated single dose at a volume of 20 mL/kg to n = 3 mice. All mice received
with fresh protein and a previously prepared fresh streptavidin matrix treatment in the fed state. Drugs were administered as solution for iv
tip. Then 5 μL of the round 2 selection output was retained to be used route in 20% encapsin and 5% DMSO and as suspension for po route in
for qPCR in order to monitor the output from this round of selection. 1% methylcellulose. Peripheral total blood was the compartment chosen
The remaining round 2 selection output was brought to 60 μL by adding for the establishment of compound concentrations. Aliquots of 25 μL of
the necessary volume of sssDNA and selection buffer. blood were taken from the lateral tail vein for each mouse at the
For selection round 3, the selection procedure was repeated with following time points: for iv route, 10, 20, and 30 min and 1, 2, 4, and 8 h;
fresh protein, a previously prepared fresh tip, and the round 2 selection for po route, 15, 30, and 45 min and 1, 2, 4, and 8 h. LCMS was used as
output. The postclear steps were not repeated at the end of round 3. the analytical method of choice for the establishment of compound
Quantitative PCR was run with the outputs from each round of selection concentration in blood with a sensitivity of LLQ = 1−5 ng/mL in 25 μL
to assess selection yields for each step. The round 3 output was of blood. The noncompartmental data analysis (NCA) was performed
sequenced using Roche/454 technology. A no-target control selection with WinNonlin Phoenix 6.3 (Pharsight, Certara L.P.), and supple-
was performed in the same fashion, using the same buffer but without mentary analysis was performed with GraphPad Prism 5 (GraphPad
the protein input. Software, Inc.).
X-ray Crystallography. InhA was expressed in E. coli and purified in In Vivo Efficacy Assessment. Specific pathogen-free, 8- to 10-
two steps by IEC and size exclusion chromatography. The protein (4.15 week-old female C57BL/6 mice were purchased from Harlan
mgs/mL in 20 mM Tris, pH 8.0, 0.1 M NaCl, 2 mM DTT) was Laboratories and were allowed to acclimate for 1 week. The
incubated with NADH and compound prior to crystallization. The experimental design has been previously described.49 In brief, mice
complex crystallized in 10% PEG 8000 and 0.1 M Tris, pH 8.5, at 20 °C were intratracheally infected with 100 000 cfu/mouse (M. tuberculosis
and was cryoprotected prior to data collection at ESRF. The structure H37Rv strain). Products were administered for 8 consecutive days
was solved by molecular replacement. More details, statistics, and starting 1 day after infection. Lungs were harvested 24 h after the last
exemplars of the density are given in the Supporting Information. administration. All lung lobes were aseptically removed, homogenized,
InhA Enzymatic Assay. Enzymatic activity was measured fluori- and frozen. Homogenates were plated in 10% OADC-7H11 medium for
metrically by following NADH oxidation at λexc = 340 nm and λem= 480 14 days at 37 °C.
nm, using 50 mM NADH and 50 mM 2-trans-dodecenoyl-CoA
(DDCoA) as substrates. Dose−response experiments to determine IC50
were performed using 5 nM InhA. The percentage of remaining

*
ASSOCIATED CONTENT
S Supporting Information
enzymatic activity (% AR) at different compound concentrations was
Additional tables, schemes, figures, protocols and experimental
calculated with the formula % AR = 100 × [(sample − control 2)/
(control 1 − control 2)] where sample is the enzymatic activity for each procedures of some assays mentioned in the main text, synthetic
compound concentration, control 1 is enzyme activity in the absence of procedures, and compound characterization. This material is
available free of charge via the Internet at http://pubs.acs.org.


any compound, and control 2 is NADH oxidation in absence of enzyme.
IC50 was calculated fitting % AR to a two parameter equation % AR =
100/[1 + (compound conc/IC50)s] where s is a slope factor. IC50 was AUTHOR INFORMATION
calculated using GraFit 5.0.12 software (Eritacus Software Ltd.). All Corresponding Authors
reactions were run in 30 mM PIPES buffer, pH 6.8, at 25 °C. *J.C.-P.: phone, +34-669-399879; fax, 34-918-070310; e-mail,
Strain and Growth Conditions. M. tuberculosis H37Rv
(ATC25618) wild type and H37Rv overexpressing InhA (HyrR) were
julia.p.castro@gsk.com.
grown in Middlebrook 7H9-ADC broth (Difco) supplemented with *G.E.: phone, 1-781-795-4423; fax, 1-781-795-4496; e-mail,
0.05% Tween 80 and on 7H10-OADC or 7H11-OADC agar (Difco) at ghotas.x.evindar@gsk.com.
37 °C. INH and Hyg were purchased from Sigma-Aldrich. Where Notes
required hygromycin (50 μg mL−1) was added to the culture medium. The authors declare no competing financial interest.


Mycobacterium tuberculosis Inhibition Assay. The measure-
ment of the minimum inhibitory concentration (MIC) for each tested ACKNOWLEDGMENTS
compound was performed in 96-well flat-bottom polystyrene microtiter
plates. Ten 2-fold drug dilutions in neat DMSO were performed. An The authors thank the Physicochemical Characterisation Group
amount of 5 μL of these drug solutions was added to 95 μL of of Analytical Chemistry, GSK Tres Cantos, Mariá Teresa Fraile-
Middlebrook 7H9 medium (lines A−H, rows 1−10 of the plate layout). Gabaldón, and Rubén Gómez for carrying out chromlog D
Isoniazid was used as a positive control. Eight 2-fold dilutions of measurements for all the compounds and formulation studies,
isoniazid starting at 160 μg/mL were prepared, and 5 μL of this control Angel Santos for protein binding measurements, and Jesús
curve was added to 95 μL of Middlebrook 7H9 medium (Difco Gómez, Ana Á lvarez, and Raquel Gabarró for their help with
catalogue ref 271310) (row 11, lines A−H). An amount of 5 μL of neat LCMS and NMR analysis. Santiago Ferrer is thanked for his
DMSO was added to row 12 (growth and blank controls). The
assistance with the pharmacology work. We acknowledge the
inoculum was standardized to approximately 1 × 107 cfu/mL mL−1 and ́
diluted 1 in 100 in Middlebrook 7H9 broth (10% ADC (Becton excellent technical expertise of Delia Blanco, Marı ́a Martinez,
Dickinson catalog ref 211887) and 0.025% Tween 80) to produce the
́ Pedro A. Torres Gómez, and Mariá José
Rubén González del Rio,
final inoculum. Then 100 μL of this inoculum was added to the entire Rebollo for biological evaluation. We gratefully acknowledge the
plate except G-12 and H-12 wells (blank controls). All plates were Laboratory Animal Science (Pre-Clinical) Department for
placed in a sealed box to prevent drying out of the peripheral wells, and essential animal lab upkeep and maintenance. The research
1286 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288
Journal of Medicinal Chemistry Article

leading to these results has received funding from the Global (12) Vilcheze, C.; Morbidoni, H. R.; Weisbrod, T. R.; Iwamoto, H.;
Alliance for TB Drug Development and from the European Kuo, M.; Sacchettini, J. C.; Jacobs, W. R., Jr. Inactivation of the inhA-
Union’s 7th Framework Program (FP7-2007-2013) under the encoded fatty acid synthase II (FASII) enoyl-acyl carrier protein
Orchid Grant Agreement No. 261378. The funders had no role reductase induces accumulation of the FASI end products and cell lysis
of Mycobacterium smegmatis. J. Bacteriol. 2000, 182, 4059−4067.
in study design, data collection, analysis, decision to publish, or
(13) Banerjee, A.; Dubnau, E.; Quemard, A.; Balasubramanian, V.; Um,
preparation of the manuscript.


K. S.; Wilson, T.; Collins, D.; de Lisle, G.; Jacobs, W. R., Jr. InhA, a gene
encoding a target for isoniazid and ethionamide in Mycobacterium
ABBREVIATIONS USED tuberculosis. Science 1994, 263, 227−230.
ADMET, absorption, distribution, metabolism, excretion, and (14) Dessen, A.; Quemard, A.; Blanchard, J. S.; Jacobs, W. R., Jr.;
toxicity; AUC, area under the curve; BOC, tert-butyloxycarbonyl; Sacchettini, J. C. Crystal structure and function of the isoniazid target of
CLND, chemiluminescent nitrogen detection; cfu, colony Mycobacterium tuberculosis. Science 1995, 267, 1638−1641.
(15) Parikh, S. L.; Xiao, G.; Tonge, P. J. Inhibition of InhA, the enoyl
forming unit; DEL, DNA-encoded chemical libraries; DIPEA,
reductase from Mycobacterium tuberculosis, by triclosan and isoniazid.
N,N-diisopropylethylamine; DMF, N,N-dimethylformamide; Biochemistry 2000, 39, 7645−7650.
DMPK, drug metabolism and pharmacokinetics; DMSO, (16) Scior, T.; Garces-Eisele, S. J. Isoniazid is not a lead compound for
dimethyl sulfoxide; ELT, encoded library technology; HATU, its pyridyl ring derivatives, isonicotinoyl amides, hydrazides, and
N-[(dimethylamino)-1H-1,2,3-triazolo[4,5-b]pyridin-1-yl- hydrazones: a critical review. Curr. Med. Chem. 2006, 13, 2205−2219.
methylene]-N-methylmethanaminium hexafluorophosphate N- (17) Argyrou, A.; Vetting, M. W.; Aladegbami, B.; Blanchard, J. S.
oxide; INH, isoniazid; InhA, Mycobacterium tuberculosis InhA; Mycobacterium tuberculosis dihydrofolate reductase is a target for
LE, ligand efficiency; LLE, ligand lipophilicity efficiency; LLEAT, isoniazid. Nat. Struct. Mol. Biol. 2006, 13, 408−413.
ligand lipophilicity efficiency Astex; MDR-TB, multidrug- (18) Schroeder, E. K.; de Souza, O. N.; Santos, D. S.; Blanchard, J. S.;
resistant tuberculosis; MIC, minimum inhibitory concentration; Basso, L. A. Drugs that inhibit mycolic acid biosynthesis in
NMR, nuclear magnetic resonance; PFI, property forecast index, Mycobacterium tuberculosis. Curr. Pharm. Biotechnol. 2002, 3, 197−225.
(19) Basso, L. A.; Santos, D. S. Drugs that inhibit mycolic acid
therapeutics; PK, pharmacokinetics; SAR, structure−activity
biosynthesis in Mycobacterium tuberculosisan update. Med. Chem. Rev.
relationship; TB, tuberculosis; TFA, trifluoroacetic acid; WHO, 2005, 2, 393−413.
World Health Organization; XDR, extensively drug-resistant (20) Hazbón, M. H.; Brimacombe, M.; Bobadilla Del Valle, M.;
tuberculosis


Cavatore, M.; Guerrero, M. I.; Varma-Basil, M.; Billman-Jacobe, H.;
Lavender, C.; Fyfe, J.; Garcia-Garcia, L.; Leon, C. I.; Bose, M.; Chaves,
REFERENCES F.; Murray, M.; Eisenach, K. D.; Sifuentes-Osornio, J.; Cave, M. D.;
(1) World Health Organisation. Tuberculosis. http://www.who.int/ Ponce de Leon, A.; Alland, D. Population genetics study of isoniazid
mediacentre/factsheets/fs104/en/index.html (accessed February 11, resistance mutations and evolution of multidrug-resistant Mycobacte-
2013). rium tuberculosis. Antimicrob. Agents Chemother. 2006, 50, 2640−2649.
(2) Becerra, M. C.; Bayona, J.; Freeman, J.; Farmer, P. E.; Kim, J. Y. (21) Kuo, M. R.; Morbidoni, H. R.; Alland, D.; Sneddon, S. F.; Gourlie,
Redefining MDR-TB transmissionhot spots’ counterpoint. Int. J. Tuberc. B. B.; Staveski, M. M.; Leonard, M.; Gregory, J. S.; Janjigian, A. D.; Yee,
Lung Dis. 2000, 4, 387−394. C.; Musser, J. M.; Kreiswirth, B.; Iwamoto, H.; Perozzo, R.; Jacobs, W.
(3) Dye, C.; Espinal, M. A.; Watt, C. J.; Mbiaga, C.; Williams, B. G. R.; Sacchettini, J. C.; Fidock, D. A. Targeting tuberculosis and malaria
Worldwide incidence of multidrug-resistant tuberculosis. J. Infect. Dis. through inhibition of enoyl reductase compound activity and structural
2002, 185, 1197−1202. data. J. Biol. Chem. 2003, 278, 20851−20859.
(4) Espinal, M. A. The global situation of MDR-TB. Tuberculosis (22) Lu, X. Y.; You, Q. D.; Chen, Y. D. Recent progress in the
(Edinburgh) 2003, 83, 44−51. identification and development of InhA direct inhibitors of
(5) Wright, A.; Zignol, M.; Van Deun, A.; Falzon, D.; Gerdes, S. R.; Mycobacterium tuberculosis. Mini-Rev. Med. Chem. 2010, 10, 182−193.
Feldman, K.; Hoffner, S.; Drobniewski, F.; Barrera, L.; van Soolingen, (23) Ballell Pages, L.; Castro Pichel, J.; Fernandez Menendez, R.;
D.; Boulabhal, F.; Paramasivan, C. N.; Kam, K. M.; Mitarai, S.; Nunn, P.; Fernandez Velando, E. P.; Gonzalez del Valle S.; Leon Diaz M. L.;
Raviglione, M. Epidemiology of antituberculosis drug resistance 2002- Mendoza Losana, A.; Wolfendale, M. J. (Pyrazol-3-yl)-1,3,4-thiadiazole-
07: an updated analysis of the global project on anti-tuberculosis drug 2-amine and (Pyrazol-3-yl)-1,3,4-thiazol-2-amine Compounds. WO/
resistance surveillance. Lancet 2009, 373, 1861−1873. 2010/118852, 2010.
(6) Chiang, C. Y.; Centis, R.; Migliori, G. B. Drug-resistant (24) Pan, P.; Tonge, P. J. Targeting InhA, the FASII enoyl-ACP
tuberculosis: past, present, future. Respirology 2010, 15, 413−432. reductase: SAR studies on novel inhibitor scaffolds. Curr. Top. Med.
(7) Multidrug and Extensively Drug-Resistant TB (M/XDR-TB): Chem. 2012, 12, 672−693.
2010 Global Report on Surveillance and Response (WHO/HTM/TB/ (25) Castro Pichel, J.; Fernandez Menendez, R.; Fernandez Velando,
2010.3). World Health Organization: Geneva, 2010. E. P.; Gonzalez del Valle S.; Mallo-Rubio, A. 3-Amino-pyrazole
(8) Shah, N. S.; Wright, A.; Bai, G. H.; Barrera, L.; Boulahbal, F.; Derivatives Useful against Tuberculosis. WO/2012/049161, 2012.
Martin-Casabona, N.; Drobniewski, F.; Gilpin, C.; Havelkova, M.; Lepe, (26) Horlacher, O. P.; Hartkoorn, R. C.; Cole, S. T.; Altmann, K.-H.
R.; Lumb, R.; Metchock, B.; Portaels, F.; Rodrigues, M. F.; Rüsch- Synthesis and antimycobacterial activity of 2,1′-dihydropyridomycins.
Gerdes, S.; Van Deun, A.; Vincent, V.; Laserson, K.; Wells, C.; Cegielski, ACS Med. Chem. Lett. 2013, 4, 264−268.
J. P. Worldwide emergence of extensively drug-resistant tuberculosis. (27) Clark, M. A.; Acharya, R. A.; Arico-Muendel, C. C.; Belyanskaya,
Emerging Infect. Dis. 2007, 13, 380−387. S. L.; Benjamin, D. R.; Carlson, N. R.; Centrella, P. A.; Chiu, C. H.;
(9) Berry, M.; Kon, O. M. Multidrug- and extensively drug-resistant Creaser, S. P.; Cuozzo, J. W.; Davie, C. P.; Ding, Y.; Franklin, G. J.;
tuberculosis: an emerging threat. Eur. Respir. Rev. 2009, 18, 195−197. Franzen, K. D.; Gefter, M. L.; Hale, S. P.; Hansen, N. J. V.; Israel, D. I.;
(10) What Is DOTS? A Guide to Understanding the WHO Jiang, J.; Kavarana, M. J.; Kelley, M. S.; Kollmann, C. S.; Li, F.; Lind, K.;
Recommended TB Control Strategy Known as DOTS. Document Mataruse, S.; Medeiros, P. F.; Messer, J. A.; Myers, P.; O’Keefe, H.; Oliff,
WHO/CDS/CPC/TB/99.270; World Health Organization: Geneva, M. C.; Rise, C. E.; Satz, A. L.; Skinner, S. R.; Svendsen, J. L.; Tang, L.;
1999. Vloten, K. V.; Wagner, R. W.; Yao, G.; Zhao, B.; Morgan, B. A. Design,
(11) Quemard, A.; Sacchettini, J. C.; Dessen, A.; Vilchèze, C.; Bittman, synthesis and selection of DNA-encoded small-molecule libraries. Nat.
R.; Jacobs, W. R.; Blanchard, J. S. Enzymic characterization of the target Chem. Biol. 2009, 5, 647−654.
for isoniazid in Mycobacterium tuberculosis. Biochemistry 1995, 34, 8235− (28) Deng, H.; O’Keefe, H.; Davie, C. P.; Lind, K. E.; Acharya, R. A.;
8241. Franklin, G. J.; Larkin, J.; Matico, R.; Neeb, M.; Thompson, M. M.; Lohr,

1287 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288


Journal of Medicinal Chemistry Article

T.; Gross, J. W.; Centrella, P. A.; O’Donovan, G. K.; Bedard, K. L.; methyltransferases define the specificity of the mycolic acid elongation
Vloten, K.-v.; Mataruse, S.; Skinner, S. R.; Belyanskaya, S. L.; Carpenter, complexes. PLoS One 2011, 6, e29564.
T. Y.; Shearer, T. W.; Clark, M. A.; Cuozzo, J. W.; Arico-Muendel, C. C.; (43) Koitka, M.; Hochel, J.; Gieschen, H.; Borchert, H. H. Improving
Morgan, B. A. Discovery of highly potent and selective small molecule the ex vivo stability of drug ester compounds in rat and dog serum:
ADAMTS-5 inhibitors that inhibit human cartilage degradation via inhibition of the specific esterases and implications on their identity. J.
encoded library technology (ELT). J. Med. Chem. 2012, 55, 7061−7079. Pharm. Biomed. Anal. 2010, 51, 664−678.
(29) Disch, J. S.; Evindar, G.; Chiu, C. H.; Blum, C. A.; Dai, H.; Jin, L.; (44) Leeson, P. D.; Springthorpe, B. The influence of drug-like
Schuman, E.; Lind, K. E.; Belyanskaya, S. L.; Deng, J.; Coppo, F.; concepts on decision-making in medicinal chemistry. Nat. Rev. Drug
Aquilani, L.; Graybill, T. L.; Cuozzo, J. W.; Lavu, S.; Mao, C.; Vlasuk, G. Discovery 2007, 6, 881−890.
P.; Perni, R. B. Discovery of thieno[3,2-d]pyrimidine-6-carboxamides as (45) Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand efficiency: a useful
potent inhibitors of SIRT1, SIRT2, and SIRT3. J. Med. Chem. 2013, 56, metric for lead selection. Drug Discovery Today 2004, 9, 430−431.
3666−3679. (46) Kuntz, I. D.; Chen, K.; Sharp, K. A.; Kollman, P. A. The maximal
(30) Graybill, T. L.; King, B. W.; Fosbenner, D. T.; Keller, P. M.;; affinity of ligands. Proc. Natl. Acad. Sci. U.S.A. 1999, 96, 9997−10002.
Morgan, B. A.; Clark, M. A.; Cuozzo, J. DNA-Encoded Libraries: A New (47) Carr, R. A. E.; Congreve, M.; Murray, C. W.; Rees, D. C.
Resource for Innovative Hit Identification. Abstract of Papers, 237th Fragment-based lead discovery: leads by design. Drug Discovery Today
National Meeting of the American Chemical Society, Salt Lake City, UT, 2005, 10, 987−992.
U.S., March 22−26, 2009; American Chemical Society: Washington, (48) Mortenson, P. N.; Murray, C. W. Assessing the lipophilicity of
fragments and early hits. J. Comput.-Aided Mol. Des. 2011, 25, 663−667.
DC, 2009; MEDI-297.
(49) Rullas, J.; Garcia, J. I.; Beltran, M.; Cardona, P. J.; Cáceres, N.;
(31) Evindar, G.; Acharya, R. A.; Arico-Muendel, C. C.; Belyanskaya, S.
García-Bustos, J. F.; Angulo-Barturen, I. Fast standardized therapeutic-
L.; Brown, J.; Clark, A. M.; Centrella, P. A.; Cuozzo, J. W.; Davie, C. P.;
efficacy assay for drug discovery against tuberculosis. Antimicrob. Agents
Ding, Y.; Franklin, G. J.; Jeffrey, W. Gross, J. W.; Israel, D. I.; Jolivette, L.
Chemother. 2010, 54, 2262−2264.
J.; Jurewicz, A. J.; Lakdawala, A. S.; Li, F.; Li, H.; Lind, K. E.; Joseph, P.;
Marino, J. P.; Morgan, B. A. Application of Encoded Library Technology
(ELT) to the Discovery of Soluble Epoxide Hydrolase Inhibitors.
Abstracts of Papers, 238th National Meeting of the American Chemical
Society, Washington, DC, United States, August 16−20, 2009; MEDI-2.
(32) Davie, C. P.; Centrella, P. A.; O’Donovan, G. K.; Evindar, G.;
O’Keefe, H.; Patel, A. M.; Clark, M. A. Design and Synthesis of a Sixteen
Million Member DNA-Encoded Library (DEL) and Discovery of
Potent Mycobacterium tuberculosis (Mtb) InhA Inhibitors. Abstracts of
Papers, 240th National Meeting of the American Chemical Society,
Boston, MA, United States, August 22−26, 2010; American Chemical
Society: Washington, DC, 2010; MEDI-150.
(33) Ding, Y.; O’Keefe, H.; Svendsen, J. L.; Franklin, G. J.; Centrella, P.
A.; Clark, M. A.; Acharya, R. A.; Li, F.; Messer, J. A.; Chiu, C. H.; Matico,
R. E.; Murray-Thompson, M. F.; Cuozzo, J. W.; Israel, D. I.; Arico-
Muendel, C.; Morgan, B. A. Discovery of Potent and Selective Inhibitors
for ADAMTS-4 through Encoded Library Technology (ELT). Abstracts
of Papers, 240th National Meeting of the American Chemical Society,
Boston, MA, United States, August 22−26, 2010; American Chemical
Society: Washington, DC, 2010; MEDI-460.
(34) Gentile, G.; Merlo, G.; Pozzan, A.; Bernasconi, G.; Bax, B.;
Bamborough, P.; Bridges, A.; Carter, P.; Neu, M.; Yao, G.; Brough, C.;
Cutler, G.; Coffin, A.; Belyanskaya, S. 5-Aryl-4-carboxamide-1,3-
oxazoles: potent and selective GSK-3 inhibitors. Bioorg. Med. Chem.
Lett. 2012, 22, 1989−1994.
(35) For ELT data analysis and terminologies refer to refs 27−29.
(36) Mendoza-Losana, A. Gordon Research Conference: Tuber-
culosis, 2011 (personal communication).
(37) Lipinski, C. A. Drug-like properties and the causes of poor
solubility and poor permeability. J. Pharmacol. Toxicol. Methods 2000,
44, 235−249.
(38) Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J.
Experimental and computational approaches to estimate solubility and
permeability in drug discovery and development settings. Adv. Drug
Delivery Rev. 2001, 46, 3−26.
(39) Lennernas, H.; Abrahamsson, B. The use of biopharmaceutic
classification of drugs in drug discovery and development: current status
and future extension. J. Pharm. Pharmacol. 2005, 57, 273−285.
(40) Dahan, A.; Miller, J. M.; Amidon, G. L. Prediction of solubility and
permeability class membership: provisional BCS classification of the
world’s top oral drugs. AAPS J. 2009, 11, 740−746.
(41) Rozwarski, D. A.; Vilcheze, C.; Sugantino, M.; Bittman, R.;
Sacchettini, J. C. Crystal structure of the Mycobacterium tuberculosis
enoyl-ACP reductase, InhA, in complex with NAD+ and a C16 fatty acyl
substrate. J. Biol. Chem. 1999, 274, 15582−15589.
(42) Cantaloube, S.; Veyron-Churlet, R.; Haddache, N.; Daffé, M.;
Zerbib, D. The Mycobacterium tuberculosis FAS-II dehydratases and

1288 dx.doi.org/10.1021/jm401326j | J. Med. Chem. 2014, 57, 1276−1288

You might also like