You are on page 1of 13

European Journal of Medicinal Chemistry 263 (2024) 115913

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: www.elsevier.com/locate/ejmech

Research paper

Discovery of 4-hydroxyl pyrazole derivatives as potent


ferroptosis inhibitors
Danzhi Ying a, 1, Xin Shen b, 1, Shuqi Wang a, Junyi Chen b, Zhenying Wu a, Wenteng Chen a,
Fudi Wang b, Junxia Min b, **, Yongping Yu a, *
a
College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
b
The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine,
Hangzhou, 310058, China

A R T I C L E I N F O A B S T R A C T

Keywords: Ferroptosis, an iron-dependent form of regulated cell death, has been well recognized as a pathogenic mechanism
Ferroptosis inhibitor in driving many diseases, such as neurodegenerative disorders, ischemia-reperfusion (I/R) injury. Blocking
4-Hydroxyl pyrazole derivatives ferroptosis has been emerging as a feasible therapeutic strategy for the prevention and treatment of these dis­
Structure-activity relationship
eases. However, novel potent ferroptosis inhibitors remain to be developed for further clinical applications. In
Radical-trapping antioxidant
this study, we screened our in-house compound libraries by phenotypic assays and identified a 4-hydroxyl
pyrazole derivative HW-3 with good ferroptosis inhibitory activity (EC50 = 120.1 ± 3.5 nM). Based on the
structure of HW-3, a series of 4-hydroxyl pyrazole derivatives were further designed and synthesized. Among
these compounds, compound 25 could significantly inhibit RSL3-induced ferroptosis with an EC50 value of 8.6 ±
2.2 nM in HT-1080 cells, which was 3-fold more potent than the classical ferroptosis inhibitor ferrostatin-1 (Fer-
1) (EC50 = 23.4 ± 1.3 nM). The potent ferroptosis inhibitory activity of compound 25 was further validated in
multiple additional cell lines. Our mechanistic study revealed that compound 25 inhibited ferroptosis via
intrinsic radical-trapping antioxidative capacity. Taken together, the findings of our study demonstrate 4-hydrox­
yl pyrazole derivative 25 is a potent ferroptosis inhibitor, which holds a great therapeutic potential for further
development.

1. Introduction In the last decade, a plenty of ferroptosis inhibitors have been


developed (Fig. 1). Ferrostatin-1 (Fer-1) and liproxstatin-1 (Lip-1) were
Ferroptosis is an iron-dependent regulated cell death (RCD) driven by the first reported ferroptosis inhibitors discovered by high-throughput
excessive lipid peroxidation and cellular metabolism [1–3]. Morpholog­ screenings of compound library [1], and have been proved to act as
ically, ferroptosis mainly manifests as obvious shrinkage of mitochondria radical-trapping antioxidants (RTAs) to suppress the accumulation of
with increased membrane density and decreased cristae [4,5], which is LPO for inhibiting ferroptosis [16–18]. CuATSM, a Cu2+ complex
distinct from other forms of RCD such as apoptosis [6] and necroptosis recently used in clinical studies for the treatment of amyotrophic lateral
[7]. A major characteristic of ferroptosis is the accumulation of lipid sclerosis (ALS), also showed anti-ferroptosis activity through quenching
peroxides (LPO) that formed through iron-catalyzed polyunsaturated the lipid radicals [19,20]. Additionally, a series of well-known RTAs [21,
fatty acids (PUFAs) autoxidation or enzyme-mediated process catalyzed 22], such as α-TOH [23], phenothiazines [24–26] as well as nitroxides
by iron-dependent lipoxygenases (LOXs) [8–11]. LPO are considered as [27,28], have been reported as ferroptosis inhibitors. Meanwhile, dex­
key inducers through their ability to crosslink ferroptosis-related proteins razoxane (DXZ), is the FDA-approved drug for preventing doxorubicin
and nucleic acids [12]. When cells fail to suppress the formation of LPO, (DOX)-induced cardiotoxicity in cancer patients [29]. Our previous
ferroptosis is triggered. It is thought as a promising strategy to inhibit research indicated that DOX-induced cell death exhibited features of
ferroptosis for the treatment of related diseases [13–15]. typical ferroptotic cell death, and DXZ displayed protective effects

* Corresponding author.
** Corresponding author.
E-mail addresses: junxiamin@zju.edu.cn (J. Min), yyu@zju.edu.cn (Y. Yu).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.ejmech.2023.115913
Received 4 September 2023; Received in revised form 11 October 2023; Accepted 24 October 2023
Available online 4 November 2023
0223-5234/© 2023 Elsevier Masson SAS. All rights reserved.
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

against DOX-induced ferroptosis [30]. In addition, iron chelators 2.2. Structure-activity relationship analysis for pyrazole derivatives
deferoxamine and deferiprone, have been proved to inhibit ferroptosis
as well [31,32]. Although a variety of ferroptosis inhibitors have been All the synthesized compounds were tested for their ferroptosis
developed so far, many of them suffer from poor activity, or unfavorable inhibitory activity with the model of RSL3 (1 μM)-induced ferroptosis in
pharmacokinetic properties, limiting their further clinical application. HT-1080 cells. The HT-1080 cell line was proved to show high sensi­
Therefore, development of new ferroptosis inhibitors for treating tivity to ferroptosis and widely used to evaluate the specificity of com­
ferroptosis-related diseases is still highly desirable. pounds to ferroptosis [16,26,27]. We firstly explored the variety of R1
Herein, we discovered a new ferroptosis inhibitor HW-3 (EC50 = and R2 on the ferroptosis inhibitory activity. As shown in Table 1,
120.1 ± 3.5 nM), a 4-hydroxy pyrazole derivative, through phenotypic compound 3 (EC50 = 69.1 ± 2.7 nM) with phenyl ring on R1 and R2
assays on 1 μM RSL3-induced HT-1080 cells ferroptosis model. Based on exhibited better inhibitory activity than HW-3, while the typical fer­
the scaffold of HW-3, a series of derivatives were further designed, roptosis inhibitor Fer-1 significantly rescued cells with an EC50 value of
synthesized, and evaluated for their ferroptosis inhibitory activity. Out 23.4 ± 1.3 nM. The activity was slightly decreased when introducing
of them, compound 25 was found to be the most potent ferroptosis in­ fluorine into the phenyl ring of R2 (4), while the phenyl ring bearing
hibitor (EC50 = 8.6 ± 2.2 nM), which was better than the classic fer­ chlorine (5) or bromine (6) led to an improved activity. The
roptosis inhibitor Fer-1 (EC50 = 23.4 ± 1.3 nM). The potent ferroptosis electron-donating group methoxy (7) was also beneficial for the fer­
inhibitory activity of compound 25 was further validated in multiple roptosis inhibition. Oppositely, substitution of the phenyl ring with
additional cell lines. Further mechanism studies demonstrated com­ furanyl (8) or n-butyl (9) group was unfavorable, leading to a signifi­
pound 25 effectively inhibited ferroptosis by intrinsic antioxidative cantly decreased activity. As for R1, 4-biphenyl substitution (10) was
capacity. more favorable than phenyl (3) group. Introduction of electron-donating
groups (11 and 12) into the phenyl ring was more beneficial than strong
2. Results and discussion electron-withdrawing groups (13 and 14). Of note, compound 15 with a
4-Cl substituted phenyl group at R1 exhibited significantly improved
2.1. Chemistry potency (EC50 = 18.9 ± 0.9 nM), which was better than Fer-1. However,
the efficacy decreased no matter changing the position (16 and 17) or
Synthetic routes for intermediates and the final products were increasing the number (18) of chlorine atom. Based on the structure of
depicted in Scheme 1 and Scheme 2. Reaction of 2-bromoketone de­ compound 15, we next explored the influence of R3 and R4 on ferroptosis
rivatives with sodium azide provided phenacyl aizdes 1a-1j. Then inhibitory activity (Table 1). Substitution of R3 with methyl or phenyl
α-azido chalcones 2a-2w were obtained via Knoevengel condensation group was well-tolerated. However, the activity was notably decreased
reaction of phenacyl aizdes with corresponding aldehydes [33]. The when the hydroxyl of R4 was replaced by amino group or hydrogen
preparations of pyrazole derivatives were performed according to our atom, demonstrating the necessity of hydroxy group at 4-position of the
previous report [34]. To be precise, treatment of α-azido chalcones with pyrazole ring for ferroptosis inhibitory activity.
corresponding hydrazine and 1,8-diazabicyclo [5.4.0]undec-7-ene Inspired by the improved ferroptosis inhibition of compound 15, we
(DBU) in acetonitrile/water afforded 4-hydroxy pyrazole derivatives further optimized the R2 moiety to improve its efficacy (Table 2). Similar
3–20, 23, 24, 27–31. Treatment of α-azido chalcone 2m with methyl­ to the above results, introduction of electron-donating group (23 and
hydrazine sulfate and sodium hydroxide in acetonitrile led to 4-amino 24) into the phenyl ring was more favorable than those with electron-
pyrazole derivative 21. Pyrazole derivative 22 was synthesized by withdrawing group (27 and 28). Of note, the efficacy of compound 25
subjecting 2m to hydrazine hydrate and DBU in acetonitrile (Scheme 1). bearing 4-hydroxy phenyl group at R2 was significantly improved (EC50
Removing the methyl group of 23 and 24 afforded phenol derivatives 25 = 8.6 ± 2.2 nM), which was 3-fold more potent than Fer-1. Compound
and 26, respectively. Hydrolysis of 31 under acid conditions afforded 26 with two hydroxyls on the phenyl ring exhibited slightly decreased
the carboxyl derivative 32, followed by condensation with corre­ activity. Replacing the phenyl ring of R2 with naphthyl (29) or imidazole
sponding amines to give amide derivatives 33, 34, 35a. Finally, the group (30) resulted in an acceptable potency. Furthermore, it was not
tetrahydropyran group of 35a was easily removed under acid condi­ beneficial for their ferroptosis inhibitory activity by introducing ester
tions, providing the target compound 35 (Scheme 2). (31), carboxy (32) or amide group (33–35) into the phenyl ring.

Fig. 1. Chemical structures of representative ferroptosis inhibitors.

2
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

Scheme 1. Reagents and conditions: (a) NaN3, acetone, rt, 3 h, 75 %–91 %. (b) acetic acid, piperidine, ethanol, rt, 12–24 h, 38 %–91 %. (c) corresponding hydrazine,
DBU, CH3CN/H2O (10/1, v/v), rt, 3 h, 22 %–96 %. (d) methylhydrazine sulfate, NaOH, CH3CN, rt, 8 h, 86 %. (e) Hydrazine hydrate, DBU, CH3CN, rt, 3 h, 22 %.

Scheme 2. (A) BBr3, CH2Cl2, 0 ◦ C to rt, 3 h, 54%–65 %. (b) concentrated HCl, reflux, 24 h, 82 %. (c) corresponding amine, HATU, Et3N, DMF, rt, 6 h, 59 %–71 %. (d)
HCl/MeOH, rt, 12 h, 88 %.

2.3. Ferroptosis inhibitory activity of compound 25 in vitro 7.2 nM, 11.3 ± 1.4 nM and 43.3 ± 6.2 nM, respectively, which were all
better than Fer-1 with EC50 values of 253.0 ± 20.1 nM, 38.2 ± 3.7 nM
Among all tested compounds, the compound 25 was found to be the and 68.6 ± 8.1 nM, respectively.
most effective for inhibiting ferroptosis induced by RSL3, and thus was The inhibition activity of compound 25 against ferroptosis was also
chosen for further evaluation. Imidazole ketone erastin (IKE), derived validated by the cell morphologic observation. As shown in Fig. 2F, 786-
from Erastin, could effectively induce ferroptosis by inhibiting system O cells became shrunken, membrane rupture and lost their normal
Xc− and preventing cystine import [35]. As shown in Fig. 2A, on morphology after treatment with RSL3. Oppositely, the cells recovered
IKE-induced HT-1080 cells ferroptosis model, compound 25 also when mixed with Fer-1 (1 μM) or compound 25 (1 μM). The similar
showed stronger protection effect than Fer-1. To determine whether the changes were also observed when the concentration of compound 25
inhibitory effect of compound 25 was cell line specific, we tested its decreased to 0.0625 μM. However, unlike compound 25, there’re no
activity on three additional cell lines: 786-O [36], BJ [37] and H9c2 significant changes when treated with 0.0625 μM Fer-1. These results
[30], which all exhibited sensitivity to ferroptosis. As shown in Table 3, further demonstrated that compound 25 had a better capacity of pro­
Fig. 2C–E, consistent with the result in HT-1080 cells (Fig. 2B), the data tecting HT-1080 cells from ferroptosis than Fer-1.
indicated that compound 25 significantly prevented 786-O, BJ and H9c2
cell lines from ferroptosis induced by RSL3 with EC50 values of 89.7 ±

3
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

Table 1 Table 2
Ferroptosis inhibitory activity of pyrazole derivatives in HT-1080 cells. Ferroptosis inhibitory activity of 4-hydroxyl pyrazole derivatives in HT-1080
cells.

Compound R1 R2 R3 R4 EC50 (nM)a


Compound R2 EC50 (nM)a
Fer-1 – – – – 23.4 ± 1.3
HW-3 4-NO2-phenyl 4-Me-phenyl H OH 120.1 ± Fer-1 – 23.4 ± 1.3
3.5 23 4-OMe-phenyl 23.1 ± 1.5
3 phenyl phenyl H OH 69.1 ± 2.7 24 3,4-di-OMe-phenyl 31.7 ± 5.1
4 phenyl 4-F-phenyl H OH 79.6 ± 2.3 25 4-OH-phenyl 8.6 ± 2.2
5 phenyl 4-Cl-phenyl H OH 30.8 ± 1.4 26 3,4-di–OH–phenyl 26.1 ± 4.3
6 phenyl 4-Br-phenyl H OH 22.6 ± 1.1 27 4-CF3-phenyl 187.2 ± 5.2
7 phenyl 4-OMe- H OH 24.8 ± 2.0 28 4-F-phenyl 52.7 ± 3.1
phenyl 29 1-naphthyl 44.8 ± 2.9
8 phenyl 2-furanyl H OH >1000 30 25.9 ± 1.2
9 phenyl n-butyl H OH 150.0 ±
5.8
10 4-biphenyl phenyl H OH 39.5 ± 2.3 31 51.1 ± 2.2
11 4-OMe-phenyl phenyl H OH 32.1 ± 1.7
12 3,4-OCH2O- phenyl H OH 27.4 ± 1.9
32b >1000
phenyl
13 4-CF3-phenyl phenyl H OH 79.6 ± 2.3
14 4-F-phenyl phenyl H OH 42.4 ± 1.3 33 81.9 ± 2.1
15 4-Cl-phenyl phenyl H OH 18.9 ± 0.9
16 2-Cl-phenyl phenyl H OH 94.9 ± 7.6
17 3-Cl-phenyl phenyl H OH 187.2 ±
5.2 34 44.8 ± 1.4
18 2,4-di-Cl-phenyl phenyl H OH 56.6 ± 3.3
19 4-Cl-phenyl phenyl Me OH 22.0 ± 0.8
20 4-Cl-phenyl phenyl phenyl OH 24.6 ± 1.2
21 4-Cl-phenyl phenyl Me NH2 720.5 ±
9.4
22 4-Cl-phenyl phenyl H H >1000 35 80.2 ± 5.6

a
Each compound was tested in three independent experiments; the values
were presented as the means ± SD. a
Each compound was tested in three independent experiments; the values
were presented as the means ± SD. bCompound was tested in the form of hy­
2.4. Mechanistic study of compound 25 protecting cells from ferroptosis drochloride salt.

It is reported that the classic ferroptosis inhibitor, Fer-1, act as a Increasing level of lipid reactive oxygen species (ROS) is a major
radical-trapping antioxidant (RTA) [16,17]. To explore the mechanism hallmark of ferroptosis. We used BODIPY 581/591C11 (BODIPY-C11)
behind the anti-ferroptosis activity of 4-hydroxy pyrazole derivatives, probe to detect levels of lipid ROS generation by flow cytometry. As
we firstly evaluated the antioxidative potency of compound 15, 21, 22, shown in Fig. 3C, RSL3 apparently induced the accumulation of lipid
25 by the 2,2-diphenyl-1-picrylhydrazyl (DPPH) bleaching method, ROS. Compound 25 exerted significantly decreased lipid peroxidation
which is widely used for the antioxidative capacity evaluation of the compared with Fer-1 treatment. In addition, we measured the intracel­
ferroptosis inhibitors [38]. As shown in Fig. 3A and Table 4, compound lular ROS level with fluorescent probe 5-(and-6)-carboxy-2′,7′-dichlor­
15, 25 were both capable of scavenging DPPH radicals in a odihydrofluorescein diacetate (DCFH-DA) by confocal laser scanning
dose-dependent manner in vitro like Fer-1, in which compound 25 (EC50 microscopy (CLSM). The results demonstrated compound 25 could
= 3.94 μM) exhibited 1.6-fold more effective than Fer-1 (EC50 = 6.48 effectively suppress the erastin-induced cytosolic ROS accumulation
μM). However, compound 21 and 22, with amino and hydrogen at R4, (Fig. 3D).
respectively, exhibited relatively poor antioxidative ability, suggesting Furthermore, iron imbalance is an essential initiator of ferroptosis
that the hydroxy at R4 plays a key role in exerting its antioxidative ca­ [1]. Thus, we evaluated the iron-chelating ability of compound 25 with
pacity, as well as ferroptosis inhibitory activity. Additionally, ABTS Fe (III) or Fe (II) by using UV–vis spectroscopy [40]. As displayed in
radical cation scavenging capacity assay under cell-free condition was Fig. 3E, compound 25 had a characteristic absorption peak at ~261 nm
performed to further validate their antioxidative capacity [39]. ABTS [2, and no new peaks were observed when Fe (III) or Fe (II) was added to the
29-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid)] radical cation solution of compound 25. Moreover, it was observed that there were no
(ABTS•+) is a blue-colored cation with absorption maxima at wave­ significant changes of UV–vis absorption at 261 nm in the presence of Fe
lengths 415 nm, and the antioxidative capacity is determined by the (III) or Fe (II) (Fig. 3F). This suggested that compound 25 had no ability
degree of blanching of ABTS•+. The results shown in Fig. 3B and Table 4 to chelate with Fe (III) or Fe (II). These results indicate that compound
were similar with the results of DPPH assays, further proving that there’s 25 mainly exerts anti-ferroptosis effect by its intrinsic antioxidative
a significant correlation between the anti-ferroptosis activity and anti­ capacity.
oxidative ability of 4-hydroxy pyrazole derivatives. In the ABTS assay, In conclusion, combining the results from both ferroptosis inhibitory
compound 25 (EC50 = 6.3 μM) exhibited nearly 8-fold stronger anti­ activity and antioxidative ability, a general SAR was summarized in
oxidative capacity than Fer-1 (EC50 = 49.8 μM). Based on the above Fig. 4. The 4-hydroxy group is necessary for both anti-ferroptosis and
results, we conclude that compound 25 holds stronger antioxidative antioxidative activity. Substitution of 1-hydrogen with methyl or phenyl
capacity than Fer-1. group is well-tolerated. As for R1 or R2, furan or alkyl group is

4
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

Fig. 2. The compound 25 shows more potent ferroptosis inhibitory activity than Fer-1 in cells.
(A) Inhibition effect of compound 25 against 2.5 μM IKE-induced ferroptosis in HT-1080 cells. (B–E) Inhibition effect of compound 25 against 1 μM RSL3-induced
ferroptosis in HT-1080 cells (B), 786-O cells (C), BJ cells (D) and H9c2 cells (E). (F) Morphology of 786-O cells treated with 1 μM RSL3 in the absence or presence of
Fer-1 or compound 25.

3. Conclusions
Table 3
Ferroptosis inhibitory activity of compound 25 and Fer-1 in different cell lines.
In this study, we firstly screen our in-house compound libraries by
No EC50 (nM) phenotypic assays and discovered a new ferroptosis inhibitor HW-3 with
HT-1080a HT-1080b 786-Ob BJb H9c2b 4-hydroxyl pyrazole scaffold. To obtain more potent compounds, we
25 48.3 ± 5.3 8.6 ± 2.2 89.7 ± 7.2 11.3 ± 1.4 43.3 ± 6.2 further designed and synthesized a series of derivatives based on the
Fer-1 75.2 ± 6.4 23.4 ± 1.3 253.0 ± 20.1 38.2 ± 3.7 68.6 ± 8.1 structure of HW-3. Among all tested compounds, compound 25 was
a identified as the most potent ferroptosis inhibitor in HT-1080 cells and
Cells were treated with 2.5 μM IKE in the presence of various concentrations
of compound 25 or Fer-1.
additional three cell lines. Notably, at the cellular level, its ferroptosis
b
Cells were treated with 1 μM RSL3 in the presence of various concentrations inhibitory activity was more potent than Fer-1. Furthermore, mecha­
of compound 25 or Fer-1. nistic studies revealed compound 25 mainly effectively inhibited fer­
roptosis by its intrinsic antioxidative capacity, which holds a great
unfavorable for ferroptosis inhibitory activity, while phenyl, naphtha­ therapeutic potential for further clinical development.
lene and imidazole are more favorable. With respect to substituents on
phenyl ring, chlorine, bromine or electron donating substituents im­
proves the anti-ferroptosis activity, while strong electron-withdrawing
group is not beneficial.

5
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

Fig. 3. The compound 25 displays higher antioxidative activity than Fer-1. The cell-free antioxidant activity of Fer-1 and compound 15, 21, 22, 25 in various
concentrations analyzed by DPPH assay (A) and ABTS assay (B). (C) Flow cytometry analyses of lipid ROS levels in HT-1080 cells treated with 0.8 μM RSL3 in the
absence or presence of Fer-1 or compound 25. (D) The confocal image of DCFH-DA in HT-1080 cells treated with 5 μM Erastin in the absence or presence of Fer-1 or
compound 25. (E) UV–vis absorption spectra of compound 25 in the absence or presence of Fe (III) or Fe (II). (F) Quantification of UV–vis absorption peaks at 261 nm
for compound 25 in the absence or presence of Fe (III) or Fe (II). Ns, not significance.

4. Experimental section
Table 4
The ferroptosis inhibitory and antioxidant activity of pyrazole derivatives.
4.1. Chemistry

4.1.1. Materials and equipment


All reagents and solvents were obtained from commercial suppliers
and used without further purification unless otherwise noted. Reactions
were monitored by TLC carried out on silica gel 60 F254 plates. Melting
points were recorded on a Büchi B-540 melting point apparatus. 1H NMR
Compound EC50 and 13C NMR spectra were recorded at 500 MHz using a Bruker AVANCE
Ferroptosis inhibition DPPH clearance ABTS clearance III spectrometer in CDCl3 or dimethyl sulfoxide (DMSO)-d6 solution. For
1
(nM) (μM) (μM) H NMR, tetramethylsilane (TMS) served as the internal standard (δ =
Fer-1 23.4 ± 1.3 6.48 49.8
0), and data are reported as follows: chemical shift, integration, multi­
15 18.9 ± 0.9 4.97 111.8 plicity (s = singlet, d = doublet, t = triplet, q = quartet, and m =
21 720.5 ± 9.4 14.61 >200 multiplet), and coupling constant(s) in Hz. For 13C NMR, TMS (δ = 0) or
22 >1000 >100 >200 CDCl3 (δ = 77.26) was used as the internal standard and spectra were
25 8.6 ± 2.2 3.94 6.3
obtained with complete proton decoupling. HPLC purity analysis and
the HRMS of all final products were confirmed on an Agilent 1290 HPLC-
6224 time-of-flight mass spectrometer using a Phenomenex Luna 5 μm
C18, 100 Å, 150 × 4.60 mm, 5 μm column at a flow rate of 0.5 mL/min
using linear gradient buffer B in A (B, CH3OH containing 0.1 % formic
acid; A, H2O containing 0.1 % formic acid). Mobile phase B was
increased linearly from 5 to 95 % over 7 min and 95 % over the next 2

6
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

Fig. 4. Summary of the SAR of the pyrazole derivatives.

min after which the column was equilibrated to 5 % for 1 min. Purity of 4.1.2.6. (Z)-2-Azido-3-(furan-2-yl)-1-phenylprop-2-en-1-one (2f).
all biologically tested compounds were determined by HPLC to be >95 Following general procedure afforded 2f as a yellow oil (Yield: 90 %). 1H
%. NMR (500 MHz, Chloroform-d) δ 7.75–7.71 (m, 2H), 7.62–7.57 (m, 1H),
7.52–7.46 (m, 3H), 7.25 (d, J = 3.6 Hz, 1H), 6.57 (dd, J = 3.9, 1.8 Hz,
4.1.2. General procedures for the synthesis of 2a-2w 1H), 6.47 (s, 1H). HRMS (ESI) m/z calcd for C13H9N3O2 [M+Na]+:
In a 100 mL round bottom flask, sodium azide (4 mmol) was added to 262.0587. Found: 262.0591.
a solution of 2-bromoketone derivatives (2 mmol) in acetone (30 mL)
with vigorous stirring at room temperature. The mixture was stirred for 4.1.2.7. (Z)-2-Azido-1-phenylhept-2-en-1-one (2g). Following general
3 h and filtered under vacuum. The filtrate was concentrated to give procedure afforded 2g as a yellow oil (Yield: 88 %). 1H NMR (500 MHz,
phenacyl azides without further purification. Aldehydes (2 mmol) were Chloroform-d) δ 7.71–7.68 (m, 2H), 7.59–7.55 (m, 1H), 7.48–7.43 (m,
added to a solution of phenacyl azides in ethanol (10 mL) at room 2H), 5.76 (t, J = 7.5 Hz, 1H), 2.32 (q, J = 7.4 Hz, 2H), 1.43–1.32 (m,
temperature. Then acetic acid (57 μL, 1 mmol) and piperidine (99 μL, 1 4H), 0.91 (t, J = 7.2 Hz, 3H). HRMS (ESI) m/z calcd for C13H15N3O
mmol) were added, and the mixture was stirred at room temperature for [M+Na]+: 252.1107. Found: 252.1114.
12 h. Next, the reaction mixture was evaporated to dryness under
reduced pressure and the residue was extracted with dichloromethane/ 4.1.2.8. (Z)-1-([1,1′-biphenyl]-4-yl)-2-azido-3-phenylprop-2-en-1-one
water. The organic phase was separated, dried over anhydrous Na2SO4, (2h). Following general procedure afforded 2h as a yellow solid (Yield:
concentrated, and purified by silica-gel column chromatography using 77 %). 1H NMR (500 MHz, Chloroform-d) δ 7.91–7.87 (m, 2H),
petroleum ether/ethyl acetate (200/1 to 100/1) as eluent to yield 7.85–7.81 (m, 2H), 7.74–7.70 (m, 2H), 7.68–7.63 (m, 2H), 7.52–7.47
α-azido chalcones. (m, 2H), 7.44–7.34 (m, 4H), 6.52 (s, 1H). HRMS (ESI) m/z calcd for
C21H15N3O [M+Na]+: 348.1113. Found: 348.1113.
4.1.2.1. (Z)-2-Azido-1,3-diphenylprop-2-en-1-one (2a). Following gen­
eral procedure afforded 2a as a yellow solid (Yield: 80 %). 1H NMR (500 4.1.2.9. (Z)-2-Azido-1-(4-methoxyphenyl)-3-phenylprop-2-en-1-one
MHz, Chloroform-d) δ 7.85–7.75 (m, 4H), 7.66–7.58 (m, 1H), 7.53–7.48 (2i). Following general procedure afforded 2i as a pale-yellow solid
(m, 2H), 7.42–7.32 (m, 3H), 6.47 (s, 1H). HRMS (ESI) m/z calcd for (Yield: 80 %). 1H NMR (500 MHz, Chloroform-d) δ 7.86–7.82 (m, 2H),
C15H11N3O [M+Na]+: 272.0794. Found: 272.0799. 7.82–7.78 (m, 2H), 7.42–7.37 (m, 2H), 7.36–7.32 (m, 1H), 7.01–6.95
(m, 2H), 6.41 (s, 1H), 3.90 (s, 3H). HRMS (ESI) m/z calcd for
4.1.2.2. (Z)-2-Azido-3-(4-fluorophenyl)-1-phenylprop-2-en-1-one (2b). C16H13N3O2 [M+Na]+: 302.0900. Found: 302.0896.
Following general procedure afforded 2b as a yellow oil (Yield: 91 %).
1
H NMR (500 MHz, Chloroform-d) δ 7.85–7.76 (m, 4H), 7.64–7.59 (m, 4.1.2.10. (Z)-2-Azido-1-(benzo[d] [1,3] dioxol-5-yl)-3-phenylprop-2-en-
1H), 7.50 (m, 2H), 7.10–7.04 (m, 2H), 6.42 (s, 1H). HRMS (ESI) m/z 1-one (2j). Following general procedure afforded 2j as a yellow solid
calcd for C15H10FN3O [M+Na]+: 290.0706. Found: 290.0711. (Yield: 85 %). 1H NMR (500 MHz, Chloroform-d) δ 7.81 (d, J = 4.5 Hz,
2H), 7.46–7.32 (m, 5H), 6.89 (d, J = 8.4 Hz, 1H), 6.44 (s, 1H), 6.12 (s,
4.1.2.3. (Z)-2-Azido-3-(4-chlorophenyl)-1-phenylprop-2-en-1-one (2c). 2H). HRMS (ESI) m/z calcd for C16H11N3O3 [M+Na]+: 316.0693.
Following general procedure afforded 2c as a yellow solid (Yield: 66 %). Found: 316.0703.
1
H NMR (500 MHz, Chloroform-d) δ 7.78 (dd, J = 15.9, 7.9 Hz, 4H),
7.65 (t, J = 7.2 Hz, 1H), 7.51 (t, J = 7.5 Hz, 2H), 7.33 (d, J = 8.3 Hz, 2H), 4.1.2.11. (Z)-2-Azido-3-phenyl-1-(4-(trifluoromethyl) phenyl) prop-2-en-
6.41 (s, 1H). HRMS (ESI) m/z calcd for C15H10ClN3O [M+Na]+: 1-one (2k). Following general procedure afforded 2k as a yellow solid
306.0410. Found: 306.0400. (Yield: 47 %). 1H NMR (500 MHz, Chloroform-d) δ 7.88 (d, J = 8.0 Hz,
2H), 7.85–7.80 (m, 2H), 7.77 (d, J = 8.1 Hz, 2H), 7.44–7.35 (m, 3H),
4.1.2.4. (Z)-2-Azido-3-(4-bromophenyl)-1-phenylprop-2-en-1-one (2d). 6.42 (s, 1H). HRMS (ESI) m/z calcd for C16H10F3N3O [M+Na]+:
Following general procedure afforded 2d as a yellow solid (Yield: 65 %). 340.0668. Found: 340.0665.
1
H NMR (500 MHz, Chloroform-d) δ 7.79 (dd, J = 8.2, 1.2 Hz, 2H), 7.68
(d, J = 8.6 Hz, 2H), 7.65–7.59 (m, 1H), 7.51 (d, J = 8.3 Hz, 4H), 6.37 (s, 4.1.2.12. (Z)-2-Azido-1-(4-fluorophenyl)-3-phenylprop-2-en-1-one (2l).
1H). HRMS (ESI) m/z calcd for C15H10BrN3O [M+Na]+: 349.9899. Following general procedure afforded 2l as a yellow oil (Yield: 38 %). 1H
Found: 349.9901. NMR (500 MHz, Chloroform-d) δ 7.87–7.77 (m, 4H), 7.42–7.34 (m, 3H),
7.22–7.15 (m, 2H), 6.42 (s, 1H). HRMS (ESI) m/z calcd for C15H10FN3O
4.1.2.5. (Z)-2-Azido-3-(4-methoxyphenyl)-1-phenylprop-2-en-1-one [M+Na]+: 290.0700. Found: 290.0697.
(2e). Following general procedure afforded 2e as a yellow oil (Yield: 91
%). 1H NMR (500 MHz, Chloroform-d) δ 7.78 (dd, J = 16.8, 7.6, 1.8 Hz, 4.1.2.13. (Z)-2-Azido-1-(4-chlorophenyl)-3-phenylprop-2-en-1-one
4H), 7.61–7.57 (m, 1H), 7.51–7.47 (m, 2H), 6.93–6.90 (m, 2H), 6.46 (s, (2m). Following general procedure afforded 2m as a pale-yellow solid
1H), 3.85 (s, 3H). HRMS (ESI) m/z calcd for C16H13N3O2 [M+Na]+: (Yield: 70 %). 1H NMR (500 MHz, Chloroform-d) δ 7.83–7.78 (m, 2H),
302.0900. Found: 302.0901. 7.77–7.72 (m, 2H), 7.50–7.46 (m, 2H), 7.42–7.34 (m, 3H), 6.42 (s, 1H).

7
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

HRMS (ESI) m/z calcd for C15H10ClN3O [M+Na]+: 306.0405. Found: 4.1.2.23. Methyl (Z)-4-(2-azido-3-(4-chlorophenyl)-3-oxoprop-1-en-1-yl)
306.0400. benzoate (2w). Following general procedure afforded 2w as a yellow
solid (Yield: 40 %). 1H NMR (500 MHz, Chloroform-d) δ 8.07–8.02 (m,
4.1.2.14. (Z)-2-Azido-1-(2-chlorophenyl)-3-phenylprop-2-en-1-one (2n). 2H), 7.88–7.84 (m, 2H), 7.79–7.75 (m, 2H), 7.52–7.48 (m, 2H), 6.40 (s,
Following general procedure afforded 2n as a yellow solid (Yield: 86 %). 1H), 3.94 (s, 3H). HRMS (ESI) m/z calcd for C17H12ClN3O3 [M+Na]+:
1 364.0459. Found: 364.0459.
H NMR (500 MHz, Chloroform-d) δ 7.80–7.76 (m, 2H), 7.49–7.44 (m,
2H), 7.42–7.34 (m, 5H), 6.32 (s, 1H). HRMS (ESI) m/z calcd for
C15H10ClN3O [M+Na]+: 306.0405. Found: 306.0407. 4.1.3. Synthesis of compounds 3–35

4.1.2.15. (Z)-2-Azido-1-(3-chlorophenyl)-3-phenylprop-2-en-1-one (2o). 4.1.3.1. General procedures for the 4-hydroxyl pyrazole derivatives. A
Following general procedure afforded 2o as a yellow oil (Yield: 80 %). mixture of vinyl azide 2 (0.5 mmol) and 80 % hydrazine hydrate (2.5
1 mmol) was stirred in acetonitrile/water (2 mL/0.2 mL) at room tem­
H NMR (500 MHz, Chloroform-d) δ 7.84–7.80 (m, 2H), 7.77 (t, J = 1.9
Hz, 1H), 7.66 (dt, J = 7.7, 1.3 Hz, 1H), 7.59 (dd, J = 8.1, 2.1, 1.1 Hz, perature. Then DBU (0.5 mmol) was added and the mixture was stirred
1H), 7.47–7.35 (m, 4H), 6.44 (s, 1H). HRMS (ESI) m/z calcd for for 5 h. The mixture was diluted with water (10 mL) and extracted three
C15H10ClN3O [M+Na]+: 306.0405. Found: 306.0400. times with ethyl acetate (10 mL). The combined organic extracts were
washed with brine, dried over anhydrous Na2SO4, concentrated, and
4.1.2.16. (Z)-2-Azido-1-(2,4-dichlorophenyl)-3-phenylprop-2-en-1-one purified by column chromatography using petroleum ether/ethyl ace­
(2p). Following general procedure afforded 2p as a yellow solid (Yield: tate (5/1 to 3/1) as eluent to afford the final compounds.
71 %). 1H NMR (500 MHz, Chloroform-d) δ 7.79 (dd, J = 7.5, 2.2 Hz,
2H), 7.51 (d, J = 1.7 Hz, 1H), 7.42–7.34 (m, 5H), 6.30 (s, 1H). HRMS 4.1.3.2. 3,5-Diphenyl-1H-pyrazol-4-ol (3). Following general procedure
(ESI) m/z calcd for C15H9Cl2N3O [M+Na]+: 340.0015. Found: afforded 3 as a white solid (Yield: 43 %). Mp 236–237 ◦ C. 1H NMR (500
340.0016. MHz, DMSO‑d6) δ 12.89 (s, 1H), 8.33 (s, 1H), 7.93 (s, 4H), 7.45 (m, J =
7.5 Hz, 4H), 7.30 (m, J = 7.4 Hz, 2H). 13C NMR (125 MHz, Acetone-d6) δ
4.1.2.17. (Z)-2-Azido-1-(4-chlorophenyl)-3-(4-(trifluoromethyl) phenyl) 135.57, 128.44, 127.07, 125.87. HRMS (ESI) m/z calcd for C15H12N2O
prop-2-en-1-one (2q). Following general procedure afforded 2q as a [M+H]+: 237.1022. Found: 237.1025.
yellow solid (Yield: 68 %). 1H NMR (500 MHz, Chloroform-d) δ
7.93–7.88 (m, 2H), 7.79–7.74 (m, 2H), 7.64 (d, J = 8.2 Hz, 2H), 4.1.3.3. 3-([1,1′-biphenyl]-4-yl)-5-phenyl-1H-pyrazol-4-ol (4). Following
7.53–7.48 (m, 2H), 6.38 (s, 1H). HRMS (ESI) m/z calcd for general procedure afforded 4 as a white solid (Yield: 52 %). Mp>250 ◦ C.
1
C16H9ClF3N3O [M+Na]+: 374.0278. Found: 374.0281. H NMR (500 MHz, DMSO‑d6) δ 12.94 (s, 1H), 8.40 (s, 1H), 7.99 (d, J =
48.5 Hz, 4H), 7.75 (m, 4H), 7.47 (m, 4H), 7.34 (m, 2H). 13C NMR (125
4.1.2.18. (Z)-2-Azido-1-(4-chlorophenyl)-3-(4-fluorophenyl) prop-2-en-1- MHz, DMSO‑d6) δ 135.98, 129.46, 129.02, 128.92, 127.90, 127.43,
one (2r). Following general procedure afforded 2r as a yellow solid 127.20, 127.19, 126.97. HRMS (ESI) m/z calcd for C21H16N2O [M+H]+:
(Yield: 85 %). 1H NMR (500 MHz, Chloroform-d) δ 7.85–7.80 (m, 2H), 313.1335. Found: 313.1328.
7.77–7.71 (m, 2H), 7.50–7.46 (m, 2H), 7.11–7.06 (m, 2H), 6.38 (s, 1H).
HRMS (ESI) m/z calcd for C16H9ClF3N3O [M+Na]+: 324.0316. Found: 4.1.3.4. 3-(4-methoxyphenyl)-5-phenyl-1H-pyrazol-4-ol (5). Following
324.0311. general procedure afforded 5 as a yellow solid (Yield: 63 %). Mp
208–210 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.73 (s, 1H), 8.17 (s, 1H),
4.1.2.19. (Z)-2-Azido-1-(4-chlorophenyl)-3-(4-methoxyphenyl) prop-2- 7.97–7.81 (m, 4H), 7.44 (t, J = 7.6 Hz, 2H), 7.30 (t, J = 7.3 Hz, 1H), 7.03
en-1-one (2s). Following general procedure afforded 2s as a yellow (d, J = 8.4 Hz, 2H), 3.80 (s, 3H)⋅13C NMR (125 MHz, Acetone-d6) δ
solid (Yield: 48 %). 1H NMR (500 MHz, Chloroform-d) δ 7.82–7.77 (m, 159.03, 134.79, 128.37, 127.19, 126.93, 125.82, 113.85, 54.67. HRMS
2H), 7.74–7.68 (m, 2H), 7.50–7.45 (m, 2H), 6.94–6.90 (m, 2H), 6.41 (s, (ESI) m/z calcd for C16H14N2O2 [M+H]+: 267.1128. Found: 267.1133.
1H), 3.85 (s, 3H). HRMS (ESI) m/z calcd for C16H12ClN3O2 [M+Na]+:
336.0510. Found: 336.0510. 4.1.3.5. 3-(benzo[d] [1,3] dioxol-5-yl)-5-phenyl-1H-pyrazol-4-ol (6).
Following general procedure afforded 6 as a white solid (Yield: 59 %).
4.1.2.20. (Z)-2-Azido-1-(4-chlorophenyl)-3-(3,4-dimethoxyphenyl) prop- Mp 211–212 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.76 (s, 1H), 8.24 (s,
2-en-1-one (2t). Following general procedure afforded 2t as a yellow 1H), 7.90 (s, 2H), 7.44 (s, 4H), 7.29 (m, 1H), 7.01 (d, J = 7.8 Hz, 1H),
solid (Yield: 77 %). 1H NMR (500 MHz, Chloroform-d) δ 7.75–7.69 (m, 6.05 (s, 2H). 13C NMR (125 MHz, Acetone-d6) δ 147.88, 146.81, 135.00,
2H), 7.52 (d, J = 2.0 Hz, 1H), 7.50–7.45 (m, 2H), 7.33 (dd, J = 8.5, 2.0 128.43, 127.04, 125.83, 119.60, 108.24, 106.35, 101.12. HRMS (ESI)
Hz, 1H), 6.87 (d, J = 8.5 Hz, 1H), 6.39 (s, 1H), 3.93 (s, 6H). HRMS (ESI) m/z calcd for C16H12N2O3 [M+H]+: 281.0921. Found: 281.0921.
m/z calcd for C17H14ClN3O3 [M+Na]+: 366.0616. Found: 366.0621.
4.1.3.6. 5-Phenyl-3-(4-(trifluoromethyl) phenyl)-1H-pyrazol-4-ol (7).
4.1.2.21. (Z)-2-Azido-1-(4-chlorophenyl)-3-(naphthalen-1-yl) prop-2-en- Following general procedure afforded 7 as a white solid (Yield: 38 %).
1-one (2u). Following general procedure afforded 2u as a yellow solid Mp>250 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 13.13 (s, 1H), 8.59 (s, 1H),
(Yield: 47 %). 1H NMR (500 MHz, Chloroform-d) δ 8.30 (d, J = 7.3 Hz, 8.17 (d, J = 34.9 Hz, 2H), 7.99–7.74 (m, 4H), 7.47 (d, J = 7.8 Hz, 2H),
1H), 7.91–7.84 (m, 4H), 7.77–7.73 (m, 1H), 7.58–7.48 (m, 5H), 7.18 (s, 7.33 (m, J = 7.4 Hz, 1H). 13C NMR (125 MHz, Acetone-d6) δ 136.24,
1H). HRMS (ESI) m/z calcd for C19H12ClN3O [M+Na]+: 356.0561. 128.62, 127.43, 126.11, 125.89, 125.30, 125.27. HRMS (ESI) m/z calcd
Found: 356.0567. for C16H11F3N2O [M+H]+: 305.0896. Found: 305.0899.

4.1.2.22. (Z)-2-Azido-1-(4-chlorophenyl)-3-(1H-imidazole-5-yl) prop-2- 4.1.3.7. 3-(4-fluorophenyl)-5-phenyl-1H-pyrazol-4-ol (8). Following


en-1-one (2v). Following general procedure afforded 2v as a yellow general procedure afforded 8 as a white solid (Yield: 64 %). Mp
solid (Yield: 74 %). 1H NMR (500 MHz, DMSO‑d6) δ 12.63 (d, J = 40.9 236–237 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.89 (s, 1H), 8.34 (s, 1H),
Hz, 1H), 8.00 (s, 1H), 7.81 (s, 1H), 7.77–7.72 (m, 2H), 7.68–7.60 (m, 7.93 (d, J = 31.7 Hz, 4H), 7.45 (m, J = 7.6 Hz, 2H), 7.35–7.21 (m, 3H).
13
2H), 6.51 (s, 1H). HRMS (ESI) m/z calcd for C12H8ClN5O [M+H]+: C NMR (125 MHz, Acetone-d6) δ 135.31, 128.52, 127.87, 127.81,
274.0496. Found: 274.0493. 127.19, 125.81, 115.22, 115.05. HRMS (ESI) m/z calcd for C15H11FN2O
[M+H]+: 255.0928. Found: 255.0931.

8
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

4.1.3.8. 3-(4-chlorophenyl)-5-phenyl-1H-pyrazol-4-ol (9). Following 4.1.3.16. 5-(Furan-2-yl)-3-phenyl-1H-pyrazol-4-ol (17). Following gen­


general procedure afforded 9 as a white solid (Yield: 78 %). Mp eral procedure afforded 17 as a black solid (Yield: 50 %). Mp
230–231 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.97 (s, 1H), 8.43 (s, 1H), 189–190 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.93 (s, 1H), 8.38 (s, 1H),
7.93 (d, J = 35.4 Hz, 4H), 7.57–7.42 (m, 4H), 7.31 (t, J = 7.3 Hz, 1H). 7.92 (s, 2H), 7.73 (s, 1H), 7.45 (t, J = 7.3 Hz, 2H), 7.31 (t, J = 7.3 Hz,
13
C NMR (125 MHz, Acetone-d6) δ 135.61, 132.13, 128.52, 128.45, 1H), 6.77 (d, J = 3.1 Hz, 1H), 6.61 (s, 1H). 13C NMR (125 MHz, Acetone-
127.37, 127.24, 125.83. HRMS (ESI) m/z calcd for C15H11ClN2O d6) δ 141.49, 135.04, 128.42, 127.07, 125.66, 111.19, 105.89. HRMS
[M+H]+: 271.0633. Found: 271.0633. (ESI) m/z calcd for C13H10N2O2 [M+H]+: 227.0815. Found: 227.0819.

4.1.3.9. 3-(2-chlorophenyl)-5-phenyl-1H-pyrazol-4-ol (10). Following 4.1.3.17. 5-Butyl-3-phenyl-1H-pyrazol-4-ol (18). Following general pro
general procedure afforded 10 as a white solid (Yield: 96 %). Mp cedure afforded 18 as a yellow solid (Yield: 47 %). Mp 211–212 ◦ C. 1H
154–155 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.86 (d, J = 169.5 Hz, NMR (500 MHz, DMSO‑d6) δ 12.73 (s, 1H), 8.17 (s, 1H), 7.87 (s, 4H),
1H), 8.41 (s, 1H), 7.92 (d, J = 29.1 Hz, 2H), 7.72 (s, 1H), 7.58–7.48 (m, 7.43 (s, 2H), 7.29 (t, J = 7.1 Hz, 1H), 7.02 (s, 2H), 3.79 (s, 3H). 13C NMR
2H), 7.44 (s, 2H), 7.28 (t, J = 7.2 Hz, 1H). 13C NMR (125 MHz, Acetone- (125 MHz, Acetone-d6) δ 205.36, 159.02, 128.37, 127.19, 126.92,
d6) δ 136.79, 134.56, 134.08, 133.33, 129.23, 128.48, 127.10, 126.95, 125.81, 113.84, 54.67. HRMS (ESI) m/z calcd for C13H16N2O [M+H]+:
125.41. HRMS (ESI) m/z calcd for C16H11ClN2O [M+H]+: 271.0633. 217.1335. Found: 217.1344.
Found: 271.0636.
4.1.3.18. 3-(4-chlorophenyl)-1-methyl-5-phenyl-1H-pyrazol-4-ol (19). A
4.1.3.10. 3-(3-chlorophenyl)-5-phenyl-1H-pyrazol-4-ol (11). Following mixture of 2m (0.5 mmol), methylhydrazine sulfate (5 mmol) was stir­
general procedure afforded 11 as a white solid (Yield: 69 %). Mp red in acetonitrile/water (2 mL/0.2 mL) at room temperature. Then DBU
208–209 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 13.02 (s, 1H), 8.51 (s, 1H), (1.0 mmol) was added and the mixture was stirred for 5 h. The mixture
8.08–7.76 (m, 4H), 7.47 (m, J = 7.8 Hz, 3H), 7.39–7.28 (m, 2H). 13C was diluted with water (10 mL) and extracted three times with ethyl
NMR (125 MHz, Acetone-d6) δ 135.84, 133.90, 130.12, 128.58, 127.34, acetate (10 mL). The combined organic extracts were washed with brine,
126.77, 125.86, 125.51, 124.12. HRMS (ESI) m/z calcd for C16H11ClN2O dried over anhydrous Na2SO4, concentrated, and purified by column
[M+H]+: 271.0633. Found: 271.0633. chromatography using petroleum ether/ethyl acetate (10/1 to 5/1) as
eluent to afford 19 as a yellow solid (Yield: 68 %). Mp 172–173 ◦ C. 1H
4.1.3.11. 3-(2,4-dichlorophenyl)-5-phenyl-1H-pyrazol-4-ol (12). Follow NMR (500 MHz, DMSO‑d6) δ 8.33 (s, 1H), 7.98 (d, J = 8.1 Hz, 2H), 7.54
ing general procedure afforded 12 as a white solid (Yield: 96 %). Mp (d, J = 7.8 Hz, 4H), 7.47 (d, J = 8.5 Hz, 3H), 3.76 (s, 3H). 13C NMR (125
184–185 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.86 (d, J = 169.5 Hz, MHz, Acetone-d6) δ 136.87, 136.37, 132.62, 131.64, 129.65, 129.23,
1H), 8.41 (s, 1H), 7.92 (d, J = 29.1 Hz, 2H), 7.72 (s, 1H), 7.58–7.48 (m, 128.69, 128.30, 128.24, 127.31, 37.47. HRMS (ESI) m/z calcd for
2H), 7.44 (s, 2H), 7.28 (t, J = 7.2 Hz, 1H). 13C NMR (125 MHz, Acetone- C16H13ClN2O [M+H]+: 285.0789. Found: 285.0795.
d6) δ 136.79, 134.56, 134.08, 133.33, 129.23, 128.48, 127.10, 126.95,
125.41. HRMS (ESI) m/z calcd for C15H10Cl2N2O [M+H]+: 305.0243. 4.1.3.19. 3-(4-chlorophenyl)-1,5-diphenyl-1H-pyrazol-4-ol (20). A mix
Found: 305.0250. ture of 2m (0.5 mmol), phenyl hydrazine (5 mmol) was stirred in
acetonitrile/water (2 mL/0.2 mL) at room temperature. Then DBU (1.0
4.1.3.12. 5-(4-fluorophenyl)-3-phenyl-1H-pyrazol-4-ol (13). Following mmol) was added and the mixture was stirred for 5 h. The mixture was
general procedure afforded 13 as a white solid (Yield: 54 %). Mp diluted with water (10 mL) and extracted three times with ethyl acetate
231–232 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.90 (s, 1H), 8.36 (s, 1H), (10 mL). The combined organic extracts were washed with brine, dried
7.94 (d, J = 31.2 Hz, 4H), 7.45 (m, 2H), 7.30 (m, 3H). 13C NMR (125 over anhydrous Na2SO4, concentrated, and purified by column chro­
MHz, Acetone-d6) δ 135.32, 128.51, 127.89, 127.82, 127.19, 125.83, matography using petroleum ether/ethyl acetate (10/1 to 8/1) as eluent
115.22, 115.05. HRMS (ESI) m/z calcd for C15H11FN2O [M+H]+: to afford 20 as a yellow solid (Yield: 45 %). Mp 181–182 ◦ C. 1H NMR
255.0928. Found: 255.0930. (500 MHz, DMSO‑d6) δ 8.66 (s, 1H), 8.06 (d, J = 8.6 Hz, 2H), 7.52 (d, J
= 8.6 Hz, 2H), 7.42–7.24 (m, 10H). 13C NMR (125 MHz, DMSO‑d6) δ
4.1.3.13. 5-(4-chlorophenyl)-3-phenyl-1H-pyrazol-4-ol (14). Following 140.53, 140.42, 138.12, 132.32, 131.97, 131.88, 129.95, 129.41,
general procedure afforded 14 as a pale-yellow solid (Yield: 79 %). Mp 129.33, 129.01, 128.95, 128.53, 128.14, 127.63, 125.00. HRMS (ESI)
240–241 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.95 (s, 1H), 8.41 (s, 1H), m/z calcd for C21H15ClN2O [M+H]+: 347.0946. Found: 347.0950.
7.93 (d, J = 26.6 Hz, 4H), 7.48 (d, J = 21.0 Hz, 4H), 7.32 (d, J = 6.5 Hz,
1H). 13C NMR (125 MHz, Acetone-d6) δ 135.59, 132.13, 131.11, 128.52, 4.1.3.20. 3-(4-chlorophenyl)-1-methyl-5-phenyl-1H-pyrazol-4-amine
128.45, 127.37, 127.24, 125.83. HRMS (ESI) m/z calcd for C15H11ClN2O (21). A mixture of 2m (0.5 mmol) and methylhydrazine sulfate (0.5
[M+H]+: 271.0633. Found: 271.0635. mmol) was stirred in acetonitrile (2 mL) at room temperature. Then
NaOH (1.0 mmol) was added and the mixture was stirred for 8 h. The
4.1.3.14. 5-(4-bromophenyl)-3-phenyl-1H-pyrazol-4-ol (15). Following mixture was diluted with water (10 mL) and extracted three times with
general procedure afforded 15 as a yellow solid (Yield: 80 %). Mp ethyl acetate (10 mL). The combined organic extracts were washed with
222–223 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.98 (s, 1H), 8.43 (s, 1H), brine, dried over Na2SO4, concentrated, and purified by column chro­
7.90 (d, J = 48.8 Hz, 4H), 7.65 (d, J = 21.1 Hz, 2H), 7.46 (s, 2H), 7.32 (s, matography using petroleum ether/ethyl acetate (5/1) as eluent to
1H). 13C NMR (125 MHz, Acetone-d6) δ 135.62, 131.44, 128.53, 127.68, afford 21 as a yellow solid (Yield: 86 %). Mp 103–104 ◦ C. 1H NMR (500
127.27, 125.84, 120.29. HRMS (ESI) m/z calcd for C15H11BrN2O MHz, DMSO‑d6) δ 7.91–7.86 (m, 2H), 7.56 (t, J = 7.5 Hz, 2H), 7.51–7.43
[M+H]+: 315.0128. Found: 315.0128. (m, 5H), 3.80 (s, 2H), 3.73 (s, 3H). 13C NMR (125 MHz, CDCl3) δ 138.72,
132.81, 132.06, 131.89, 129.43, 129.36, 129.25, 128.91, 128.57,
4.1.3.15. 5-(4-methoxyphenyl)-3-phenyl-1H-pyrazol-4-ol (16). Following 127.84, 124.42, 37.62. HRMS (ESI) m/z calcd for C16H14ClN3 [M+H]+:
general procedure afforded 16 as a yellow solid (Yield: 61 %). Mp 284.0958. Found: 284.0954.
220–221 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.73 (s, 1H), 8.17 (s, 1H),
7.87 (s, 4H), 7.43 (s, 2H), 7.29 (t, J = 7.1 Hz, 1H), 7.02 (s, 2H), 3.79 (s, 4.1.3.21. 3-(4-chlorophenyl)-5-phenyl-1H-pyrazole (22). A mixture of
3H). 13C NMR (125 MHz, Acetone-d6) δ 205.36, 159.02, 128.37, 127.19, vinyl azide 2m (0.5 mmol) and 80 % hydrazine hydrate (2.5 mmol) was
126.92, 125.81, 113.84, 54.67. HRMS (ESI) m/z calcd for C16H14N2O2 stirred in acetonitrile (2 mL) at room temperature. Then DBU (0.5
[M+H]+: 267.1128. Found: 267.1135. mmol) was added and the mixture was stirred for 5 h. The mixture was

9
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

diluted with water (10 mL) and extracted three times with ethyl acetate 125.37, 125.34. HRMS (ESI) m/z calcd for C16H10ClF3N2O [M+H]+:
(10 mL). The combined organic extracts were washed with brine, dried 339.0507. Found: 339.0506.
over anhydrous Na2SO4, concentrated, and purified by column chro­
matography using petroleum ether/ethyl acetate (5/1) as eluent to 4.1.3.27. 3-(4-chlorophenyl)-5-(4-fluorophenyl)-1H-pyrazol-4-ol (28).
afford 22 as a white solid (Yield: 22 %). Mp 215–216 ◦ C. 1H NMR (500 Following general procedure afforded 28 as a white solid (Yield: 67 %).
MHz, DMSO‑d6) δ 13.45 (s, 1H), 7.85 (d, J = 22.6 Hz, 4H), 7.48 (s, 4H), Mp>250 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.97 (s, 1H), 8.45 (s, 1H),
7.36 (s, 1H), 7.23 (s, 1H). 13C NMR (125 MHz, Acetone-d6) δ 132.87, 7.94 (s, 4H), 7.52 (s, 2H), 7.30 (s, 2H)⋅13C NMR (125 MHz, Acetone-d6) δ
128.87, 128.79, 128.01, 126.89, 125.31, 99.71. HRMS (ESI) m/z calcd 135.43, 132.28, 128.55, 127.91, 127.84, 127.34, 115.36, 115.18. HRMS
for C15H11ClN2 [M+H]+: 255.0684. Found: 255.0694. (ESI) m/z calcd for C15H10ClFN2O [M+H]+: 289.0538. Found:
289.0541.
4.1.3.22. 3-(4-chlorophenyl)-5-(4-methoxyphenyl)-1H-pyrazol-4-ol
(23). Following general procedure afforded 23 as a white solid (Yield: 4.1.3.28. 3-(4-chlorophenyl)-5-(naphthalen-1-yl)-1H-pyrazol-4-ol (29).
54 %). Mp 239–240 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.78 (s, 1H), Following general procedure afforded 29 as a yellow solid (Yield: 62 %).
8.24 (s, 1H), 7.89 (d, J = 70.6 Hz, 4H), 7.49 (d, J = 7.2 Hz, 2H), 7.02 (d, Mp 180–181 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.98 (d, J = 132.0 Hz,
J = 8.0 Hz, 2H), 3.79 (s, 3H). 13C NMR (125 MHz, DMSO‑d6) δ 163.67, 1H), 8.42 (s, 1H), 8.03 (d, J = 29.3 Hz, 5H), 7.70–7.42 (m, 6H). 13C NMR
136.45, 132.44, 132.27, 132.09, 131.91, 60.38. HRMS (ESI) m/z calcd (125 MHz, Acetone-d6) δ 137.07, 133.93, 131.94, 131.85, 128.74,
for C16H13ClN2O2 [M+H]+: 301.0738. Found: 301.0738. 128.46, 128.22, 128.16, 127.14, 126.38, 126.20, 126.07, 125.42. HRMS
(ESI) m/z calcd for C19H13ClN2O [M+H]+: 321.0789. Found: 321.0788.
4.1.3.23. 3-(4-chlorophenyl)-5-(3,4-di-methoxyphenyl)-1H-pyrazol-4-ol
(24). Following general procedure afforded 24 as a white solid (Yield: 4.1.3.29. 3-(4-chlorophenyl)-5-(1H-imidazole-5-yl)-1H-pyrazol-4-ol
65 %). Mp 221–222 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.84 (s, 1H), (30). Following general procedure using dichloromethane/methanol
8.35 (s, 1H), 7.98 (s, 2H), 7.49 (s, 4H), 7.04 (d, J = 7.7 Hz, 1H), 3.80 (d, (100/1) as eluent afforded 30 as a yellow solid (Yield: 45 %). Mp>
J = 12.9 Hz, 6H). 13C NMR (125 MHz, Acetone-d6) δ 149.49, 148.98, 250 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.75 (s, 1H), 12.36 (s, 1H),
135.04, 132.03, 128.42, 127.37, 118.48, 111.91, 109.74, 55.22, 55.16. 8.01–7.90 (m, 2H), 7.84 (s, 1H), 7.48 (d, J = 8.0 Hz, 2H), 7.41 (s, 1H).
HRMS (ESI) m/z calcd for C17H15ClN2O3 [M+H]+: 331.0844. Found: 13
C NMR (125 MHz, DMSO‑d6) δ 140.50, 140.43, 136.12, 131.79,
331.0846. 131.75, 131.66, 131.60. HRMS (ESI) m/z calcd for C12H9ClN4O
[M+H]+: 261.0538. Found: 261.0528.
4.1.3.24. 3-(4-chlorophenyl)-5-(4-hydroxylphenyl)-1H-pyrazol-4-ol
(25). Under N2 atmosphere, 23 (0.5 mM) was stirred in dried CH2Cl2 (5 4.1.3.30. Methyl 4-(3-(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl) ben­
mL) at 0 ◦ C, then BBr3 in CH2Cl2 (2.5 mM, 1.0 M) was added slowly. The zoate (31). Following general procedure afforded 31 as a white solid
reaction was allowed to continue at room temperature for another 3 h. (Yield: 65 %). Mp 237–238 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 13.18 (s,
Then water (20 mL) was added and the mixture was extracted with ethyl 1H), 8.69 (s, 1H), 8.01 (m, 6H), 7.53 (s, 2H), 3.87 (s, 3H). 13C NMR (125
acetate (20 mL × 2). The combined organic extracts were washed with MHz, DMSO‑d6) δ 141.63, 136.82, 134.75, 133.87, 132.97, 132.61,
saturated NaHCO3 (aq.) and brine. The organic phase was dried over 132.48, 132.08, 130.34, 130.10, 57.27. HRMS (ESI) m/z calcd for
anhydrous Na2SO4, concentrated, and purified by column chromatog­ C17H13ClN2O3 [M+H]+: 329.0687. Found: 329.0682.
raphy using petroleum ether/ethyl acetate (3/1 to 2/1) as eluent to
afford compound 25 as a white solid (Yield: 54 %). Mp 240–241 ◦ C. 1H 4.1.3.31. 4-(3-(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl) benzoic acid
NMR (500 MHz, DMSO‑d6) δ 12.73 (s, 1H), 9.57 (s, 1H), 8.20 (s, 1H), hydrochloride (32). In a 50 mL round bottom flask, methyl 4-(3-(4-
7.95 (s, 2H), 7.69 (s, 2H), 7.49 (d, J = 7.8 Hz, 2H), 6.84 (d, J = 8.4 Hz, chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl) benzoate (31) (2 mmol) was
2H). 13C NMR (125 MHz, DMSO‑d6) δ 157.14, 131.61, 128.91, 127.59, stirred in a solution of concentrated hydrochloride (20 mL) under reflux
127.52, 127.47, 127.35, 115.82. HRMS (ESI) m/z calcd for condition for 24 h. The mixture was filtered under vacuum, washed with
C15H11ClN2O2 [M+H]+: 287.0582. Found: 287.0588. water, and dried to yield the crude product, which was further purified
by treating with ethyl acetate to afford 32 as a white solid (Yield: 82 %).
4.1.3.25. 3-(4-chlorophenyl)-5-(3,4-di-hydroxylphenyl)-1H-pyrazol-4-ol Mp>250 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 13.06 (s, 2H), 8.66 (s, 1H),
(26). Under N2 atmosphere, 24 (0.5 mM) was stirred in dried CH2Cl2 (5 8.03 (m, 4H), 7.94 (d, J = 8.1 Hz, 2H), 7.53 (d, J = 8.2 Hz, 2H). 13C NMR
mL) at 0 ◦ C, and then BBr3 in CH2Cl2 (2.5 mM, 1.0 M) was added slowly. (125 MHz, DMSO‑d6) δ 167.77, 136.75, 132.00, 130.13, 129.71,
The reaction was allowed to continue at room temperature for another 3 129.09, 127.55, 125.56. HRMS (ESI) m/z calcd for C16H11ClN2O3
h. Then water (20 mL) was added and the mixture was extracted with [M+H]+: 315.0531. Found: 315.0533.
ethyl acetate (20 mL × 2). The combined organic extracts were washed
with saturated NaHCO3 (aq.) and brine, dried over anhydrous Na2SO4, 4.1.3.32. 4-(3-(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl)-N-ethyl­
concentrated, and purified by column chromatography using petroleum benzamide (33). In a 25 mL round bottom flask, 4-(3-(4-chlorophenyl)-
ether/ethyl acetate (3/1 to 1/1) as eluent to afford 26 as a gray solid 4-hydroxy-1H-pyrazol-5-yl) benzoic acid hydrochloride (32) (176 mg,
(Yield: 65 %). Mp>250 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 12.65 (s, 0.5 mmol) and HATU (285 mg, 0.75 mmol) was dissolved in dried DMF
1H), 8.97 (s, 2H), 8.15 (s, 1H), 7.94 (s, 2H), 7.48 (d, J = 7.7 Hz, 2H), (3 mL) for 10 min. Then ethylamine hydrochloride (49 mg, 0.6 mmol)
7.32 (s, 1H), 7.11 (s, 1H), 6.83–6.75 (m, 1H). 13C NMR (125 MHz, and triethylamine (139 μL, 1.0 mmol) were added after the mixture was
DMSO‑d6) δ 145.69, 145.31, 134.87, 131.58, 128.90, 117.46, 116.13, cooling to 0 ◦ C. The reaction was warmed to room temperature and
113.84. HRMS (ESI) m/z calcd for C15H11ClN2O3 [M+H]+: 303.0531. stirred for 3 h. Then water (10 mL) was added and the mixture was
Found: 303.0529. extracted three times with ethyl acetate (10 mL). The combined organic
phases were washed with saturated NaHCO3 (aq.) and brine, dried over
4.1.3.26. 3-(4-chlorophenyl)-5-(4-(trifluoromethyl)phenyl)-1H-pyrazol-4- anhydrous Na2SO4, concentrated, and purified by column chromatog­
ol (27). Following general procedure afforded 27 as a white solid raphy using petroleum ether/ethyl acetate (1/2) as eluent to afford 33 as
(Yield: 57 %). Mp 247–248 ◦ C. 1H NMR (500 MHz, DMSO‑d6) δ 13.20 (s, a pale-yellow solid (Yield: 71 %). Mp>250 ◦ C. 1H NMR (500 MHz,
1H), 8.70 (s, 1H), 8.23–7.77 (m, 6H), 7.54 (d, J = 20.8 Hz, 2H). 13C NMR DMSO‑d6) δ 13.08 (s, 1H), 8.53 (d, J = 48.9 Hz, 2H), 7.95 (m, 6H), 7.52
(125 MHz, Acetone-d6) δ 136.40, 132.55, 128.64, 127.44, 126.13,

10
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

(s, 2H), 3.34–3.26 (m, 2H), 1.15 (t, J = 7.2 Hz, 3H). 13C NMR (125 MHz, H9c2 cells were cultured in DMEM (Gibco) media; BJ cell was cultured
DMSO‑d6) δ 168.21, 136.46, 133.35, 131.88, 129.16, 128.98, 127.90, in α-MEM (Gibco) media with 1 % 100 × nonessential amino acid so­
127.05, 125.71, 124.93, 34.52, 15.34. HRMS (ESI) m/z calcd for lution, 1 mM sodium pyruvate. All media were supplemented with 10 %
C18H16ClN3O2 [M+H]+: 342.1004. Found: 342.1000. fetal bovine serum (Yeasen) and 1 % penicillin/streptomycin (Corning).
Cells were passaged by dissociation with 0.05 % trypsin - 0.2 % EDTA
4.1.3.33. 4-(3-(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl)-N-(3-(dime­ (Gibco) every other day.
thylamino)propyl)benza-mide (34). In a 25 mL round bottom flask, 4-(3-
(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl) benzoic acid hydrochlo­ 4.2.2. Cell viability assay
ride (32) (176 mg, 0.5 mmol) and HATU (285 mg, 0.75 mmol) was For cellular viability assays, cells were seeded in 96-well plates
dissolved in dried DMF (3 mL) for 10 min. Then 3-dimethylaminopro­ (Corning) at 2000–5000 cells per well. Then 24 h after seeding, cells
pylamine (76 μL, 0.6 mmol) and triethylamine (83 μL, 0.6 mmol) were treated with compounds at the indicated concentrations for 24 h.
were added after cooling to 0 ◦ C. The reaction was warmed to room Cellular ATP levels were quantified using CellTiter-Glo (Promega).
temperature and stirred for 3 h. Then water (10 mL) was added and the Relative viability was normalized to the respective DMSO-treated con­
mixture was extracted three times with ethyl acetate (10 mL). The dition. For data presentation, the mean and standard deviation for the
combined organic phases were washed with saturated NaHCO3 (aq.) and three biological replicates of each data point in a representative exper­
brine, dried over anhydrous Na2SO4, concentrated, and purified by iment were plotted in Prism 8 (Graphpad) and presented. Sigmoidal
column chromatography using petroleum ether/ethyl acetate (1/3) as nonlinear regression models were used to compute the regression fit
eluent to afford 34 as a yellow solid (Yield: 67 %). Mp 240–241 ◦ C. 1H curves.
NMR (500 MHz, DMSO‑d6) δ 12.95 (s, 1H), 8.51 (t, J = 5.3 Hz, 1H), 7.98
(dd, J = 22.7, 8.2 Hz, 4H), 7.88 (d, J = 8.3 Hz, 2H), 7.47 (d, J = 8.3 Hz, 4.2.3. Morphological observation
2H), 3.29 (q, J = 6.6 Hz, 2H), 2.26 (t, J = 7.0 Hz, 2H), 2.14 (s, 6H), For cell morphology observation, 5000 786-O cells were seeded per
1.70–1.63 (m, 2H). 13C NMR (125 MHz, DMSO‑d6) δ 166.24, 133.06, well in 96-well plates. 1 μM RSL3 mixed with different concentration of
131.64, 128.93, 127.81, 127.40, 125.18, 57.47, 45.69, 38.27, 27.62. compound 25 and Fer-1 (1 μM,0.0625 μM) were added 24 h after
HRMS (ESI) m/z calcd for C21H23ClN4O2 [M+H]+: 399.1582. Found: seeding. After one day of incubation, cells were observed by light
399.1584. microscopy.

4.1.3.34. 4-(3-(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl)-N-((tetrahy­ 4.2.4. DPPH assay


dro-2H-pyran-2-yl) oxy) benzamide (35a). In a 25 mL round bottom DPPH solution (498 μL, 40.2 μM) in methanol was mixed with the
flask, 4-(3-(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl) benzoic acid tested compound (2 μL of compound solution in DMSO). The final re­
hydrochloride (32) (176 mg, 0.5 mmol) and HATU (285 mg, 0.75 mmol) action system contained 40 μM DPPH and different concentrations of
was dissolved in dried DMF (3 mL) for 10 min. Then O-(tetrahydro-2H- target compounds. The zero control (DMSO) and blank control (meth­
pyran-2-yl) hydroxylamine (70 mg, 0.6 mmol) and triethylamine (83 μL, anol) were set up. After incubating at room temperature for 1 h, the
0.6 mmol) were added after cooling to 0 ◦ C. The reaction was warmed to absorbance was measured at 517 nm, and the percentage scavenging
room temperature and stirred for 3 h. Then water (10 mL) was added was calculated as follows: Scavenging % = [Ab (DMSO)-Ab (sample solu­
and the mixture was extracted three times with ethyl acetate (10 mL). tion)]/[Ab (DMSO)-Ab (methanol)] × 100 %
The combined organic phases were washed with saturated NaHCO3 (aq.)
and brine, dried over anhydrous Na2SO4, concentrated, and purified by 4.2.5. ABTS assay
column chromatography using petroleum ether/ethyl acetate (1/1) as 2,2′-azinobis-(3-ethylbenzothiazoline-6-sulphonic acid diammonium
eluent to afford 35a as a white solid (Yield: 59 %), Mp>250 ◦ C. 1H NMR salt (ABTS) was dissolved in double-distilled water to 14 mM. ABTS•+
(500 MHz, DMSO‑d6) δ 12.95 (s, 1H), 8.01 (d, J = 8.2 Hz, 2H), was produced by reacting ABTS stock solution with potassium persulfate
7.99–7.94 (m, 2H), 7.82 (d, J = 8.4 Hz, 2H), 7.47 (d, J = 8.3 Hz, 2H), (2.45 mM, final concentration) and allowing the mixture to incubate in
5.01 (s, 1H), 4.10–4.00 (m, 1H), 3.53 (m, 2H), 1.73 (m, 3H), 1.56 (m, the dark at room temperature for 18 h. The solution of ABTS•+ was
3H). 13C NMR (125 MHz, DMSO‑d6) δ 128.86, 127.81, 127.30, 125.01, diluted with sodium phosphate buffer (50 mM, pH 7.4) to 100 μM.
101.41, 61.98, 28.59, 25.29, 19.06. HRMS (ESI) m/z calcd for Tested compounds (1–100 μM, 50 μL) were added to 150 μL of 100 μM
C21H20ClN3O4 [M+H]+: 414.1215. Found: 414.1208. ABTS•+ solution. The optical absorbance of ABTS•+ at 415 nm was
measured after incubated at 30 ◦ C for 6 min.
4.1.3.35. 4-(3-(4-chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl)-N-hydrox­
ybenzamide (35). To a solution of hydrochloric in methanol, 4-(3-(4- 4.2.6. Measurement of lipid ROS
chlorophenyl)-4-hydroxy-1H-pyrazol-5-yl)-N-((tetrahydro-2H-pyran-2- HT-1080 cells were cultured in a 6-well plate at the density of 4 ×
yl) oxy) benzamide (35a) was added and the reaction was stirred at 105/well overnight, which made the cells adherent. Cells were co-
room temperature overnight. The mixture was concentrated. The res­ treated with ferroptosis inducer (0.8 μM RSL3) and either vehicle
idue was washed with saturated NaHCO3 (aq.) and brine, dried over (DMSO) or a ferroptosis inhibitor (1 μM Fer-1 or 1 μM compound 25),
anhydrous Na2SO4, concentrated, and purified by column chromatog­ then incubated for 8 h. The medium was discarded and the cells were
raphy using petroleum ether/ethyl acetate (1/2) as eluent to afford 35 as washed twice with PBS. Treated cells were incubated with BODIPY lipid
a white solid (Yield: 88 %), Mp>250 ◦ C. 1H NMR (500 MHz, DMSO‑d6) probe 581/591C11 (Invitrogen, USA) at a final concentration of 5 μM at
δ 11.37 (s, 1H), 8.81 (s, 3H), 8.04–7.97 (m, 4H), 7.85 (d, J = 8.5 Hz, 2H), 37 ◦ C for 1 h. Following incubations, cells were harvested for FITC
7.54–7.50 (m, 2H). 13C NMR (125 MHz, DMSO‑d6) δ 164.32, 136.44, fluorescence imaging and analysis to measure the intracellular lipid ROS
132.03, 131.47, 129.03, 127.70, 127.66, 125.61. HRMS (ESI) m/z calcd level.
for C16H12ClN3O3 [M+H]+: 330.0640. Found: 330.0645.
4.2.7. Measurement of intracellular ROS
DCFH-DA, a total ROS assay kit (Solarbio, Beijing), was used to assess
4.2. Biology the intracellular ROS generation. Briefly, HT-1080 cells were seeded on
glass coverslips at the density of 4 × 105/well overnight, which made
4.2.1. Cell lines and culture conditions the cells adherent. Cells were co-treated with ferroptosis inducer (5 μM
All cells were cultured in a humidified incubator at 37 ◦ C with 5 % Erastin) and either vehicle (DMSO) or a ferroptosis inhibitor (1 μM Fer-1
CO2. 786-O cell was cultured in RPMI-1640 (Gibco) media; HT-1080 and or 1 μM compound 25), then incubated for 24 h. The medium was

11
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

discarded and the cells were washed twice with PBS. Treated cells were [9] B.R. Stockwell, X. Jiang, The chemistry and biology of ferroptosis, Cell Chem. Biol.
27 (2020) 365–375, https://doi.org/10.1016/j.chembiol.2020.03.013.
incubated with DCFH-DA probe at a final concentration of 10 μM at
[10] X. Fang, H. Ardehali, J. Min, F. Wang, The molecular and metabolic landscape of
37 ◦ C for 20 min. Then, the medium was removed. Cells were washed iron and ferroptosis in cardiovascular disease, Nat. Rev. Cardiol. 20 (2023) 7–23,
with PBS three times, and finally observed by CLSM. https://doi.org/10.1038/s41569-022-00735-4.
[11] J. Chen, X. Li, C. Ge, J. Min, F. Wang, The multifaceted role of ferroptosis in liver
disease, Cell Death Differ. 29 (2022) 467–480, https://doi.org/10.1038/s41418-
4.2.8. Iron chelating assay 022-00941-0.
The chelating interaction of compounds with FeCl3 or FeSO4 was [12] X. Fang, Z. Cai, H. Wang, D. Han, Q. Cheng, P. Zhang, F. Gao, Y. Yu, Z. Song,
determined using UV–vis spectrophotometer TU-1810. To investigate Q. Wu, P. An, S. Huang, J. Pan, H.Z. Chen, J. Chen, A. Linkermann, J. Min,
F. Wang, Loss of cardiac ferritin H facilitates cardiomyopathy via slc7a11-mediated
the iron ion binding ability, the tested compounds were dissolved in ferroptosis, Circ. Res. 127 (2020) 486–501, https://doi.org/10.1161/
MeOH at a concentration of 20 mM, followed by mixing with FeCl3 or CIRCRESAHA.120.316509.
FeSO4 at a concentration of 10 mM. Then, the absorbance value was [13] Y. Jin, Y. Zhuang, M. Liu, J. Che, X. Dong, Inhibiting ferroptosis: a novel approach
for stroke therapeutics, Drug Discov. Today 26 (2021) 916–930, https://doi.org/
measured after dilution with MeOH/H2O. 10.1016/j.drudis.2020.12.020.
[14] Y. Gu, Y. Li, J. Wang, L. Zhang, J. Zhang, Y. Wang, Targeting ferroptosis: paving
4.2.9. Statistical analysis new roads for drug design and discovery, Eur. J. Med. Chem. 247 (2023), 115015,
https://doi.org/10.1016/j.ejmech.2022.115015.
Statistical analyses were performed by Prism 9 (GraphPad Software). [15] X. Wang, Y. Zhou, J. Min, F. Wang, Zooming in and out of ferroptosis in human
For statistical comparison, we performed two-tailed Student’s t-test to disease, Front. Med. 17 (2023) (2023) 173–206, https://doi.org/10.1007/s11684-
analyze two groups with normally distributed data and one-way ANOVA 023-0992-z.
[16] R. Skouta, S.J. Dixon, J. Wang, D.E. Dunn, M. Orman, K. Shimada, P.A. Rosenberg,
for multiple comparisons with Gaussian distribution. P < 0.05 was
D.C. Lo, J.M. Weinberg, A. Linkermann, B.R. Stockwell, Ferrostatins inhibit
considered significant (represented as × p < 0.05, **p < 0.01, ***p < oxidative lipid damage and cell death in diverse disease models, J. Am. Chem. Soc.
0.001, ****p < 0.0001). 136 (2014) 4551–4556, https://doi.org/10.1021/ja411006a.
[17] O. Zilka, R. Shah, B. Li, J.P. Friedmann Angeli, M. Griesser, M. Conrad, D.A. Pratt,
On the mechanism of cytoprotection by ferrostatin-1 and liproxstatin-1 and the
role of lipid peroxidation in ferroptotic cell death, ACS Cent. Sci. 3 (2017)
Declaration of competing interest 232–243, https://doi.org/10.1021/acscentsci.7b00028.
[18] G. Miotto, M. Rossetto, M.L. Di Paolo, L. Orian, R. Venerando, A. Roveri, A.
M. Vuckovic, V. Bosello Travain, M. Zaccarin, L. Zennaro, M. Maiorino, S. Toppo,
The authors declare that they have no known competing financial
F. Ursini, G. Cozza, Insight into the mechanism of ferroptosis inhibition by
interests or personal relationships that could have appeared to influence ferrostatin-1, Redox Biol. 28 (2020), 101328, https://doi.org/10.1016/j.
the work reported in this paper. redox.2019.101328.
[19] Omkar Zilka, Jia-Fei Poon, Derek A. Pratt, Radical-trapping antioxidant activity of
copper and nickel bis(thiosemicarbazone) complexes underlies their potency as
Data availability inhibitors of ferroptotic cell death, J. Am. Chem. Soc. 143 (2021) 19043–19057,
https://doi.org/10.1021/jacs.1c08254.
Data will be made available on request. [20] A. Southon, K. Szostak, K.M. Acevedo, K.A. Dent, I. Volitakis, A.A. Belaidi, K.
J. Barnham, P.J. Crouch, S. Ayton, P.S. Donnelly, A.I. Bush, Cu(II) (atsm) inhibits
ferroptosis: implications for treatment of neurodegenerative disease, Br. J.
Acknowledgments Pharmacol. 177 (2020) 656–667, https://doi.org/10.1111/bph.14881.
[21] K.U. Ingold, D.A. Pratt, Advances in radical-trapping antioxidant chemistry in the
21st century: a kinetics and mechanisms perspective, Chem. Rev. 114 (2014)
This project was supported from the Zhejiang Provincial Natural 9022–9046, https://doi.org/10.1021/cr500226n.
Science Foundation of China under Grant No. LY20H300001 (Yongping [22] E. Sassetti, M.H. Clausen, L. Laraia, Small-molecule inhibitors of reactive oxygen
species production, J. Med. Chem. 64 (2021) 5252–5275, https://doi.org/
Yu).
10.1021/acs.jmedchem.0c01914.
[23] R. Shah, M.S. Shchepinov, D.A. Pratt, Resolving the role of lipoxygenases in the
Appendix A. Supplementary data initiation and execution of ferroptosis, ACS Cent. Sci. 4 (2018) 387–396, https://
doi.org/10.1021/acscentsci.7b00589.
[24] R. Shah, K. Margison, D.A. Pratt, The potency of diarylamine radical-trapping
Supplementary data to this article can be found online at https://doi. antioxidants as inhibitors of ferroptosis underscores the role of autoxidation in the
org/10.1016/j.ejmech.2023.115913. mechanism of cell death, ACS Chem. Biol. 12 (2017) 2538–2545, https://doi.org/
10.1021/acschembio.7b00730.
[25] J. Liu, I. Bandyopadhyay, L. Zheng, O.M. Khdour, S.M. Hecht, Antiferroptotic
References activity of phenothiazine analogues: a novel therapeutic strategy for oxidative
stress related disease, ACS Med. Chem. Lett. 11 (2020) 2165–2173, https://doi.
org/10.1021/acsmedchemlett.0c00293.
[1] S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev, C.E. Gleason,
[26] W. Yang, X. Liu, C. Song, S. Ji, J. Yang, Y. Liu, J. You, J. Zhang, S. Huang,
D.N. Patel, A.J. Bauer, A.M. Cantley, W.S. Yang, B. Morrison 3rd, B.R. Stockwell,
W. Cheng, Z. Shao, L. Li, S. Yang, Structure-activity relationship studies of
Ferroptosis: an iron-dependent form of nonapoptotic cell death, Cell 149 (2012)
phenothiazine derivatives as a new class of ferroptosis inhibitors together with the
1060–1072, https://doi.org/10.1016/j.cell.2012.03.042.
therapeutic effect in an ischemic stroke model, Eur. J. Med. Chem. 209 (2021),
[2] W.S. Yang, R. SriRamaratnam, M.E. Welsch, K. Shimada, R. Skouta, V.
112842, https://doi.org/10.1016/j.ejmech.2020.112842.
S. Viswanathan, J.H. Cheah, P.A. Clemons, A.F. Shamji, C.B. Clish, L.M. Brown, A.
[27] T. Krainz, M.M. Gaschler, C. Lim, J.R. Sacher, B.R. Stockwell, P. Wipf,
W. Girotti, V.W. Cornish, S.L. Schreiber, B.R. Stockwell, Regulation of ferroptotic
A mitochondrial-targeted nitroxide is a potent inhibitor of ferroptosis, ACS Cent.
cancer cell death by GPX4, Cell 156 (2014) 317–331, https://doi.org/10.1016/j.
Sci. 2 (2016) 653–659, https://doi.org/10.1021/acscentsci.6b00199.
cell.2013.12.010.
[28] M. Griesser, R. Shah, A.T. Van Kessel, O. Zilka, E.A. Haidasz, D.A. Pratt, The
[3] Y. Yu, L. Jiang, H. Wang, Z. Shen, Q. Cheng, P. Zhang, J. Wang, Q. Wu, X. Fang,
catalytic reaction of nitroxides with peroxyl radicals and its relevance to their
L. Duan, S. Wang, K. Wang, P. An, T. Shao, R.T. Chung, S. Zheng, J. Min, F. Wang,
cytoprotective properties, J. Am. Chem. Soc. 140 (2018) 3798–3808, https://doi.
Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis,
org/10.1021/jacs.8b00998.
Blood 136 (2020) 726–739, https://doi.org/10.1182/blood.2019002907.
[29] R.L. Jones, Utility of dexrazoxane for the reduction of anthracycline-induced
[4] J. Li, F. Cao, H.L. Yin, Z.J. Huang, Z.T. Lin, N. Mao, B. Sun, G. Wang, Ferroptosis:
cardiotoxicity, Expert Rev. Cardiovasc Ther. 6 (2008) 1311–1317, https://doi.org/
past, present and future, Cell Death Dis. 11 (2020) 88, https://doi.org/10.1038/
10.1586/14779072.6.10.1311.
s41419-020-2298-2.
[30] X. Fang, H. Wang, D. Han, E. Xie, X. Yang, J. Wei, S. Gu, F. Gao, N. Zhu, X. Yin,
[5] X. Yang, N.K. Kawasaki, J. Min, T. Matsui, F. Wang, Ferroptosis in heart failure,
Q. Cheng, P. Zhang, W. Dai, J. Chen, F. Yang, H.T. Yang, A. Linkermann, W. Gu,
J. Mol. Cell. Cardiol. 173 (2022) 141–153, https://doi.org/10.1016/j.
J. Min, F. Wang, Ferroptosis as a target for protection against cardiomyopathy,
yjmcc.2022.10.004.
Proc. Natl. Acad. Sci. U. S. A. 116 (2019) 2672–2680, https://doi.org/10.1073/
[6] E. Solary, L. Dubrez, B. Eymin, The role of apoptosis in the pathogenesis and
pnas.1821022116.
treatment of diseases, Eur. Respir. J. 9 (1996) 1293–1305, https://doi.org/
[31] X. Yao, Y. Zhang, B.-Y. Fan, Y.-L. Pang, W.-Y. Shen, X. Wang, C.-X. Zhao, W.-X. Li,
10.1183/09031936.96.09061293.
C. Liu, X.-H. Kong, G.-Z. Ning, S.-Q. Feng, Neuroprotective effect of deferoxamine
[7] J. Seo, Y.W. Nam, S. Kim, D.B. Oh, J. Song, Necroptosis molecular mechanisms:
on erastin-induced ferroptosis in primary cortical neurons, Neural Regen. Res. 15
recent findings regarding novel necroptosis regulators, Exp. Mol. Med. 53 (2021)
(2020) 1539–1545, https://doi.org/10.4103/1673-5374.274344.
1007–1017, https://doi.org/10.1038/s12276-021-00634-7.
[32] A. Rayatpour, F. Foolad, M. Heibatollahi, K. Khajeh, M. Javan, Ferroptosis
[8] M. Conrad, D.A. Pratt, The chemical basis of ferroptosis, Nat. Chem. Biol. 15 (2019)
inhibition by deferiprone, attenuates myelin damage and promotes
1137–1147, https://doi.org/10.1038/s41589-019-0408-1.

12
D. Ying et al. European Journal of Medicinal Chemistry 263 (2024) 115913

neuroprotection in demyelinated optic nerve, Sci. Rep. 12 (2022), 19630, https:// [37] M.H. Larraufie, W.S. Yang, E. Jiang, A.G. Thomas, B.S. Slusher, B.R. Stockwell,
doi.org/10.1038/s41598-022-24152-2. Incorporation of metabolically stable ketones into a small molecule probe to
[33] U.D. Newar, S. Borra, R.A. Maurya, Visible-light 2,4-dinitrophenol-mediated increase potency and water solubility, Bioorg. Med. Chem. Lett. 25 (2015)
photoannulation of α-azidochalcones into 2,5-diaryloxazoles, Org. Lett. 24 (2022) 4787–4792, https://doi.org/10.1016/j.bmcl.2015.07.018.
4454–4458, https://doi.org/10.1021/acs.orglett.2c01691. [38] M. Conlon, C.D. Poltorack, G.C. Forcina, D.A. Armenta, M. Mallais, M.A. Perez,
[34] W. Huang, S. Liu, B. Chen, X. Guo, Y. Yu, Synthesis of polysubstituted 4-amino­ A. Wells, A. Kahanu, L. Magtanong, J.L. Watts, D.A. Pratt, S.J. Dixon,
pyrazoles and 4-hydroxypyrazoles from vinyl azides and hydrazines, RSC Adv. 5 A compendium of kinetic modulatory profiles identifies ferroptosis regulators, Nat.
(2015) 32740–32743, https://doi.org/10.1039/C5RA04371A. Chem. Biol. 17 (2021) 665–674, https://doi.org/10.1038/s41589-021-00751-4.
[35] Y. Zhang, H. Tan, J.D. Daniels, F. Zandkarimi, H. Liu, L.M. Brown, K. Uchida, O. [39] R. Sheng, L. Tang, L. Jiang, L. Hong, Y. Shi, N. Zhou, Y. Hu, Novel 1-Phenyl-3-
A. O’Connor, B.R. Stockwell, Imidazole ketone erastin induces ferroptosis and hydroxy-4-pyridinone derivatives as multifunctional agents for the therapy of
slows tumor growth in a mouse lymphoma model, Cell Chem. Biol. 26 (2019) alzheimer’s disease, ACS Chem. Neurosci. 7 (2016) 69–81, https://doi.org/
623–633, https://doi.org/10.1016/j.chembiol.2019.01.008. 10.1021/acschemneuro.5b00224.
[36] Y. Du, H.C. Zhao, H.C. Zhu, Y. Jin, L. Wang, Ferroptosis is involved in the anti- [40] H. Zhu, J. Cen, C. Hong, H. Wang, Y. Wen, Q. He, Y. Yu, J. Cao, W. Chen, Targeting
tumor effect of lycorine in renal cell carcinoma cells, Oncol. Lett. 22 (2021) 781, labile iron-mediated ferroptosis provides a potential therapeutic strategy for
https://doi.org/10.3892/ol.2021.13042. rhabdomyolysis-induced acute kidney injury, ACS Chem. Biol. 18 (2023)
1294–1304, https://doi.org/10.1021/acschembio.2c00914.

13

You might also like