Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Journal Pre-proofs

Identifying new lead structures to enhance tolerance towards drought stress


via high-throughput screening giving crops a quantum of solace

Jens Frackenpohl, Linn Schneider, Luka J.B. Decker, Jan Dittgen, Franz
Fenkl, Christian Fischer, Jana Franke, Joerg Freigang, Rahel Getachew,
Susana M. Gonzalez Fernandez-Nino, Hendrik Helmke, Martin J. Hills,
Sabine Hohmann, Jochen Kleemann, Karoline Kurowski, Gudrun Lange,
Peter Luemmen, Nicole Meyering, Fabien Poree, Dirk Schmutzler, Sebastian
Wrede

PII: S0968-0896(19)31027-2
DOI: https://doi.org/10.1016/j.bmc.2019.115142
Reference: BMC 115142

To appear in: Bioorganic & Medicinal Chemistry

Received Date: 12 July 2019


Revised Date: 19 September 2019
Accepted Date: 25 September 2019

Please cite this article as: J. Frackenpohl, L. Schneider, L.J.B. Decker, J. Dittgen, F. Fenkl, C. Fischer, J. Franke,
J. Freigang, R. Getachew, S.M. Gonzalez Fernandez-Nino, H. Helmke, M.J. Hills, S. Hohmann, J. Kleemann, K.
Kurowski, G. Lange, P. Luemmen, N. Meyering, F. Poree, D. Schmutzler, S. Wrede, Identifying new lead
structures to enhance tolerance towards drought stress via high-throughput screening giving crops a quantum of
solace, Bioorganic & Medicinal Chemistry (2019), doi: https://doi.org/10.1016/j.bmc.2019.115142

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover
page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version
will undergo additional copyediting, typesetting and review before it is published in its final form, but we are
providing this version to give early visibility of the article. Please note that, during the production process, errors
may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

© 2019 Elsevier Ltd. All rights reserved.


Graphical Abstract
To create your abstract, type over the instructions in the template box below.
Fonts or abstract dimensions should not be changed or altered.

Identifying new lead structures to enhance Leave this area blank for abstract info.
tolerance towards drought stress via high-
throughput screening giving crops a
quantum of solace
Jens Frackenpohla,, Linn Schneiderb, Luka J. B. Deckera, Jan Dittgena, Franz Fenkla, Christian
Fischera, Jana Frankea, Joerg Freigangc, Susana M. Gonzalez Fernandez-Ninoa, Hendrik Helmkea,
Martin J. Hillsa, Sabine Hohmanna, Rahel Getachewa, Jochen Kleemannc, Karoline Kurowskia, Gudrun
Langea, Peter Luemmena, Nicole Meyeringb, Fabien Poreea, Dirk Schmutzlera and Sebastian Wredeb
a
Research & Development, Weed Control - Bayer AG, Crop Science Division, Industriepark Höchst, D-65926 Frankfurt am Main
b
Research & Development, Lead Discovery – Bayer AG, Pharmaceutical Division, Aprather Weg 18a, D-42096 Wuppertal
c
Research & Development - Bayer AG, Crop Science Division, Gebäude 6240, Alfred-Nobel-Straße 50 ,D-40789 Monheim

High-throughput screening
Bioorganic & Medicinal Chemistry

Identifying new lead structures to enhance tolerance towards drought stress via high-
throughput screening giving crops a quantum of solace
Jens Frackenpohla, , Linn Schneiderb, Luka J. B. Deckera, Jan Dittgena, Franz Fenkla, Christian Fischera,
Jana Frankea, Joerg Freigangc, Rahel Getachewa, Susana M. Gonzalez Fernandez-Ninoa, Hendrik Helmkea,
Martin J. Hillsa, Sabine Hohmanna, Jochen Kleemannc, Karoline Kurowskia, Gudrun Langea, Peter
Luemmena, Nicole Meyeringb, Fabien Poreea, Dirk Schmutzlera and Sebastian Wredeb
a
Research & Development, Weed Control - Bayer AG, Crop Science Division, Industriepark Höchst, D-65926 Frankfurt am Main
b
Research & Development, Lead Discovery – Bayer AG, Pharmaceutical Division, Aprather Weg 18a, D-42096 Wuppertal
c
Research & Development - Bayer AG, Crop Science Division, Alfred-Nobel-Straße 50 ,D-40789 Monheim

ARTICLE INFO ABSTRACT

Article history: Novel synthetic lead structures interacting with RCAR/(PYR/PYL) receptor proteins were
Received identified based on the results of a high-throughput screening campaign of a large compound
Received in revised form library followed by focused SAR studies of the three most promising hit clusters. Whilst
Accepted indolinylmethyl sulfonamides 8y,z and phenylsulfonyl ethylenediamines 9y,z showed strong
Available online affinities for RCAR/ (PYR/PYL) receptor proteins in wheat, thiotriazolyl acetamides 7f,s
exhibited promising efficacy against drought stress in vivo (wheat, corn and canola) combined
Keywords: with confirmed target interaction in wheat and arabidopsis thaliana. Remarkably, binding
phytochemistry affinities of several representatives of 8 and 9 were on the same level or even better than the
drought stress essential plant hormone abscisic acid (ABA).
high throughput screening
aryl sulfonamides 2009 Elsevier Ltd. All rights reserved.
thiotriazolyl acetamides

1. Introduction proteins as soluble ABA-receptors.10,11 It was shown via crystal


structural analysis that binding of ABA to AtRCAR12 (cf.
Agricultural crops face tremendous losses due to biotic stress AtPYL1) induces a conformational change in the highly
such as pests, diseases, and weed damages, but abiotic stress conserved ABA receptor proteins giving rise to an interaction
adversely affects crop production even further in various parts of with phosphoprotein phosphatases 2C (PP2Cs) thereby inhibiting
the world, decreasing average yields for most of the crops their activity. Hence, these findings have granted new insights
significantly.1 Among various abiotic stresses affecting into the structure activity relationship (SAR) of ABA whilst its
agricultural production, drought stress is considered to be the catabolism had been well explored prior to identifying the
main source of crop losses around the globe.2 There are various RCAR/(PYR/PYL) receptor proteins.
strategies for reducing the impact that drought has on crop yield.
These include exploiting beneficial effects of crop protection ABA’s principal pathway of oxidation is through
agents,3 developing drought tolerant crops through transgenic monooxygenase-mediated hydroxylation, i.e. by the CYP707A-
approaches or breeding,4,5 but also exploring novel chemical family, of the 8’-methyl group affording 8’-hydroxy-ABA which
active ingredients inspired by naturally occurring plant is in equilibrium with the cyclized form phaseic acid. 12-15 Whilst
hormones. Abscisic acid (1, S-(+)-ABA), a chiral sesquiterpenoid stabilizing or further substituting the cyclohexenone moiety has
first discovered in the 1960s,6-9 is one the most important plant thus attracted significant interest,16-22 surprisingly few approaches
hormones regulating signals in plants such as seed maturation, have been made to identify open-chain analogues of ABA with
dormancy or water use. It has been commercialized, e.g. for promising target affinity. Recently, it was shown that cyano-
enhancing colour development in red table grapes, but it also cyclopropyl groups in 2a-c served as suitable replacements of the
mediates the adaption of plants to different types of cyclohexenone moiety leading to ABA analogues with strong
environmental abiotic stress such as drought, heat or salinity activity in vitro and in vivo.23 Furthermore, the first synthetic
stress. Studies on ABA mediated signaling have progressed compounds interacting with RCAR/(PYR/PYL) receptor proteins
have been prepared.24-26 In our view this indicates that we can
———
rapidly since the recent discovery of RCAR/(PYR/PYL) receptor

 Corresponding author. Building G836 Industriepark Höchst, D- 65926 Frankfurt am Main; E-mail: jens.frackenpohl@bayer.com
6
8´ 9´
5
3

2
5´ 6´
4
1

2´ 7´

3a X = CH, R1 = Br 5a R1 = n-Pr, R2 = Me
(S)-(+)-ABA 1 3b X = N, R1 = F 5b R1 = allyl, R2 = Me
5c R1 = F3C-CH2-, R2 = Cl/CN

2a R2 = Et, R3 = H 4a X = C-CH3, Y = S, R1 = Br 6a X = Me, R1 = n-propyl, R2 = Me


2b R2 = Et, R3 = CH3 4b X = N-Et, Y = CH, R1 = H 6b X = OMe, R1 = c-Pr-CH2-, R2 = Me
2c R2 = CF3, R3 = Me

Figure 1. Abscisic acid and selected terpenes, sulfonamides and phosphonamides interacting with RCAR/(PYR/PYL) receptor proteins

identify ABA analogues with more profound structural changes


that exhibit good receptor binding and promising in vivo efficacy.
2.2. High-throughput screening
Substituted aryl sulfonamides, e. g. pyrabactin 3a,27,28 closely
related pyrimidine 3b, as well as thiazoles and pyrazoles 4a-b,29 A biochemical assay was developed, probing for compounds
have shown promising initial receptor affinity and beneficial inducing the inhibitory complex of wheat phosphatase TaABI1
effects in in vivo tests (Fig. 1). More recently, tetrahydro- and co-regulator TaRCAR14. TaABI1 activity was detected by
quinolinyl sulfonamides 5a-c have shown improved in vitro dephosphorylation of 4-methylumbelliferyl phosphate (MUP).
activity.30-33 Interestingly, it was further demonstrated that the The assay was successfully adapted to the 1536 well plate format
sulfonamide unit in these hit structures could be replaced by and fully automated processing with the robotic screening
phosphonamides 6a-b preserving binding affinity and thus system. Sensitivity towards compounds inducing the inhibitory
emphasizing opportunities to introduce further structural motifs complex was assessed using ABA as the reference compound,
into ABA-related structures.34 Herein, we outline how we which was also continuously used to monitor the assay
identified new lead structures interacting with RCAR/(PYR/ performance and the constancy of the primary screening
PYL) receptor proteins via high-throughput screening (HTS) campaign. On each robotic test day full kinetic traces were
using a wheat RCAR and subsequent focused SAR studies. determined to confirm the overall enzymatic activity. A total of
3,691,096 tested compounds were classified by the change in
fluorescence intensity of the substrate from an initial
2. Results and discussion measurement directly after pipetting (M1, Figure 2b) to an end
point of the enzymatic reaction after 80 min at 32 °C (M2).
2.1. Receptor protein identification and isolation Change in fluorescence intensity in the presence of reference
In earlier studies we solved crystal structures with close compound ABA or the test compounds (termed M2-M1) was
analogs of ABA (1) in complex with receptor protein AtRCAR11 normalized by the negative control DMSO, which is set to 100 %
(cf. AtPYR1) and the PP2C phosphatase AtABI1 isolated from (Figure 2a). Primary hits were selected by activity values below
Arabidopsis thaliana (At).35 In line with results observed within DMSO scattering with a z-score < -8 (Figure 2a). This hit limit is
our in vivo screening campaign of terpenoid-based ABA-analogs also annotated in the dose response curve (DRC) of ABA (Figure
it was shown that structural variations (i) of the diene side chain 2c). With the selected hit limit, the high assay sensitivity allowed
or (ii) of key elements responsible for receptor binding (e.g. the to resolve an activity of about 15 nM ABA, a concentration
cyclohexenone carbonyl group) had a stronger impact on receptor eliciting ~20 % of the maximum response.
binding in AtRCAR11 than changes in the absolute configuration A B
of stereocentres in the cyclohexanone moiety. In order to explore
unprecedented synthetic lead structures we decided not to use the
model plant Arabidopsis thaliana, but to investigate RCAR
receptor proteins in a relevant target crop as the basis for a high-
throughput screening campaign, i.e. wheat (Triticum aestivum,
Ta). Therefore, the orthologous genes of AtRCAR11 and AtABI1 C
coding for the proteins were identified in Triticum aestivum. To
do so, a reciprocal BLAST search was performed using the
Arabidopsis sequences as the query. TaABI1 (AB238930) and
TaRCAR14 (KD239761) were identified. Synthetic coding genes
were ordered and cloned in the expression vector pQE70 (Life
Techn.). Proteins were produced in Escherichia coli and
subsequently purified. Thereafter, their activity was measured
and compared to the activity of the corresponding Arabidopsis Figure 2. Primary HTS scatter plot: Phosphatase activity, determined by
protein as previously described.35 Likewise, the effects of a change in fluorescence intensity of the substrate from an initial measurement
selected set of compounds having emerged from our recent directly after pipetting to an end point of the reaction after 80 min at 32 °C
synthesis programmes23, 29, 32, 33, 35 in the field of ABA signaling (M2-M1) (B), is plotted on the y-axis of the scatter plot (A). Values were
were tested. Significant differences could not be observed normalized to DMSO, which is set to 100. Compounds were tested at 10 or 5
between both series of proteins. Thus, TaABI1 and TaRCAR14 µM and the reference compound ABA was included as DRC. For hit
isolated from wheat could be used to start the high-throughput selection, a hit limit was set to z-score < -8. This hit limit is also annotated in
screening campaign. ABA DRCs of the robotic run (C) (brown dotted line). Compounds affecting
the fluorescence intensity independent of the enzymatic activity were
deselected by the change in initial fluorescence intensity plotted at the x-axis
of the scatter plot (A).
As a result of the in-depth hit evaluation, three promising hit
clusters were identified with several closely related members
Among these hits a set of compounds affecting the (clusters I – III, Figure 4). Compounds from clusters II and III
fluorescence intensity independent of enzymatic activity was showed strong initial binding affinities, whereas moderate
deselected by the respective increase or decrease of the initial affinities were observed for the members of cluster I. These
measurement directly after pipetting (M1 % Figure 2a). findings prompted us to investigate structural features of the
Additionally, obvious frequent hitters were excluded based on scaffolds and their respective in vitro and in vivo efficacies in
their uHTS data base profile, indicating non-specific broad detail.
activity on biochemical assays. In total, 3,908 compounds were Cluster I Cluster III
selected for secondary HTS.
For the selected hit compounds EC50 values were determined.
Based on a compound concentration of 10 µM, as used for
primary screening, an effect above 20% of the maximum
response could be confirmed for 2,988 compounds. This re- 7a R1 = Et, R2 = H, R3 = o-Cl 9a R1 = CF3, R2 = H
confirmation rate of 76 % evaluates the quality and precision of 7b R1 = c-Pr, R2 = Me, R3 = p-CO2Me
9b R1 = CF3, R2 = p-F
7c R1 = p-MeO-Bn, R2 = Me, R3 = m-SMe
the uHTS campaign. To distinguish compounds inducing the 9c R1 = H, R2 = p-Br
9d R1 = CF3, R2 = p-OMe
TaRCAR-TaABI1 inhibitory complex from complex-indepen- 9e R1 = CF3, R2 = m-Cl
Cluster II
dent compound effects, like direct inhibition of TaABI1 9f R1 = CF3, R2 = o-Cl
phosphatase activity, a counter assay on TaABI1 in the absence 9g R1 = CF3, R2 = m-F
9h R1 = CF3, R2 = m-OMe
of TaRCAR14 was performed. Figure 3 shows the correlation of
EC50 values in presence and absence of TaRCAR14. Most
compounds directly inhibit TaABI1 phosphatase activity, leading
8a R1 = Cl, R2 = Me
to comparable potency in both assay formats (Figure 3). Final 8b R1 = Br, R2 = Et
hits were selected by EC50 values below 10 µM in the presence of 8c R1 = Cl, R2 = Ph
8d R1 = Br, R2 = Ph
TaRCAR14 and no detectable direct effect on phosphatase
activity. The inset of Figure 3 shows the DRCs for an exemplary Figure 4. Promising hit clusters (all EC50 values below 10 µM@TaRCAR14)
hit compound. The final hit list with 127 compounds was then with selected representative members chosen for further exploration.
submitted to chemical inspection and subsequent hit evaluation.

2.3. Modeling studies


As a starting point for our SAR investigations selected
members of the three most potent hit clusters were docked into
the ABA binding site of the crystal structure of ABA-bound
AtRCAR11 and related phosphatase AtHAB1. Resulting poses
were then scored using the HYDE scoring function.36
Unfortunately, for hit cluster I no convincing docking poses
were identified. In contrast, for hit clusters II and III, reasonable
docking poses were obtained. The carbonyl group within the
indolinyl amide moiety of hit structure 8b forms an H-bond to
the conserved water molecule at the interface between
AtRCAR11 and AtHAB1 adopting the role of the carbonyl
Figure 3. EC50 correlation and HTS overview: For EC50 determination oxygen in ABA. This essential water molecule forms all four
hits were retested in 8 concentrations in the primary HTS assay format and in
absence of TaRCAR14 to deselect compounds with direct effects on possible H-bonds including H-bonds to the backbone oxygen of
phosphatase activity. All in all, 127 final hits were selected, showing no proline 115 and backbone amide of arginine 143 from
direct phosphatase inhibition. The inset shows DRCs of an exemplary final AtRCAR11 and NE1 of tryptophan 385 from AtHAB1, thus
hit. representing a key element in the protein–protein interaction

a c

b e

10 11 12a-i 7d-x

12a R = CH3, R = CH3


1 2
f h
12b R1 = CH3, R2 = i-Pr
12c R1 = c-Bu, R2 = CH3
12d R1 = CF3, R2 = CH3
12e R1 = furan-2-yl, R2 = CH3
12f R1 = 4-Cl-Ph, R2 = c-Pr g
12g R1 = CH3, R2 = furan-2-ylmethyl
12h R1 = H, R2 = furan-2-ylmethyl
12i R1 = c-Pr, R2 = CH3

Scheme 1. Synthesis of triazolyl acetamides 7; a) 1. (COCl)2,or SOCl2, N,N-DMF, 0 °C – r.t., 2. N2H4-H2O; b) EtOH, 80 °C; c) 1. aq. NaOH (1M), reflux, 2.
aq. HCl (1M); d) R3-Ph-NH2, DMA, 0 °C, r.t.; e) MeCN, K2CO3, reflux; f) MeCN, K2CO3, g) LiOH, H2O; h) R3-Ph-NH2, HOBt, EDC, (i-Pr)2NEt CH2Cl2.
a c

13 14 15

d d

h i e

17a-f 16
g j

f
17a R2 = CH3
17b R2 = Et
17c R2 = Ph

17d R2 = 2-Me-prop-1-yl
17e R2 = i-Pr
17f R2 = 2,2,2-trifluoroethyl

8e-z

Scheme 2. Synthesis of indolinylmethyl sulfonamides 8; a) formic acid (5 equiv.), toluene, reflux, 5h; b) conc. H2SO4, 0 °C – r.t., 18h; c) aq. HCl (1M),
MeOH, reflux, 4h; d) Et3N, DMAP, CH2Cl2, 0 °C – r.t., 4h; e) N2H4-H2O, CH2Cl2/MeOH, r.t., 12h; f) pyridine (3 equiv.), MeCN, 70 °C, 5h; g) formylation
failed; h) 1. N-Bromosuccinimide, CH2Cl2, r.t., 4h; 2. CuCN, CuI, N,N-DMF, reflux, 4d; i) H2 (40 bar), Raney-Ni/H2O, NH3 in MeOH, r.t.; j) NaBH3CN

between the RCARs and the corresponding phosphatases.


Furthermore, the sulfonamide moiety in 8b adopts the role of
A similar binding mode can be observed for the cyanophenyl
ABA’s terminal carboxylic acid group with one of the SO2-
aminoethyl sulfonamide 9b (from cluster III), with the nitrile
oxygens forming a second H-bond to a crucial water molecule
moiety of 9b forming an H-bond to the essential water molecule
(Figure 5a).
at the interface between AtRCAR11 and AtHAB1 adopting the
role of the carbonyl oxygen in ABA or the amide C=O group in
5b) 8b from hit cluster II (Figure 5b). Interestingly, the free electron
5a)
pairs serving as the H-bond donors in ABA and 9b, i.e. the
carbonyl oxygen of ABA’s cyclohexenone moiety and the nitrile
group in 9b, are not located at the same place due to differences
in binding geometry. The docking poses suggest in both cases
that there is additional space around the phenyl or heteroaryl
moiety for further exploration. In addition, there is some space
for small hydrophobic moieties at position R2 for compound
cluster II and at position R1 for III, respectively (cf. Fig. 4).
The beneficial impact of p-cyano aniline moieties on the
9b interaction of agonists with ABA receptor proteins has also been
8b observed in a structural analysis of the binding mode of
sulfonamide-based agonist cyanabactin with PYR1.37, 38 In good
correlation with X-ray analyses of cyano cyclopropyl analogues
of ABA (cf. 2a-c),23 this study showed that a 3-cyclopropyl-4-
Figure 5. Superposition of the best ranked docking poses of sulfonamides cyanoaniline moiety mimics the oxygen of ABA’s
8b and 9b (purple) with ABA (green) bound to AtRCAR11 and AtABI1 cyclohexenone engaging the key components required to stabilize
[special colour: O(red), S(yellow), N(blue), F(light green)]. the activated receptors.

a b

18 9i-z

c d e

19 20a-h

Scheme 3. Synthesis of phenylsulfonyl ethylenediamines 9 and 20; a) 2-aminoethyl amine, Et3N, MeCN, 50 °C, 4h; b) Arylsulfonylchloride (1 equiv.), (i-
Pr)2NEt, CH2Cl2, r.t., 20h; c) 2-bromoethyl amine, Et3N, CH2Cl2, 5h; d) KOH, H2O, toluene, 1.5h; e) aniline (1 equiv.), SiO2, H2O, 3d.
amines 17a-f in hand, a set of desired sulfonamides 8e-z was
prepared using pyridine in acetonitrile as the base.43
2.4. Chemistry
Substituted phenylsulfonyl ethylenediamines could be easily
Based on the results from high-throughput screening and prepared using two alternative approaches, (i) via nucleophilic
modeling studies versatile synthetic approaches were investigated aromatic substitution and subsequent formation of the
with emphasis on structural variations of key moieties crucial for sulfonamide or (ii) via formation of an aziridinyl sulfonamide,
good receptor binding. Substituted thiotriazolyl acetamides followed by ring opening using a substituted aniline.44 The first
which share key structural features with hit structures 7a-c (hit approach proved to be particularly suitable for 2-trifluoromethyl-
cluster I) have been described in earlier studies as active 4-fluorobenzonitrile, which reacted easily with 2-aminoethyl
ingredients against insect odorant sensory receptors. 39 We thus amine to afford intermediate 18. The desired 4-cyanophenyl
explored and optimized the established general synthetic aminoethylsulfonamides 9i-z were then obtained in good yields
approach to prepare a diverse set of thiotriazolyl acetamides 7. using the corresponding sulfonyl chloride with diisopropyl ethyl-
Starting from readily available carboxylic acids intermediate amine as base. In our hands, the second pathway turned out to
1,2,4-triazole-3-thiols 12a-i were obtained in 4 steps via oxalyl work well with anilines carrying less electron-withdrawing
chloride-mediated formation of hydrazides 10, followed by groups enabling us to explore a wider range of substituents. SiO2-
coupling with a suitable isothiocyanate and subsequent base- mediated ring opening of intermediate aziridinyl sulfonamides 19
mediated cyclization of the thiosemicarbazides 11. In our hands, in water afforded phenylsulfonyl ethylenediamines 20a-h as the
cyclizations worked best using aq. KOH or NaOH at elevated target compounds, thus broadening our SAR-study (Scheme 3).
temperatures affording desired intermediates 12a-i in 75 to 94%
yield. 1,2,4-Triazole-3-thiols 12a-i were then converted into
desired thiotriazolyl acetamides 7d-x via (i) direct base-mediated 2.5. SAR study
coupling of a freshly prepared halogen acetanilide, preferably
using K2CO3 or Cs2CO3 as base or via (ii) formation of a suitable All compounds prepared to explore the SAR of hit clusters I-II
triazolylthio acetic acid ester with subsequent ester cleavage and were tested for target affinity, as well as for beneficial effects in
amide formation. Following the second approach good yields vivo under drought stress conditions upon foliar application on
were obtained uing HOBt, EDC and Hünig’s base as amide crops. Within cluster III selected representatives were tested in
coupling reagents (Scheme 1).40 dedicated greenhouse trials. Wheat and Canola were chosen as
model plants for monocotyledonous and dicotyledonous species,
Indolinylmethyl sulfonamides which had emerged within hit
whereas in vitro tests were carried out for thiotriazolyl
cluster II of our high throughput screening were synthesized
acetamides 7 both at the ABA-receptor system AtRCAR11 and
based on two complementing routes starting from commercially
the orthologous ABA-receptor system in wheat, i.e. TaRCAR14.
available indoline 13 (Scheme 2). After introducing a N-formyl
As these results were nicely correlating, measurements for
group to protect the indoline nitrogen via formic acid-mediated
compounds 8 and 9 were only carried out on TaRCAR14.
formylation, the indolinylmethyl phthalimide 14 was formed via
Tscherniac-Einhorn reaction using concd sulfuric acid as Good receptor affinities could be observed for substituted
catalyst.41,42 Subsequent indoline N-deprotection, N-acylation thiotriazolyl acetamides carrying 4-cyano phenyl acetamide
and hydrazine-mediated phthalimide cleavage afforded the key moieties combined with small alkyl and haloalkyl substituents in
indolinylmethyl amine intermediates 17a-f, albeit in low overall the triazolyl unit, albeit with lower pI50 values than abscisic acid
yields (26-41%). Alternatively, indolinylmethyl amines 17 could 1 as the internal standard (Table 1). Other anilide substituents
be obtained with improved yield (up to 57%) using an optimized afforded weaker target affinities emphasizing the role of the 4-
four-step protocol via N-acylation of 13, followed by cyano group in receptor binding. In particular, target compounds
bromination, Cu-salt mediated cyanation and Ni-mediated carrying a 4-CN-phenyl acetamide moiety together with c-Pr (7x,
hydrogenation in NH3/MeOH under elevated pressure (40 bar). A Table 1, entry 21), c-Bu (7f, entry 3) and CF3-substituents (7j,
further approach via formylation and reductive amination did not entry 7) at position R1 showed good target affinities together with
afford desired key intermediates 17. With indolinylmethyl good efficacy against drought stress in vivo, particularly in wheat.

Table 1. SAR-results of selected thiotriazolyl acetamides 7

7d-v

Substituents[a] In vitro activity – AtABI1/TaABI1[b] In vivo efficacy vs. drought stress


[250 g/ha][c]

En- No. R1 R2 R3 Activity Activity pI50 Wheat[c] Canola[c]


try (TaRCAR14) (AtRCAR11)
[%] 5 M [%] 5 M

1 7d CH3 CH3 4-CN + ++ 4.2 ++ +

2 7e CH3 i-Pr 4-CN + - n.d. ++ ++

3 7f c-Bu CH3 4-CN +++ +++ 4.7 +++ ++


4 7g c-Bu CH3 2,4-F2 + - n.d. ++ 0

5 7h CF3 CH3 3-Cl - + n.d. + ++

6 7i CF3 CH3 2,4-F2 + + n.d. ++ ++

7 7j CF3 CH3 4-CN ++ +++ 4.8 +++ ++

8 7k Furan-2-yl CH3 3-CF3 + + n.d. + +

9 7l Thiophen-2-yl CH3 4-CN + + n.d. ++ +

10 7m 4-Cl-C6H4 c-Pr 4-CN n.d. ++ n.d. ++ +

11 7n 4-Cl-C6H4 c-Pr 2-Cl + ++ n.d. ++++ ++

12 7o CH3 Furan-2-ylmethyl 4-Cl + + n.d. ++ +

13 7p CH3 Furan-2-ylmethyl 4-CN +++ + n.d. ++ +

14 7q H Furan-2-ylmethyl 4-Cl +++ +++ 4.6 + +++

15 7r H Furan-2-ylmethyl 3-CN n.d. + n.d. + +

16 7s H Furan-2-ylmethyl 4-CN ++++ ++++ 5.3 ++ +++

17 7t H Furan-2-ylmethyl 3-Cl ++ ++ 4.5 + +

18 7u H Furan-2-ylmethyl 2,4-Cl2 ++ +++ 4.4 ++ ++

19 7v H Furan-2-ylmethyl 2-Cl - - n.d. + 0

20 7w c-Pr c-Pr 2-Cl - - n.d. + 0

21 7x c-Pr c-Pr 4-CN ++ + 4.2 +++ +

22 1 Abscisic acid ++++ ++++ 7.1 +++ +++


a
c-Pr = cyclopropyl, c-Bu = cyclobutyl, i-Pr = iso-propyl; b a final expert assessment of target affinity was made according to the following classification: “-”: <
30% ; “+” = 30% < activity < 50%; “++” = 50% < activity < 70%; “+++” = 70% < activity < 90%; “++++” = activity > 90%; c standard application rate for crop
protection greenhouse trials; d a final expert assessment of efficacies was made according to the following classification: “0” = no effect, “+” = slight beneficial
effect, “++” = significant beneficial effect, “+++” = strong beneficial effect against drought stress, “++++” = very strong beneficial effect superior to internal
standard ABA (comparative visual assessment of greenmass)

Whilst other variations at R1 such as methyl, furan-2-yl, the hydrogen at position R1 as also 4-Cl-, 3-Cl- and 2,4-Cl2-
thiophen-2-yl or 4-Cl-phenyl groups afforded weaker effects, anilides 7q, 7t, 7u exhibited good receptor affinities in wheat and
both in vitro and in vivo (cf. Table 1, entries 1, 8, 9, 10), Arabidopsis thaliana (cf. Table 1, entries 14, 17, 18). To further
thiotriazolyl acetamide 7n carrying a p-Cl-phenyl group at R1 validate our initial in vivo screening results further, we selected
together with a c-Pr group at R2 showed strong effects against 7n (with moderate target affinity, but surprisingly strong efficacy
drought stress in wheat. Hence, a focused variation of R2 showed in wheat) for advanced greenhouse trials (i.e. more replicates,
that furan-2ylmethyl groups had a beneficial impact on binding additional crops such as corn and barley, different corn varieties
affinity with 7s (entry 16) exhibiting the best target affinity of all and moderate stress levels). Accordingly, significant beneficial
thiotriazolyl acetamides investigated within our SAR study. effects against drought stress were observed in wheat compared
to untreated controls upon treatment with 7n (Figure 6).
Exploring hit cluster II gave a similar picture as observed for
the SAR study of cluster I. In line with results observed for
thiotriazolyl acetamides 7d-7x, receptor binding affinities of
indolinylmethyl sulfonamides 8e-8z varied strongly upon
changes in substituents R1 and R2 giving rise to a narrow SAR.
Halogen-substituted thiophen-2-yl moieties as sulfonyl
substituents R1 proved to afford the best binding affinities when
combined with small alkyl groups at R2. 5-Me-, 5-Br-, 5-Cl- and
4,5-dibromo substituents in the thiophen-2-ylsulfonamide moiety
were well tolerated since indolinylmethyl sulfonamides 8h, 8i,
8k, and 8r (Table 2, entries 4, 5, 7, and 8) showed good in vitro
activity, as well as promising in vivo efficacy against drought
stress in wheat. Interestingly, compounds 8e and 8g bearing 4-
Figure 6. Advanced drought stress trials with 7n (application rate: 25 g/ha
with replicates) in wheat and barley under moderate water stress conditions,
cyano- and 4-bromo phenyl groups as R1 substituents still gave
i.e. 42% damage for barley and 48% for wheat, respectively in comparison good results in vivo, but showed weaker target affinity.
with untreated, unstressed control plants. Furthermore, replacing halogen thiophenyl groups by 4-CF3- and
4-MeO-phenyl groups led to significantly weaker target affinities
combined with a loss of efficacy in vivo against drought stress
Interestingly, a broader variety of anilide substituents R3 was (cf. cpds. 8l, 8n, Table 2, entries 8, and 10). These observations
tolerated in combination with furan-2ylmethyl groups at R2 and are in line with results from dedicated modeling studies
indicating the privileged role of halogen-substituted thiophene Whilst small alkyl groups such as methyl (cf. 8r, Table 2,
moieties in receptor binding of indolinylmethyl sulfonamides 8. entry 14,), ethyl (e.g. 8h, entry 4) and iso-propyl (e.g. 8p, entry
Introducing a benzthiophen-2-yl group as part of the sulfonyl 12) were well tolerated as substituents R2, introducing haloalkyl
moiety also afforded weaker effects in vitro and in vivo (8m, or phenyl groups led to a loss of target affinity, and of in vivo
entry 9, table 2). efficacy against drought stress.

Table 2. SAR-results of indolinylmethyl sulfonamides 8. Table 3. SAR-results of phenylsulfonyl ethylenediamines.

Substituentsa In vitro
Substituents a
In vitro In vivo activity b
activity b efficacy TaABI1
TaABI1 vs. drought
stress En- No. R1 R2 R3 TaRCAR14b
try [%] [5 M]
En- No. R1 R2 TaRCAR14 b Wheatc
try [%] [5 M] [250 g/ha] d 1 9i CF3 2-Cl-C6H4 CN ++++

1 8e 4-CN-C6H4 Et ++ ++ 2 9j CF3 4-Cl-C6H4 CN ++++

2 8f thiophen-2-yl Et + + 3 9k CF3 4-Me-C6H4 CN ++++

3 8g 4-Br-C6H4 Et + ++ 4 9l H 4-Cl-C6H4 CN +++

4 8h 5-Cl-thiophen-2-yl Et +++ ++ 5 9m H 4-F-C6H4 CN +++

5 8i 4,5-Br2-thiophen-2-yl Et +++ + 6 9n H 3-F-C6H4 CN +

6 8j 4-Cl-C6H4 Et + + 7 9o MeO 4-F-C6H4 CN ++++

7 8k 5-Me-thiophen-2-yl Et +++ + 8 9p MeO 4-CH3-C6H4 CN ++++

8 8l 4-CF3-C6H4 Et + 0 9 9q Cl 4-Cl-C6H4 CN ++++

9 8m benzthiophen-2-yl Et + + 10 9r Cl 2-Cl-C6H4 CN ++++

10 8n 4-MeO-C6H4 Et + 0 11 9s Cl 4.H3CO-C6H4 CN ++++

11 8o thiophen-2-yl i-Pr ++ + 12 9t Me 4-H3CO-C6H4 CN ++++

12 8p 5-Br-thiophen-2-yl i-Pr +++ ++ 13 9u Me 2-Cl-C6H4 CN ++++

13 8q 5-Cl-thiophen-2-yl i-Pr ++ + 14 9v Me 3-Cl-C6H4 CN ++++

14 8r 5-Br-thiophen-2-yl CH3 +++ ++ 15 9w Me 4-Cl-C6H4 CN ++++

15 8s thiophen-2-yl F3C-CH2- + 0 16 9x Me 4-F-C6H4 CN ++++

16 8t 5-Br-thiophen-2-yl F3C-CH2- ++ ++ 17 9y Me 4-Me-C6H4 CN ++++

18 9z Me 1-methyl-1H- CN ++++
17 8u 5-Cl-thiophen-2-yl F3C-CH2- + +
imidazol-4-yl
18 8v 5-Me-thiophen-2-yl F3C-CH2- + 0
19 20a CF3 4-Cl-C6H4 F ++
19 8w 5-Me-thiophen-2-yl CH3 ++ +
20 20b CF3 4-CH3-C6H4 F +++
20 8x 5-Me-thiophen-2-yl C6H5 + 0
21 20c CF3 4-H3CO-C6H4 F +++
21 8y 5-Br-thiophen-2-yl c-Pr-CH2 +++ ++
22 20d CF3 2-F-C6H4 F +++
22 8z 5-Cl-thiophen-2-yl c-Pr-CH2 +++ +
23 20e CF3 4-Cl-C6H4 H 0
a
Me = CH3, Et = ethyl, i-Pr = iso-propyl, c-Pr = cyclopropyl; b a final expert
assessment of target affinity was made according to the following 24 20f CF3 4-H3CO-C6H4 Cl +
classification: “-”: < 30%; “+” = 30% < activity < 50%; “++” = 50% <
activity < 70%; “+++” = 70% < activity < 90%,; “++++” = activity > 90%; c 25 20g CF3 3-Cl-C6H4 Cl 0
standard application rate for crop protection greenhouse trials; d a final expert
assessment of efficacies was made according to the following classification: 26 20h CF3 4-F-C6H4 OMe 0
“0” = no effect; “+” = slight beneficial effect; “++” = significant beneficial a
effect; “+++” = strong beneficial effect against drought stress; “++++” = very Me = CH3, Et = ethyl, i-Pr = iso-propyl; b a final expert assessment of target
strong beneficial effect superior to internal standard ABA (comparative visual affinity was made according to the following classification: “-”: < 30% ; “+”
assessment of greenmass). = 30% < activity < 50%; “++” = 50% < activity < 70%; “+++” = 70% <
activity < 90%; “++++” = activity > 90%.
Interestingly, cyclopropylmethyl groups at R 2 afforded some leaf stage (BBCH10 – BBCH13). To ensure uniform water
of the best in vitro results observed for indolinylmethyl supply before commencement of drought stress, the potted plants
sulfonamides when combined with halogen-substituted thiophen- were supplied with the maximum amount of water immediately
2-ylsulfonyl moieties (cf. 8y, 8z, Table 2, entries 21, 22,). beforehand by dam irrigation and, after application, transferred
Compound 8y also exhibited promising efficacy against drought into plastic inserts in order to prevent subsequent, excessively
stress in wheat. rapid drying. The respective compounds, formulated in the form
of wettable powders (WP) or emulsion concentrates (EC), were
The detailed SAR study of phenylsulfonyl ethylenediamines 9
sprayed onto the green parts of the plants as an aqueous
and 20 (Table 3) afforded highly consistent results nicely
suspension at an equivalent water application rate of 600 l/ha
complementing results obtained for our largest hit cluster III. As
with addition of 0.2% wetting agent (agrotin). Substance
observed for thiotriazolyl acetamides 7d-7x, anilines 9 carrying a
application is followed immediately by drought stress treatment
p-CN group at R3 showed high target affinities (e.g. 9i, 9j, 9k,
of the plants. Drought stress was induced by gradual drying out
and 9s, entries 2, 3, 4, and 11), albeit a second substituent R1 in
under the following conditions: “day” = 14 hours with
the 3-position of the aniline appeared to be mandatory for strong
illumination at 26 °C; “night” = 10 hours without illumination at
in vitro activity. This observation is based on weaker binding
18 °C. The duration of the respective stress phases was guided
affinities of compounds 9l, 9m, and 9n bearing a hydrogen atom
mainly by the state of the untreated (= treated with blank
at R1. Furthermore, p-cyano aniline moieties with an additional
formulation but without test compound), stressed control plants
substituent at R1 such as CF3, methyl or methoxy exhibited a
and thus varied from wheat to canola. It was ended (by re-
good tolerance of changes in substitution of the sulfonamide
irrigating or transfer to a greenhouse with standard growth
moiety (i.e. R2). In particular, 2-Cl-, 3-Cl- and 4-Cl-phenyl
conditions) as soon as irreversible damage was observed on the
sulfonyl ethylene-diamines 9u, 9v and 9w gave consistently high
untreated, stressed control plants. In the case of dicotyledonous
target affinities. Remarkably, phenylsulfonyl ethylenediamines 9j
crops, for example canola, the duration of the drought stress
and 9k exhibited higher target affinities (pI50 7.32 and 7.27) than
phase varied between 3 and 5 days; in the case of
reference compound ABA (pI50 7.11). To validate our promising
monocotyledonous crops, for example wheat, it varied between 6
in vitro screening results, we selected 9j and 9k for advanced
and 10 days. The end of the stress phase was followed by an
greenhouse trials. Accordingly, beneficial effects against drought
approx. 5-7-day recovery phase, during which the plants were
stress were observed in wheat and canola compared to untreated
once again kept under good growth conditions in a greenhouse.
controls upon treatment, e.g. results for 9j: wheat(250 g/ha) ++,
In order to rule out any influence of the effects observed by any
canola(250 g/ha) +++; for 9k: wheat(250 g/ha) ++, canola(250
fungicidal action of the test compounds, it was additionally
g/ha) ++.
ensured that the tests proceeded without fungal infection and
3. Conclusions without infection pressure. After the recovery phase had ended,
the intensities of damage were rated visually in comparison to
We have identified and confirmed three unprecedented lead untreated, unstressed controls of the same age (in the case of
structures interacting with RCAR/(PYR/PYL) receptor proteins drought stress) or the same growth stage (in the case of cold
in wheat via high-throughput screening of our corporate stress). The intensity of damage was first recorded as a
compound library and subsequent dedicated SAR studies. percentage (100% = plants have died, 0% = like control plants).
Receptor protein TaRCAR14 and the PP2C phosphatase TaABI1 These values were then used to calculate the efficacy of the test
were isolated from wheat and chosen for the screening campaign. compounds (= percentage reduction in the intensity of damage as
Independently, a transgenic approach has demonstrated that ABA a result of substance application), and a final assessment of the
receptor overexpression in wheat improves grain production respective efficacy was made, i.e. “0” = no effect, “+” = slight
under drought underlining the potential for the use of new beneficial effect, “++” = significant beneficial effect, “+++” =
chemical lead structures that interact with relevant receptor strong beneficial effect against drought stress, “++++” = very
proteins in wheat.45 Whilst phenylsulfonyl ethylenediamines (e.g. strong beneficial effect superior to internal standard ABA.
9j and 9k) showed consistent strong target affinities in vitro on
the same level or even better than the plant hormone abscisic acid 4.1.2. In vitro - AtABI1-(AtRCAR11) / TaABI1-(TaRCAR14) -
(ABA), several thiotriazolyl acetamides (e.g. 7f and 7s) exhibited The assays described hereinafter utilize the inhibition of the
promising efficacy against drought stress mainly in wheat phosphatase AtABI1 via the co-regulator RCAR11/PYR1 from
combined with good target interaction. In all three compound Arabidopsis thaliana or the inhibition of the wheat TaABI1 via
classes particular structural features such as 4-cyanoaniline or 5- co-regulator RCAR14 from Triticum aestivum. Expression and
halogenthiophen-2-yl moieties were explored which had an purification of RCARs and PP2Cs was performed as described.10
essential impact on target affinity in vitro and in vivo efficacy. A concise overview of the correlation between PYR/PYL and
Our results demonstrate that p-cyano aniline moieties are RCAR numbering can be found in reference [46] by Grill et al. 46
effective bioisosters of the carbonyl group in ABA’s For the determination of activity, the dephosphorylation of 4-
cyclohexenone headgroup and in tetrahydroquinolinyl methylumbelliferyl phos-phate (MUP) was measured at 460 nm.
sulfonamides. Furthermore, indolinylmethyl sulfonamides 8 and The in vitro assay was conducted in Greiner 96-well, 384-well or
phenylsulfonyl ethylenediamines 9 represent two novel classes of 1536-well PS microplates, using two controls: a) dimethyl
sulfonamides interacting with RCAR/(PYR/PYL) receptor sulfoxide (DMSO) 0.5% (f.c.) and b) 5 M (f.c.) abscisic acid
proteins, thus nicely complementing earlier work in this field. (ABA). For high throughput screening 1 µg/ml ABI1 and 0,5
µg/ml RCAR11 were incubated with 100 µM MUP in reaction
4. Experimental buffer (50 mM Tris/HCl pH 7.8, 50 mM NaCl, 0.3 mM MnCl2,
0.01 % Tween, 0.01 % BSA) at 32°C for 80 minutes.
4.1. Biology Fluorescence intensity was detected by a BMG Labtech Pherastar
using a 340 nm excitation filter and a 460 nm emission filter. For
4.1.1. In vivo studies - Seeds of monocotyledonous and
follow up compound characterization the assay was conducted
dicotyledonous crop plants were laid out in sandy loam in wood-
with substance concentrations of the appropriate chemical test
fiber pots, covered with soil and cultivated in a greenhouse under
substances in a concentration range of 0.1 M to 100 M in a
good growth conditions. The test plants were treated at the first
solution of DMSO and water. The substance solution thus The resulting reaction mixture was heated under reflux
obtained, if necessary, was stirred with esterase from porcine conditions for 3-5 h. After cooling to room temperature, ethanol
liver (EC 3.1.1.1) at room temperature for 3 h and centrifuged at was removed carefully under reduced pressure, followed by
4000 rpm for 30 min. A total volume of 45 l was introduced addition of dichloromethane and thorough extraction. The
into each cavity of the microplate, having the following combined organic layer was dried over magnesium sulfate and
composition: 1. 5l of substance solution, i.e. a) DMSO 5% or b) concentrated under reduced pressure to afford the desired acid
abscisic acid solution or c) the corresponding example compound hydrazide.
of the general formula (I) dissolved in 5% DMSO. 2. 20l of
Step b: The requisite acid hydrazide (1.0 equiv) was dissolved
enzyme buffer mix, composed of a) 40% by vol. of enzyme
or suspended in ethanol (1.5 mL/mmol) at r.t. The appropriate
buffer [10 ml contain equal proportions by volume of 500 mM
isothiocyanate (1.0 equiv) was added in one portion. The reaction
Tris-HCl pH8, 500 mM NaCl, 3.33 mM MnCl2, 40 mM
flask was immersed in an oil bath and stirred at 80 °C until the
dithiothreitol (DTT)], b) 4% by vol. of AtABI1 or TaABI1
reaction was completed (2-3 h, TLC: DCE/MeOH 5:1). After
dilution (protein stock solution was diluted so as to give, after
cooling to room temperature, the product precipitated, which was
addition, a final concentration in the assay of 0.15 g
filtered off, washed with cold ethanol (1 mL/mmol) and dried
ABI1/well), c) 4% by vol. of AtRCAR11 or TaRCAR14 dilution
thoroughly under reduced pressure. The product was TLC pure
(enzyme stock was diluted so as to give, on addition of the
(average yield: 65-85%).
dilution to the enzyme buffer mix, a final concentration in the
assay of 0.30 g enzyme/well), d) 5% by vol. of Tween20 (1%), Step c: The appropriate thiosemicarbazide derivative (1.0
e) 47% by vol. H2O bi-dist. 3. 20l of substrate mix, composed equiv) was suspended thereafter in 1M aq. NaOH solution (1
of a) 10% by vol. of 500 mM Tris-HCl pH8, b) 10% by vol. of mL/mmol). The mixture was heated at reflux temperature until
500 mM NaCl, c) 10% by vol. of 3.33 mM MnCl 2, d) 5% by vol. the starting thiosemicarbazide derivative was no longer
of 25 mM MUP, 5% by vol. of Tween20 (1%), 60% by vol. of detectable by TLC (n-heptane/EtOAc 1:1 or CHCl3:MeOH 10:1).
H2O bi-dist. Enzyme buffer mix and substrate mix were made up After completion of the reaction, the mixture was cooled to 5 °C
5 minutes prior to the addition and warmed to a temperature of and neutralized with 1M aq. HCl solution to pH ~6-7. The
35°C. On completion of pipetting of all the solutions and on precipitated product was filtered off, washed with water (2×1
completion of mixing, the plate was incubated at 35°C for 20 mL/mmol) and dried in a vacuum desiccator over P2O5/KOH
minutes. Finally, a relative fluorescence measurement was made (average yield: 65-95%). Only in few cases the resulting 1,2,4-
at 35°C with a BMG Labtech "POLARstar Optima" microplate triazole-3-thiol had to be purified via flash chromatography
reader using a 340/10 nm excitation filter and a 460 nm emission (gradient EtOAc/n-heptane).
filter. An expert assessment of target affinity was made according
to the following classification: “-”: < 30% , “+” = 30% < activity Step d: DMA (3.3 mL) was cooled to 0 °C, then
< 50%, “++” = 50% < activity < 70%,, “+++” = 70% < activity < chloroacetylchloride (10 mmol, 1.0 equiv) was added and the
90%,, “++++” = activity > 90%. solution was stirred for 10 minutes at 0 °C. A solution of the
appropriate aniline (10 mmol, 1.0 equiv) in DMA (3.3 mL) was
4.2. Chemistry added dropwise and the reaction mixture was stirred for 2-4
hours at rt. The reaction was monitored by TLC (n-
4.2.1. General – All reagent-grade solvents and chemicals heptane:EtOAc 1:1). Water (12 mL) was added and the reaction
were purchased from standard commercial suppliers and used mixture became a thick crystalline slurry. The crude product was
without further purification. All non-aqueous reactions were isolated by filtration, washed with water (10 mL) and then dried
carried out under anhydrous conditions using dry solvents. at 50 °C. Purification via flash chromatography afforded the
Reactions were monitored by LC-MS or TLC carried out on 0.25 desired chloro-acetamide in average yield from 60 to 90%.
mm silica gel plates (60F-254). TLC plates were visualized using
UV light. Flash chromatography was carried out using Biotage Step e: The appropriate 1,2,4-triazole-3-thiol derivative (1.0
Isolera One systems with pre-packed column cartridges (Biotage equiv), acetonitrile (4 mL/mmol) and freshly powdered
KP-Sil [40+M] or KP-Sil [25+M]). The 1H NMR, 13C NMR and potassium carbonate (2.0 equiv) were placed in a 5 mL vial. The
19
F NMR spectroscopy data which are reported for the chemical appropriate chloro-acetanilide (1.07 equiv) was then added in one
examples described below (400 MHz for 1H NMR and 150 MHz portion at r.t. The reaction mixture was heated at 80 °C for 1-2 h
for 13C NMR and 375 MHz for 19F NMR, solvent: CDCl3, (TLC: CHCl3/MeOH 5:1). After cooling to r.t., the mixture was
CD3OD or d6-DMSO, internal standard: tetramethylsilane δ = concentrated, dissolved in dichloromethane (4.0 mL/mmol) and
0.00 ppm), were obtained on a Bruker instrument, and the signals extracted with water (2×2 mL/mmol). The phases were separated,
listed have the meanings given below: br = broad; s = singlet, d = and the organic layer was dried, filtered and evaporated.
doublet, t = triplet, dd = doublet of doublets, ddd = doublet of a Purification by flash chromatography or prep HPLC afforded the
doublet of doublets, m = multiplet, q = quartet, quint = quintet, desired S-alkylated 1,2,4-triazole derivative (av. yield: 30-70%).
sext = sextet, sept = septet, dq = doublet of quartets, dt = doublet
Step f: The appropriate 1,2,4-triazole-3-thiol derivative (1.0
of triplets. The abbreviations used for chemical groups or
equiv), acetonitrile (4 mL/mmol) and freshly powdered
dedicated hydrogen atoms are defined as follows: Me = CH3, Et
potassium carbonate (2.0 equiv) were placed in a 5 mL vial. The
= CH2CH3, t-Hex = C(CH3)2CH(CH3)2, t-Bu = C(CH3)3, n-Bu =
appropriate chloro- or bromo acetic acid ester (1.07 equiv) was
unbranched butyl, n-Pr = unbranched propyl, c-Hex =
then added in one portion at r.t. The reaction mixture was stirred
cyclohexyl, ArH = aromatic hydrogen, HetH = heteroaromatic
at r.t. for 3-4 h, then concentrated under reduced pressure,
hydrogen. In the case of diastereomeric mixtures, either the
dissolved in dichloromethane (4.0 mL/mmol) and extracted
significant signals for each of the diastereomers or the
thoroughly with water (3×2 mL/mmol). The combined organic
characteristic signal of the main diastereomer is/are reported.
layer was dried, filtered and evaporated under reduced pressure.
4.2.2. General Procedure A for the preparation of thio- Purification by flash chromatography or prep HPLC afforded the
triazolyl acetamides 7d-x. Step a: The appropriate carboxylic desired substituted 1,2,4-triazolylthio acetic acid ester derivative
acid ester (1.0 equiv.) was dissolved either in N,N-DMF of in (average yield: 65-90%).
ethanol (2 ml/mmol), and hydrazine hydrate (5 equiv) was added.
Step g: The appropriate 1,2,4-triazol ethyl ester (1.0 equiv) 4.2.2.5 N-(3-chlorophenyl)-2-{[4-methyl-5-trifluoromethyl-
was diluted with ethanol (2 mL), then water (1 mL) and solid 4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7h). Waxy colourless
NaOH or LiOH (1.0 equiv) were added. The solution was stirred solid; yield 31%; 1H NMR (400 MHz, CDCl3)  9.98 (s, 1H,
at r.t. for 2-4 h (TLC: DCM/EtOH 5:1). Ethanol was removed NH), 7.73 (s, 1H, ArH), 7.36 (d, J = 2 Hz, 1H, ArH), 7.20 (m,
under reduced pressure, the remaining residue was diluted with 1H, ArH), 7.08 (m, 1H, ArH), 4.00 (s, 2H, CH2), 3.70 (s, 3H,
water (ca. 2 mL) and acidified with 10% aq. HCl solution to pH CH3). LCMS (ESI, m/z): [M+H]+ 351.0.
~1. The precipitated carboxylic acid product was filtered off,
4.2.2.6. N-(2,4-difluorophenyl)-2-{[4-methyl-5-trifluorome-
washed with water (2×2 mL) and dried. The crude product was
thyl-4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7i). Waxy
purified by flash chromatography or preparative HPLC to give
colourless solid; yield 82%; 1H NMR (400 MHz, CDCl3)  10.12
the desired product (average yield: 45-75%).
(s, 1H, NH), 7.96 (s, 1H, ArH), 7.65 (d, J = 2 Hz, 1H, ArH), 7.44-
Step h: The appropriate 1,2,4-triazolylthio acetic acid (1 7.37 (m, 1H, ArH), 4.03 (s, 2H, CH2), 3.71 (s, 3H, CH3). LCMS
equiv.) was dissolved in dry dichloromethane (7 mL/mmol) and (ESI, m/z): [M+H]+ 353.2.
1-hydroxy-1H-benzotriazol (1.3 equiv.), as well as 1-ethyl-3-(3-
4.2.2.7. N-(4-cyanophenyl)-2-{[4-methyl-5-(4trifluoromethyl-
dimethylaminopropyl)carbodiimide (1.3 equiv.) were added. The
4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7j). Waxy colourless
resulting reaction mixture was stirred at r.t. for 10 min, followed
solid; yield 59%; 1H NMR (400 MHz, CDCl3)  10.47 (s, 1H,
by addition of the corresponding substituted aniline (1.1 equiv.).
NH), 7.71 (d, J = 2 Hz, 2H, ArH), 7.60 (d, J = 2 Hz, 2H, ArH),
After further stirring at r.t. for 10 min triethyl amine (3 equiv.)
4.01 (s, 2H, CH2), 3.71 (s, 3H, CH3). LCMS (ESI, m/z): [M+H]+
was added, and the reaction mixture was stirred at r.t. for 12 h.
342.1.
Thereafter, the reaction mixture was filtered, diluted with CH2Cl2
and water, followed by thorough extraction of the aq. layer with 4.2.2.8. N-(3-trifluoromethylphenyl)-2-{[4-methyl-5-(furan-2-
CH2Cl2 (3×10 mL/mmol). The combined organic layer was dried, yl)-4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7k). White solid;
filtered and evaporated under reduced pressure. Purification by yield 92%; 1H NMR (400 MHz, CDCl3)  10.70 (s, 1H, NH),
flash chromatography or prep HPLC afforded the desired 8.01 (s, 1H, HetH), 7.70 (m, 1H, ArH), 7.62 (m, 1H, HetH), 7.40
thiotriazolyl acetamide 7d-x. (m, 1H, ArH), 7.33 (m, 1H, ArH), 7.11 (m, 1H, HetH), 6.61 (m,
1H, HetH), 3.99 (s, 2H, CH2), 3.80 (s, 3H, CH3). LCMS (ESI,
4.2.2.1 N-(4-cyanophenyl)-2-{[4,5-dimethyl-4H-1,2,4-triazol-
m/z): [M+H]+ 383.1.
3-yl]sulfanyl}acetamide (7d). Waxy colourless solid; yield 72%;
1
H NMR (600 MHz, CDCl3)  11.23 (s, 1H, NH), 7.73 (d, J = 2 4.2.2.9. N-(4-cyanophenyl)-2-{[4-methyl-5-(thiophen-2-yl)-
Hz, 2H, ArH), 7.58 (d, J = 2 Hz, 2H, ArH), 3.91 (s, 2H, CH2), 4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7l). White solid; yield
3.50 (s, 3H, CH3), 2.47 (s, 3H, CH3). 13C NMR (150 MHz, 79%; 1H NMR (600 MHz, CDCl3)  11.08 (s, 1H, NH), 7.75 (d, J
CDCl3) 166.79, 153.09, 148.18, 143.04, 133.35, 119.19, 118.97, = 2 Hz, 2H, ArH), 7.58 (m, 3H, ArH+HetH), 7.48 (m, 1H, HetH),
105.19, 37.66, 30.03, 10.49. LCMS (ESI, m/z): [M+H]+ 288.3. 7.22 (m, 1H, HetH), 3.99 (s, 2H, CH2), 3.73 (s, 3H, CH3). 13C
HRMS (ESI, m/z): calcd. for C13H14N5OS, 288.0919 [M+H]+; NMR (150 MHz, CDCl3) 167.03, 152.94, 151.51, 142.34,
found 288.0924. 133.17, 129.07, 128.33, 128.04, 126.84, 119.75, 118.96, 107.03,
106.88, 36.61, 31.94. LCMS (ESI, m/z): [M+H] + 340.4. HRMS
4.2.2.2 N-(4-cyanophenyl)-2-{[4-(prop-2-yl)-5-methyl-4H-
(ESI, m/z): calcd. for C16H14N5OS2, 356.0640 [M+H]+; found
1,2,4-triazol-3-yl]sulfanyl}acetamide (7e). Waxy colourless
356.0635.
solid; yield 72%; 1H NMR (600 MHz, CDCl3)  11.23 (s, 1H,
NH), 7.74 (d, J = 2 Hz, 2H, ArH), 7.58 (d, J = 2 Hz, 2H, ArH), 4.2.2.10. N-(4-cyanophenyl)-2-{[4-cyclopropyl-5-(4-chloro-
4.42 (sept, 1H, i-Pr), 3.93 (s, 2H, CH2), 2.53 (s, 3H, CH3), 1.55 phenyl)-4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7m). White
(d, J = 3 Hz, 6H, CH3). 13C NMR (150 MHz, CDCl3) 167.43, solid; yield 81%; 1H NMR (400 MHz, CDCl3)  11.00 (s, 1H,
152.67, 150.67, 142.51, 133.15, 119.64, 119.04, 106.82, 48.94, NH), 7.74 (br. d, 4H, ArH), 7.59 (d, J = 2 Hz, 2H, ArH), 7.52 (d,
36.94, 21.23, 12.37. LCMS (ESI, m/z): [M+H] + 288.3. HRMS J = 2 Hz, 2H, ArH), 4.01 (s, 2H, CH2), 3.22 (m, 1H, c-Pr), 1.14
(ESI, m/z): calcd. for C15H18N5OS, 288.0919 [M+H]+; found (m, 2H, c-Pr), 0.79 (m, 2H, c-Pr). LCMS (ESI, m/z): [M+H]+
288.092 410.1.
4.2.2.3 N-(4-cyanophenyl)-2-{[4-methyl-5-cyclobutyl-4H- 4.2.2.11. N-(2-chlorophenyl)-2-{[4-cyclopropyl-5-(4-chloro-
1,2,4-triazol-3-yl]sulfanyl}acetamide (7f). Waxy colourless solid; phenyl)-4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7n). White
yield 77%; 1H NMR (600 MHz, CDCl3)  11.29 (s, 1H, NH), solid; yield 80%; 1H NMR (400 MHz, CDCl3)  9.92 (s, 1H,
7.76 (d, J = 2 Hz, 2H, ArH), 7.59 (d, J = 2 Hz, 2H, ArH), 3.97 (s, NH), 8.32 (m, 1H, ArH), 7.73 (d, J = 2 Hz, 2H, ArH), 7.49 (d, J =
2H, CH2), 3.56-3.51 (m, 1H, c-Bu), 3.42 (s, 3H, CH3), 2.56-2.50 2 Hz, 2H, ArH), 7.32 (m, 1H, ArH), 7.22 (m, 1H, ArH), 7.03 (m,
(m, 2H, c-Bu), 2.48-2.43 (m, 2H, c-Bu), 2.21-2.13 (m, 1H, c-Bu), 1H, ArH), 4.17 (s, 2H, CH2), 3.20 (m, 1H, c-Pr), 1.13 (m, 2H, c-
2.09-2.04 (m, 1H, c-Bu). 13C NMR (150 MHz, CDCl3) 167.36, Pr), 0.79 (m, 2H, c-Pr). LCMS (ESI, m/z): [M+H]+ 419.1.
159.36, 151.73, 142.49, 133.14, 119.77, 119.04, 106.90, 36.74,
30.28, 30.01, 26.58, 18.91. LCMS (ESI, m/z): [M+H] + 328.2. 4.2.2.12. N-(4-chlorophenyl)-2-{[4-(2-furylmethyl)-5-methyl-
HRMS (ESI, m/z): calcd. for C16H18N5OS, 328.1232 [M+H]+; 4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7o). White solid; yield
found 328.1227. 39%; 1H NMR (400 MHz, CDCl3)  10.22 (s, 1H, NH), 7.54 (d, J
= 3 Hz, 2H, ArH), 7.43 (m, 1H, HetH), 7.23 (d, J = 3 Hz, 2H,
4.2.2.4 N-(2,4-difluorophenyl)-2-{[4-methyl-5-cyclobutyl-4H- ArH), 6.46 (m, 1H, HetH), 6.37 (m, 1H, HetH), 5.05 (s, 2H,
1,2,4-triazol-3-yl]sulfanyl}acetamide (7g). Waxy colourless CH2), 3.92 (s, 2H, CH2), 2.49 (s, 3H, CH3). LCMS (ESI, m/z):
solid; yield 82%; 1H NMR (400 MHz, CDCl3)  10.42 (s, 1H, [M+H]+ 363.1.
NH), 7.96 (m, 1H, ArH), 7.65 (d, J = 2 Hz, 1H, ArH), 7.44-7.37
(m, 1H, ArH), 3.99 (s, 2H, CH2), 3.54-3.50 (m, 1H, c-Bu), 3.46 4.2.2.13. N-(4-cyanophenyl)-2-{[4-(2-furylmethyl)-5-methyl-
(s, 3H, CH3), 2.53-2.48 (m, 2H, c-Bu), 2.46-2.41 (m, 2H, c-Bu), 4H-1,2,4-triazol-3-yl]sulfanyl}acetamide (7p). White solid; yield
2.22-2.15 (m, 1H, c-Bu), 2.07-2.01 (m, 1H, c-Bu). LCMS (ESI, 46%; 1H NMR (400 MHz, CDCl3)  11.04 (s, 1H, NH), 7.72 (d, J
m/z): [M+H]+ 338.4. = 2 Hz, 2H, ArH), 7.59 (d, J = 2 Hz, 2H, ArH), 7.42 (m, 1H,
HetH), 6.45 (m, 1H, HetH), 6.37 (m, 1H, HetH), 5.05 (s, 2H,
CH2), 3.92 (s, 2H, CH2), 2.51 (s, 3H, CH3),. LCMS (ESI, m/z): 1H, ArH), 7.32 (m, 1H, ArH), 7.20 (m, 1H, ArH), 6.99 (m, 1H,
[M+H]+ 354.2. ArH), 4.07 (s, 2H, CH2), 3.05-3.01 (m, 1H, c-Pr), 1.99-1.94 (m,
1H, c-Pr), 1.21-1.11 (m, 6H, c-Pr), 1.09-1.04 (m, 2H, c-Pr).
4.2.2.14. N-(4-chlorophenyl)-2-{[4-(2-furylmethyl)-4H-1,2,4-
LCMS (ESI, m/z): [M+H]+ 349.2.
triazol-3-yl]sulfanyl}acetamide (7q). White solid; yield 68%; 1H
NMR (600 MHz, CDCl3)  10.46 (s, 1H, NH), 8.19 (s, 1H, 4.2.2.21. N-(4-cyanophenyl)-2-{[4,5-bis-cyclopropyl-4H-
HetH), 7.54 (d, J = 3 Hz, 2H, ArH), 7.42 (m, 1H, HetH), 7.23 (d, 1,2,4-triazol-3-yl]sulfanyl}acetamide (7x). White solid; yield
J = 3 Hz, 2H, ArH), 6.45 (m, 1H, HetH), 6.38 (m, 1H, HetH), 91%; 1H NMR (400 MHz, CDCl3)  11.11 (s, 1H, NH), 7.71 (d, J
5.05 (s, 2H, CH2), 3.94 (s, 2H, CH2). 13C NMR (150 MHz, = 3 Hz, 2H, ArH), 7.58 (d, J = 3 Hz, 2H, ArH), 3.91 (s, 2H, CH2),
CDCl3) 166.43, 151.18, 145.97, 144.54, 144.13, 136.84, 130.51, 3.06-3.03 (m, 1H, c-Pr), 2.02-1.98 (m, 1H, c-Pr), 1.23-1.18 (m,
128.99, 120.98, 110.93, 110.79, 41.63, 36.92. LCMS (ESI, m/z): 2H, c-Pr), 1.16-1.09 (m, 6H, c-Pr). LCMS (ESI, m/z): [M+H]+
[M+H]+ 349.2. HRMS (ESI, m/z): calcd. for C15H14N4O2SCl, 340.1.
349.0526 [M+H]+; found 349.0528.
4.2.3. General Procedure B for the preparation of
4.2.2.15. N-(3-cyanophenyl)-2-{[4-(2-furylmethyl)-4H-1,2,4- indolinylmethyl sulfonamides 8e-z. Step a: Indoline (2.00 g, 16.7
triazol-3-yl]sulfanyl}acetamide (7r). White solid; yield 73%; 1H mmol, 1 eq) was dissolved in abs. toluene (50 mL), followed by
NMR (600 MHz, CDCl3)  10.85 (s, 1H, NH), 8.21 (s, 1H, addition of formic acid (3.17 mL, 83.9 mmol, 5 eq). The resulting
HetH), 8.06 (s, 1H, ArH), 7.71 (m, 1H, ArH), 7.43 (m, 1H, reaction mixture was refluxed for 5h using a Dean-Stark-
HetH), 7.39-7.35 (m, 2H, ArH), 6.46 (m, 1H, HetH), 6.39 (m, apparatus. After cooling to room temperature, the solvent was
1H, HetH), 5.06 (s, 2H, CH2), 3.94 (s, 2H, CH2). 13C NMR (150 removed under reduced pressure and chloroform was added.
MHz, CDCl3) 166.88, 151.30, 145.87, 144.60, 144.21, 139.11, After washing with NaOH-soln. (2N) the organic layer was dried
129.69, 127.63, 123.86, 122.84, 118.56, 112.97, 110.96, 110.86, over sodium sulfate and concentrated under reduced pressure to
41.69, 36.78. LCMS (ESI, m/z): [M+H]+ 340.4. HRMS (ESI, afford 1-formyl indoline (1.81 g, 73%) which was used in the
m/z): calcd. for C16H14N5O2S, 340.0868 [M+H]+; found next step without further purification. 1H-NMR (400 MHz,
340.0865. CDCl3 , ppm) 8.94 (s, 1H), 7.15-7.26 (m, 3H), 7.00-7.08 (m,
1H), 4.05-4.14 (m, 2H), 3.14-3.22 (m, 2H).
4.2.2.16. N-(4-cyanophenyl)-2-{[4-(2-furylmethyl)-4H-1,2,4-
triazol-3-yl]sulfanyl}acetamide (7s). White solid; yield 70%; 1H Step b: 1-Formyl indoline (22.52 g, 0.15 mol, 1 eq) was
NMR (600 MHz, CDCl3)  11.01 (s, 1H, NH), 8.21 (s, 1H, dissolved in conc. sulfuric acid (200 mL) at 0 °C, and N-
HetH), 7.73 (d, J = 2 Hz, 2H, ArH), 7.59 (d, J = 2 Hz, 2H, ArH), (hydroxymethyl)phthalimide (27.11 g, 153.0 mol) was added
7.43 (m, 1H, HetH), 6.46 (m, 1H, HetH), 6.39 (m, 1H, HetH), portionwise. The resulting reaction mixture was stirred at 0 °C
5.05 (s, 2H, CH2), 3.93 (s, 2H, CH2). 13C NMR (150 MHz, for 6h and was allowed to warm to room temperature thereafter
CDCl3) 166.97, 151.18, 145.97, 144.61, 144.23, 133.17, 119.71, with stirring being continued continued for 12h. Then, the
118.96, 110.96, 110.88, 106.88, 41.70, 36.85. LCMS (ESI, m/z): reaction mixture was poured carefully on ice water. The
[M+H]+ 340.4. HRMS (ESI, m/z): calcd. for C16H14N5O2S, precipitate formed was filtered off and dissolved in a mixture of
340.0868 [M+H]+; found 340.0871. water and chloroform, followed by adjusting to pH10 by addition
of sodium carbonate. The aqueous layer was extracted thoroughly
4.2.2.17. N-(3-chlorophenyl)-2-{[4-(2-furylmethyl)-4H-1,2,4-
with chloroform, and the combined organic layer was dried over
triazol-3-yl]sulfanyl}acetamide (7t). White solid; yield 49%; 1H
Na2SO4 and concentrated under reduced pressure. The remaining
NMR (400 MHz, CDCl3)  10.48 (s, 1H, NH), 8.18 (s, 1H,
crude product was dissolved in a mixture of dichloromethane and
HetH), 8.06 (m, 1H, ArH), 7.43 (m, 1H, HetH), 7.38 (m, 1H,
acetone (1:1) and refluxed for 15 min. After cooling to room
ArH), 7.23 (m, 1H, ArH), 6.47 (m, 1H, HetH), 6.39 (m, 1H,
temperature, a colourless precipitate was formed which was
HetH), 5.05 (s, 2H, CH2), 3.99 (s, 2H, CH2). LCMS (ESI, m/z):
filtered off, washed with acetone and dried to afford 5-[(1,3-
[M+H]+ 349.2. HRMS (ESI, m/z): calcd. for C15H14N4O2SCl,
dioxoisoindolin-2-yl)methyl]-1-formyl indoline 14 (29.8 g, 57%).
349.0526 [M+H]+; found 349.0524. 1
H-NMR (400 MHz, CDCl3 , ppm) 8.89 (s, 1H), 7.82-7.87 (m,
4.2.2.18. N-(2,4-dichlorophenyl)-2-{[4-(2-furylmethyl)-4H- 2H), 7.69-7.74 (m, 2H), 7.26-7.34 (m, 2H), 7.08-7.10 (m, 1H),
1,2,4-triazol-3-yl]sulfanyl}acetamide (7u). White solid; yield 4.80 (s, 2H), 4.01-4.11 (m, 2H), 3.09-3.17 (m, 2H).
65%; 1H NMR (600 MHz, CDCl3)  10.05 (s, 1H, NH), 8.27 (d, J
Step c: 5-[(1,3-Dioxoisoindolin-2-yl)methyl]-1-formyl
= 2 Hz, 1H, ArH), 8.18 (s, 1H, HetH), 7.43 (m, 1H, HetH), 7.35
indoline 14 (29.8 g, 0.097 mol, 1 eq) was then dissolved in
(m, 1H, ArH), 7.21 (m, 1H, ArH), 6.44 (m, 1H, HetH), 6.38 (m,
methanol (300 mL) and HCl (5.5N soln. in iso-propanol, 50 mL)
1H, HetH), 5.03 (s, 2H, CH2), 4.08 (s, 2H, CH2). 13C NMR (150
was added. The reaction mixture was refluxed for 4 h and
MHz, CDCl3) 166.87, 150.40, 146.04, 144.68, 144.08, 133.86,
concentrated under reduced pressure after cooling to room
129.46, 129.00, 127.47. 124.72, 123.01, 110.90, 110.69, 41.51,
temperature. The residue was taken up with satd. sodium
36.45. LCMS (ESI, m/z): [M+H]+ 383.2. HRMS (ESI, m/z):
carbonate solution and CH2Cl2, followed by thorough extraction
calcd. for C15H13N4O2SCl2, 383.0136 [M+H]+; found 383.0130.
of the aqueous layer with CH2Cl2. The combined organic layer
4.2.2.19. N-(2-chlorophenyl)-2-{[4-(2-furylmethyl)-4H-1,2,4- was dried over Na2SO4 and concentrated under reduced pressure
triazol-3-yl]sulfanyl}acetamide (7v). White solid; yield 37%; 1H to afford 2-(indolin-5-ylmethyl)isoindoline-1,3-dione 15 (25.7 g,
NMR (400 MHz, d6-DMSO)  9.94 (s, 1H, NH), 7.79 (m, 1H, 90%) as colourless solid. 1H-NMR (400 MHz, CDCl3 , ppm)
ArH), 7.64 (s, 1H, HetH), 7.53 (m, 1H, HetH), 7.48 (m, 1H, 7.80-7.84 (m, 2H), 7.67-7.70 (m, 2H), 7.22-7.26 (m, 1H), 7.11-
HetH), 7.33 (m, 1H, ArH), 7.19 (m, 1H, ArH), 6.49 (m, 1H, 7.14 (m, 1H), 6.54-6.56 (d, 1H), 4.73 (s, 2H), 3.50-3.54 (m, 2H),
HetH), 6.44 (m, 1H, HetH), 5.22 (s, 2H, CH2), 4.12 (s, 2H, CH2). 2.96-3.00 (m, 2H).
LCMS (ESI, m/z): [M+H]+ 349.2. HRMS (ESI, m/z): calcd. for
Steps d,e: 2-(Indolin-5-ylmethyl)isoindoline-1,3-dione 15 (1.0
C15H14N4O2SCl, 349.0526 [M+H]+; found 349.0524.
g, 3.59 mmol, 1 eq), Et3N (0.75 mL, 5.39 mmol, 1.5 eq) and
4.2.2.20. N-(2-chlorophenyl)-2-{[4,5-bis-cyclopropyl-4H- DMAP (4 mg, 0.03 mmol, 0.01 eq) were dissolved in CH2Cl2 (25
1,2,4-triazol-3-yl]sulfanyl}acetamide (7w). White solid; yield mL) and cooled to 0 °C. Thereafter, the corresponding
77%; 1H NMR (400 MHz, CDCl3)  9.96 (s, 1H, NH), 8.26 (m, acylchloride (4.31 mmol, 1.2 eq), dissolved in CH2Cl2 (5 mL),
was added dropwise to the reaction mixture at 0 °C. Thereafter, temperature of 50 °C for 90 min. After cooling to room
the reaction mixture was allowed to warm to room temperature. temperature and full pressure release the reaction mixture was
Stirring was continued for 4h, then water was added, and the filtered and concentrated under reduced pressure. The remaining
aqueous layer was extracted with CH2Cl2. The combined organic residue was purified via column chromatography
layer was dried over Na2SO4 and concentrated under reduced (heptane/EtOAc, gradient 10:0 to 1:1) to afford the desired
pressure. The remaining crude product was dissolved in Et2O and intermediate 17. 17a-f (0.77 mmol, 1 eq) and pyridine (0.19 mL,
refluxed for 10 min. After cooling to room temperature, a 2.32 mmol, 3 eq) were dissolved in dry MeCN (10 mL) and the
colourless precipitate was formed which was filtered off, washed desired aryl or heteroaryl sulfonyl chloride (0.10 mL, 0.77 mmol,
with cold Et2O and dried to afford the desired acylated 2- 1 eq) was added. The resulting reaction mixture was stirred at 70
(indolin-5-ylmethyl)isoindoline-1,3-dione 16 which was then in °C for 5h. After cooling to room temperature CH2Cl2 (10 mL)
dissolved in step e in a mixture of CH2Cl2 and MeOH (5 was added. The organic layer was washed with HCl (10%, 10
mL/mmol, 1.25:1). Hydrazine hydrate (3.2 eq) was added mL), dried over Na2SO4 and concentrated under reduced
carefully, and the resulting reaction mixture was stirred at room pressure. The residue was purified via column chromatography
temperature for 12 h. The precipitate formed was filtered off and (heptane/EtOAc, gradient 10:0 to 1:1) to afford desired
washed with CH2Cl2/MeOH. The filtrate was diluted with CH2Cl2 indolinylmethyl sulfonamide 8e-z.
and washed with water. The combined organic layer was dried
4.2.3.1. 4-Cyano-N-[(1-propionyl-2,3-dihydro-1H-indol-5-yl)-
over Na2SO4 and concentrated under reduced pressure to afford
methyl]benzenesulfonamide (8e). White solid; yield 32%; 1H
the desired intermediate 17a-f.
NMR (400 MHz, CDCl3)  8.12-8.14 (m, 1H), 7.92-7.94 (m,
Step f: 17a-f (0.77 mmol, 1 eq) and pyridine (0.19 mL, 2.32 2H), 7.76-7.79 (m, 2H), 6.98-7.00 (m, 2H), 4.73 (m, 1H), 4.14-
mmol, 3 eq) were dissolved in abs. MeCN (10 mL) and the 4.16 (d, 2H), 4.04 (m, 2H), 3.10-3.12 (m, 2H), 2.44-2.46 (m, 2H),
desired aryl or heteroaryl sulfonyl chloride (0.10 mL, 0.77 mmol, 1.21-1.26 (m, 3H). LCMS (ESI, m/z): [M+H]+ 370.3. HRMS
1 eq) was added. The resulting reaction mixture was stirred at 70 (ESI, m/z): calcd. for C19H19N3O3S, 369.4384 [M+H]+; found
°C for 5h. After cooling to room temperature CH2Cl2 (10 mL) 369.4379.
was added. The organic layer was washed with HCl (10%, 10
4.2.3.2. N-[(1-Propionyl-2,3-dihydro-1H-indol-5-yl)-methyl]-
mL), dried over Na2SO4 and concentrated under reduced
thiophen-2-ylsulfonamide (8f). White solid; yield 32%; 1H NMR
pressure. The residue was purified via column chromatography
(400 MHz, CDCl3)  8.09-8.14 (m, 1H), 7.59-7.63 (m, 2H), 7.09-
(heptane/EtOAc, gradient 10:0 to 1:1) to afford desired
7.13 (m, 2H), 6.96-7.02 (m, 1H), 4.66-4.73 (m, 1H), 4.16-4.17
indolinylmethyl sulfonamide 8e-z.
(m, 2H), 4.02-4.08 (m, 2H), 3.14-3.18 (m, 2H), 2.41-2.46 (m,
General Procedure C for the preparation of indolinylmethyl 2H), 1.17-1.28 (m, 3H). LCMS (ESI, m/z): [M+H]+ 351.4.
sulfonamides 8e-z. Step d: Indoline (1 eq), Et3N (1.5 eq) and
4.2.3.3. 4-Bromo-N-[(1-propionyl-2,3-dihydro-1H-indol-5-yl)-
DMAP (0.01 eq) were dissolved in CH2Cl2 (2 mL/mmol) and
methyl]benzenesulfonamide (8g). White solid; yield 22%; 1H
cooled to 0 °C. Thereafter, the corresponding acylchloride (4.31
NMR (400 MHz, CDCl3)  8.13-8.15 (m, 1H), 7.70 (d, 2H), 7.64
mmol, 1.2 eq), dissolved in CH2Cl2 (5 mL), was added dropwise
(d, 2H), 6.94-7.00 (m, 2H), 4.67 (m, 1H), 4.09-4.12 (d, 2H),
to the reaction mixture at 0 °C. Thereafter, the reaction mixture
4.01-4.05 (m, 2H), 3.09-3.13 (m, 2H), 2.42-2.47 (m, 2H), 1.21-
was allowed to warm to room temperature. Stirring was
1.26 (m, 3H). LCMS (ESI, m/z): [M+H]+ 422.9/424.9, logp 2.62.
continued for 4h, then water was added, and the aqueous layer
was extracted with CH2Cl2. The combined organic layer was 4.2.3.4. 5-Chloro-N-[(1-propionyl-2,3-dihydro-1H-indol-5-
dried over Na2SO4 and concentrated under reduced pressure. The yl)-methyl]-thiophen-2-ylsulfonamide (8h). White solid; yield
residue was purified via column chromatography 29%; 1H NMR (400 MHz, CDCl3)  8.08-8.11 (m, 1H), 7.38 (m,
(heptane/EtOAc, gradient 10:0 to 1:1) to afford the acylated 1H), 7.13 (m, 1H), 6.94 (m, 1H), 6.90 (m, 1H), 4.70 (m, 1H),
indoline which was then dissolved in dichloromethane (1 eq in 4.20 (m, 2H), 4.03-4.09 (m, 2H), 3.15-3.20 (m, 2H), 2.44-2.40
5ml/mmol). (m, 2H), 1.21-1.26 (m, 3H). LCMS (ESI, m/z): [M+H]+ 384.9.
Step h: N-Bromosuccinimide was added (1.1 equiv) and the 4.2.3.5. 4,5-Dibromo-N-[(1-propionyl-2,3-dihydro-1H-indol-
resulting reaction mixture was stirred at r.t. for 90 min. Water 5-yl)-methyl]-thiophen-2-ylsulfonamide (8i). White solid; yield
was added, and the aqueous layer was extracted with CH2Cl2. 19%; 1H NMR (400 MHz, CDCl3)  8.18 (m, 1H), 7.27-7.30 (m,
The combined organic layer was dried over Na2SO4 and 1H), 6.98-7.06 (m, 2H), 4.80 (m, 1H), 4.21 (m, 2H), 4.04-4.09
concentrated under reduced pressure to afford the crude (m, 2H), 3.14-3.20 (m, 2H), 2.42-2.50 (m, 2H), 1.20-1.25 (m,
brominated acyl indoline which was used in the next step without 3H). LCMS (ESI, m/z): [M+2H]+ 508.7.
further purification. The corresponding brominated acyl indoline
(1 eq) was dissolved in abs. N,N-dimethylformamide (4 4.2.3.6. 4-Chloro-N-[(1-propionyl-2,3-dihydro-1H-indol-5-
ml/mmol), Copper(I)cyanide (2.5 eq) and copper(I)iodide (0.1 yl)-methyl]benzenesulfonamide (8j). White solid; yield 54%; 1H
eq) were added, and the resulting reaction mixture was stirred NMR (400 MHz, CDCl3)  8.12-8.14 (m, 1H), 7.78 (d, 2H), 7.48
under reflux conditions for 4 d. After cooling to r.t. ethylacetate (d, 2H), 6.95-7.02 (m, 2H), 4.61 (m, 1H), 4.11 (d, 2H), 4.02-4.07
and water were added, and the aqueous layer was extracted with (m, 2H), 3.09-3.14 (m, 2H), 2.41-2.47 (m, 2H), 1.21-1.26 (m,
ethyl acetate. The combined organic layer was dried over Na2SO4 3H). LCMS (ESI, m/z): [M+H]+ 379.0, logp 2.56.
and concentrated under reduced pressure. The remaining residue 4.2.3.7. 5-Methyl-N-[(1-propionyl-2,3-dihydro-1H-indol-5-
was purified via column chromatography (heptane/EtOAc, yl)-methyl]-thiophen-2-ylsulfonamide (8k). White solid; yield
gradient 10:0 to 1:1) to afford the desired acylated cyano 22%; 1H NMR (400 MHz, CDCl3)  8.13-8.15 (m, 1H), 7.44 (m,
indoline. 1H), 7.11 (m, 1H), 6.97-7.00 (m, 1H), 6.76 (m, 1H), 4.58 (br. t,
Steps i, f: The corresponding acylated cyano indoline (1 eq) 1H, NH), 4.14 (d, 2H), 4.02-4.08 (m, 2H), 3.14-3.18 (m, 2H),
was dissolved in a high pressure reactor in NH3/MeOH (20 2.55 (s, 3H), 2.47-2.41 (m, 2H), 1.21-1.25 (m, 3H). LCMS (ESI,
ml/mmol) and Raney-Nickel in H2O was added. Then, the reactor m/z): [M+H]+ 364.9, logp 2.38.
was closed, and a pressure of 40 bar was established by adding 4.2.3.8. 4-Trifluoromethyl-N-[(1-propionyl-2,3-dihydro-1H-
hydrogen gas. The reaction mixture was agitated at 40 bar at a indol-5-yl)-methyl]benzenesulfonamide (8l). White solid; yield
43%; 1H NMR (400 MHz, CDCl3)  8.11-8.14 (m, 1H), 7.97 (d, 4.2.3.18. 5-Methyl-N-[(1-isobutyryl-2,3-dihydro-1H-indol-5-
2H), 7.76 (d, 2H), 6.95-7.01 (m, 2H), 4.69 (m, 1H), 4.15 (d, 2H), yl)methyl]-thiophen-2-ylsulfonamide (8v). White solid; yield
4.02-4.07 (m, 2H), 3.08-3.13 (m, 2H), 2.41-2.47 (m, 2H), 1.21- 54%; 1H NMR (400 MHz, CDCl3)  8.16-8.18 (m, 1H), 7.43 (m,
1.25 (m, 3H). LCMS (ESI, m/z): [M+H]+ 412.1, logp 2.80. 1H), 7.13 (m, 1H), 6.97-7.00 (m, 1H), 6.75 (m, 1H), 4.56 (br. t,
1H, NH), 4.11-4.17 (m, 4H), 3.14-3.18 (m, 2H), 2.73-2.81 (m,
4.2.3.9. N-[(1-Propionyl-2,3-dihydro-1H-indol-5-yl)-methyl]-
1H), 2.55 (s, 3H), 1.23 (d, 6H). LCMS (ESI, m/z): [M+H]+ 379.0,
benzthiophen-2-ylsulfonamide (8m). White solid; yield 32%; 1H
logp 2.68.
NMR (400 MHz, CDCl3)  8.10-8.12 (m, 1H), 7.81-7.88 (m,
3H), 7.45-7.52 (m, 2H), 6.99-7.05 (m, 2H), 4.79 (m, 1H), 4.22 (d, 4.2.3.19. 5-Methyl-N-[(1-acetyl-2,3-dihydro-1H-indol-5-yl)-
2H), 3.93-3.97 (m, 2H), 2.99-3.05 (m, 2H), 2.38-2.44 (m, 2H), methyl]-thiophen-2-ylsulfonamide (8w). White solid; yield 27%;
1.19-1.24 (m, 3H). LCMS (ESI, m/z): [M+H]+ 401.0, logp 2.71. 1
H NMR (400 MHz, CDCl3)  8.09-8.11 (m, 1H), 7.44 (m, 1H),
7.13 (m, 1H), 6.97-7.00 (m, 1H), 6.76 (m, 1H), 4.61 (br. t, 1H,
4.2.3.10. 4-Methoxy-N-[(1-propionyl-2,3-dihydro-1H-indol-5-
NH), 4.13-4.17 (m, 2H), 4.04-4.07 (m, 2H), 3.14-3.19 (m, 2H),
yl)-methyl]benzenesulfonamide (8n). White solid; yield 43%; 1H
2.55 (s, 3H), 2.22 (s, 3H). LCMS (ESI, m/z): [M+H]+ 351.2, logp
NMR (400 MHz, CDCl3)  8.11-8.13 (m, 1H), 7.81 (d, 2H), 7.07
2.06.
(m, 1H), 7.00 (d, 2H), 6.92-6.96 (m, 1H), 4.47 (m, 1H), 4.01-4.08
(m, 4H), 3.88 (s, 3H), 3.12-3.17 (m, 2H), 2.42-2.47 (m, 2H), 4.2.3.20. 5-Methyl-N-[(1-benzoyl-2,3-dihydro-1H-indol-5-yl)-
1.21-1.26 (m, 3H). LCMS (ESI, m/z): [M+H]+ 375.1, logp 2.18. methyl]-thiophen-2-ylsulfonamide (8x). White solid; yield 25%;
1
H NMR (400 MHz, CDCl3)  7.52-7.56 (m, 2H), 7.44-7.47 (m,
4.2.3.11. N-[(1-Isobutyryl-2,3-dihydro-1H-indol-5-yl)methyl]-
3H), 7.31 (m, 2H), 7.17 (m, 1H), 6.99 (m, 1H), 6.77 (m, 1H),
thiophene-2-sulfonamide (8o). White solid; yield 39%; 1H NMR
4.58 (br. t, 1H, NH), 4.14-4.17 (m, 2H), 4.07-4.11 (m, 2H), 3.08-
(400 MHz, CDCl3)  8.15-8.17 (m, 1H), 7.60-7.64 (m, 2H), 7.09-
3.13 (m, 2H), 2.55 (s, 3H). LCMS (ESI, m/z): [M+H]+ 413.0,
7.13 (m, 2H), 6.96-7.00 (m, 1H), 4.66 (br. t, 1H, NH), 4.10-4.18
logp 2.77.
(m, 4H), 3.14-3.18 (m, 2H), 2.73-2.81 (m, 1H), 1.20-1.28 (m,
6H). LCMS (ESI, m/z): [M+H]+ 364.9, logp 2.40. 4.2.3.21. 5-bromo-N-{[1-(cyclopropylacetyl)-2,3-dihydro-1H-
indol-5-yl]methyl}thiophene-2-sulfonamide (8y). White solid;
4.2.3.12. 5-Bromo-N-[(1-isobutyryl-2,3-dihydro-1H-indol-5-
yield 27%; 1H NMR (400 MHz, CDCl3)  8.16-8.18 (m, 1H),
yl)-methyl]-thiophen-2-ylsulfonamide (8p). White solid; yield
7.35 (m, 1H), 7.06-7.09 (m, 2H), 7.00-7.04 (m, 1H), 4.74 (br. t,
29%; 1H NMR (400 MHz, CDCl3)  8.17-8.19 (m, 1H), 7.35 (m,
1H, NH), 4.17 (d, 2H), 4.01-4.05 (m, 2H), 3.13-3.17 (m, 2H),
1H), 7.09 (m, 1H), 7.05 (m, 1H), 7.01 (m, 1H), 4.67 (m, 1H),
2.36-2.39 (m, 2H), 1.16-1.21 (m, 1H), 0.59-0.65 (m, 2H), 0.18-
4.11-4.21 (m, 4H), 3.15-3.19 (m, 2H), 2.78 (m, 1H), 1.17-1.27
0.23 (m, 2H). LCMS (ESI, m/z): [M+H]+ 454.9, logp 2.97.
(m, 6H). LCMS (ESI, m/z): [M+H]+ 443.0, logp 3.00.
4.2.3.22. 5-chloro-N-{[1-(cyclopropylacetyl)-2,3-dihydro-1H-
4.2.3.13. 5-Chloro-N-[(1-isobutyryl-2,3-dihydro-1H-indol-5-
indol-5-yl]methyl}thiophene-2-sulfonamide (8z). White solid;
yl)-methyl]-thiophen-2-ylsulfonamide (8q). White solid; yield
yield 27%; 1H NMR (400 MHz, CDCl3)  8.17-8.19 (m, 1H),
37%; 1H NMR (400 MHz, CDCl3)  8.16-8.18 (m, 1H), 7.39 (m,
7.38 (m, 1H), 7.09 (m, 1H), 7.01 (m, 1H), 6.91 (m, 1H), 4.73 (br.
1H), 7.09 (m, 1H), 7.00 (m, 1H), 6.91 (m, 1H), 4.78 (m, 1H),
t, 1H, NH), 4.17 (d, 2H), 4.01-4.05 (m, 2H), 3.13-3.17 (m, 2H),
4.11-4.20 (m, 4H), 3.14-3.18 (m, 2H), 2.79 (m, 1H), 1.21-1.23 (d,
2.36-2.38 (m, 2H), 1.16-1.20 (m, 1H), 0.61-0.65 (m, 2H), 0.18-
6H). LCMS (ESI, m/z): [M+H]+ 399.1, logp 2.97.
0.23 (m, 2H). LCMS (ESI, m/z): [M+H]+ 411.1, logp 2.93.
4.2.3.14. 5-Bromo-N-[(1-methyl-2,3-dihydro-1H-indol-5-yl)-
4.2.4. General Procedure D for the preparation of
methyl]-thiophen-2-ylsulfonamide (8r). White solid; yield 45%;
phenylsulfonyl ethylenediamines 9i-z. 2-Aminoethyl amine (1
1
H NMR (400 MHz, CDCl3)  8.15-8.17 (m, 1H), 7.35 (m, 1H),
equiv.) and 2-trifluormethyl-4-fluoro benzonitrile (1 equiv.) were
7.09 (m, 1H), 7.05 (m, 1H), 7.01 (m, 1H), 4.77 (m, 1H), 4.11-
dissolved under argon in dry MeCN, followed by addition of
4.20 (m, 4H), 3.14-3.18 (m, 2H), 2.30 (s, 3H). LCMS (ESI, m/z):
Et3N (1.5 equiv). The resulting reaction mixture was stirred at 50
[M+H]+ 414.9, logp 2.29.
°C for 4h. After cooling to room temperature water and CH 2Cl2
4.2.3.15. N-[(1-(3,3,3-Trifluoropropionyl)-2,3-dihydro-1H- were added, followed by thorough extraction. The combined
indol-5-yl)methyl]-thiophene-2-sulfonamide (8s). White solid; organic layer was dried over Na2SO4, filtered and concentrated
yield 13%; 1H NMR (400 MHz, CDCl3)  8.13-8.15 (m, 1H), under reduced pressure to afford desired 4-[(2-aminoethyl)-
7.61-7.65 (m, 2H), 7.16 (m, 1H), 7.12 (m, 1H), 7.01 (m, 1H), amino]-2-(trifluormethyl)benzonitrile. 4-[(2-aminoethyl)-amino]-
4.67 (br. m, 1H, NH), 4.19 (m, 2H), 4.09-4.14 (m, 2H), 3.30-3.38 2-(trifluormethyl)benzonitrile (160 mg, 0.69 mmol) was
(m, 2H), 3.20-3.26 (m, 2H). LCMS (ESI, m/z): [M+H]+ 405.0, dissolved in a flame-dried round-bottomed flask in CH2Cl2 (20
logp 2.35. mL) under argon and diisopropylethyl amine (0.37 mL, 2.09
mmol) was added. After stirring at room temperature for 5 min a
4.2.3.16. 5-Bromo-N-[(1-(3,3,3-trifluoropropionyl)-2,3-di- solution of the corresponding aryl- or hetarylsulfonyl chloride
hydro-1H-indol-5-yl)-methyl]-thiophen-2-ylsulfonamide (8t). (0.69 mmol) in dry CH2Cl2 (5 mL) was added, and the resulting
White solid; yield 34%; 1H NMR (400 MHz, CDCl3)  8.16-8.18 reaction mixture was stirred at room temperature for 20h. H2O
(m, 1H), 7.35 (m, 1H), 7.12 (m, 1H), 7.02-7.07 (m, 2H), 4.70 (br. and CH2Cl2 were added, followed by thorough extraction. The
m, 1H, NH), 4.19 (m, 2H), 4.09-4.14 (m, 2H), 3.30-3.39 (m, 2H), combined organic layer was dried over Na2SO4, filtered,
3.20-3.26 (m, 2H). LCMS (ESI, m/z): [M+H]+ 482.8, logp 2.93. concentrated under reduced pressure and purified via column
4.2.3.17. 5-Chloro-N-[(1-(3,3,3-trifluoropropionyl)-2,3-di- chromatography (heptane/EtOAc, gradient 10:0 to 1:1) to afford
hydro-1H-indol-5-yl)-methyl]-thiophen-2-ylsulfonamide (8u). desired N-(2-{[4-cyano-3-(trifluormethyl)phenyl]amino}-ethyl)-
White solid; yield 33%; 1H NMR (400 MHz, CDCl3)  8.16-8.18 aryl-/hetaryl sulfonamide 9i-z.
(m, 1H), 7.40 (m, 1H), 7.14 (m, 1H), 7.06 (m, 1H), 6.93 (m, 1H), 4.2.4.1. N-(2-{[4-Cyano-3-(trifluormethyl)phenyl]amino}-
4.70 (br. m, 1H, NH), 4.19 (m, 2H), 4.09-4.14 (m, 2H), 3.29-3.37 ethyl)-2-chlorobenzene sulfonamide (9i). White solid; yield 49%;
(m, 2H), 3.21-3.26 (m, 2H). LCMS (ESI, m/z): [M+H]+ 438.9, 1
H NMR (400 MHz, d6-DMSO)  7.99-7.94 (m, 2H), 7.70 (d,
logp 2.90. 1H), 7.61-7.57 (m, 2H), 7.54-7.48 (m, 1H), 7.21-7.16 (m, 1H),
6.92 (m, 1H), 6.76 (m, 1H), 3.27-3.22 (m, 2H), 3.03-2.97 (m,
2H). LCMS (ESI, m/z): [M+H]+ 404.0, logp 2.74. HRMS (ESI, 6.43-6.38 (m, 2H), 3.82 (s, 3H), 3.17-3.12 (m, 2H), 2.87-2.81 (m,
m/z): calcd. for C16H13ClF3N3O2S, 403,0369 [M+H]+; found 2H), 2.30 (s, 3H).. LCMS (ESI, m/z): [M+H]+ 346.2, logp 2.83.
403,0307.
4.2.4.12. N-(2-{[4-Cyano-3-(methyl)phenyl]amino}-ethyl)-2-
4.2.4.2. N-(2-{[4-Cyano-3-(trifluormethyl)phenyl]amino}- chlorobenzene sulfonamide (9u). White solid; yield 77%; 1H
ethyl)-4-chlorobenzene sulfonamide (9j). White solid; yield 49%; NMR (400 MHz, d6-DMSO)  7.98-7.95 (m, 2H), 7.65-7.61 (m,
1
H NMR (400 MHz, d6-DMSO)  7.88 (br. t, 1H, NH), 7.78-7.76 2H), 7.53-7.49 (m, 1H), 7.35 (d, 1H), 6.52-6.49 (br. t, 1H, NH),
(d, 2H), 7.70 (d, 1H), 7.63 (d, 2H), 7.21-7.18 (br. t, 1H, NH), 6.42-6.36 (m, 2H), 3.17-3.12 (m, 2H), 3.00-2.95 (m, 2H), 2.30 (s,
6.96 (m, 1H), 6.79-6.76 (dd, 1H), 3.27-3.22 (m, 2H), 2.93-2.88 3H). LCMS (ESI, m/z): [M+H]+ 350.1, logp 3.01.
(m, 2H). LCMS (ESI, m/z): [M+H]+ 404.0, logp 2.91.
4.2.4.13. N-(2-{[4-Cyano-3-(methyl)phenyl]amino}-ethyl)-3-
4.2.4.3. N-(2-{[4-Cyanophenyl]amino}-ethyl)-4-chloroben- chlorobenzene sulfonamide (9v). White solid; yield 81%; 1H
zene sulfonamide (9l). White solid; yield 91%; 1H NMR (400 NMR (400 MHz, d6-DMSO)  7.92 (br. t, 1H, NH), 7.79-7.70
MHz, d6-DMSO)  7.87-7.74 (br. m, 3H, 2H+NH), 7.65 (d, 2H), (m, 3H), 7.63-7.60 (m, 1H), 7.36 (d, 1H), 6.55-6.52 (br. t, 1H,
7.42 (d, 2H), 6.65-6.62 (br. t, 1H, NH), 6.55 (d, 2H), 3.19-3.14 NH), 6.45-6.40 (m, 2H), 3.19-3.14 (m, 2H), 2.94-2.89 (m, 2H),
(m, 2H), 2.93-2.87 (m, 2H). LCMS (ESI, m/z): [M+H]+ 336.1, 2.30 (s, 3H). LCMS (ESI, m/z): [M+H]+ 350.1, logp 3.19.
logp 2.98.
4.2.4.14. N-(2-{[4-Cyano-3-(methyl)phenyl]amino}-ethyl)-4-
4.2.4.4. N-(2-{[4-Cyanophenyl]amino}-ethyl)-4-fluoroben- chlorobenzene sulfonamide (9w). White solid; yield 81%; 1H
zene sulfonamide (9m). White solid; yield 87%; 1H NMR (400 NMR (400 MHz, d6-DMSO)  7.84 (br. m, 1H, NH), 7.78 (d,
MHz, d6-DMSO)  7.87-7.80 (br. m, 3H, 2H+NH), 7.44-7.38 (m, 2H), 7.65 (d, 2H), 7.36 (d, 1H), 6.53-6.50 (br. t, 1H, NH), 6.44-
4H), 6.67-6.64 (br. t, 1H, NH), 6.55 (d, 2H), 3.19-3.14 (m, 2H), 6.38 (m, 2H), 3.18-3.13 (m, 2H), 2.93-2.87 (m, 2H), 2.30 (s, 3H).
2.92-2.85 (m, 2H). LCMS (ESI, m/z): [M+H]+ 320.1, logp 2.51. LCMS (ESI, m/z): [M+H]+ 350.1, logp 3.19.
4.2.4.5. N-(2-{[4-Cyanophenyl]amino}-ethyl)-3-fluoroben- 4.2.4.15. N-(2-{[4-Cyano-3-(methyl)phenyl]amino}-ethyl)-4-
zene sulfonamide (9n). White solid; yield 81%; 1H NMR (400 fluorbenzene sulfonamide (9x). White solid; yield 84%; 1H NMR
MHz, d6-DMSO)  7.89 (m, 1H), 7.69-7.65 (m, 2H), 7.59 (m, (400 MHz, d6-DMSO)  7.87-7.84 (m, 2H), 7.79-7.76 (m, 1H),
1H), 7.52 (m, 1H), 7.42 (d, 1H), 6.67-6.64 (br. t, 1H, NH), 6.56 7.44-7.38 (m, 2H), 7.37-7.34 (m, 1H), 6.54-6.51 (br. t, 1H, NH),
(d, 2H), 3.20-3.15 (m, 2H), 2.93-2.88 (m, 2H). LCMS (ESI, m/z): 6.43-6.39 (m, 2H), 3.18-3.13 (m, 2H), 2.89-2.84 (m, 2H), 2.30 (s,
[M+H]+ 320.1, logp 2.66. 3H).. LCMS (ESI, m/z): [M+H]+ 334.2, logp 2.89
4.2.4.6. N-(2-{[4-Cyano-3-(methoxy)phenyl]amino}-ethyl)-4- 4.2.4.16. N-(2-{[4-Cyano-3-(methyl)phenyl]amino}-ethyl)-4-
fluorobenzene sulfonamide (9o). White solid; yield 62%; 1H methylbenzene sulfonamide (9y). White solid; yield 77%; 1H
NMR (400 MHz, d6-DMSO)  7.88-7.82 (m, 3H), 7.45-7.38 (m, NMR (400 MHz, d6-DMSO)  7.69-7.63 (m, 3H), 7.38-7.34 (m,
2H), 7.28 (m, 1H), 7.51 (d, 1H), 6.67 (br. t, 1H, NH), 6.19-6.13 3H), 6.54-6.51 (br. t, 1H, NH), 6.44-6.38 (m, 2H), 3.18-3.12 (m,
(m, 2H), 3.80 (s, 3H), 3.22-3.17 (m, 2H), 2.92-2.87 (m, 2H). 2H), 2.86-2.81 (m, 2H), 2.37 (s, 3H), 2.30 (s, 3H). LCMS (ESI,
LCMS (ESI, m/z): [M+H]+ 350.0, logp 2.13. m/z): [M+H]+ 330.2, logp 3.19.
4.2.4.7. N-(2-{[4-Cyano-3-(methoxy)phenyl]amino}-ethyl)-4- 4.2.4.17. N-[2-(4-Cyano-3-methylanilino)ethyl]-1-methyl-1H-
methylbenzene sulfonamide (9p). White solid; yield 78%; 1H imidazole-4-sulfonamide (9z). White solid; yield 31%; 1H NMR
NMR (400 MHz, d6-DMSO)  7.69-7.66 (m, 3H), 7.38 (d, 2H), (400 MHz, d6-DMSO)  7.78 (s, 1H), 7.73 (s, 1H), 7.53 (br. m,
7.28 (d, 1H), 6.66 (br. t, 1H, NH), 6.18-6.13 (m, 2H), 3.79 (s, 1H, NH), 7.38 (d, 1H), 6.57-6.53 (br. t, 1H, NH), 6.48 (d, 1H),
3H), 3.21-3.16 (m, 2H), 2.90-2.84 (m, 2H), 2.37 (s, 3H). LCMS 6.44-6.40 (dd, 1H), 3.68 (s, 3H), 3.20-3.14 (m, 2H), 2.99-2.94
(ESI, m/z): [M+H]+ 346.0, logp 2.26. (m, 2H). LCMS (ESI, m/z): [M+H]+ 319.4, logp 1.88.
4.2.4.8. N-(2-{[4-Cyano-3-(chloro)phenyl]amino}-ethyl)-4- 4.2.5. Preparation of phenylsulfonyl ethylenediamines 20a-h
chlorobenzene sulfonamide (9q). White solid; yield 45%; 1H via sulfonyl aziridines – all cpds were prepared following the
NMR (400 MHz, d6-DMSO)  7.99-7.94 (m, 2H), 7.86 (m, 1H), protocols outlined for the synthesis of 20a,b:
7.78 (d, 2H), 7.51 (d, 1H), 7.01-6.96 (m, 1H), 6.69 (m, 1H), 6.55
4.2.5.1. N-(2-{[4-Fluoro-3-(trifluormethyl)phenyl]amino}-
(m, 1H), 3.22-3.17 (m, 2H), 2.92-2.87 (m, 2H). LCMS (ESI,
ethyl)-4-chlorobenzene sulfonamide 20a. 2-Bromoethyl amine (1
m/z): [M+H]+ 369.9, logp 2.71.
equiv.) and 4-Chlorphenylsulfonylchloride (1 equiv.) were
4.2.4.9. N-(2-{[4-Cyano-3-(chloro)phenyl]amino}-ethyl)-2- dissolved in dry CH2Cl2 under argon and triethyl amine (1.5
chlorobenzene sulfonamide (9r). White solid; yield 38%; 1H equiv) was added. The resulting reaction mixture was stirred at
NMR (400 MHz, d6-DMSO)  7.96 (m, 2H), 7.62 (m, 2H), 7.54- room temperature for 5 h. Then, water and CH 2Cl2 were added,
7.49 (m, 2H), 6.96 (m, 1H, NH), 6.66 (d, 1H), 6.52 (m, 1H), followed by thorough extraction of the aqueous layer with
3.20-3.16 (m, 2H), 3.00-2.96 (m, 2H). LCMS (ESI, m/z): CH2Cl2. The combined organic layer was dried over Na2SO4 and
[M+H]+ 369.9, logp 2.56. concentrated under reduced pressure. The residue was purified
via column chromatography (heptane/EtOAc, gradient 10:0 to
4.2.4.10. N-(2-{[4-Cyano-3-(chloro)phenyl]amino}-ethyl)-4-
1:1) to afford desired N-(2-bromoethyl)-4-chlorobenzene
methoxybenzene sulfonamide (9s). White solid; yield 51%; 1H
sulfonamide as intermediate which was dissolved in toluene (15
NMR (400 MHz, d6-DMSO)  7.72 (d, 2H), 7.57 (br. t, 1H, NH),
mL/mmol). Thereafter, a solution of KOH (3 equiv) in water (1
7.51 (d, 1H), 7.09 (d, 2H), 6.98 (br. t, 1H, NH), 6.69 (d, 1H),
mL/mmol) was added. The resulting reaction mixture was stirred
6.54 (dd, 1H), 3.83 (s, 3H), 3.21-3.15 (m, 2H), 2.85-2.82 (m,
at room temperature for 90 min. After complete conversion,
2H). LCMS (ESI, m/z): [M+H]+ 365.1, logp 2.38.
water and ethyl acetate were added, followed by thorough
4.2.4.11. N-(2-{[4-Cyano-3-(methyl)phenyl]amino}-ethyl)-4- extraction of the aqueous layer with EtOAc. The combined
methoxybenzene sulfonamide (9t). White solid; yield 93%; 1H organic layer was dried over Na2SO4 and concentrated under
NMR (400 MHz, d6-DMSO)  7.73 (d, 2H), 7.61-7.54 (br. m, reduced pressure. The residue was purified via column
1H, NH), 7.36 (d, 1H), 7.10 (d, 2H), 6.54-6.51 (br. t, 1H, NH), chromatography (heptane/EtOAc, gradient) to afford desired 1-
[(4-chlorophenyl)sulfonyl]aziridine as intermediate. 1-[(4-
chlorophenyl)sulfonyl]aziridine (150 mg, 0.69 mmol), 4-fluoro- 70%; 1H NMR (400 MHz, d6-DMSO)  8.01 (br. t, 1H, NH),
3-trifluoromethyl aniline (123 mg, 0.69 mmol) and SiO2 (100 7.82-7.78 (m, 1H), 7.70-7.65 (m, 1H), 7.42-7.35 (m, 2H), 7.20-
mg, 1.66 mmol) were then suspended in H2O (2 mL), and the 7.16 (m, 1H), 6.78-6.73 (m, 2H), 6.01-5.98 (br. t, 1H, NH), 3.16-
resulting reaction mixture was treated with ultra sound for 90 min 3.10 (m, 2H), 3.02-2.97 (m, 2H). LCMS (ESI, m/z): [M+2H]+
and was stirred at room temperature for 20h. Upon addition of 382.2, logp 3.07.
water and ethyl acetate the aqueous layer was extracted
4.2.5.5. N-(2-{[3-(Trifluormethyl)phenyl]amino}-ethyl)-4-
thoroughly with ethyl acetate. The combined organic layer was
chlorobenzene sulfonamide (20e). White solid; yield 63%; 1H
dried over Na2SO4 and concentrated under reduced pressure.
NMR (400 MHz, d6-DMSO)  7.86 (m, 1H), 7.81-7.77 (m, 2H),
Purification of the remaining residue via column chromatography
7.65-7.61 (m, 2H), 7.28-7.24 (m, 1H), 6.81 (m, 1H), 6.74 (m,
(heptane/EtOAc, gradient) afforded N-(2-{[4-fluoro-3-(tri-
2H), 6.12-6.08 (m, 1H), 3.17-3.12 (m, 2H), 2.91-2.87 (m, 2H).
fluormethyl)phenyl]amino}ethyl)-4-chlorobenzene sulfonamide
LCMS (ESI, m/z): [M+H]+ 379.7, logp 3.48.
20a (210 mg, 77 %) as colourless solid. 1H-NMR (400 MHz,
CDCl3 , ppm) 7.80 (d, 2H), 7.48 (d, 2H), 7.00 (m, 1H), 6.70- 4.2.5.6. N-(2-{[4-Chloro-3-(trifluormethyl)phenyl]amino}-
6.64 (m, 2H), 4.92 (br. t, 1H, NH), 4.02 (br. m, 1H, NH), 6.79 ethyl)-4-methoxybenzene sulfonamide (20f). White solid; yield
(m, 1H), 3.30-3.25 (m, 2H), 3.22-3.17 (m, 2H). LCMS (ESI, 64%; 1H NMR (400 MHz, d6-DMSO)  7.72 (d, 2H), 7.56 (m,
m/z): [M+H]+ 396,7, logp 2.98. HRMS (ESI, m/z): calcd. for 1H), 7.32 (m, 1H), 7.09 (d, 2H), 6.90 (m, 1H), 6.73 (m, 1H),
C15H13ClF4N2O2S, 396,0322 [M+H]+; found 396,0341. 6.30-6.25 (m, 1H), 3.82 (s, 3H), 3.16-3.11 (m, 2H), 2.85-2.80 (m,
2H). LCMS (ESI, m/z): [M+2H]+ 410.9, logp 3.33.
4.2.5.2. N-(2-{[4-Fluoro-3-(trifluormethyl)phenyl]amino}-
ethyl)-4-methylbenzolsulfonamide 20b. 2-Bromoethyl amine (1 4.2.5.7. N-(2-{[4-Chloro-3-(trifluormethyl)phenyl]amino}-
equiv.) and 4-methylphenylsulfonylchloride (1 equiv.) were ethyl)-3-chlorobenzene sulfonamide (20g). White solid; yield
dissolved in dry CH2Cl2 under argon and triethyl amine (1.5 42%; 1H NMR (400 MHz, d6-DMSO)  7.89 (m, 1H), 7.78 (m,
equiv) was added. The resulting reaction mixture was stirred at 1H), 7.74 (m, 1H), 7.70 (m, 1H), 7.60 (m, 1H), 7.33-7.30 (m,
room temperature for 5 h. Then, water and CH 2Cl2 were added, 1H), 6.90 (m, 1H), 6.73 (m, 1H), 6.31-6.26 (m, 1H), 3.17-3.12
followed by thorough extraction of the aqueous layer with (m, 2H), 2.94-2.88 (m, 2H). LCMS (ESI, m/z): [M+2H]+ 414.5,
CH2Cl2. The combined organic layer was dried over Na2SO4 and logp 3.75.
concentrated under reduced pressure. The residue was purified
via column chromatography (heptane/EtOAc, gradient 10:0 to 4.2.5.8. N-(2-{[4-Methoxy-3-(trifluormethyl)phenyl]amino}-
1:1) to afford desired N-(2-bromoethyl)-4-methylbenzene ethyl)-4-fluorobenzene sulfonamide (20h). White solid; yield
sulfonamide (1.00 g, 3.54 mmol) as intermediate which was 61%; 1H NMR (400 MHz, d6-DMSO)  7.87-8.83 (m, 2H), 7.76
dissolved in toluene (100 mL). Thereafter, a solution of KOH (br. m, 1H, NH), 7.42-7.38 (m, 2H), 7.03-7.00 (m, 1H), 6.74-6.69
(1.19 g, 21.27 mmol) in water (15 mL) was added. The resulting (m, 2H), 5.57-5.54 (br. t, 1H, NH), 3.74 (s, 2H), 3.11-3.05 (m,
reaction mixture was stirred at room temperature for 90 min. 2H), 2.90-2.85 (m, 2H). LCMS (ESI, m/z): [M+H]+ 393.2, logp
After complete conversion, water and ethyl acetate were added, 2.90.
followed by thorough extraction of the aqueous layer with 4.3. Modelling studies
EtOAc. The combined organic layer was dried over Na2SO4 and
concentrated under reduced pressure. The residue was purified Modelling was done using the crystal structure of ABA-bound
via column chromatography (heptane/EtOAc, gradient) to afford AtRCAR11 and related phosphatase AtHAB1 (3QN1 – F.
desired 1-[(4-methylphenyl)sulfonyl]aziridine (781 mg, 98 %) as Dupeux et al.).47 Docking poses were generated using SeeSAR
intermediate. 1H-NMR (400 MHz, CDCl3 , ppm) 7.85 (d, 2H), from BioSolveIT and scored using the HYDE scoring function.36
7.37 (d, 1H), 2.45 (s, 3H), 2.42-2.30 (m, 4H). 1-[(4-
methylphenyl)sulfonyl]aziridine (120 mg, 0.61 mmol), 4-fluoro- Acknowledgments
3-trifluoromethyl aniline (109 mg, 0.61 mmol) and SiO2 (110
mg, 1.83 mmol) were then suspended in H2O (2 mL), and the We would like to thank Matthias Jank, Susanne Ries, Gudrun
resulting reaction mixture was stirred at room temperature for Fey, Peter Zöllner and Martin Annau for valuable analytical
20h. Upon addition of water and ethyl acetate the aqueous layer support. Furthermore, we would like to thank David Barber for
was extracted thoroughly with ethyl acetate. The combined fruitful discussions during the preparation of the manuscript.
organic layer was dried over Na2SO4 and concentrated under
reduced pressure. Purification of the remaining residue via References
column chromatography (heptane/EtOAc, gradient) afforded N-
(2-{[4-fluoro-3-(trifluormethyl)phenyl]amino}ethyl)-4-methyl- 1. D. Li, T. T. M. Luong, W.-J. Dan, Y. Ren, H. X. Nien, A.-L.
benzene sulfonamide 20b (70 mg, 31 %) as colourless solid. 1H- Zhang, J.-M. Gao, Bioorg. Med. Chem. 2018, 26, 386.
2. S. Y. Salehi-Lisar, H. Bakhshayeshan-Agdam in Drought Stress
NMR (400 MHz, CDCl3 , ppm) 7.75 (d, 2H), 7.31 (d, 2H), 7.00 Tolerance in Plants, Vol 1; Physiology and Biochemistry (Eds.:
(m, 1H), 6.68-6.64 (m, 2H), 4.79 (br. t, 1H, NH), 4.05 (br. m, 1H, M. A. Hossain, S. H. Wani, S. Bhattacharjee, D. J. Burritt, L.-S. P.
NH), 6.79 (m, 1H), 3.25-3.16 (m, 4H), 2.43 (s, 3H). LCMS (ESI, Tran), Springer-Verlag, Berlin Heidelberg, 2016, pp. 1-16.
m/z): [M+H]+ 396,7, logp 2.98. HRMS (ESI, m/z): calcd. for 3. D. S. Battisti, R. L. Naylor, Science 2009, 323, 240;
C16H16F4N2O2S, 376,0869 [M+H]+; found 376,0856. 4. H. Campos, M. Cooper, J. E. Habben, G. O. Edmeades, J. R.
Schussler, Field Crops Res 2004, 90, 19.
4.2.5.3. N-(2-{[4-Fluoro-3-(trifluormethyl)phenyl]amino}- 5. K. S. Nemali, C. Bonin, F. G. Dohlemann, M. Stephens, W. R.
ethyl)-4-Methoxybenzene sulfonamide (20c). White solid; yield Reeves, D. E. Nelson, P. Castiglioni, J. E. Whitsel, B. Sammons,
R. A. Silady, D. Anstrom, R. E. Sharp, O. R. Patharkar, D. Clay,
41%; 1H NMR (400 MHz, d6-DMSO)  7.72 (d, 2H), 7.55 (m, M. Coffin, M. A. Nemeth, M. E. Leibman, M. Luethy, M.
1H), 7.20-7.14 (m, 1H), 7.08 (d, 2H), 6.77-6.73 (m, 2H), 6.00- Lawson, Plant Cell Environ. 2015, 38, 1866.
5.96 (m, 1H), 3.82 (s, 3H), 3.13-3.08 (m, 2H), 2.85-2.81 (m, 2H). 6. F. T. Addicott, J. L. Lyon, Annu. Rev. Plant Physiol. 1969, 20,
LCMS (ESI, m/z): [M+2H]+ 394.1, logp 3.09. 139.
7. B. V. Milborrow, Annu. Rev. Plant Physiol. 1974, 25, 259
4.2.5.4. N-(2-{[4-Fluoro-3-(trifluormethyl)phenyl]amino}- 8. S. R. Cutler, P. L. Rodriguez, R. R. Finkelstein, S. R. Abrams,
ethyl)-2-fluorobenzene sulfonamide (20d). White solid; yield Annu. Rev. Plant Biol. 2010, 61, 651.
9. E. Nambara, A. Marion-Poll, Annu. Rev. Plant Biol. 2005, 56, 30. S. R. Cutler, M. Okamoto, WO2013/148339, 2013.
165. 31. S. V. Wendeborn, P. J. Jung, M. D. Lachia, R. Dumeunier, S. R.
10. Y. Ma, I. Szostkiewicz, A. Korte, D. Moes, Y. Yang, A. Cutler, WO2014/210555, 2014.
Christmann, E. Grill, Science 2009, 324, 1064. 32. J. Frackenpohl, G. Bojack, H. Helmke, S. Lehr, T. Müller, L.
11. S.-Y. Park, P. Fung, P. Fung, N. Nishimura, D. R. Jensen, H. Fujii, Willms, H. Dietrich, D. Schmutzler, R. Baltz, U. Bickers,
Y. Zhao, S. Lumba, J. Santiago, A. Rodriguez, T.-F. F. Chow, S. WO2015/155154, 2015.
E. Alfred, D. Bonetta, R. Finkelstein, N. J. Provart, D. Desveaux, 33. J. Frackenpohl, G. Bojack, H. Helmke, L. Willms, S. Lehr, T.
P. L. Rodriguez, P. McCourt, J.-K. Zhu, J. I. Schroeder, B. F. Müller, J. Dittgen, D. Schmutzler, R. Baltz, U. Bickers,
Volkman, S. R. Cutler, Science 2009, 324, 1068. WO2016/128365, 2016.
12. K. Ueno, Y. Todoroki, Current Med. Chem. 2010, 17, 3230. 34. H. Helmke, J. Frackenpohl, J. Franke, G. Bojack, J. Dittgen, D.
13. A. J. Cutler, J. E. Krochko, Trends Plant Sci 1999, 4, 472. Schmutzler, U. Bickers, F. Poree, F. Roth, J.-P. Vors,
14. R. Zhou, A. J. Cutler, S. J. Ambrose, M. M. Galka, K. M. Nelson, WO2017/009321, 2017.
T. M. Squires, M. K. Loewen, A. S. Jadhav, A. R. S. Ross, D. C. 35. J. Frackenpohl, E. Grill, G. Bojack, R. Baltz, M. Busch, J. Dittgen,
Taylor, S. R. Abrams, Plant Physiol. 2004, 134, 361. J. Franke, J. Freigang, S. Gonzalez, I. Heinemann, H. Helmke, M.
15. J. J. Balsevich, A. J. Cutler, N. Lamb, L. J. Friesen, E. U. Kurz, Hills, S. Hohmann, P. von Koskull-Döring, J. Kleemann, G.
M. R. Perras, S. R. Abrams, Plant Physiol. 1994, 106, 135. Lange, S. Lehr, T. Müller, E. Peschel, F. Poree, D. Schmutzler, A.
16. J. M. Nyangulu, K. M. Nelson, P. A. Rose, Y. Gai, M. Loewen, B. Schulz, L. Willms, C. Wunschel, Eur. J. Org. Chem. 2018, 1403.
Lougheed, J. W. Quail, A. J. Cutler, S. A. Abrams, Org. Biomol. 36. I. Reulecke., G. Lange, J. Albrecht, R. Klein, M. Rarey,
Chem. 2006, 4, 1400. ChemMedChem 2008, 3,:885-897.
17. X. Han, L. Jiang, C. Che, C. Wan, H. Lu, Y. Xiao, Y. Xu, Z. 37. A. S. Vaidya, F. C. Peterson, D. Yarmolinsky, E. Merilo, I.
Chem, Z. Qin, Sci. Rep. 2017, 7, 43863. Verstraeten, S.-Y. Park, D. Elzinga, A. Kaundal, J. Helander, J.
18. J. Takeuchi, M. Okamoto, T. Akiyama, T. Muto, S. Yajima, M. Lozano-Juste, M. Otani, K. Wu, D. R. Jensen, H. Kollist, B. F.
Sue, M. Seo, Y. Kanno, T. Kamo, A. Endo, E. Nambara, N. Hirai, Volkman, S. R. Cutler, ACS Chem. Biol. 2017, 12, 2842.
T. Ohnishi, S. R. Cutler, Y. Todoroki, Nat. Chem. Biol. 2014, 10, 38. D. Elzinga, E. Sternburg, D. Sabbadin, M. Bartsch, S.-Y. Park, A.
477. Vaidya, A. Mosquna, A. Kaundal, S. Wendeborn, M. Lachia, F.
19. G. T. Wang, D. Heiman, G. D. Venburg, J. H. Lustig, M. A. V. Karginov, S. R. Cutler, ACS Chem. Biol. 2019, 14, 332-336
Surpin, R. E. Fritts, F. P. Silverman, D. D. Woolard, 39. L. Zwiebel, G. M. Pask, D. C. Rinker, I. M. Romaine, G. A.
WO2016/168535, 2016. Sulikowski, P. R. Reid, A. G. Waterson, K. Kim, P. L. Jones, R.
20. G. T. Wang, D. Heiman, G. D. Venburg, E. Nagano, M. A. W. Taylor, WO2012/154403, 2012
Surpin, J. H. Lustig, WO2016/007587, 2016. 40. J. Frackenpohl, G. Bojack, H. Helmke, S. Lehr, I. Heinemann, K.
21. F. P. Silverman, G. Wang, K. A. Falco, D. D. Woolard, D. O. Minn, J. Dittgen, D. Schmutzler, U. Bickers, M. J. Hills, F. Roth,
Wilson, D. C. Leep, G. D. Venburg, WO2016/187370, 2016. F. Poree, EP3210469, 2017.
22. G. Wang, R. Hopkins, D. C. Leep, D, D. Woolard, G. D. Venburg, 41. J. Tscherniac, DE134979, 1901.
WO2016/187370, 2016. 42. A. Einhorn, E. Bischkopff, B. Szelinski, G. Schupp, E. Spröngerts,
23. J. Frackenpohl, G. Bojack, R. Baltz, U. Bickers, M. Busch, J. C. Ladisch, T. Mauermayer, Liebigs Ann. Chem. 1905, 343, 207.
Dittgen, J. Franke, J. Freigang, E. Grill, S. Gonzalez, H. Helmke, 43. J. Franke, J. Frackenpohl, H. Helmke, D. Schmutzler, P. Lümmen,
M. J. Hills, S. Hohmann, P. von Koskull-Döring, J. Kleemann, G. F. Poree, S. M. Gonzalez Fernandez-Nino, L. Schneider,
Lange, S. Lehr, D. Schmutzler, A. Schulz, K. Walther, L. Willms WO/2018/108627, 2018.
and C. Wunschel, Eur. J. Org. Chem. 2018, 1416. 44. H. Helmke, J. Frackenpohl, J. Franke, D. Schmutzler, P. Lümmen,
24. J. Takeuchi, T. Ohnishi, M. Okamoto, Y. Todoroki, Bioorg. Med. F. Poree, S. M. Gonzalez Fernandez-Nino, L. Schneider, M.
Chem. Lett. 2015, 25, 3507. Koppitz, WO 2019/025153, 2019.
25. T. Miyakawa, M. Tanokura, Biophysics 2011, 7, 123. 45. R. Mega, F. Abe, J.-S. Kim, Y. Tsuboi, K. Tanaka, H. Kobayashi,
26. J. D. M. Helander, A. S. Vaidya, S. R. Cutler, Bioorg. Med. Chem. Y. Sakata, K. Hanada, H. Tsujimoto, J. Kikuchi, S. R. Cutler, M.
2016, 24, 493. Okamoto Nature Plants, 2019, 5, 153.
27. S. R. Cutler, S.-Y. Park, A. Defries, WO2010/093954, 2010. 46. A. S. Raghanvendra, V. K. Gonugunta, A. Christmann, E. Grill,
28. F. C. Peterson, E. S. Burgie, S.-Y. Park, D. R. Jensen, J. J. Weiner, Trends Plant Sci. 2010, 15, 395.
C. A. Bingman, C.-E. A. Chang, S. R. Cutler, G. N. Phillips, B. F. 47. F. Dupeux, R. Antoni, K. Betz, J. Santiago, M. Gonzalez-Guzman,
Volkman, Nature Struct. Mol. Biol. 2010, 17, 1109. L. Rodriguez, S. Rubio, S.-Y. Park, S. R. Cutler, P. L. Rodriguez,
29. J. Frackenpohl, I. Heinemann, T. Müller, P. v. Koskull-Döring, J. J. A. Márquez, Plant Physiol. 2011, 156, 106.
Dittgen, D. Schmutzler, C. H. Rosinger, I. Häuser-Hahn, M. J.
Hills, WO2011/113861, 2011.
Graphical Abstract – J. Frackenpohl

Identifying new lead structures to enhance Leave this area blank for abstract info.
tolerance towards drought stress via high-
throughput screening giving crops a
quantum of solace
Jens Frackenpohla,, Linn Schneiderb, Luka J. B. Deckera, Jan Dittgena, Franz Fenkla, Christian
Fischera, Jana Frankea, Joerg Freigangc, Susana M. Gonzalez Fernandez-Ninoa, Hendrik Helmkea,
Martin J. Hillsa, Sabine Hohmanna, Rahel Getachewa, Jochen Kleemannc, Karoline Kurowskia, Gudrun
Langea, Peter Luemmena, Nicole Meyeringb, Fabien Poreea, Dirk Schmutzlera and Sebastian Wredeb
a Research & Development, Weed Control - Bayer AG, Crop Science Division, Industriepark Höchst, D-65926 Frankfurt am Main
b Research & Development, Lead Discovery – Bayer AG, Pharmaceutical Division, Aprather Weg 18a, D-42096 Wuppertal
c Research & Development - Bayer AG, Crop Science Division, Gebäude 6240, Alfred-Nobel-Straße 50 ,D-40789 Monheim

H 3
N R
High-throughput screening N N
S O
1
R N
2
R

O O
2 S
R N
H N
O
1
R
3
R
2
H R
N
S
O O N 1
H R
BMC_2019_046

Declaration of interests

☒ The authors declare that they have no known competing financial interests or personal relationships that
could have appeared to influence the work reported in this paper.

☐The authors declare the following financial interests/personal relationships which may be considered as potential
competing interests:

You might also like