Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Articles

pubs.acs.org/acschemicalbiology

Identification and Further Development of Potent TBK1 Inhibitors


André Richters,† Debjit Basu,† Julian Engel,† Meryem S. Ercanoglu,‡ Hyatt Balke-Want,‡ Roberta Tesch,†,§
Roman K. Thomas,‡ and Daniel Rauh*,†

Department of Chemistry and Chemical Biology, Technical University of Dortmund, Otto-Hahn-Straße 6, 44227 Dortmund,
Germany

Department of Translational Genomics, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
§
Programa de Pós-Graduaçaõ em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio
de Janeiro, Rio de Janeiro, 21941-901 Brazil
*
S Supporting Information
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: The cytosolic Ser/Thr kinase TBK1 was


discovered to be an essential element in the mediation of
signals that lead to tumor migration and progression. These
Downloaded via 112.211.6.52 on January 5, 2022 at 10:36:58 (UTC).

findings meet the need for the identification of novel tool


compounds and potential therapeutics to gain deeper insights
into TBK1 related signaling and its relevance in tumor
progression. Herein, we undertake the activity-based screening
for unique inhibitors of TBK1 and their subsequent
optimization. Initial screening approaches identified a selection
of TBK1 inhibitors that were optimized using methods of
medicinal chemistry. Variations of the structural characteristics
of a representative 2,4,6-substituted pyrimidine scaffold resulted in improved potency. Prospective use as tool compounds or
basic contributions to drug design approaches are anticipated for our improved small molecules.

T ANK-binding kinase 1 (TBK1) is a critical regulator of the


antiviral response and initiates type I interferon (IFN)
transcription through an endosomal toll-like receptor 3 (TLR3)
chemokine 5 (CCL5) and intereukin 6 (IL-6) further
substantiate a potential role in cancer therapy.8 However,
TBK1 was also demonstrated to participate in tumor metastasis
pathway. Subsequently, the interferon regulatory factors 3 and and epithelial−mesenchymal transition (EMT), and therefore,
7 (IRF3/7) are activated and then translocated to the nucleus modulating TBK1 activity may curtail tumor growth and
to initiate IFN gene transcription.1−4 Alternatively, cytosolic propagation. Prevailing findings support this assumption, since
activation occurs by the recognition of infiltrated viral RNA small molecule inhibitors have been discovered that block
through sensor proteins such as melanoma differentiation- TBK1 transferase activity and thereby hinder the ability of
associated protein 5 (MDA5) or the retinoic acid-inducible tumor cells to further metastasize, proliferate, or undergo EMT
gene 1 protein (RIG-I). Consecutive TBK1 activation is processes.9 Recently, TBK1 has been shown to activate
induced by adapter protein mediated phosphorylation of TBK1, estrogen receptor α (ERα) by phosphorylation, thereby
thereby initiating the same downstream cascades of IFN stabilizing ERα and inducing transactivational activity and
production as in the endosomal pathway. TBK1 is involved in modulating breast cancer growth. Ectopic expression of TBK1
the formation of a higher ordered multi enzyme complex and in this context renders breast cancer cells resistant to tamoxifen
interacts with several adapter proteins such as nuclear factor κB while suppression of TBK1 activity by treatment with BX795
(NFκB)-activating kinase-associated protein 1 (NAP1), TBK1- (Figure 1),10,11 a known 3-phosphoinositide-dependent protein
binding protein 1 (SINTBAD), and TRAF family member- kinase 1 (PDK1) and TBK1 inhibitor, in combination with
associated NFκB activator (TANK) in an orchestrated tamoxifen leads to synergistic antiproliferative effects in breast
trafficking network.1,5 Besides its implication in the mediation cancer cells. Therefore, TBK1 may serve as a predictive marker
of antiviral responses, TBK1 has been proposed as a target for for tamoxifen resistance and as a potential therapeutic target in
cancer therapy since TBK1 knockout studies led to breast cancer.12 Additionally, TBK1 is involved in mouse
antiproliferative effects in KRAS-mutated human alveolar double minute 2 (MDM2) mediated repression of estrogen
adenocarcinoma cells (A549).6 Based on this analysis, TBK1
has become a focus of medicinal chemistry approaches to
Special Issue: New Frontiers in Kinases
decipher the complex regulation and activation mechanism and
its associated signaling pathway.1,7 Constitutive explorations Received: November 7, 2014
investigating the role of TBK1 in promoting KRAS-driven Accepted: December 26, 2014
tumorigenicity and survival by activating autocrine C−C motif Published: December 26, 2014

© 2014 American Chemical Society 289 DOI: 10.1021/cb500908d


ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

potential target in cancer therapy.8,14 Since TBK1 may serve as


a potential therapeutic target, numerous studies have been
conducted to identify and develop new TBK1 inhibitors such as
(2- and 6-aryl-)azabenzimidazoles, 6-aminopyrazolopyrimi-
dines, and pyrimidines, respectively (Figure 1).15−18
The recently reported crystal structure of the full-length
TBK1 kinase in complex with the type I inhibitor BX795 has
provided the structural basis for medicinal chemistry
approaches.19 TBK1 consists of three superordinate domains:
a catalytic domain facilitating Ser/Thr kinase activity, a
ubiquitin-like domain (ULD) for substrate specificity, and a
scaffolding and dimerization domain (SDD) for structural
Figure 1. Representative scaffolds (6-aminopyrazolopyrimidine (1), 2-
integrity and mediating adapter protein interactions to form a
aryl-azabenzimidazole (2), and pyrimidines (BX795, CYT387)) of
currently known TBK1 inhibitors revealing broad target specificity. multi-enzyme complex.19 Moreover, the presence of SDD and
ULD provided evidence for TBK1 existing as a homo-
dimer.1,20,21 Although TBK1 was proposed as a target in
induced protein kinase B (Akt) activation by facilitating various cancer types and particularly in facilitating EMT
phosphorylation induced degradation of hematopoietic PBX- processes, its associated tumor biology to large extent remains
interaction protein (HPIP).13 Broader studies indicate potential unclear.9,22 Therefore, tool compounds for deciphering TBK1
therapeutic use of TBK1 inhibitors, since they attenuate biology as well as specific inhibitors are needed to more deeply
radiation-induced EMT of human lung cancer cells (A549) investigate its function with respect to the orchestrated network
via activation of glycogen synthase kinase-3 β (GSK3β) and of cellular signaling of the antiviral response, as well as its role
simultaneous repression of zinc finger E-box-binding homeobox in cancer progression and metastasis.
1 (ZEB1).9 Similarly, studies on the impact of TBK1 as a Herein, we report the identification of a new class of
therapeutic target in HER2 positive breast cancer and KRAS- pyrimidine-based inhibitors of TBK1 by activity-based screen-
driven tumorigenicity further emphasized that TBK1 is a ing approaches. Subsequent ligand optimization by using

Figure 2. (A) Activity-based screening results as cluster analysis according to inhibitory effect and structural similarity. Colors encode for the
remaining TBK1 activity elicited at 13.2 μM compound concentrations. Ring sizes represent the discontinuity of binding: Marginal differences in the
compound’s structural composition result in dramatic differences of the inhibitory effect. The clustering was performed using SARANEA.36 (B)
Simplified screening workflow for Collections I and II as well as illustration of redundant scaffolds revealing divergent substitution patterns for
subsequent SAR studies. (C) Validated hit compounds derived from Collection I (% remaining activity in gray).

290 DOI: 10.1021/cb500908d


ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

Table 1. Collection of Tozasertib Derivatives and Screening Hits and Related IC50 Valuesa

compd R1 R2 R3 X IC50 [μM]


13 H H NO2 O 2.35 ± 0.28
14 H NO2 H O 1.41 ± 0.46
15a NO2 H H O 0.06 ± 0.03
15b t
Bu H H O 2.66 ± 0.11
15c i
Pr H H O 0.34 ± 0.07
15d Me H H O 0.38 ± 0.10
15e OMe H H O 0.65 ± 0.21
15f F H H O 0.84 ± 0.36
15g Cl H H O 1.19 ± 0.12
15h CF3 H H O 0.29 ± 0.11
15i Cl F H O 0.72 ± 0.05
15j Cl CF3 H O 3.00 ± 1.22
15k NH2 H H O 0.73 ± 0.30
22 H H NHCOCH2CH3 O nib
23 H NHCOCH2CH3 H O >10
24 NHCOCH2CH3 H H O 2.50 ± 0.39
tozasertib NHCO(cyclopropane) H H S 6.22 ± 2.86
axitinib 0.47 ± 0.04
bosutinib 0.67 ± 0.03
dovitinib 0.06 ± 0.01
erlotinib 2.20 ± 0.48
K252a <0.001
sunitinib 0.22 ± 0.01
tofacitinib 5.76 ± 0.36
a
IC50 values were determined in three independent duplicate experiments. bni = no inhibitory effect at 50 μM compound concentration

methods of medicinal chemistry and drug design led to Activity-Based Screening for TBK1 Inhibitors and Hit
promising TBK1 inhibitors with activities in the nanomolar Validation. We initially screened two compound libraries,
range. In addition, we screened focused compound collections comprised of 839 (Collection I) and 968 (Collection II)
to identify and validate further TBK1 inhibitors that were molecules, for inhibitors of TBK1 using an activity-based assay
provided by GSK and ROCHE in the course of promoting the that quantified TBK1-dependent substrate phosphorylation in
idea of public private partnerships among academic and presence and absence of test compound (see Methods). With
industrial research facilities.23 respect to Collection I, each small molecule that reduced TBK1


activity to 40% at 13.2 μM test concentration was considered as
RESULTS AND DISCUSSION a hit and subjected to further validation studies (Figure 2A,B).
A Z′-factor26 of 0.79, based on the consideration of at least
Current studies in TBK1 biology have provided evidence for eight positive (BX795, 13.2 μM) and negative (DMSO)
this enzyme’s involvement in the progression and metastasis of controls, was calculated for the entire screen. In Collection I,
breast cancer cells as well as an implication in various lung and we identified eight compounds of commercial origin as
colon cancers.9,12,24 Since TBK1 may represent a future drug promising TBK1 inhibitors (Figure 2C). Validation studies
target in cancer therapy, the identification and development of confirmed their inhibitory impact and the staurosporine
novel inhibitors to modulate TBK1 kinase activity is an analogue K252a, exhibiting an IC50 value <0.001 μM, was
important area of research. Currently known inhibitors such as determined to be the most potent compound followed by
BX79510,11 or CYT38725 (Figure 1) were potent TBK1 dovitinib (0.06 μM) and sunitinib (0.2 μM, Table 1).
inhibitors but lacked specificity, which rendered them Collection II originated 52 hits, which significantly reduced
unfeasible for application as molecular probes to decipher TBK1 activity to less than 30% (Z′-factor: 0.68). This
TBK1 biology. We therefore sought to screen a wide array of compilation contained oxindoles (17), pyrimidines (12),
compounds, seeking to identify promising enzyme binders by pyridopyrimidinones (5), commercial inhibitors (4), benzo-
utilizing activity-based assay systems. Our ultimate goal was to thiophenes (3), quinolines (2), flavones (2), and other
improve the inhibitory effects of the identified hits against (unique) scaffolds (7). Of these, seven selected scaffolds and
TBK1 for potential application to investigate TBK1 signaling derivatives thereof were subjected to further validation studies
and for the development of more potent inhibitors or starting and confirmed as hits in activity-based assays with IC50 values
compounds for constitutive drug design approaches. of 0.14 to 15 μM (Supporting Information Figure S1). Of note,
291 DOI: 10.1021/cb500908d
ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

Figure 3. SARANEA-supported cluster analysis of activity-based screening results according to inhibitory effect and structural similarity of the hit
compounds of the GSK- (A) and ROCHE-library (B), respectively. Representative derivatives of selected scaffolds are illustrated and assigned to the
respective cluster centers (roman numerals). The color code illustrates remaining TBK1 activity elicited at 10 μM compound concentrations.

we removed flavone scaffolds from the panel as they are appropriate cores were redundant throughout the libraries and
frequent hitters in HTS campaigns and recently were in-depth therefore were represented by sundry derivatives, we
described in the literature as pan assay interference compounds immediately analyzed the respective hits with respect to the
(PAINS).27−29 However, we identified a pyrimidine scaffold corresponding structure−activity relationships (SARs). Con-
(Scaffold I, Figure 2B, IC50 = 8.6 μM), which illustrated a cerning the GSK library, we identified 14 compounds (Figure
similar substitution pattern as well as analog inhibitory potency 4) represented by substituted imidazopyridines (25−29) and
relative to tozasertib30 (IC50 = 6.2 μM). Comparative analysis oxindoles (30−34) as well as maleimide(-like) structures (35,
of the crystal structures of tozasertib in complex with Aurora A 36) and pyrimidines (37, 38). Of note, we generated IC50
(PDB-code: 3E5A) and TBK1 in complex with BX795 (PDB- values within the range 0.14−1.6 μM for the imidazopyridines
code: 4EUU) revealed that TBK1 and Aurora A share common 25−29, which are structurally related to the known Akt
structural features with respect to the hinge region (Supporting
inhibitor GSK 690693.31 In detail, molecule 28 leads to the
Information Figure S2). Therefore, we focused our optimiza-
most potent inhibition of TBK1 (IC50 = 0.14 μM) due to the
tion efforts on the design of further analogues of Scaffold I.
substitutions in 2- and 6-position, which seem to be superior to
In a second screening approach, we utilized compound
collections provided by GSK (Boston, MA; 360 compounds) the coincidental decoration of 2- and 5-position (25−27, IC50 =
and ROCHE (Basel; 235 compounds) that were comprised of 0.72−0.92 μM) as well as solely 5-position decorated
inhibitors and intermediates that were already part of compounds as 29 (IC50 = 1.6 μM). However, the 2-
pharmaceutical medicinal chemistry endeavors in the past. methylbut-3-yn-2-ol decoration in 2-position of the imidazo-
Since these libraries contain high affinity inhibitors, we set the pyridines appears to be crucial for gaining inhibitory activity,
cutoff to pick a hit compound to 10% remaining kinase activity since 29 remarkably drops in its effect toward TBK1. With
at 10 μM compound concentration (Supporting Information respect to the identified oxindoles, we likewise observed SARs.
Figure S3). We determined several core-structures (Figure 3) Among these, the entity of hits was decorated with divergent
revealing fairly divergent chemotypes that were shown to motifs on both 3- and 5-position and except 30 evoked IC50
significantly modulate TBK1 in activity-based assays. Since the values in the range 0.07 μM (33) to 0.28 μM (31). Thereby, 33
292 DOI: 10.1021/cb500908d
ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

Figure 4. GSK screening hits and corresponding IC50 values (determined in at least two independent duplicate experiments).

represented the most potent TBK1 inhibitor among the series moderate inhibitory impact (IC50 = 0.40−6.1 μM). Further-
of identified GSK screening hits. more, we identified substituted oxindoles (71−74, 77) as
Based on the ROCHE library, we were able to originate 39 effective modulators of TBK1 at which 77 (IC50 = 0.003 μM)
hits (Supporting Information Figure S4) that were significantly was shown to be the most potent inhibitor among the
characterized by a plethora of substituted bis-aryl maleimides collectivity of hits derived from the ROCHE screening library.
(39−63). Additionally, we detected several quinolines (64−70, Design of Derivatives with Improved Inhibitory
75), substituted oxindoles (71−74, 77), and an aminothiazole Efficiency. Based on our screening results, we identified the
(76). Interestingly, the vast majority of substitutions that was 2,4,6-substituted pyrimidine tozasertib30 and the structurally
found on the maleimide core translated into moderate to strong related Scaffold I derived from our in-house library as
TBK1 inhibition; thus, differences in potency could be linked to candidates for subsequent medicinal chemistry approaches
the divergent substitution patterns (2- and 3-position). While a involving inhibitor design and synthesis to aim for compounds
1-methyl-1H-indole was found to be present among the entity with enhanced inhibitory properties. Moderate inhibition of
of maleimides, the majority of compounds were additionally TBK1 activity was shown for tozasertib and Scaffold I resulting
decorated with a further 1H-indole (linked through indole 3- in IC50 values of 6.2 μM and ∼8.6 μM, respectively. Since these
position), which was variably substituted at its amine function molecules differ solely in the substituents attached to the 2-
thereby accounting for discriminative inhibitory effects. position of the pyrimidine while retaining the same inhibitory
Essentially, we observed a broad range of IC50 values starting effect, we anticipated that this position would be productive for
from 0.03 μM and ending up into 5.6 μM for 59 and 61 derivatization. We chemically optimized this scaffold, paying
respectively. Among these structures with linear extensions on particular attention to the alteration of the para-linked
the indole’s amine functionality, we illustrated that 59, 48, 58, cyclopropylamide of tozasertib for SAR studies and to improve
55, and 53 were most potent in our activity-based studies (IC50 the inhibitory effect. A crystal structure of tozasertib in complex
= 0.03 μM, 0.27 μM, 0.38 μM, 0.46 μM, and 0.47 μM, with Aurora A (PDB code: 3E5A) was used for in silico
respectively). Notably, the most potent maleimide 59 features a superposition with TBK1 (PDB code: 4EUT) revealing that a
nitroguanidine moiety, which was already described to similar binding mode may be adopted (Supporting Information
introduce a large dipole moment to the compound, and this Figure S2). As a first design approach, we replaced the
property was thereby speculated to account for its remarkable arylthioether by an arylether and converted the cyclo-
potency against protein kinase C (PKC).32 propylamide moiety into the corresponding propylamide,
However, compound 39 decorated with a small aniline which was attached to the accordant ortho-, meta-, and para-
substituent instead of a second 1H-indole moiety on the positions. We then focused on the derivatization of the para-
maleimide’s 4-position represents an exception from the rule position, since biochemical characterizations (see SAR Studies)
and elicited an IC50 value of 0.23 μM. On the contrary, exhibited loss of inhibitory activity in decreasing order for the
substituents with larger spatial arrangement as shown for 60 meta- and ortho- substitutions. Various alternative substituents
(naphthyl) and 63 (tetrahydropyrazinoindole) were, in general, along with electron withdrawing as well as donating properties
found to be less potent on TBK1 (IC50 ≥ 20 μM). Besides were introduced at the para-position including −NO2, −NH2,
these, we found several quinolines (64, 65, 67-70, 75) with −Cl, −F or −CF3, as well as −OMe. We therefore synthesized
293 DOI: 10.1021/cb500908d
ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

Scheme 1. Synthetic Procedure for the Preparation of Tozasertib Derivativesa

a
Reagents and conditions: (i) mCPBA, DCM, 0 °C−room temperature (RT), 72 h, 86%; (ii) substituted phenol and (a) NaH, THF, 0 °C, 30−240
min or (b) NaOAc, tBuOH, 90 °C, 40 min, 73−95% (9c−f: crude); (iii) 5-methyl-1H-pyrazol-3-amine, DIPEA, DMF, NaI (optional), 85 °C, 4−12
h, 69−83% (12c−f: 13−28%, 2 steps); (iv) 1-methylpiperazine, 110 °C, 15−30 min, 48−84%; (v) 5% Pd/C, NH4HCO2, EtOH, 95 °C, 1 h, 69%;
(vi) Boc2O, Et3N, MeOH, RT, 12 h, 56−66%; (vii) 5% Pd/C, NH4HCO2, EtOH, 95 °C, 30 min, 82−87%; (viii) propionyl chloride in THF, DIPEA,
THF, 0 °C, 30−60 min, crude; (ix) 25% TFA in DCM, RT, 45−60 min, 58−93% (2 steps).

a small focused library of tozasertib derivatives for subsequent (58−92%, two steps). Our focused library, comprised of 16
use in inhibition and SAR studies. derivatives, was subsequently applied to biochemical character-
Synthesis of a Focused Library of Tozasertib izations with respect to TBK1 inhibition utilizing the activity-
Derivatives. We synthesized a compound collection of based assay systems. Detailed synthetic procedures are
tozasertib derivatives (15a−k) following a general procedure described in the Supporting Information Methods.
(Scheme 1). Initially, 4,6-dichloro-2-(methylthio)pyrimidine 5 Activity-Based In Vitro Characterization and SAR
was oxidized using mCPBA in order to generate an appropriate Studies. To investigate structure−activity relationships, we
leaving group leading to the corresponding sulfone 6 in good conducted analyses by applying serial dilutions (0−20 μM) of
yield (86%). Subsequent introduction of a para-modified each synthesized compound to active TBK1. The observed
phenyl by nucleophilic aromatic substitution resulted in inhibitory effects were shown to be dependent on the position
intermediates 9a−j. Due to the substitution pattern of the of derivatization as well as the size of the introduced substituent
applied phenols, we observed differing reaction times and and its electron withdrawing or donating properties (Table 1).
correspondingly different yields in the range 45−95%. In the In particular, we demonstrated weak inhibition when the
initial effort to alter the position of phenyl substituents, we terminal cyclopropylamide moiety was replaced by an open
introduced a 2-nitrophenol group (7, 27%) and a 3-nitrophenol chain propylamide (24, IC50 = 2.5 μM). Studies comparing the
group (8, 84%). One of the 2,4-connected chlorines was connection positions (ortho, meta) illustrated that decorations
substituted with 5-methyl-1H-pyrazol-3-amine using DIPEA in at the para site revealed the most significant inhibitory effects.
DMF resulting in intermediates 10, 11, and 12a−j in yields of Propylamide substitution in the meta and para locations led to
70−80%. Finally, we introduced the methylpiperazine moiety at remarkably reduced inhibitory effects exhibiting IC50 values of
the pyrimidine’s 6-position by refluxing the respective precursor >10 μM and no inhibition, respectively. Further structural
in 1-methylpiperazine, forming compounds 13, 14, and 15a−j improvements were therefore focused on the derivatization of
in good yields (74−84%). To generate the p-amino substituted the para-phenyl position. Replacing the terminal propylamide
analogue, we reduced the respective p-nitro intermediate (15a) with a tert-butyl group did not improve the inhibitory effects
to the corresponding amine (15k) using Pd/C and NH4HCO2 (15b, 2.7 μM), while a decrease in the spatial arrangement of
in moderate yield (69%). the terminal functional group, for example, introducing
In a continued synthetic approach, we introduced propyl- isopropyl (15c) or methyl (15d) substituents, led to improved
amine moieties in the ortho, meta, and para positions by starting inhibitory activity (IC50 = 0.34 μM and 0.38 μM, respectively).
from the corresponding nitro-substituted intermediates (13, 14, Introduction of a methoxy group (15e) as well as halogen
15a) of which the secondary pyrazole amine was first Boc substitutions as in the case of 15f (p-F) and 15g (p-Cl) slightly
protected using Boc2O (yield: 55−67%). We reduced the nitro decreased the inhibitory activity, leading to IC50 values in the
group to obtain the corresponding amines 19, 20, and 21 range 0.65−1.2 μM. Strong electron donating effects as for
(Yield: 60−83%), which were then converted into propiony- amino substitution (15k, 0.73 μM) did not improve the
lamides by SN2 substitution using propionyl chloride. inhibition. In contrast, electron withdrawing groups led to
Successive deprotection using 25% TFA in DCM led to the consistent inhibitory properties as compared to isopropyl and
desired compounds (22, 23, 24) in moderate to good yield methyl substitution (15c,d) as demonstrated for trifluorometh-
294 DOI: 10.1021/cb500908d
ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

yl (15h, 0.29 μM). Notably, nitro decoration (15a) induced a and coincidently, it does not significantly affect the IFN-β
strong beneficial effect in terms of inhibition toward TBK1 pathway in cellular studies. On the contrary, 15f and 15c as less
possessing an IC50 value of 0.06 μM corresponding to a 100- effective inhibitors illustrate the opposed effect. One explan-
fold improvement in inhibition versus the initial screening hit ation might be justified in their different chemical properties
tozasertib. Taking into account the most potent compound (nitro- vs isopropyl- vs fluorine-substituents), which might
(15a) of our focused library, we likewise explored different interfere with cell penetration of the compound. However,
nitro attachment positions as was done for the cyclo- further analyses and studies need to be performed to deeper
propylamide decoration (22 vs 23 vs 24) and observed analog investigate these opposing results and the specific properties of
adverse inhibitory effects as illustrated before (13 vs 14 vs 15a, the designed inhibitors.
IC50 = 2.4, 1.4, and 0.06 μM, respectively). Activity-Based Profiling and Docking Studies. To
Evaluation of 15a, 15c, and 15f in RAW Macrophages. investigate the selectivity of 2,4,6-substituted pyrimidine-based
Since the potential oncogenic role of TBK1 is not proven, we inhibitor 15a, we analyzed its inhibitory activity with respect to
set out to utilize qPCR-coupled analyses of IFN-β expression in a panel of 80 representative kinases at a screening
macrophages in order to investigate the inhibitory effect of 15a concentration of 1 μM (Supporting Information Table S1,
on the relevant TBK1-associated signaling cascade in a cellular SelectScreen Kinase Profiling Service from Life Technologies).
setup. The expression of IFNs correlates with the affection of We identified 25 kinases that were significantly inhibited in
the TBK1 pathway to trigger antiviral and TLR-mediated their catalytic activity by more than 70%. Tyrosine kinases such
immune responses and therefore monitoring IFN expression as Abelson kinase (99% inhibition) and four representatives of
levels by qPCR represents a powerful tool to scrutinize the the fibroblast growth factor receptor (FGFR) and vascular
impact of our TBK1 inhibitors in cell-based studies as was done endothelial growth factor receptor (VEGFR) families were
before.33 Remarkably, we did not find compound 15a to most effectively inhibited (>95%). Notably, the ten prime
significantly affect the IFN-pathway in RAW macrophages, targets of 15a within this panel are represented by tyrosine
whereas 15c and 15f were illustrated to reveal more dramatic kinases, and GSK3β (91% inhibition) is the most effectively
impact on the respective signaling cascade. In detail, LPS inhibited Ser/Thr kinase followed by Aurora A (88%). TBK1
stimulated cells were shown to increase IFN-expression to was likewise demonstrated to be efficiently inhibited by 15a
about 10-fold as compared to unstimulated cells (Figure 5). (79%), but the vast diversity of addressed targets illustrates that
selective inhibition of TBK1 is challenging. Remarkably, PDK1
(33% inhibition) as well as IKKε (13% inhibition), which
represent prime targets of the known TBK1 inhibitor BX795,
were not significantly impaired.
In addition to profiling studies, we further investigated the
binding mode of 15a within the catalytic domain of TBK1 by
performing docking studies using the free available receptor−
ligand docking server DockThor (Supporting Information
Figure S5).34 These analyses support our initially proposed
binding mode based on the superposition of tozasertib in
complex with Aurora A and TBK1. According to our
assumption that the pyrazole moiety accounts for the formation
of hydrogen interactions to the backbone amides within the
hinge region a similar binding mode was observed. However,
the docking studies solely revealed the formation of two instead
of three hydrogen bond interactions to the peptide backbone of
Glu87 and Phe88. In addition, we identified a vice versa
Figure 5. qRT-PCR-based examination of IFN-pathway interference orientation of the piperazine and para-nitrophenyl moieties
in RAW macrophages rendered by compounds BX795, 15a, 15c, and
most likely due to the formation of favorable ionic interactions
15f at 1 μM compound concentration (ctrl: unstimulated; DMSO:
LPS-induced). of the piperazine with Asp157 and due to the lack of an
aromatic amino acid within the glycine-rich loop that allows for
flexible ligand rearrangement (see Supporting Information
Notably, for 15a, we observed a 6-fold increased mRNA level of Figure S5 for detailed information).
IFN-β at 1 μM compound concentration. The respective Conclusions. In the present study, we report the successful
pathway was apparently still induced and therefore 15a was identification of TBK1 inhibitors by activity-based screening
expulsed from representing a potent cellular modulator of endeavors. Compilations of industry provided compounds
TBK1 and its associated signaling cascade. In contrast, 15c and (GSK and ROCHE) were scrutinized for inhibitory activity
15f, previously highlighted as moderate TBK1 inhibitors in with respect to TBK1. Since this collection was comprised of
activity-based analyses, were capable of blocking the pathway molecules that were already subject to drug development
activation more dramatically. In detail, 15c merely led to a 2- endeavors, we derived a plethora of compounds with
fold increase in mRNA levels as compared to unstimulated cells remarkable inhibitory effects and were able to retrace SARs
though 15f likewise impaired the pathway activation ending up among the identified scaffolds. Among these, oxindole 77 (IC50
into 4-fold IFN-β expression above control level. Hereby, we = 0.003 μM) from ROCHE and imidazopyrimidine 28 (IC50 =
confirmed that our inhibitors significantly impact the TBK1 0.14 μM) from GSK were shown to constitute the most potent
associated signaling cascade on a cellular level. We can only representatives of each library.
speculate on the reasons for the converse result of 15a In addition, two independently compiled small molecule in-
representing the most potent inhibitor in activity-based assays, house libraries afforded 2,4,6-substituted pyrimidine scaffolds
295 DOI: 10.1021/cb500908d
ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

(tozasertib and Scaffold I) that were considered to serve as the fluorophore XL665 were added. FRET between europium cryptate
basis for further rational design approaches to improve these and XL665 was measured to quantify the phosphorylation of the
core structures with respect to their inhibitory activity toward substrate peptide. ATP concentrations were adjusted to 30 μM while a
TBK1. Consecutive validation studies and optimization of substrate concentration of 1 μM was used. Kinase and inhibitor were
preincubated for 30 min before the reaction was started by addition of
tozasertib resulted in the linear synthesis of a small focused ATP and substrate peptide. A Tecan infinite M1000 plate reader was
library of derivatives (13−15a−k and 22−24). We paid used to measure the fluorescence of the samples at 620 nm (Eu-
particular attention to the derivatization of the para-phenyl labeled antibody) and 665 nm (XL665 labeled streptavidin) 60 μs after
position, which was found to be crucial for modulating TBK1 excitation at 317 nm. The quotient of both intensities for reactions
activity in vitro. Candidates in our focused library were then made with eight different inhibitor concentrations was fit to a Hill
tested for enhanced inhibitory effects resulting in distinct SARs. four-parameter equation to determine IC50 values. Each reaction was
Notably, the nature of the para-connected functional group performed in duplicate, and at least three independent determinations
with respect to its spatial arrangement combined with electron of each IC50 were made.
For screening purposes, we utilized the Cisbio system HTRF
withdrawing and, in turn, electron donating properties was
KinEASE-STK as stated above and TBK1 from Millipore (#14-628,
pivotal for the evoked inhibitory effect. We illustrated the ability Lot# D9SN044N-A). Robotics assisted compound transfers (2 × 6.6
of terminal tert-butyl moieties to exhibit weak inhibition (15b, nL for 13.2 μM screening concentration) were processed using a
2.7 μM), which was significantly improved by decreasing the PinTool system from Tecan, or compounds were manually transferred
spatial arrangement with isopropyl and methyl derivatives for where appropriate (10 μM screening concentration). At least 8 data
improved potency (15c and 15d; 0.34 and 0.38 μM, points for positive and negative controls were included for each assay
respectively). The most significant perturbation of TBK1 plate using BX795 (13.2 μM or 10 μM) as 0% and DMSO as 100%
revealed an inhibitory effect within the midnanomolar range TBK1 kinase activity control. The assay readout was performed using a
(IC50 = 0.06 μM) and was elicited by nitro substituted Tecan infinite M1000 plate reader, and a Z′-factor of 0.81 was
determined for the entire screen.
compound 15a providing evidence for the advantageous effect qRT-PCR in RAW Macrophages. RAW 264.7 macrophages were
of electron withdrawing functional groups. preincubated for 1 h with DMSO or TBK1 inhibitors as indicated and
Ultimately, we conducted cellular studies using RAW then stimulated with 100 ng/mL LPS (Invivogen) for 2 h. Hereafter,
macrophages and methods of qPCR to investigate the RNA was extracted using Quiageńs RNeasy Mini Kit. cDNA was
compounds impact on the expression of interferons as a prepared from 500 ng RNA using the Superscript III reverse
marker for the TBK1-pathway modulation. Significant down- transcriptase (Invitrogen #18064). qRT-PCR was performed using a
regulation of the IFN-β expression was confirmed for 7300 Real-Time PCR System (Applied Biosystems) and Power SYBR
compounds 15c and 15f as the copy number of IFN-β was Green PCR Master Mix (Applied Biosystems) with primers for mouse
decreased, at 1 μM compound concentrations, to approximately GAPDH and Interferon-β (both at 60 °C) as described by Orzalli et
al.33 ΔCt-values were determined using the 7300 System Software
2- and 4-fold, respectively, as compared to the relative (Applied Biosystems) using GAPDH as reference control and gene
expression in nonstimulated macrophages. expression was calculated by the ΔΔCt-method.35


Consecutive profiling studies revealed a plethora of kinases
that is significantly inhibited by the most potent compound 15a ASSOCIATED CONTENT
and underline the need for further design approaches to tune *
S Supporting Information
the specificity of our 2,4,6-substituted pyrimidine-based Detailed experimental procedures for compound synthesis,
inhibitors. Docking studies further illuminated the potential experimental screening workflows, cluster analysis of screening
binding mode of 15a and provide a substantial basis for broader hits from Collection II, superposition of Aurora A and TBK1,
design endeavors. qPCR validation studies, docking studies, profiling studies as
In conclusion, our activity-based screening procedure well as IC50 data for identified screening hits derived from GSK-
allowed us to identify compounds that inhibited TBK1. and ROCHE-libraries. This material is available free of charge
These compounds were then optimized by using methods of via the Internet at http://pubs.acs.org.


medicinal chemistry. Potential use of these compounds to
decipher TBK1 related signaling or to provide starting materials AUTHOR INFORMATION
for subsequent drug design approaches is anticipated for our
Corresponding Author
optimized derivatives.
*Phone: +49 (0)231−755 7080. Fax: +49 (0)231−755 7082.

■ METHODS
Reagents and Materials. Unless otherwise noted, all reagents and
E-mail: daniel.rauh@tu-dortmund.de.
Notes
The authors declare no competing financial interest.


solvents were purchased from Acros, Fluka, Sigma-Aldrich, or Merck
and used without further purification. Small volume (20 μL fill ACKNOWLEDGMENTS
volume) white round-bottom 384-well plates were obtained from
Greiner Bio-One GmbH (Solingen, Germany). All supplies for the This work was cofunded by the German federal state North
TBK1 HTRF assay kit were purchased from CisBio (Bagnols-sur- Rhine Westphalia (NRW) and the European Union (European
Cèze, France). Axitinib, bosutinib, dovitinib, erlotinib, K252a, Regional Development Fund: Investing in Your Future), the
sunitinib, tofacitinib, and tozasertib were purchased from LC German Federal Ministry for Education and Research
Laboratories (Woburn, MA). (NGFNPlus and e:Med) (Grant No. BMBF 01GS08104,
Activity-Based Assay for IC50 Determination and Screening. 01ZX1303C). R.T. is funded and supported by CAPES and
IC 50 determinations for TBK1 (Millipore, #14-628M, Lot# FAPERJ.


D9SN044N-B) were performed with the KinEASE-STK assay from
Cisbio according to the manufacturer’s instructions. A biotinylated
ABBREVIATIONS
substrate peptide (STK3 from Cisbio) was phosphorylated by TBK1.
After completion of the reaction, an antiphosphoserine/-threonine Akt, protein kinase B; CCL5, C−C motif chemokine 5; FGFR,
antibody labeled with europium cryptate and streptavidin labeled with fibroblast growth factor receptor; GSK3β, glycogen synthase
296 DOI: 10.1021/cb500908d
ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

kinase-3 β; HER2, receptor tyrosine-protein kinase erbB-2; kinase mediates Ser-172 phosphorylation and activation. J. Biol. Chem.
HPIP, hematopoietic PBX-interaction protein; IL-6, intereukin 284, 14136−14146.
6; MDA5, melanoma differentiation-associated protein 5; (12) Wei, C., Cao, Y., Yang, X., Zheng, Z., Guan, K., Wang, Q., Tai,
NAP1, NFκB-activating kinase-associated protein 1; NFκB, Y., Zhang, Y., Ma, S., Ge, X., Xu, C., Li, J., Yan, H., Ling, Y., Song, T.,
Zhu, L., Zhang, B., Xu, Q., Hu, C., Bian, X. W., He, X., and Zhong, H.
nuclear factor NFκB p105 subunit; PBX, pre-B-cell leukemia
(2014) Elevated expression of TANK-binding kinase 1 enhances
transcription factor; PKC, protein kinase C; RIG-I, retinoic tamoxifen resistance in breast cancer. Proc. Natl. Acad. Sci. U.S.A. 111,
acid-inducible gene 1 protein; RTK, receptor tyrosine kinase; E601−610.
SAR, structure−activity relationship; SINTBAD, TBK1-binding (13) Shostak, K., Patrascu, F., Göktuna, S. I., Close, P., Borgs, L.,
protein 1; TANK, TRAF family member-associated NFκB Nguyen, L., Olivier, F., Rammal, A., Brinkhaus, H., Bentires-Alj, M.,
activator; TBK1, TANK binding kinase 1; TNF, tumor necrosis Marine, J. C., and Chariot, A. (2014) MDM2 restrains estrogen-
factor; TRAF, TNF receptor associated factor; qRT-PCR, mediated AKT activation by promoting TBK1-dependent HPIP
quantitative real-time polymerase chain reaction; VEGFR, degradation. Cell Death Differ. 21, 811−824.
vascular endothelial growth factor receptor; ZEB1, zinc finger (14) Deng, T., Liu, J. C., Chung, P. E., Uehling, D., Aman, A., Joseph,
E-box-binding homeobox 1 B., Ketela, T., Jiang, Z., Schachter, N. F., Rottapel, R., Egan, S. E., Al-


Awar, R., Moffat, J., and Zacksenhaus, E. (2014) shRNA kinome
screen identifies TBK1 as a therapeutic target for HER2+ breast
REFERENCES cancer. Cancer Res. 74, 2119−2130.
(1) Goncalves, A., Bürckstümmer, T., Dixit, E., Scheicher, R., Górna, (15) Wang, T., Block, M. A., Cowen, S., Davies, A. M., Devereaux, E.,
M. W., Karayel, E., Sugar, C., Stukalov, A., Berg, T., Kralovics, R., Gingipalli, L., Johannes, J., Larsen, N. A., Su, Q., Tucker, J. A.,
Planyavsky, M., Bennett, K. L., Colinge, J., and Superti-Furga, G. Whitston, D., Wu, J., Zhang, H. J., Zinda, M., and Chuaqui, C. (2012)
(2011) Functional dissection of the TBK1 molecular network. PLoS Discovery of azabenzimidazole derivatives as potent, selective
One 6, e23971. inhibitors of TBK1/IKKε kinases. Bioorg. Med. Chem. Lett. 22,
(2) Pichlmair, A., and Reis e Sousa, C. (2007) Innate recognition of 2063−2069.
viruses. Immunity 27, 370−383. (16) Johannes, J. W., Chuaqui, C., Cowen, S., Devereaux, E.,
(3) Fitzgerald, K. A., McWhirter, S. M., Faia, K. L., Rowe, D. C., Latz, Gingipalli, L., Molina, A., Wang, T., Whitston, D., Wu, X., Zhang, H. J.,
E., Golenbock, D. T., Coyle, A. J., Liao, S. M., and Maniatis, T. (2003) and Zinda, M. (2014) Discovery of 6-aryl-azabenzimidaoles that
IKKε and TBK1 are essential components of the IRF3 signaling inhibit the TBK1/IKK-ε kinases. Bioorg. Med. Chem. Lett. 24, 1138−
pathway. Nat. Immunol. 4, 491−496. 1143.
(4) Kawai, T., and Akira, S. (2007) Signaling to NF-κB by Toll-like (17) Ou, Y. H., Torres, M., Ram, R., Formstecher, E., Roland, C.,
receptors. Trends Mol. Med. 13, 460−469. Cheng, T., Brekken, R., Wurz, R., Tasker, A., Polverino, T., Tan, S. L.,
(5) Häcker, H., and Karin, M. (2006) Regulation and function of IKK and White, M. A. (2011) TBK1 directly engages Akt/PKB survival
and IKK-related kinases. Sci. STKE 2006, re13. signaling to support oncogenic transformation. Mol. Cell 41, 458−470.
(6) Barbie, D. A., Tamayo, P., Boehm, J. S., Kim, S. Y., Moody, S. E., (18) McIver, E. G., Bryans, J., Birchall, K., Chugh, J., Drake, T., Lewis,
Dunn, I. F., Schinzel, A. C., Sandy, P., Meylan, E., Scholl, C., Fröhling, S. J., Osborne, J., Smiljanic-Hurley, E., Tsang, W., Kamal, A., Levy, A.,
S., Chan, E. M., Sos, M. L., Michel, K., Mermel, C., Silver, S. J., Weir, B. Newman, M., Taylor, D., Arthur, J. S., Clark, K., and Cohen, P. (2012)
A., Reiling, J. H., Sheng, Q., Gupta, P. B., Wadlow, R. C., Le, H., Synthesis and structure−activity relationships of a novel series of
Hoersch, S., Wittner, B. S., Ramaswamy, S., Livingston, D. M., Sabatini, pyrimidines as potent inhibitors of TBK1/IKKε kinases. Bioorg. Med.
D. M., Meyerson, M., Thomas, R. K., Lander, E. S., Mesirov, J. P., Chem. Lett. 22, 7169−7173.
Root, D. E., Gilliland, D. G., Jacks, T., and Hahn, W. C. (2009) (19) Ma, X., Helgason, E., Phung, Q. T., Quan, C. L., Iyer, R. S., Lee,
Systematic RNA interference reveals that oncogenic KRAS-driven M. W., Bowman, K. K., Starovasnik, M. A., and Dueber, E. C. (2012)
cancers require TBK1. Nature 462, 108−112. Molecular basis of Tank-binding kinase 1 activation by trans-
(7) Newman, A. C., Scholefield, C. L., Kemp, A. J., Newman, M., autophosphorylation. Proc. Natl. Acad. Sci. U.S.A. 109, 9378−9383.
McIver, E. G., Kamal, A., and Wilkinson, S. (2012) TBK1 kinase (20) Larabi, A., Devos, J. M., Ng, S. L., Nanao, M. H., Round, A.,
addiction in lung cancer cells is mediated via autophagy of Tax1bp1/ Maniatis, T., and Panne, D. (2013) Crystal structure and mechanism
Ndp52 and non-canonical NF-κB signalling. PLoS One 7, e50672. of activation of TANK-binding kinase 1. Cell Rep. 3, 734−746.
(8) Zhu, Z., Aref, A. R., Cohoon, T. J., Barbie, T. U., Imamura, Y., (21) Tu, D., Zhu, Z., Zhou, A. Y., Yun, C. H., Lee, K. E., Toms, A. V.,
Yang, S., Moody, S. E., Shen, R. R., Schinzel, A. C., Thai, T. C., Reibel, Li, Y., Dunn, G. P., Chan, E., Thai, T., Yang, S., Ficarro, S. B., Marto, J.
J. B., Tamayo, P., Godfrey, J. T., Qian, Z. R., Page, A. N., Maciag, K., A., Jeon, H., Hahn, W. C., Barbie, D. A., and Eck, M. J. (2013)
Chan, E. M., Silkworth, W., Labowsky, M. T., Rozhansky, L., Mesirov, Structure and ubiquitination-dependent activation of TANK-binding
J. P., Gillanders, W. E., Ogino, S., Hacohen, N., Gaudet, S., Eck, M. J., kinase 1. Cell Rep. 3, 747−758.
Engelman, J. A., Corcoran, R. B., Wong, K. K., Hahn, W. C., and (22) Kim, J. Y., Beg, A. A., and Haura, E. B. (2013) Non-canonical
Barbie, D. A. (2014) Inhibition of KRAS-driven tumorigenicity by IKKs, IKK, and TBK1, as novel therapeutic targets in the treatment of
interruption of an autocrine cytokine circuit. Cancer Discovery 4, 452− non-small cell lung cancer. Expert Opin. Ther. Targets 17, 1109−1112.
465. (23) Knapp, S., Arruda, P., Blagg, J., Burley, S., Drewry, D. H.,
(9) Liu, W., Huang, Y. J., Liu, C., Yang, Y. Y., Liu, H., Cui, J. G., Edwards, A., Fabbro, D., Gillespie, P., Gray, N. S., Kuster, B., Lackey,
Cheng, Y., Gao, F., Cai, J. M., and Li, B. L. (2014) Inhibition of TBK1 K. E., Mazzafera, P., Tomkinson, N. C., Willson, T. M., Workman, P.,
attenuates radiation-induced epithelial−mesenchymal transition of and Zuercher, W. J. (2013) A public−private partnership to unlock the
A549 human lung cancer cells via activation of GSK-3β and repression untargeted kinome. Nat. Chem. Biol. 9, 3−6.
of ZEB1. Lab. Invest. 94, 362−370. (24) Shen, R. R., and Hahn, W. C. (2011) Emerging roles for the
(10) Feldman, R. I., Wu, J. M., Polokoff, M. A., Kochanny, M. J., non-canonical IKKs in cancer. Oncogene 30, 631−641.
Dinter, H., Zhu, D., Biroc, S. L., Alicke, B., Bryant, J., Yuan, S., (25) Pardanani, A., Lasho, T., Smith, G., Burns, C. J., Fantino, E., and
Buckman, B. O., Lentz, D., Ferrer, M., Whitlow, M., Adler, M., Finster, Tefferi, A. (2009) CYT387, a selective JAK1/JAK2 inhibitor: In vitro
S., Chang, Z., and Arnaiz, D. O. (2005) Novel small molecule assessment of kinase selectivity and preclinical studies using cell lines
inhibitors of 3-phosphoinositide-dependent kinase-1. J. Biol. Chem. and primary cells from polycythemia vera patients. Leukemia 23,
280, 19867−19874. 1441−1445.
(11) Clark, K., Plater, L., Peggie, M., and Cohen, P. (2009) Use of (26) Zhang, J. H., Chung, T. D., and Oldenburg, K. R. (1999) A
the pharmacological inhibitor BX795 to study the regulation and Simple statistical parameter for use in evaluation and validation of high
physiological roles of TBK1 and IκB kinase ε: A distinct upstream throughput screening assays. J. Biomol. Screen. 4, 67−73.

297 DOI: 10.1021/cb500908d


ACS Chem. Biol. 2015, 10, 289−298
ACS Chemical Biology Articles

(27) Baell, J. B., and Holloway, G. A. (2010) New substructure filters


for removal of pan assay interference compounds (PAINS) from
screening libraries and for their exclusion in bioassays. J. Med. Chem.
53, 2719−2740.
(28) Baell, J. B. (2013) Broad coverage of commercially available
lead-like screening space with fewer than 350 000 compounds. J. Chem.
Inf. Model. 53, 39−55.
(29) Baell, J., and Walters, M. A. (2014) Chemistry: Chemical con
artists foil drug discovery. Nature 513, 481−483.
(30) Harrington, E. A., Bebbington, D., Moore, J., Rasmussen, R. K.,
Ajose-Adeogun, A. O., Nakayama, T., Graham, J. A., Demur, C.,
Hercend, T., Diu-Hercend, A., Su, M., Golec, J. M., and Miller, K. M.
(2004) VX-680, a potent and selective small-molecule inhibitor of the
Aurora kinases, suppresses tumor growth in vivo. Nat. Med. 10, 262−
267.
(31) Rhodes, N., Heerding, D. A., Duckett, D. R., Eberwein, D. J.,
Knick, V. B., Lansing, T. J., McConnell, R. T., Gilmer, T. M., Zhang, S.
Y., Robell, K., Kahana, J. A., Geske, R. S., Kleymenova, E. V.,
Choudhry, A. E., Lai, Z., Leber, J. D., Minthorn, E. A., Strum, S. L.,
Wood, E. R., Huang, P. S., Copeland, R. A., and Kumar, R. (2008)
Characterization of an Akt kinase inhibitor with potent pharmacody-
namic and antitumor activity. Cancer Res. 68, 2366−2374.
(32) Davis, P. D., Elliott, L. H., Harris, W., Hill, C. H., Hurst, S. A.,
Keech, E., Kumar, M. K., Lawton, G., Nixon, J. S., and Wilkinson, S. E.
(1992) Inhibitors of protein kinase C. 2. Substituted bisindolylmalei-
mides with improved potency and selectivity. J. Med. Chem. 35, 994−
1001.
(33) Orzalli, M. H., DeLuca, N. A., and Knipe, D. M. (2012) Nuclear
IFI16 induction of IRF-3 signaling during herpes viral infection and
degradation of IFI16 by the viral ICP0 protein. Proc. Natl. Acad. Sci.
U.S.A. 109, E3008−3017.
(34) de Magalhães, C. S., Almeida, D. M., Barbosa, H. J. C., and
Dardenne, L. E. (2014) A dynamic niching genetic algorithm strategy
for docking highly flexible ligands. Inf. Sci. 289, 206−224.
(35) Pfaffl, M. W. (2001) A new mathematical model for relative
quantification in real-time RT-PCR. Nucleic Acids Res. 29, e45.
(36) Lounkine, E., Wawer, M., Wassermann, A. M., and Bajorath, J.
(2010) SARANEA: A freely available program to mine structure−
activity and structure−selectivity relationship information in com-
pound data sets. J. Chem. Inf. Model. 50, 68−78.

298 DOI: 10.1021/cb500908d


ACS Chem. Biol. 2015, 10, 289−298

You might also like