Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

European Journal of Medicinal Chemistry 264 (2024) 116037

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


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

Review article

Click chemistry-aided drug discovery: A retrospective and


prospective outlook
Rui Zhao a, b, 1, Junlong Zhu a, b, 1, Xiaoying Jiang a, b, Renren Bai a, b, *
a
School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China
b
Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines,
Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China

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

Keywords: Click chemistry has emerged as a valuable tool for rapid compound synthesis, presenting notable advantages and
Click chemistry convenience in the exploration of potential drug candidates. In particular, in situ click chemistry capitalizes on
In situ click chemistry enzymes as reaction templates, leveraging their favorable conformation to selectively link individual building
Compound library
blocks and generate novel hits. This review comprehensively outlines and introduces the extensive use of click
1,2,3-Triazole
chemistry in compound library construction, and hit and lead discovery, supported by specific research exam­
Drug discovery
ples. Additionally, it discusses the limitations and precautions associated with the application of click chemistry
in drug discovery. Our intention for this review is to contribute to the development of a modular synthetic
approach for the rapid identification of drug candidates.

1. Introduction is frequently applied as a linker of proteolysis targeting chimeras


(PROTACs) [17]. More importantly, large or small compound libraries
Researchers have always been trying to find reliable solutions to can be quickly constructed through click chemistry reactions, providing
break through technological barriers and thus shorten the drug discov­ a large library for screening active compounds [18]. For a specific drug
ery process for decades. Fortunately, the emergence and ongoing target, click chemistry combines two similar or different pharmacophore
development of artificial intelligence (AI), computer-aided drug design groups through a specific skeleton, the triazole heterocyclic ring, to
(CADD), and click chemistry may improve the efficiency and accelerate obtain a compound with high affinity (Fig. 2) [19,20]. This approach
the process of drug discovery (Fig. 1) [1–5]. also makes click chemistry a viable lead compound discovery strategy
Click chemistry is a combinatorial method initially proposed by [21]. If lead compounds or pharmacophores are unavailable for a certain
Professor Sharpless for the rapidly synthesis C-X-C atoms [6]. The most target like enzyme, in situ click chemistry can be used to directly
widely used click chemical reaction in organic chemistry, chemical generate hits with research value in the binding pocket of the target. The
biology, and biomedical fields is the Husigen 1,3-dipole cycloaddition process of synthesizing and screening enzyme inhibitors can be
reaction (CuAAC) between azides and acetylene. The reaction has streamlined [22].
become a classic click chemistry reaction [7–10]. Undertaking a comprehensive review of the complete applications of
Click chemistry has a wide range of applications in the field of drug click chemistry in medicinal chemistry is a daunting task requiring
discovery, especially medicinal chemistry. Compared to conventional extensive content and ample examples. Due to limitations in space, word
radiolabeling strategies, click reactions exhibited many advantages, count and journal scope, this review will primarily focus on the signif­
including increased radiochemical yield [11], convenience connecting icant aspects of click chemistry in drug discovery, with an emphasis on
complex radiotracers [12], and significantly improved availability of medicinal chemistry. Specifically, we summarized its applications in the
radiopharmaceuticals [13,14]. Moreover, click chemistry also plays a generation of compound libraries, as well as the discovery of leads and
primary role in protein-lipid modification. It also serves as reasonable hits. Furthermore, we also discussed the challenges and prospects
linkers for conjugates or probes [9,15,16]. For example, click chemistry encountered in the utilization of click chemistry in drug discovery.

* Corresponding author. School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China.
E-mail address: renrenbai@hznu.edu.cn (R. Bai).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.ejmech.2023.116037
Received 22 October 2023; Received in revised form 20 November 2023; Accepted 8 December 2023
Available online 12 December 2023
0223-5234/© 2023 Elsevier Masson SAS. All rights reserved.
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

Ultimately, this review aims to provide pharmaceutical chemists with a


valuable point of reference for their future research endeavors involving
click chemistry.

2. Rapid construction of compound libraries assisted by click


chemistry

The hit-to-lead stage is a crucial and time-consuming aspect of drug


discovery, also the most expensive [23]. However, the introduction of
CuAAC click chemistry and in situ screening has expedited this process
while enhancing both its efficiency and the quality of drug discovery
[24,25]. This approach offers the advantage of rapidly and efficiently
obtaining lead compounds and testing their activity results. After the
discussion of the structure-activity relationship (SAR), the lead com­
pounds were screened again to form a virtuous circle [24]. This
approach had great potential in the field of medicinal chemistry (Fig. 3)
[26].
Chan’s group reported the preparation of flavonoid dimers by
CuAAC reaction, which was used as the MPR1 inhibitors. Compound 1
(Fig. 4) exhibited significant inhibitory activity and included two
identical flavonoid moieties. Moreover, PEG was used as the linker to
couple two identical moieties, showing potent inhibitory activity. Af­
terward, this group prepared a library of triazole compounds with two
distinct flavonoid fragments. A 300-member MPR1 inhibitors library
was assembled in microtiter plates without any purification, among Fig. 2. Summary of click chemistry application in medicinal chemistry.
which compound 2 has shown a potent inhibitory effect (EC50 = 53 nM)
[27]. click chemistry in 2013. In this work, all library compounds were easily
HDAC3 is a member of the HDAC family and belongs to Type I HDAC prepared by treating nine alkynes with fifty-five azides, and evaluated
(1, 2, 3 and 8). Abnormal increases in HDAC3 are involved in the for inhibitory effect on HDAC3 and two compounds (7 and 8) displayed
development of many diseases, including cancer [28], inflammation potent and selective HDAC3 inhibitory effects [18].
[29], and neurodegenerative diseases [30]. In 2012, Miyata and col­ Previous investigations have shown that quinone-containing frag­
leagues designed a library of HDAC8 inhibitors via click chemistry. ments play an important role in inhibiting Cdc25 phosphatase activity.
Further primary screening at a concentration of 3 μM of the product To find Cdc25 phosphatase inhibitors with strong inhibitory activity and
revealed that compounds 3–6 displayed considerable inhibition on high subtype selectivity, Zhan et al. designed a quinone derivatives li­
HDAC8 with IC50 values of 0.35, 0.18, 0.10, and 0.007 μM, respectively. brary of candidate Cdc25-selective inhibitors, in which two types of
At first, the CuAAC reaction between eight alkynes and fifteen azides alkynes and forty-eight azides were connected by a triazole-containing
provides a 120-member library. Fortunately, without undergoing puri­ linker. Finally, a 96-member Cdc25-selective inhibitors library was
fication, two crude compounds 3 and 4 exhibited potent inhibitory ac­ assembled in microtiter plates using the CuAAC reaction. As shown in
tivity against HDAC8. Subsequently, to prepare HDAC8-selective Fig. 3, compound 9 exerted an obvious inhibitory effect on the Cdc25
inhibitors with more significant activity than the control 4, they subtype with an IC50 value of 0.09 μM [32].
generated a 31-trizole library by click chemistry. Notably, compounds 5 For further structural optimization, this group used click chemistry
and 6 show more potent inhibitory activity than compound 4 at a con­ between the key intermediates alkynes and a set of 37 azides gave a
centration of 0.2 μM. The triazole products were confirmed by TLC and library including quinone fragments. Then, those crude compounds
LCMS [31]. were tested for activity. Notably, compound 10 showed more potent
On the base of the previous investigations, this group further re­ inhibition with an IC50 value of 0.06 ± 0.04 μM, which was equal to the
ported a 504-member library of compound HDAC3 inhibitors through control NSC663284 (IC50 = 0.23 ± 0.01 μM) [33].

Fig. 1. Click chemistry serves as a useful strategy and accelerates the process of hits and leads discovery.

2
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

Fig. 3. Compound library rapid generation by click chemistry.

Fig. 4. The chemical structures of hits 1–10 targeting MPR1, HDAC, and Cdc25.

3. Hit discovery aided by in situ click chemistry chemistry technique, in contrast to conventional fragment-based drug
design, can use biological targets to chemically construct their inhibitors
Target-guided synthesis (TGS) is a synthesis method of enzyme in­ in a templated way by creating covalent links between fragments.
hibitors based on the selective assembly of complementary functional Developing novel, extremely effective inhibitors of AChE and CA has
groups of biological targets (e.g. enzymes) as templates, mainly divided shown in situ click chemistry to be a revolutionary drug discovery
into dynamic combinational chemistry (DCC) and dynamic TGS [34]. technique to synthesize more effective self-enzyme inhibition under the
The application of copper-catalyzed cycloaddition reactions of azides physiological activity of enzymes (Fig. 5) [36]. In 2002, Sharpless’s
and alkynes in TGS is named in situ click chemistry [35]. The in situ click group first discovered and demonstrated an effective AChE inhibitor

3
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

Fig. 5. Discovery of potent “hit” by in situ click chemistry stratergy.

through in situ click chemistry [37]. related to the activity [39].

3.1. Hits targeting acetylcholinesterase 3.2. Hits targeting cyclooxygenase

The first target for the application of in situ click chemistry was COX is a heme protein located in the cell membrane, existing in two
AChE. distinct subtypes: constitutive COX-1 and inducible COX-2. COX-2 plays
In 2005, Hartmuth et al. discovered a potent AChE inhibitor by in a critical role in promoting inflammation.
situ click chemistry, which confirmed the great potential of in situ click Wuest et al. reported a series of COX-2 inhibitor derivatives, which
chemistry in medicinal chemistry (Fig. 5) [22]. Inspired by were prepared by treatment azide and alkyne groups at the COX-2
AChE-selective double binding sites, Renard’s group designed novel binding site by in situ click chemistry. Two potent COX-2 inhibitory
hupine inhibitor heterodimers. Compound 11 (Fig. 6) showed potent and selective compounds, 13 (triacoxib, IC50 = 90 nM) and 14 (IC50 =
inhibitory activity against recombinant human-AChE (rh-AChE) ligand, 50 nM), were successfully identified, which exhibited anti-inflammatory
with an IC50 value of 0.43 nM. Notably, the synthesis of enzyme in­ activity similar to the control (celecoxib, IC50 = 70 nM). In the
hibitors was thermodynamically driven to control the regional selec­ carrageenan-induced rat paw edema model, compounds 13 (ED50 =
tivity (syn or anti) of Huisgen reactions, leading to the successful 0.44 mg/kg) and 14 (ED50 = 0.12 mg/kg) showed potent anti-
identification of effective enzyme inhibitors [38]. Akbarzadeh and his inflammatory activity, superior to celecoxib (ED50 = 10.8 mg/kg)
colleagues discovered a series of novel 1,2,3-triazole derivatives by in [40]. Similarly, the same group reported radiotracer [18F]triacoxib (15)
situ click chemistry. Among these, compound 12 demonstrated obvious for PET imaging of COX-2 by in situ click chemistry. The
inhibition against AChE (IC50 = 7.31 μM). The results of molecular anti-inflammatory activity of compound 15 was similar to [18F]pyr­
docking showed that the planar structure of 1,2,3-triazole was closely icoxib both in vitro and in vivo. In HCA-7 tumor-bearing mice model,

Fig. 6. The chemical structures of hits 11–23 targeting AChE and COX.

4
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

compound 15 was found to bind specifically to COX-2, confirming that HIV-1-Pr, G48V, V82F, and V82A were as low as nanomolar (25: IC50 =
compound 15 was an excellent radiotracer [41]. 6 ± 0.5 nM, 19 ± 1 nM, 39 ± 1 nM, 46 ± 2 nM, 27: IC50 = 13 ± 0.5 nM,
Based on the structure of COX-2 inhibitors, it was found that most of 24 ± 1 nM, 17 ± 1 nM, 52 ± 2 nM, respectively) [47].
the compounds contained five-membered heterocyclic rings. Wuest Non-nucleoside reverse transcriptase inhibitors (NNRTIs) occupy a
et al. synthesized five 1,4-diaryl-1,2,3-triazole derivatives by Cu(I) large proportion in the treatment of HIV disease due to their high
catalyzing the cycloaddition reaction between 1-azido-4-methane sul­ antiviral ability. Targeting HIV-1 reverse transcriptase (RT), Kang’s
fonyl benzene and phenylacetylene containing different p-substituents. group screened out 22 compounds that displayed markedly suppressed
Among these, phenyl substituted derivative 16, fluorophenyl substituted HIV NNRTI by in situ click chemical. Within this series, compounds
derivative 17, and chlorophenyl derivative 18 exerted promising ac­ comprising 1,2,3-triazole exhibited strong activity (EC50 = 3.28–10.4
tivities, with IC50 values ranging from 0.15 to 0.21 μM. More impor­ nM) superior to the control NVP (EC50 = 163 nM). In particular, com­
tantly, the substitution of 1,2,3-triazole of the central cyclopene portion pound 29 demonstrated the best inhibition against HIV-1 NNRTI (IC50
can reduce the lipophilicity of the compound and thus serve as a tracer = 3.28 nM) and the lowest cytotoxicity (CC50 > 210 μM) [48].
of in vivo radioactivity by reducing non-specific binding in vivo [42].
Kharbanda’s group also identified a series of 1,2,3-triazole-contain­ 3.4. Hits targeting abelson tyrosine kinase
ing compounds as inhibitors of COX through in situ click chemistry.
Treatment of 2-mercaptobenzooxazolyl compounds and various The earliest Bcr-abelson (Abl) tyrosine kinase inhibitors (TKIs) were
substituted aromatic azides gave a library which compounds containing used for the treatment of chronic myelogenous leukemia and acute
a double heterocyclic ring structure. Further biological evaluation lymphoblastic leukemia. However, due to the existence of drug resis­
proved that compounds 19, 20, 21, 22, and 23 significantly reduced tance and mutants, the development of new TKIs is of great clinical
inflammation and the absence of gastric ulceration. The result indicated significance.
that they demonstrated potential anti-inflammatory medication prop­ The CuAAC reaction between azide and acetylene allowed Peruz­
erties. Furthermore, compounds 19 and 21 had a higher COX-2 selec­ zotti’s group to generate a small candidate library, which can bind to the
tivity (IC50 = 3.8 μM and 2.8 μM, SI = 64.79 and 66.47, respectively) active position site of Abl. Within the series, compound 30 (Fig. 8)
[43]. exhibited inhibitory activity against K-562 cell line and tyrosine kinase
with IC50 values of 0.89 μM and 0.9 ± 0.1 μM, respectively [49].
Kalesh et al. designed a library of candidate Abl inhibitors containing
3.3. Hits targeting HIV-1 about 344 compounds using in situ click chemistry by treating ADP-
alkynes and azides. The SAR study of the library showed that the
HIV-1 protease is a target for treating acquired immunodeficiency short-chain C-2 position of azides demonstrated remarkably inhibitory
syndrome (AIDS). Inhibiting HIV protease activity disrupts HIV’s ability activity. The previous result showed that the piperazine fragment of
to replicate and infect. imatinib did not show a significant contribution to the inhibition of Abl.
Using in situ click chemistry, Whiting et al. synthesized a triazole Consequently, based on the structural information of imatinib and the
compound 24 (Fig. 7), which used to be an inhibitor of HIV-1-Pr and its primary screening of products, 90 triazole compounds were synthesized,
mutants (IC50 = 6 nM, Ki = 1.7 nM) [44]. However, single-target drug containing the core structure of the imatinib. 11 compounds exhibited
has the disadvantage of easy drug resistance. To address this issue, the effective inhibition against Abl over Src kinase. Compounds 31 and 32
application of dual-target or multi-target provides a new idea for the displayed remarked inhibition against Abl (IC50 = 0.70 μM and 1.12 μM,
discovery of anti-HIV drugs. Brik et al. retained the core structure of respectively), while the inhibition of the parent of compounds 31 and 32
hydroxyethylamine in HIV-1-Pr marketed drugs (e.g. Amprenavir [45] with IC50 values of 1.36 μM and 2.73 μM, respectively [50].
and Nelfinavir [46]) and used it to generate novel inhibitors. The
promising compounds 25–28 were obtained from a 100-member library
that showed potent inhibitory activity against HIV-1-Pr at the concen­
tration of 10 nM. Notably, the inhibition of compounds 25 and 27 on

Fig. 7. The chemical structures of hits 24–29 targeting HIV-1.

5
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

Fig. 8. The chemical structures of hits 30–46 targeting Abl, CA, Chi, and OGA.

3.5. Hits targeting carbonic anhydrase 3.6. Hits targeting chitinase

CA is a zinc-containing enzyme catalyzing the mutual conversion of Chitinase (Chi) is the main component of fungal cell walls, and Chi
bicarbonate and carbon dioxide. Abnormal elevation of CA has been inhibitors have the potential of bactericidal, insecticidal, and asthmatic
observed in a variety of diseases, including tumors and AD. in chemotherapy.
Kugler et al. utilized in situ click chemistry to design five inhibitors Argifin (37) is a cyclic pentapeptide natural product, which was
targeting hCA-II. Among these, compound 33 exerted potent inhibitory proved to be an effective Chi inhibitor (SmChiA: IC50 = 0.025 μM;
activity (Ki = 2.5 ± 1.4 nM and SI = 4). Notably, the Ki of compound 33 SmChiB: IC50 = 6.4 μM). In particular, Nw-methylcarbamoyl-L-arginine
was higher than the parent (20-fold) [51]. scaffold was considered as the key moiety of Argifin, which can be used
Sulfonamide and glycoconjugates were identified as pharmaco­ as a model to obtain more potent inhibitory activity than Argifin [56].
phores for CA inhibitors. Poulsen’s group designed a series of potential Derivative 38 displayed potent inhibitory activity against SmChiB with
CA inhibitors. 12 compounds presented potent inhibition against bCA-II an IC50 value of 0.58 ± 0.044 μM. Subsequently, using in situ click
(Kds = 0.2–7.1 nM). Notably, compounds also showed higher affinity chemistry, Hirose et al. constructed a 1,2,3-triazole candidates library.
due to the higher selection of the enzyme [52]. Wilkinson et al. syn­ Compound 39 was prepared by compound 38 and alkyne comprising
thesized a series of glycoconjugate benzene sulfonamide compounds dibenzylamide analog and displayed significant Chi inhibitory against
using in situ click chemistry, which treated the carbohydrate azides and SmChiB (IC50 = 0.022 μM), superior to the azide precursor. Further
arylsulfonamide alkynyl. Within the series, compounds 34 and 35 dis­ X-ray crystallography and the control assay without the presence of Chi
played potent suppressed hCA-II isozyme activity (Ki = 7.5 nM and 2.3 showed that SmChiB can promote the formation of compound 39 [56,
nM, respectively), superior to the parent alkyne (Ki = 5.1 nM) [53]. 57].
Based on these results, Michaela et al. developed a PET [18F] mo­ The CuAAC reaction was chosen as the linking method to couple
lecular imaging probe compound 36 using in situ click chemistry, which erythronolide A oxime alkyne with azide comprising N-methyl­
can confirm the expression of CA-II in vivo [54]. Additionally, an inte­ carbamoylguanidinyl fragment to generate compound 40 at C3 position
grated microfluidic device for large-scale in situ click chemistry can by Sunazuka’s et al. Simultaneously, compounds with C5 and C9 sub­
remarkably decrease reagent consumption and screening time [55]. stitution positions were synthesized by the same procedure. The results
indicated that compounds with C3, C5, and C9 substitution exhibited
remarkably Chi inhibitory activity against SmChiB, with IC50 values of

6
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

3.3 μM, 11.9 μM, and 24.1 μM, respectively. Notably, C3 substitution 4. Click chemistry-aided drug discovery
induced potent inhibitory activity [58].
4.1. Anti-Alzheimer’s disease leads
3.7. Hits targeting O-GlcNAc
AD is a neurodegenerative disease commonly observed in the
O-GlcNAc (OGA) plays an important role in many basic cellular elderly. The possible pathogenesis is downregulation of acetylcholine
processes, and its dysregulation has been linked to the causes of car­ (ACh) level, activation of oxidative stress, deposition of beta-amyloid
diovascular diseases, type 2 diabetes, cancer, and neurological diseases. protein (Aβ), and hyperphosphorylation of tan protein. AChE in­
Li et al. reported an efficient synthetic method for the preparation of hibitors (tacrine) are widely accepted by researchers for the treatment of
a series of 1,2,3-triazole candidates using in situ click chemistry. Further AD. Coumarin exerts a wide range of biological activities, such as anti-
inhibitory investigation of human OGA revealed that most of the com­ inflammatory, anti-tumor, and anti-AD [62]. Studies have revealed
pounds showed effective inhibitory activity against OGlcNAcase. In that coumarin can inhibit Aβ-protein aggregation, oxidative stress, and
particular, compound 41 demonstrated the best potency and lowest ACh degradation. Oxidative damage and neurotoxicity created by H2O2
toxicity, with an inhibition of 81.3 % (Ki = 185.6 μM). Notably, the are considered as most important factors controlling the progress of
triazole fragment of compound 41 enhanced interaction with biological neurodegenerative disorder [63].
targets via hydrogen bonding and dipole-dipole [59]. Foroumadi et al. recently reported that coumarin-lipoic acid conju­
By use of in situ click chemistry, a series of glucopyranoside triazole gate 47 (Fig. 9) exerted AChE-selective inhibitory activity with an IC50
derivatives library was constructed by Carvalho’s group, which were of 16.4 μM, and exhibited neuroprotective effect against H2O2-induced
prepared from sugar azides and terminal alkynes. Within the series, oxidative in PC12 cells and Aβ1-42-induced cytotoxicity in SH-SY5Y cells
compounds 42–44 exhibited significant inhibitory and selective activity [64]. Saeedi and co-workers synthesized a series of 1,2,3-triazole-chro­
against OGA, with IC50 values of 0.50 ± 0.02 μM, 0.52 ± 0.01 μM, and menone carboxamides derivatives by click chemistry. Among these
0.72 ± 0.02 μM, respectively. Moreover, compounds were shown to be compounds, compound 48 was demonstrated to be a potent AChE and
nontoxic at the concentration of 10 μM. Based on the structure infor­ beta-secretase 1 (BACE1) inhibitor with IC50 values of 1.8 μM and 21.13
mation, the presence of heteroatoms (N or O) and the two carbons bridge μM, respectively. Compound 48 also displayed significant neuro­
between the aromatic ring and five-membered heterocyclic ring may be protectivity and showed good metal-chelating properties to Cu2+ and
the key pharmacophore to the increased activity [60]. Zn2+ ions [65].
Zhang et al. used in situ click chemistry to generate a triazole library Based on the CuAAC reaction between five alkynes and nine
and screened for novel O-GlcNAc transferase (OGT)-selective inhibitors, tryptamine-derived azides, a series of derivatives were prepared.
among which compounds 45 and 46 showed potent inhibitory against Further biological evaluation proved that compound 49 exhibited
OGT on COS-7 cells in vitro (IC50 = 66.7 ± 0.8 μM and 139 ± 14 μM, potent AChE inhibitory activity (IC50 = 18 nM). Interestingly, the linker
respectively). Their deacetylate precursors displayed more significant triazole moiety increased hAChE activity [66]. Muńoz-Torrero et al.
cytotoxicity in vitro. Besides, they showed no effect on the viability of synthesized a series of huprine derivatives by click chemistry with
normal cells. It was noteworthy that the inhibitory activity of compound cholinesterase (ChE) inhibitor huprine Y and capasaicin, targeting
45 increased more than 60-fold comparable with the parent alkynes and oxidative stress-related diseases and neuroinflammation diseases.
azides, which was comparable to positive drug BZX in vivo [61]. Among the hybrids, compound 50 exerted potent ChE inhibitory activity
and antioxidant activity, with hAChE and hBChE inhibition of 1.06 nM
and 7.3 nM, respectively. Additionally, compound 50 attained a higher
brain concentration superior to the reference drug donepezil [67].

Fig. 9. The chemical structures of anti-AD leads 47–55.

7
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

Carvalho’s group reported a series of aryl-1,2,3-triazyl benzylpiperidine transporter SVCT2 to cross the blood-brain barrier (BBB) and enter the
inhibitors. Further investigation revealed that compound 51 presented brain. Additionally, 55 showed moderate amyloid aggregation inhibi­
significant inhibition (IC50 = 0.17 nM) and selective (>58,000-fold) of tory comparable to the control group (71.79 ± 1.42 % inhibition at a
hBChE compared to donepezil (IC50 = 9.14 μM) without cytotoxicity concentration of 100 μM) and markedly antioxidant effect (IC50 = 72.26
[68]. Khoobi et al. prepared a series of multi-target directed ligand μM). Moreover, the expression of COX-2 and iNOS genes was effectively
(MTDL) compounds based on chromones and lipoic acid. Results indi­ reduced by compound 55 [21].
cated that compound 52 exhibited inhibitory activity against BChE
(IC50 = 7.55 μM) and potent neuroprotection activity by remarkably 4.2. Anti-inflammatory leads
decreasing reactive oxygen species (ROS). As reported before, amid and
disulfide bonds of chelating sites lead to antioxidant effects of compound A series of thioquinazolinone molecules were prepared from 2-Mer­
52 [69]. capto-3-substituted phenyl-quinazlin-4(3H)-ones intermediates and ar­
Miri and co-workers reported that iminochromene-2H-carboxamide omatic azides using click chemistry. Compound 56 (Fig. 10)
53 showed high selective BACE1 inhibitory activity and neuroprotective demonstrated a significant inhibitory effect against COX-1 and COX-2
activity, with IC50 values of 2.2 μM and 2.2 μM, respectively, superior to (IC50 = 2.97 μM and 0.11 μM, respectively, SI = 27) and moderate
the positive control (caffeic acid, IC50 = 75.8 μM) [70]. inhibitory activity against 15-LOX (IC50 = 7.26 μM). Further monocyte-
Xie’s group prepared a series of dual-target coumarin derivatives to-macrophage differentiation inhibitory tests revealed that compound
against AD. The most potent compound 54 markedly inhibited the iron 56 exhibited a potent effect in comparison to diclofenac (IC50 = 0.11 μM
chelation activity (pFe3+ = 17.4) and anti-monoamine oxidase-B (MAO- and 0.8 μM, respectively). Additionally, in vivo Formalin-induced rat
B) activity (IC50 = 0.68 μM) [71]. paw edema evaluation showed that compound 56 demonstrated
Vajragupta’s group used click chemistry to prepare a series of doubled inhibition compared to celecoxib [72].
ascorbic derivatives through an ascorbic core and tryptoline or phenolic Compound 57, prepared by Kharbanda et al., was a 2-mercaptoben­
moieties. Investigation on sodium-dependent vitamin C transporter-2 zoxazole derivative containing 1,2,3-triazoles. Compound 57 displayed
(SVCT2) inhibitory activity and targeting BBB permeation of all ascor­ significant inhibition on COX-2 (IC50 = 3.8 μM and SI = 64.79) and the
bic derivatives revealed that compound 55 might interact with the level of Nitric Oxide (NO). In particular, 57 decreased the expression of

Fig. 10. The chemical structures of anti-inflammatory leads 56–66.

8
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

COX-2 genes (0.94-fold) and exhibited effective gastric mucosa toler­ conjugates. These favorable findings indicated that Ibu conjugates
ance. In particular, it showed potent anti-inflammatory activity with demonstrated potent effects, superior to indomethacin conjugates.
66.66 % inhibition [43]. Among these Ibu-triazole conjugates, compounds 63–66 showed sig­
Han’s group synthesized a series of hybridizable compounds based nificant anti-inflammatory, peripheral analgesic, and central analgesic
on tryptamine and paeonol by click chemistry. Compound 58 was effects. Noticeably, compounds 63, 64, and 66 were potent and selective
demonstrated to be a potent generation inhibitor for NO and STAT-3 in a inhibitors of COX-2 (IC50 = 0.743 ± 0.02 μM, 12.35 ± 0.35 μM, and
dose-dependent manner in an LPS-induced BV2 cell inflammatory 4.467 ± 0.13 μM, respectively) with the SI values ranged from 2.15 to
model. More importantly, compound 58 exerted lower toxicity associ­ 23.10 [77].
ated with the free phenolic hydroxyl moiety of peaonol [73].
Haider et al. reported a novel anti-inflammation compound 59 based 4.3. Anti-virus leads
on piperine. Anti-inflammation evaluation revealed that compound 59
showed potent in vivo anti-inflammatory activity in a carrageenan- A series of 1,2,3-triazole glycosides comprising benzimidazole,
induced rat paw edema model (80.40 % inhibition) and decreased benzoxazole, and benzotriazole complexes were prepared through click
TNF-α level in RAW 264.7 cell lines in vitro (73.73 % inhibition), which chemistry by Ali’s group. All the compounds were evaluated for antiviral
was superior to the control piperine (54.72 % and 49.67 % inhibition, activity of H1N1, H5N1wild, H5N1V116A, and H5N1N295S. Compound 67
respectively). Simultaneously, compound 59 demonstrated remarked (Fig. 11) displayed a potent antiviral activity with IC50 values of 1.64,
analgesic and ulcerogenic activity, without cytotoxicity [74]. 2.25, 1.65, and 55.09 μM, respectively. It was also suggested that
Serafini’s group synthesized a series of biphenyl triazoles based on compound 67 was a potent and selective H1N1 inhibitor with an SI
the structure of Synta66 for the treatment of acute pancreatitis. Com­ value of 184.1. In further investigation of plaque reduction in vitro and
pound 60, with a 3-carboxyphenyl key moiety, showed a potential mice models, compound 67 showed potent antiviral activity. Further­
inhibitory activity (87.8 ± 2.9 % inhibition) comparable to Synta66. For more, the antiviral activity and safe profiles of compound 67 were su­
further structural optimization obtained compound 61, displaying perior to the reference drug oseltamivir [78].
effective inhibitory in HEK cells with an IC50 value of 851 ± 54 nM. Kataev’s group reported a series of 1,2,3-triazolyl nucleoside ana­
Moreover, compound 61 did not show cytotoxicity at the concentration logues using CuAAC reaction. The promising compound 68 demon­
of 60 μM and high aqueous solubility. Surprisingly, the 1,2,3-triazole strated a significant anti-H1N1 activity (IC50 = 15 μM) and effectively
fragment increased the targeting of the compound [75]. selective index (SI = 5) but showed moderate cytotoxicity (CC50 = 79
Compound 62 was a dual steric/bitopic agent, synthesized by Man­ μM). This effect could depend on the polymerase acidic protein of RNA-
era et al. Compound 62 inhibited the production of pro-inflammatory dependent RNA polymerase (RdRp). Notably, 1,2,3-triazole nucleoside
media IL-6 release and increased the expression of IL-10. The anti- derivatives containing 6-menthyluracil and luinazoline-2,4-dione exer­
inflammation activity and neuroprotection of compound 62 were ted significant antiviral effects, while uridine and thymidine derivatives
confirmed in HMC3 cells and a mouse model of nociceptive behavior. In demonstrated lower activity against the influenza virus [79].
particular, it was not cytotoxic in microglia [76]. In 2022, this group further synthesized a series of 1,2,3-triazolyl
Ghanim’s group designed a series of anti-inflammation candidate nucleoside analogues and their 5-phosphorylated derivatives. The
inhibitors based on ibuprofen (Ibu) and indomethacin triazole promising compound 69 showed potent and selective inhibition against

Fig. 11. The chemical structures of anti-virus leads 67–79.

9
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

the H1N1 influenza virus (IC50 = 25 μM, SI = 21), while its parent suppressed HIV-1 (IC50 = 10 nM, H9 cell line), which was superior to the
compound absence of activity. The result also indicated that 5-triphos­ original lead molecule [84].
phate derivative demonstrated significant anti-H1V1 activity, Zhan et al. used CuAAC reaction to prepare 1,2,3-triazole-containing
compared to the nucleoside analogues [80]. phenylalanine derivatives, among which one potent anti-HIV-1 inhibitor
He et al. prepared a series of 1,2,3-triazole rupestonic aid derivatives 75 was identified. Compound 75 exhibited significant anti-HIV-1 NL4-3
through click chemistry. Further in vivo biological evaluation proved virus activity with an EC50 value of 4.33 μM (SI > 13.33), comparable to
that compound 70 demonstrated a significant antiviral effect against the original lead compound PF-74. Further surface plasmon resonance
influenza A virus H1N1 (IC50 = 2.82 ± 0.36 μg/mL) and compound 71 assay showed that compound 75 show potent activity via inhibiting the
showed a potent antiviral effect against influenza B (IC50 = 1.71 μg/mL), early and late stages replication of HIV-1 [85].
which were superior to the reference drugs oseltamivir (IC50 = 2.81 μg/ Compound 76 was a diarylpyrimidine derivative reported by Liu’s
mL) and ribavirin (IC50 = 7.84 ± 4.41 μg/mL) [81]. group, showing promising and selective inhibitory activity against wild-
Yu et al. successfully obtained a series of oleanolic acid (OA)- type HIV-1 strain (IC50 = 0.013 μM, SI > 1000). Additionally, it also
saccharide complexes by click chemistry, connected by a triazole- displayed higher water solubility and lower cytotoxicity [86].
containing linker. All productions were employed for the anti- Based on the structure of dUY11, a series of 1,2,3-triazole derivatives
influenza assay, and compound 72 showed a significant inhibitory ef­ were prepared by Korshun’s group, which converse the linker of the
fect against A/WSN/33 virus (IC50 = 5.47 μM) and displayed a wide triple bond to 1,2,3-triazole ring. Among those, compounds 77 and 78
range of biological activities against OSV-p resistant viruses. Moreover, showed significant activities against tick-borne encephalitis virus
compound 72 did not show cytotoxicity at the concentration of 100 μM (TBEV), with EC50 values of 0.031 ± 0.013 and 0.023 ± 0.012 μM,
[82]. respectively. In particular, the perylene residue-contained triazolyl
Liu et al. designed and synthesized a series of dihydro-alkylthio- heterocycle was then selected as an antiviral drug fragment for further
benzyl-oxopyrimidines (S-DABOs) derivatives incorporating 1,2,3-tria­ study [87].
zole by click chemistry. Among these, compound 73 (EC50 = 3.22 μM) Aouad et al. synthesized phenylpyrazolone derivatives consisting of
showed the best activity against HIV-1 in comparison to reference drug triazole ring and lipophilic aromatic terminals. Within the series, com­
3 TC (EC50 = 2.24 μM). In terms of chemical structures, methyl and pound 79 exhibited effective inhibitory activity against SARS-CoV-2
sulfonamide moieties of compound 73 were associated with anti-HIV-1 Main protease (IC50 = 3.16 ± 1.2 μM) and Vero E6 cells (IC50 = 22.1
activity [83]. μM). Furthermore, compound 79 displayed essentially lower cytotoxic
Rana’s group synthesized a series of 1,2,3-triazoles based on the and higher antiviral effects than the positive drug GC-376 (IC50 = 12.85
structure of RN-18-based viral infectivity factor (VIF). It was noticed μM) at the concentration of 10 μM [88].
that the 1,4- disubstituede-1,2,3-triazole analogue showed the most
significant anti-HIV activity, with an IC50 value of 1.2 μM. Further
structural modifications provided compound 74, displaying potently

Fig. 12. The chemical structures of anti-infective leads 80–87.

10
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

4.4. Anti-infective leads 4.5. Anti-cancer leads

Zhang et al. reported a series of novel compounds, with 1,2,3-tria­ Xu et al. applied click chemistry to couple 1,2,3-triazole fragments
zoles linked to (2R,3S)-2-(2,4-difluorophenyl)-3-menthyl-1-(1H-1,2,4- with different C6 substituted quinazoline scaffolds antiproliferative
triazol-1-yl)pent-4-yn-2-ol moiety. The promising compound 80 evaluation revealed that compound 88 (Fig. 13) showed significant
(Fig. 12) demonstrated in vitro potent antifungal activity against antitumor activity against human lung cancer A549 and colorectal
C. albicans SC5314, C. neoformans H99, and A. fumigatus 7544 (MIC = cancer HCT116 cells in vitro, with IC50 values of 1.18 μM and 0.36 μM,
0.0313 μM, 0.0625 μM, and 1.0 μg/mL, respectively). It also exhibited respectively. Besides, compound 88 also exerted a broad-spectrum
potent broad-spectrum and protection in vivo. Moreover, it showed low antiproliferative activity against various cell lines. Results also showed
cytotoxicity and good safety [89]. compound 88 inhibited tumor growth by tethering RNF186 with P62
Bhattacharya et al. designed a series of novel compounds with 1,2,3- [97].
triazoles linked to glycoconjugate moieties for further antifungal He et al. designed a calothrixin A (CAA) compound library, prepared
investigation. Compound 81 displayed excellent antifungal activity from various sugar/polyhydroxy azides and CAA using click chemistry.
against A.fumigatus, with an IC50 value of 5.42 μM and an MIC value of The most active compound 89 displayed potent antiproliferation activ­
10.86 μM. Additionally, compound 81 was not cytotoxic on an un­ ity against A357 and HCT116 cell lines, superior to the control CPT and
transformed cell line with a CC50 value of 43.46 μM and an SI value of VP-16, with IC50 values of 20 nM, 50 nM, 660 nM, and 280 nM,
8.01 [90]. respectively. Additionally, the results also showed that compound 89
Shen et al. chose click chemistry as a linking method to prepare enhanced antitumor activity and water solubility compared to the
triazole-piperidine side chains derivatives The result indicated that most parent compound CAA [98].
of the derivatives demonstrated broad-spectrum antifungal activity. The Zhang et al. generated 31 triazole compounds processing mel­
promising compound 82 showed remarkably antifungal activity in vitro, ampomagnolide B (MMB) fragments via click chemistry. The promising
superior to the control fluconazole and itraconazole, with MIC80 values compound 90 exhibited potent inhibition on the growth and migration
from 0.0125 to 2 μg/mL, against Candida albicans, Candida parapsilosis, against HCT116 cell line, superior to the parent compound MMB (IC50 =
and Cryptococcus neoformans strains. It was noteworthy that compound 0.43 μM and 4.93 μM, respectively). Additionally, it also demonstrated
82 showed potent antifungal activity due to the triazole ring being effective antitumor activity against Bel704, PANC1, A549, and U87 cell
associated with the coordination bond and hydrophobic interaction of lines, while did not significantly affect the viability of normal cells [99].
CACYP51 [91]. He et al. identified a novel series of indoleamine 2,3-dioxygenase-1
1-alkyl-1H-1,2,3-triazole-4-carboxylic acids (ATCs) incorporating (IDO1) and indoleamine 2,3-dioxygenase-2 (IDO2)-selective inhibitors
1,2,3-triazole were prepared based on the structure of cis-2-dodecenoic by using click chemistry, generating the promising candidate compound
acid (BDSF) by Wang’s group. Compound 83 exerted a potent antifungal 91. It exhibited significant antitumor activity against IDO1 and IDO2 in
activity effect against C. albicans yeast-cell growth and germ-tube for­ vitro, with IC50 values of 28 nM and 144 nM, respectively. In the CT26
mation in vitro (95 % inhibition at the concentration of 200 μM and xenograft tumor mouse nude model, compound 91 exhibited potent
length remarkably reduced from 71.9 to 53.3 μM, respectively). The suppression of tumor growth (TGI = 69.7 %) and was nontoxic [100].
1,2,3-triazole hybridized structure increases the antifungal properties of Bhadra et al. prepared a series of quinazolinone derivatives incor­
BDSF analogues [92]. porating triazole fragments for further antiproliferative investigation.
Kumar et al. designed a library of antibacterial candidates in which Among which, compound 92 presented significant anticancer against
C12-sphinganine and 1,2,3-triazole analogues were connected by a HeLa cell line (IC50 = 5.94 μM and SI = 43.09) and moderate activity
triazole-containing linker. The most active compound 84 demonstrated over MCF-7, K562, and HEK cell lines (IC50 = 8.0 μM, 20.16 μM, and
potent antibacterial activity against C. albicans MTCC 227, C. albicans 256 μM, respectively). Notably, compound 92 exhibited an improved
854 (MIC = 3.9 μg/ml and 3.9 μg/ml, respectively), and potent sup­ antitumor activity due to the triazole ring interacting with Zn2+-medi­
pression on biofilm formation against C. albicans MTCC 227, staphylo­ ated HDACs [101].
coccus aureus MTCC 96 (IC50 = 1.9 μg/ml and 2.9 μg/ml, respectively) Zhu’s group synthesized a series of HDAC8-selective inhibitors based
[93]. on PCI-34051 and 1,2,3-triazole rings. Within the series, compound 93
A small library of novel 1,2,3-triazoles bearing β-D-ribofuranosyl displayed considerable inhibition on A549 cells (IC50 = 9.55 μM)
were synthesized by Prasad’s group. Within this series, compound 85 through reducing proliferation. Furthermore, it also remarkably
exhibited potent inhibitory activity against DNA gyrase at the concen­ decreased HDAC8 protein levels in A549 cells (DC50 = 0.59 ± 0.06 μM).
tration of 0.66 μM (IC50 = 0.8 μM) and InhA enzyme at the concentration In particular, co-treatment with compound 93 and irradiation (IR)
of 5 μM. Furthermore, it showed good safety and selectivity compared to demonstrated a significant antiproliferative effect (IC50 = 6.04 μM) su­
streptomycin in the human THP-1 cell line [94]. perior to either compound 93 or PCI-34051 alone [102].
Kumar et al. designed a series of chalcone triazole derivatives using The synthesis of a library of 1,2,3-triazole derivatives was conducted
click chemistry. The promising compound 86 showed significant anti­ to find potent and selective antitumor agents by Raić-Malić et al. Among
bacterial activity against E.coli, superior to the positive drug fluconazole these, compound 94 showed more potent and selective inhibition
(MIC = 0.0032 μmol/mL and 0.0102 μmol/mL, respectively). Notice­ against non-small cell lung cancer A549 cells, with an IC50 value of 70
ably, molecular docking showed that its potent antibacterial effect nM, which was superior to the 5-FU (IC50 = 2.80 μM). Besides, com­
depended on the DNA topoisomerase inhibition of the chalcone triazole pound 94 was not cytotoxic in a submicromolar range and suppressed
structure [95]. the TGM2, CDK9, SK1, and p38 MAPK protein kinases [103]. In 2019,
The pleuromutilin complexes comprising 1,2,3-triazole fragments the same group synthesized a series of benzimidazo[1,2-α]quinoline
were prepared through click chemistry by Jin’s group. Among which, complexes with 1,2,3-triazole. Compound 95 exhibited potent anti­
compound 87 demonstrated potent antibacterial activity against proliferative activity against HCT116 and H460 cell lines (IC50 = 0.5 μM
multidrug-resistant Staphylococcus aureus (MRSA) (MIC = 0.25 μg/ and 0.7 μM, respectively) and significant selection over HaCaT cells
mL). In the thigh infection and MRSA infection mouse model, compound (IC50 = 3.5 μM and SI > 7) [104]. Subsequently, In 2019, Raić-Malić
87 further showed effective amelioration compared to the standard drug et al. generated a library of anticancer compounds, namely 4-substituted
tiamulin [96]. 6-(1,2,3-triazolyl)-2,3-dibenzyl-L-ascorbic acid derivatives. Among
these, compound 96 demonstrated a significant inhibitory effect against
breast adenocarcinoma (MCF-7) cells (IC50 = 0.08 μM) through the
HIF-1α signaling pathway. Besides, it also showed the best lipophilic

11
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

Fig. 13. The chemical structures of anti-cancer leads 88–103.

(clog P = 5.25) property and selectivity against tumor cells [105]. μM). Further biological evaluation proved that the antiproliferative ac­
Additionally, compound 97, one of the coumarin analogues comprising tivity of compound 98 depended on disrupting the microtubule orga­
the 1,2,3-triazole group, was identified. It exhibited the greatest potency nization [107]. Subsequently, a series of podophyllotoxin derivatives
and high selectivity against Hepatocellular carcinoma (HepG2) cells with 4β-amidotriazole were synthesized. The further results showed that
with an IC50 value of 0.9 μM and an SI value of 50. Besides, it also compound 99 potently suppressed human tumor cell proliferation
moderately suppressed over other tumor cell lines with IC50 values against DU-145, Hela, HepG2, and MCF-7 cell lines (IC50 = 0.7 μM, 0.78
ranging from 17.48 to 45.33 [106]. μM, 0.78 μM, and 0.97 μM, respectively) [108].
The imidazo[2,1-b]thiazole complexes were prepared through click Zhao et al. designed a library of novel triazole candidates against
chemistry reported by Kamal’s group. The most potent compound 98 dehydrogenase (hDHODH), containing 1,4-benzoquinone fragments.
significantly inhibited the A549 and MCF-7 cell lines (IC50 = 0.778 μM Compound 100, with a diphenyl skeleton, displayed remarkable inhi­
and 1.013 μM, respectively), superior to the control group (IC50 = 1.57 bition on hDHODH with an IC50 value of 4.5 nM. It also demonstrated

12
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

significant proliferation inhibition over human cancer cell lines with for potential drug candidates (Fig. 14). However, the application of click
IC50 values ranging from 0.02 to 9.61 μM. Furthermore, nanocrystalline chemistry in drug discovery also presents several challenges and cannot
samples of compound 100 exhibited a higher safety profile and anti­ be considered as a universal solution. When generating a large number
tumor activity in vivo [109]. of compounds at a fast pace, it is unclear whether click chemistry en­
Crook’s group reported a series of MMB derivatives with 1,2,3-tria­ sures the desired drug-like properties in the resulting compounds.
zole rings. Compound 101 showed significant antitumor activity Firstly, it is important to note that all click chemistry reactions yield
against the RXF393 renal cancer cell line (GI50 = 20 nM). Besides, it also products with a triazole core structure, leading to significant enhance­
suppressed the growth of colon cancer, melanoma, renal cancer, and ment of the lipophilicity and consequently reducing the solubility of the
breast cancer sub-panels [110]. compounds. Poor solubility, a key negative determinant of drug-like
To discover new MLL1-WDR5 inhibitors, Gao et al. provide a triazole properties, can compromise bioavailability and hinder the further
library containing fragments of phenyl-triazole. All derivatives were development of a compound. Additionally, a review of synthesized
evaluated for antiproliferation activity. Compound 102 exhibited potent compounds reveals that the majority of click chemistry-assisted prod­
and selective inhibitory against MV4-11 and MOLM-13 cell lines (IC50 = ucts possess larger structures, which negatively impacts their drug-like
8.0 μM and 9.9 μM, respectively), with little impact on normal cells characteristics and further dissolution, penetration, and absorption.
(IC50 > 100 μM). Moreover, it also demonstrated remarkably binding Lastly, despite the decades since its discovery, click chemistry has pri­
affinity (IC50 = 8.6 ± 1.3 nM, Kd = 11.6 ± 4.0 nM), proper solubility, marily aided researchers in identifying multiple hits and leads for spe­
and permeability. The further in vivo results showed that compound 102 cific targets, but few marketed drugs were successfully discovered by
displayed potent antitumor activity via reducing the level of H3K4me click chemistry. Consequently, the application of click chemistry in
and the expression of HOXA9 and Meis1 genes [111]. medicinal chemistry and drug discovery presents both advantages and
The 1,2,3-triazole-tethered dehydroabietic acid derivatives were disadvantages, along with numerous challenges. Well understanding the
discovered through click chemistry by Xu’s group. The most selective project objectives in utilizing click chemistry is therefore crucial.
compound 103 displayed potent antitumor activity against MDA-MB-
231 cells (IC50 = 0.7 μM), superior to positive drug 5-FU. Furthermore, it 6. Conclusion and perspectives
also exhibited a moderate antiproliferative effect on various human
tumor cells with IC50 values ranging from 0.6 to 1.8 μM, including In traditional drug discovery, the design and synthesis of a large
MCF7, HCT116, PC-3, and SKOV-3 cell lines. Notably, the SAR study number of compounds typically entail significant time, manpower, and
indicated that the potent antiproliferative activity was due to the pres­ financial resources, leading to lengthy drug development cycles. How­
ence of 1,2,3-triazole fragment and its C-4 substituents [112]. ever, the utilization of click chemistry in compound discovery has
considerably reduced the time needed for synthesis. Click chemistry
5. Challenges of click chemistry-aided drug discovery offers several advantages, including resistance to oxygen and water,
high yields, regioselectivity, and stereoselectivity. These attributes
Click chemistry has demonstrated significant advantages in the rapid enable researchers to rapidly prepare target compounds and build both
synthesis of compounds, offering considerable convenience in the search small and large compound libraries. Additionally, in situ click chemistry
exploits enzymes as reaction templates, capitalizing on their favorable
conformation in physiologically active conditions. This approach
selectively connects individual building blocks to synthesize novel in­
hibitors of the enzyme.
Nevertheless, when employing click chemistry in drug discovery,
careful and deep consideration must be given to balancing and opti­
mizing both biological activity and drug-like properties. Such optimi­
zation ensures that the synthesized active compounds possess desirable
physical and chemical characteristics. Only when this equilibrium is
attained can the compound exhibit favorable dissolution and absorp­
tion, thereby warranting further development. Furthermore, as addi­
tional types of click chemistry reactions are discovered, researchers will
be empowered to swiftly prepare compounds with diverse and intricate
structures, surpassing those solely containing triazoles. This will expand
the chemical space of drug-like compounds and significantly enhance
the potential for identifying promising hits and leads in drug discovery.

CRediT authorship contribution statement

Rui Zhao: Data curation, Investigation, Writing – original draft.


Junlong Zhu: Data curation, Resources, Writing – original draft.
Xiaoying Jiang: Writing – review & editing, Funding acquisition.
Renren Bai: Conceptualization, Data curation, Funding acquisition,
Supervision, Writing – original draft, Writing – review & editing.

Declaration of competing interest

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.

Fig. 14. Sources and clinical applications of triazole heterocycles.

13
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

Data availability [7] A.K. Agrahari, P. Bose, M.K. Jaiswal, S. Rajkhowa, A.S. Singh, S. Hotha,
N. Mishra, V.K. Tiwari, Cu(I)-Catalyzed click chemistry in glycoscience and their
diverse applications, Chem. Rev. 121 (2021) 7638–7956.
No data was used for the research described in the article. [8] A. Angeli, C.T. Supuran, Click chemistry approaches for developing carbonic
anhydrase inhibitors and their applications, J. Enzym. Inhib. Med. Chem. 38
(2023), 2166503.
Acknowledgements [9] M. Gehringer, S.A. Laufer, Click chemistry: novel applications in cell biology and
drug discovery, Angew. Chem., Int. Ed. Engl. 56 (2017) 15504–15505.
This project was supported by the National Natural Science Foun­ [10] J. Kaur, M. Saxena, N. Rishi, An overview of recent advances in biomedical
applications of click chemistry, Bioconjugate Chem. 32 (2021) 1455–1471.
dation of China (82304284) and the Hangzhou Normal University
[11] Z.-B. Li, Z. Wu, K. Chen, F.T. Chin, X. Chen, Click chemistry for 18F-labeling of
Startup Fund (2021QDL026 and KYQD-2023-175). RGD peptides and microPET imaging of tumor integrin αvβ3 expression,
bioconjugate chemistry, Bioconjugate Chem. 18 (2007) 1987–1994.
[12] N. Walter, J. Bertram, B. Drewes, V. Bahutski, M. Timmer, M.B. Schütz,
Abbreviations F. Krämer, F. Neumaier, H. Endepols, B. Neumaier, B.D. Zlatopolskiy, Convenient
PET-tracer production via SuFEx 18F-fluorination of nanomolar precursor
ACh acetylcholine amounts, Eur. J. Med. Chem. 5 (2022), 114383.
[13] B. Gröner, M. Willmann, L. Donnerstag, E.A. Urusova, F. Neumaier, S. Humpert,
AChE acetylcholinesterase H. Endepols, B. Neumaier, B.D. Zlatopolskiy, 7-[18F]Fluoro-8-azaisatoic
AD alzheimer’s disease anhydrides: versatile prosthetic groups for the preparation of PET tracers, J. Med.
AI artificial intelligence Chem. 66 (2023) 12629–12644.
[14] C. Timperanza, H. Jensen, T. Bäck, S. Lindegren, E. Aneheim, Pretargeted alpha
AIDS acquired immunodeficiency syndrome
therapy of disseminated cancer combining click chemistry and astatine-211,
ATCs 1-alkyl-1H-1,2,3-triazole-4-carboxylic acids Pharmaceuticals 16 (2023) 595.
Aβ beta-amyloid protein [15] K.F. Suazo, K.-Y. Park, M.D. Distefano, A not-so-ancient grease history: click
chemistry and protein lipid modifications, Chem. Rev. 121 (2021) 7178–7248.
BACE1 beta secretase
[16] M. Baalmann, L. Neises, S. Bitsch, H. Schneider, L. Deweid, P. Werther,
BBB blood-brain barrier N. Ilkenhans, M. Wolfring, M.J. Ziegler, J. Wilhelm, H. Kolmar, R. Wombacher,
CA carbonic anhydrase A bioorthogonal click chemistry toolbox for targeted synthesis of branched and
CAA calothrixin A well-defined protein-protein conjugates, Angew. Chem., Int. Ed. Engl. 59 (2020)
12885–12893.
CADD computer-aided drug design [17] R.P. Wurz, K. Dellamaggiore, H. Dou, N. Javier, M.-C. Lo, J.D. McCarter, D. Mohl,
ChE cholinesterase C. Sastri, J.R. Lipford, V.J. Cee, A “click chemistry platform” for the rapid
Chi chitinase synthesis of bispecific molecules for inducing protein degradation, J. Med. Chem.
61 (2018) 453–461.
COX cyclooxygenase [18] T. Suzuki, Y. Kasuya, Y. Itoh, Y. Ota, P. Zhan, K. Asamitsu, H. Nakagawa,
DCC dynamic combinational chemistry T. Okamoto, N. Miyata, Identification of highly selective and potent histone
HIV-1 human immunodeficiency virus-1 deacetylase 3 inhibitors using click chemistry-based combinatorial fragment
assembly, PLoS One 8 (2013), e68669.
Ibu ibuprofen [19] P. Prasher, M. Sharma, Tailored therapeutics based on 1,2,3-1H-triazoles: a mini
IDO1 indoleamine 2,3-dioxygenase-1 review, Medchemcomm 10 (2019) 1302–1328.
IDO2 indoleamine 2,3-dioxygenase-2 [20] A. Rani, G. Singh, A. Singh, U. Maqbool, G. Kaur, J. Singh, CuAAC-ensembled
1,2,3-triazole-linked isosteres as pharmacophores in drug discovery: review, RSC
IR irradiation
Adv. 10 (2020) 5610–5635.
KTGs target-guided synthesis [21] J. Jiaranaikulwanitch, H. Pandith, S. Tadtong, P. Thammarat, S. Jiranusornkul,
MAO-B monoamine oxidase-B N. Chauthong, S. Nilkosol, O. Vajragupta, Novel multifunctional ascorbic triazole
derivatives for amyloidogenic pathway inhibition, anti-inflammation, and
MMB melampomagnolide B
neuroprotection, Molecules 26 (2021) 1562.
MRSA multidrug-resistant staphylococcus aureus [22] A. Krasiński, Z. Radic, R. Manetsch, J. Raushel, P. Taylor, K.B. Sharpless, H.
MTDL multi-target directed ligand C. Kolb, In situ selection of lead compounds by click chemistry: target-guided
NNRTI non-nucleoside reverse transcriptase inhibitor optimization of acetylcholinesterase inhibitors, J. Am. Chem. Soc. 127 (2005)
6686–6692.
NO nitric oxide; [23] P. Kirsch, V. Jakob, K. Oberhausen, S.C. Stein, I. Cucarro, T.F. Schulz,
OA oleanolic acid M. Empting, Fragment-based discovery of a qualified hit targeting the latency-
SAR structure-activity relationship associated nuclear antigen of the oncogenic kaposi’s sarcoma-associated
herpesvirus/human herpesvirus 8, J. Med. Chem. 62 (2019) 3924–3939.
RdRp RNA-dependent RNA polymerase [24] P. Gehrtz, S. Marom, M. Bührmann, J. Hardick, S. Kleinbölting, A. Shraga,
ROS reactive oxygen species C. Dubiella, R. Gabizon, J.N. Wiese, M.P. Müller, G. Cohen, I. Babaev,
RT reverse transcriptase K. Shurrush, L. Avram, E. Resnick, H. Barr, D. Rauh, N. London, Optimization of
covalent MKK7 inhibitors via crude nanomole-scale libraries, J. Med. Chem. 65
S-DABOs dihydro-alkylthio-benzyl-oxopyrimidines (2022) 10341–10356.
SI selective index [25] X. Wang, B. Huang, X. Liu, P. Zhan, Discovery of bioactive molecules from CuAAC
SVCT2 sodium-dependent vitamin C transporter-2 click-chemistry-based combinatorial libraries, Drug Discov. Today 21 (2016)
118–132.
TBEV tick-borne encephalitis virus
[26] M. Martinez-Bailen, A.T. Carmona, E. Moreno-Clavijo, I. Robina, D. Ide, A. Kato,
TGS target-guided synthesis A.J. Moreno-Vargas, Tuning of β-glucosidase and α-galactosidase inhibition by
TKIs tyrosine kinase inhibitors generation and in situ screening of a library of pyrrolidine-triazole hybrid
molecules, Eur. J. Med. Chem. 138 (2017) 532–542.
VIF vital infectivity factor
[27] I.L.K. Wong, X. Zhu, K.-F. Chan, M.C. Law, A.M.Y. Lo, X. Hu, L.M.C. Chow, T.
H. Chan, Discovery of novel flavonoid dimers to reverse multidrug resistance
References protein 1 (MRP1, ABCC1) mediated drug resistance in cancers using a high
throughput platform with “click chemistry”, J. Med. Chem. 61 (2018)
9931–9951.
[1] H.C. Kolb, K.B. Sharpless, The growing impact of click chemistry on drug
[28] J.M. Mariadason, Dissecting HDAC3-mediated tumor progression, Cancer Biol.
discovery, Drug Discov. Today 8 (2003) 1128–1137.
Ther. 7 (2014) 1581–1583.
[2] H. Zhu, Big data and artificial intelligence modeling for drug discovery, Annu.
[29] X. Chen, I. Barozzi, A. Termanini, E. Prosperini, A. Recchiuti, J. Dalli, F. Mietton,
Rev. Pharmacol. Toxicol. 60 (2020) 573–589.
G. Matteoli, S. Hiebert, G. Natoli, Requirement for the histone deacetylase Hdac3
[3] N. Maharao, V. Antontsev, M. Wright, J. Varshney, Entering the era of
for the inflammatory gene expression program in macrophages, Proc. Natl. Acad.
computationally driven drug development, Drug Metab. Rev. 52 (2020) 283–298.
Sci. U. S. A. 109 (2012) E2865–E2874.
[4] D. Vemula, P. Jayasurya, V. Sushmitha, Y.N. Kumar, V. Bhandari, CADD, AI and
[30] C. Xu, E. Soragni, C.J. Chou, D. Herman, H.L. Plasterer, J.R. Rusche, J.
ML, Drug discovery: a comprehensive review, Eur. J. Pharmaceut. Sci. 181
M. Gottesfeld, Chemical probes identify a role for histone deacetylase 3 in
(2023), 106324.
Friedreich’s ataxia gene silencing, Chem. Biol. 16 (2009) 980–989.
[5] J.H. Cai, X.Z. Zhu, P.Y. Guo, P. Rose, X.T. Liu, X. Liu, Y.Z. Zhu, Recent updates in
[31] T. Suzuki, Y. Ota, M. Ri, M. Bando, A. Gotoh, Y. Itoh, H. Tsumoto, P.R. Tatum,
click and computational chemistry for drug discovery and development, Front.
T. Mizukami, H. Nakagawa, S. Iida, R. Ueda, K. Shirahige, N. Miyata, Rapid
Chem. 11 (2023), 1114970.
discovery of highly potent and selective inhibitors of histone deacetylase 8 using
[6] H.C. Kolb, M.G. Finn, K.B. Sharpless, Click chemistry: diverse chemical function
from a few good reactions, Angew. Chem., Int. Ed. Engl. 40 (2001) 2004–2021.

14
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

click chemistry to generate candidate libraries, J. Med. Chem. 55 (2012) [55] Y. Wang, W.-Y. Lin, K. Liu, R.J. Lin, M. Selke, H.C. Kolb, N. Zhang, X.-Z. Zhao, M.
9562–9575. E. Phelps, C.K.F. Shen, K.F. Faull, H.-R. Tseng, An integrated microfluidic device
[32] L. Jing, G. Wu, X. Hao, F.A. Olotu, D. Kang, C.H. Chen, K.H. Lee, M.E.S. Soliman, for large-scale in situ click chemistry screening, Lab Chip 9 (2009) 2281–2285.
X. Liu, Y. Song, P. Zhan, Identification of highly potent and selective Cdc25 [56] T. Hirose, T. Sunazuka, A. Sugawara, A. Endo, K. Iguchi, T. Yamamoto, H. Ui,
protein phosphatases inhibitors from miniaturization click-chemistry-based K. Shiomi, T. Watanabe, K.B. Sharpless, S. Omura, Chitinase inhibitors: extraction
combinatorial libraries, Eur. J. Med. Chem. 183 (2019), 111696. of the active framework from natural argifin and use of in situ click chemistry,
[33] Y. Tao, X. Hao, L. Jing, L. Sun, S. Cherukupalli, S. Liu, G. Wu, S. Xu, X. Zhang, J. Antibiot. (Tokyo) 62 (2009) 277–282.
X. Shi, Y. Song, X. Liu, P. Zhan, Discovery of potent and selective Cdc25 [57] T. Hirose, N. Maita, H. Gouda, J. Koseki, T. Yamamoto, A. Sugawara, H. Nakano,
phosphatase inhibitors via rapid assembly and in situ screening of Quinonoid- S. Hirono, K. Shiomi, T. Watanabe, H. Taniguchi, K.B. Sharpless, S. Omura,
focused libraries, Bioorg. Chem. 115 (2021), 105254. T. Sunazuka, Observation of the controlled assembly of preclick components in
[34] D. Bosc, J. Jakhlal, B. Deprez, R. Deprea-Poulain, Kinetic target-guided synthesis the in situ click chemistry generation of a chitinase inhibitor, Proc. Natl. Acad.
in drug discovery and chemical biology: a comprehensive facts and figures Sci. U. S. A. 110 (2013) 15892–15897.
survey, Future Med. Chem. 8 (2016) 381–404. [58] A. Sugawara, N. Maita, H. Gouda, T. Yamamoto, T. Hirose, S. Kimura, Y. Saito,
[35] S.K. Mamidyala, M.G. Finn, In situ click chemistry: probing the binding H. Nakano, T. Kasai, H. Nakano, K. Shiomi, S. Hirono, T. Watanabe, H. Taniguchi,
landscapes of biological molecules, Chem. Soc. Rev. 39 (2010) 1252–1261. S. Omura, T. Sunazuka, Creation of customized bioactivity within a 14-membered
[36] R. Manetsch, A. Krasiński, Z. Radić, Jessica Raushel, P. Taylor, K.B. Sharpless, H. macrolide scaffold: design, synthesis, and biological evaluation using a family-18
C. Kolb, In situ click chemistry: enzyme inhibitors made to their own chitinase, J. Med. Chem. 58 (2015) 4984–4997.
specifications, J. Am. Chem. Soc. 126 (2004) 12809–12818. [59] T. Li, L. Guo, Y. Zhang, J. Wang, Z. Li, L. Lin, Z. Zhang, L. Li, J. Lin, W. Zhao, J. Li,
[37] W.G. Lewis, L.G. Green, F. Grynszpan, Z. Radić, P.R. Carlier, P. Taylor, M.G. Finn, P.G. Wang, Design and synthesis of O-GlcNAcase inhibitors via ‘click chemistry’
K.B. Sharpless, Click chemistry in situ: acetylcholinesterase as a reaction vessel and biological evaluations, Carbohydr. Res. 346 (2011) 1083–1092.
for the selective assembly of a femtomolar inhibitor from an array of building [60] M.O. Igual, P.S.G. Nunes, R.M. da Costa, S.P. Mantoani, R.C. Tostes, I. Carvalho,
blocks, Angew. Chem., Int. Ed. Engl. 41 (2002) 1053–1057. Novel glucopyranoside C2-derived 1,2,3-triazoles displaying selective inhibition
[38] E. Oueis, G. Santoni, C. Ronco, O. Syzgantseva, V. Tognetti, L. Joubert, of O-GlcNAcase (OGA), Carbohydr. Res. 471 (2019) 43–55.
A. Romieu, M. Weik, L. Jean, C. Sabot, F. Nachon, P.Y. Renard, Reaction site- [61] Y. Wang, J. Zhu, L. Zhang, Discovery of cell-permeable O-GlcNAc transferase
driven regioselective synthesis of AChE inhibitors, Org. Biomol. Chem. 12 (2014) inhibitors via tethering in situ click chemistry, J. Med. Chem. 60 (2017) 263–272.
156–161. [62] C. Zhang, Y. Lv, R. Bai, Y. Xie, Structural exploration of multifunctional
[39] M. Mohammadi-Khanaposhtani, M. Saeedi, N.S. Zafarghandi, M. Mahdavi, monoamine oxidase B inhibitors as potential drug candidates against Alzheimer’s
R. Sabourian, E.K. Razkenari, H. Alinezhad, M. Khanavi, A. Foroumadi, disease, Bioorg. Chem. 114 (2021), 105070.
A. Shafiee, T. Akbarzadeh, Potent acetylcholinesterase inhibitors: design, [63] R. Bai, J. Guo, X.-Y. Ye, Y. Xie, T. Xie, Oxidative stress: the core pathogenesis and
synthesis, biological evaluation, and docking study of acridone linked to 1,2,3- mechanism of Alzheimer’s disease, Ageing Res. Rev. 77 (2022), 101619.
triazole derivatives, Eur. J. Med. Chem. 92 (2015) 799–806. [64] L. Jalili-Baleh, H. Forootanfar, T.T. Küçükkılınç, H. Nadri, Z. Abdolahi, A. Ameri,
[40] A. Bhardwaj, J. Kaur, M. Wuest, F. Wuest, In situ click chemistry generation of M. Jafari, B. Ayazgok, M. Baeeri, M. Rahimifard, S.N. Abbas Bukhari,
cyclooxygenase-2 inhibitors, Nat. Commun. 8 (2017) 1. M. Abdollahi, M.R. Ganjali, S. Emami, M. Khoobi, A. Foroumadi, Design,
[41] M. Litchfield, M. Wuest, D. Glubrecht, F. Wuest, Radiosynthesis and biological synthesis and evaluation of novel multi-target-directed ligands for treatment of
evaluation of [18F]triacoxib: a new radiotracer for PET imaging of COX-2, Mol. Alzheimer’s disease based on coumarin and lipoic acid scaffolds, Eur. J. Med.
Pharm. 17 (2020) 251–261. Chem. 152 (2018) 600–614.
[42] F. Wuest, X. Tang, T. Kniess, J. Pietzsch, M. Suresh, Synthesis and cyclooxygenase [65] A. Rastegari, H. Nadri, M. Mahdavi, A. Moradi, S.S. Mirfazli, N. Edraki, F.
inhibition of various (aryl-1,2,3-triazole-1-yl)-methanesulfonylphenyl H. Moghadam, B. Larijani, T. Akbarzadeh, M. Saeedi, Design, synthesis and anti-
derivatives, Bioorg. Med. Chem. 17 (2009) 1146–1151. Alzheimer’s activity of novel 1,2,3-triazole-chromenone carboxamide derivatives,
[43] S. Haider, M.S. Alam, H. Hamid, S. Shafi, A. Dhulap, F. Hussain, P. Alam, S. Umar, Bioorg. Chem. 83 (2019) 391–401.
M.A.Q. Pasha, S. Bano, S. Nazreen, Y. Ali, C. Kharbanda, Synthesis of novel 2- [66] Z. Molęda, A. Zawadzka, Z. Czarnocki, L. Monjas, A.K.H. Hirsch,
mercaptobenzoxazole based 1,2,3-triazoles as inhibitors of proinflammatory A. Budzianowski, J.K. Maurin, "Clicking" fragment leads to novel dual-binding
cytokines and suppressors of COX-2 gene expression, Eur. J. Med. Chem. 81 cholinesterase inhibitors, Bioorg. Med. Chem. 42 (2021), 116269.
(2014) 204–217. [67] E. Viayna, N. Coquelle, M. Cieslikiewicz-Bouet, P. Cisternas, C.A. Oliva,
[44] M. Whiting, J. Muldoon, Y.C. Lin, S.M. Silverman, W. Lindstrom, A.J. Olson, H. E. Sánchez-López, M. Ettcheto, M. Bartolini, A. De Simone, M. Ricchini,
C. Kolb, M.G. Finn, K.B. Sharpless, J.H. Elder, V.V. Fokin, Inhibitors of HIV-1 M. Rendina, M. Pons, O. Firuzi, B. Pérez, L. Saso, V. Andrisano, F. Nachon,
protease by using in situ click chemistry, Angew. Chem., Int. Ed. Engl. 45 (2006) X. Brazzolotto, M.L. García, A. Camins, I. Silman, L. Jean, N.C. Inestrosa, J.-
1435–1439. P. Colletier, P.-Y. Renard, D. Munoz-Torrero, Discovery of a potent dual inhibitor
[45] S. Blaine-Sauer, T.L. Samuels, K. Yan, N. Johnston, The protease inhibitor of acetylcholinesterase and butyrylcholinesterase with antioxidant activity that
amprenavir protects against pepsin-induced esophageal epithelial barrier alleviates alzheimer-like pathology in old APP/PS1 mice, J. Med. Chem. 64
disruption and cancer-associated changes, Int. J. Mol. Sci. 24 (2023) 6765. (2021) 812–839.
[46] Z. Liu, X. Guo, A. Guo, S. Zhang, Y. Zou, Y. Wang, X. Li, W. He, L. Pu, S. Zhang, [68] P. de Andrade, S.P. Mantoani, P.S. Goncalves Nunes, C.R. Magadán, C. Pérez, D.
Q. Zeng, X. Cai, S. Wang, HIV protease inhibitor nelfinavir is a potent drug J. Xavier, E.T.S. Hojo, N.E. Campillo, A. Martínez, I. Carvalho, Highly potent and
candidate against echinococcosis by targeting Ddi1-like protein, EBioMedicine 82 selective aryl-1,2,3-triazolyl benzylpiperidine inhibitors toward
(2022), 104177. butyrylcholinesterase in Alzheimer’s disease, Bioorg. Med. Chem. 27 (2019)
[47] A. Brik, J. Muldoon, Y.-C. Lin, J.H. Elder, D.S. Goodsell, A.J. Olson, V.V. Fokin, K. 931–943.
B. Sharpless, C.-H. Wong, Rapid diversity-oriented synthesis in microtiter plates [69] L. Jalili-Baleh, H. Nadri, H. Forootanfar, T.T. Küçükkılınç, B. Ayazgök,
for in situ screening of HIV protease inhibitors, Chembiochem 4 (2003) M. Sharifzadeh, M. Rahimifard, M. Baeeri, M. Abdollahi, A. Foroumadi,
1246–1248. M. Khoobi, Chromone-lipoic acid conjugate: neuroprotective agent having
[48] D. Kang, D. Feng, L. Jing, Y. Sun, F. Wei, X. Jiang, G. Wu, E.D. Clercq, acceptable butyrylcholinesterase inhibition, antioxidant and copper-chelation
C. Pannecouque, P. Zhan, X. Liu, In situ click chemistry-based rapid discovery of activities, Daru 29 (2021) 23–38.
novel HIV-1 NNRTIs by exploiting the hydrophobic channel and tolerant regions [70] A. Iraji, O. Firuzi, M. Khoshneviszadeh, M. Tavakkoli, M. Mahdavi, H. Nadri,
of NNIBP, Eur. J. Med. Chem. 193 (2020), 112237. N. Edraki, R. Miri, Multifunctional iminochromene-2H-carboxamide derivatives
[49] C. Peruzzotti, S. Borrelli, M. Ventura, R. Pantano, G. Fumagalli, M. containing different aminomethylene triazole with BACE1 inhibitory,
S. Christodoulou, D. Monticelli, M. Luzzani, A.L. Fallacara, C. Tintori, M. Botta, neuroprotective and metal chelating properties targeting Alzheimer’s disease,
D. Passarella, Probing the binding site of abl tyrosine kinase using in situ click Eur. J. Med. Chem. 141 (2017) 690–702.
chemistry, ACS Med. Chem. Lett. 4 (2013) 274–277. [71] Z. Mi, B. Gan, S. Yu, J. Guo, C. Zhang, X. Jiang, T. Zhou, J. Su, R. Bai, Y. Xie, Dual-
[50] K.A. Kalesh, K. Liu, S.Q. Yao, Rapid synthesis of Abelson tyrosine kinase target anti-Alzheimer’s disease agents with both iron ion chelating and
inhibitors using click chemistry, Org. Biomol. Chem. 7 (2009) 5129–5136. monoamine oxidase-B inhibitory activity, J. Enzym. Inhib. Med. Chem. 34 (2019)
[51] M. Kugler, M. Hadzima, R. Dzijak, R. Rampmaier, P. Srb, L. Vrzal, Z. Voburka, 1489–1497.
P. Majer, P. Řezáčová, M. Vrabel, Identification of specific carbonic anhydrase [72] G. Moussa, R. Alaaeddine, L.M. Alaeddine, R. Nassra, A.S.F. Belal, A. Ismail, A.
inhibitors via in situ click chemistry, phage-display and synthetic peptide F. El-Yazbi, Y.S. Abdel-Ghany, A. Hazzaa, Novel click modifiable
libraries: comparison of the methods and structural study, RSC Med. Chem. 14 thioquinazolinones as anti-inflammatory agents: design, synthesis, biological
(2023) 144–153. evaluation and docking study, Eur. J. Med. Chem. 144 (2018) 635–650.
[52] V.P. Mocharla, B. Colasson, L.V. Lee, S. Röper, K.B. Sharpless, C.-H. Wong, H. [73] E.-H. Jung, J.-S. Hwang, M.-Y. Kwon, K.-H. Kim, H. Cho, I.K. Lyoo, S. Shin, J.-
C. Kolb, In situ click chemistry: enzyme-generated inhibitors of carbonic H. Park, I.-O. Han, A tryptamine-paeonol hybridization compound inhibits LPS-
anhydrase II, Angew. Chem., Int. Ed. Engl. 44 (2004) 116–120. mediated inflammation in BV2 cells, Neurochem. Int. 100 (2016) 35–43.
[53] B.L. Wilkinson, A. Innocenti, D. Vullo, C.T. Supuran, S.-A. Poulsen, Inhibition of [74] Y. Ali, M.S. Alam, H. Hamid, A. Husain, S. Bano, A. Dhulap, C. Kharbanda,
carbonic anhydrases with glycosyltriazole benzene sulfonamides, J. Med. Chem. S. Nazreen, S. Haider, Design, synthesis and biological evaluation of piperic acid
51 (2008) 1945–1953. triazolyl derivatives as potent anti-inflammatory agents, Eur. J. Med. Chem. 92
[54] V.P. Mocharla, J.C. Walsh, H.C. Padgett, H. Su, B. Fueger, W.A. Weber, J. Czernin, (2015) 490–500.
H.C. Kolb, From in situ to in vivo: an in situ click-chemistry-derived carbonic [75] M. Serafini, C. Cordero-Sanchez, R. Di Paola, I.P. Bhela, S. Aprile, B. Purghè,
anhydrase II imaging agent for positron emission tomography, ChemMedChem 8 R. Fusco, S. Cuzzocrea, A.A. Genazzani, B. Riva, T. Pirali, Store-operated calcium
(2013) 43–48. entry as a therapeutic target in acute pancreatitis: discovery and development of
drug-like SOCE inhibitors, J. Med. Chem. 63 (2020) 14761–14779.

15
R. Zhao et al. European Journal of Medicinal Chemistry 264 (2024) 116037

[76] F. Gado, R. Ferrisi, B. Polini, K.A. Mohamed, C. Ricardi, E. Lucarini, S. Carpi, potential antimicrobial and anti-biofilm agents, Eur. J. Med. Chem. 118 (2016)
F. Domenichini, L.A. Stevenson, S. Rapposelli, G. Saccomanni, P. Nieri, G. Ortore, 98–106.
R.G. Pertwee, C. Ghelardini, L. Di Cesare Mannelli, G. Chiellini, R.B. Laprairie, [94] S. Srivastava, D. Bimal, K. Bohra, B. Singh, P. Ponnan, R. Jain, M. Varma-Basil,
C. Manera, Design, synthesis, and biological activity of new CB2 receptor ligands: J. Maity, M. Thirumal, A.K. Prasad, Synthesis and antimycobacterial activity of 1-
from orthosteric and allosteric modulators to dualsteric/bitopic ligands, J. Med. (β-d-Ribofuranosyl)-4-coumarinyloxymethyl-/-coumarinyl-1,2,3-triazole, Eur. J.
Chem. 65 (2022) 9918–9938. Med. Chem. 150 (2018) 268–281.
[77] R.M. Bokhtia, S.S. Panda, A.S. Girgis, N. Samir, M.F. Said, A. Abdelnaser, S. Nasr, [95] P. Yadav, K. Lal, L. Kumar, A. Kumar, A. Kumar, A.K. Paul, R. Kumar, Synthesis,
M.S. Bekheit, A.S. Dawood, H. Sharma, M. Wade, S.K. Sharma, A.M. Ghanim, crystal structure and antimicrobial potential of some fluorinated chalcone-1,2,3-
New NSAID conjugates as potent and selective COX-2 inhibitors: synthesis, triazole conjugates, Eur. J. Med. Chem. 155 (2018) 263–274.
molecular modeling and biological investigation, Molecules 28 (2023) 1945. [96] Z. Zhang, K. Li, G.-Y. Zhang, Y.-Z. Tang, Z. Jin, Design, synthesis and biological
[78] O. Kutkat, A. Kandeil, Y. Moatasim, Y.A.M.M. Elshaier, W.A. El-Sayed, S. activities of novel pleuromutilin derivatives with a substituted triazole moiety as
T. Gaballah, A. El Taweel, M.N. Kamel, M. El Sayes, M.A. Ramadan, R. El- potent antibacterial agents, Eur. J. Med. Chem. 204 (2020), 112604.
Shesheny, F.M.E. Abdel-Megeid, R. Webby, G. Kayali, M.A. Ali, In Vitro and in [97] F.-C. Wang, B. Peng, T.-T. Ren, S.-P. Liu, J.-R. Du, Z.-H. Chen, T.-T. Zhang, X. Gu,
vivo antiviral studies of new heteroannulated 1,2,3-triazole glycosides targeting M. Li, S.-L. Cao, X. Xu, A 1,2,3-triazole derivative of quinazoline exhibits
the neuraminidase of influenza A viruses, Pharmaceuticals 15 (2022) 351. antitumor activity by tethering RNF168 to SQSTM1/P62, J. Med. Chem. 65
[79] O.V. Andreeva, B.F. Garifullin, V.V. Zarubaev, A.V. Slita, I.L. Yesaulkova, L. (2022) 15028–15047.
F. Saifina, M.M. Shulaeva, M.G. Belenok, V.E. Semenov, V.E. Kataev, Synthesis of [98] X. Yang, Z.-P. Wang, S. Xiang, D. Wang, Y. Zhao, D. Luo, Y. Qiu, C. Huang, J. Guo,
1,2,3-triazolyl nucleoside analogues and their antiviral activity, Mol. Divers. 25 Y. Dai, S.-L. Zhang, Y. He, Optimization of the natural product calothrixin A to
(2021) 473–490. discover novel dual topoisomerase I and II inhibitors with improved anticancer
[80] D.A. Tatarinov, B.F. Garifullin, M.G. Belenok, O.V. Andreeva, I.Y. Strobykina, A. activity, J. Med. Chem. 65 (2022) 8040–8061.
V. Shepelina, V.V. Zarubaev, A.V. Slita, A.S. Volobueva, L.F. Saifina, M. [99] Y. Ding, H. Guo, W. Ge, X. Chen, S. Li, M. Wang, Y. Chen, Q. Zhang, Copper(I)
M. Shulaeva, V.E. Semenov, V.E. Kataev, The first 5’-phosphorylated 1,2,3-tria­ oxide nanoparticles catalyzed click chemistry based synthesis of
zolyl nucleoside analogues with uracil and quinazoline-2,4-dione moieties: a melampomagnolide B-triazole conjugates and their anti-cancer activities, Eur. J.
synthesis and antiviral evaluation, Molecules 27 (2022) 6214. Med. Chem. 156 (2018) 216–229.
[81] Y.-W. He, C.-Z. Dong, J.-Y. Zhao, L.-L. Ma, Y.-H. Li, H.A. Aisa, 1,2,3-Triazole- [100] X. He, G. He, Z. Chu, H. Wu, J. Wang, Y. Ge, H. Shen, S. Zhang, J. Shan, K. Peng,
containing derivatives of rupestonic acid: click-chemical synthesis and antiviral Z. Wei, Y. Zou, Y. Xu, Q. Zhu, Discovery of the first potent Ido1/Ido2 dual
activities against influenza viruses, Eur. J. Med. Chem. 76 (2014) 245–255. inhibitors: a promising strategy for cancer immunotherapy, J. Med. Chem. 64
[82] Y. Su, L. Meng, J. Sun, W. Li, L. Shao, K. Chen, D. Zhou, F. Yang, F. Yu, Design, (2021) 17950–17968.
synthesis of oleanolic acid-saccharide conjugates using click chemistry [101] R. Venkatesh, M.J. Ramaiah, H.K. Gaikwad, S. Janardhan, R. Bantu, L. Nagarapu,
methodology and study of their anti-influenza activity, Eur. J. Med. Chem. 182 G.N. Sastry, A.R. Ganesh, M. Bhadra, Luotonin-A based quinazolinones cause
(2019), 111622. apoptosis and senescence via HDAC inhibition and activation of tumor suppressor
[83] Z. Fang, D. Kang, L. Zhang, B. Huang, H. Liu, C. Pannecouque, E. De Clercq, proteins in HeLa cells, Eur. J. Med. Chem. 94 (2015) 87–101.
P. Zhan, X. Liu, Synthesis and biological evaluation of a series of 2-((1- [102] J. Huang, J. Zhang, W. Xu, Q. Wu, R. Zeng, Z. Liu, W. Tao, Q. Chen, Y. Wang, W.-
substituted-1H-1,2,3-triazol-4-yl)methylthio)-6-(naphthalen-1-ylmethyl) G. Zhu, Structure-based discovery of selective histone deacetylase 8 degraders
pyrimidin-4(3H)-one as potential HIV-1 inhibitors, Chem. Biol. Drug Des. 86 with potent anticancer activity, J. Med. Chem. 66 (2023) 1186–1209.
(2015) 614–618. [103] A. Bistrovic, L. Krstulovic, A. Harej, P. Grbcic, M. Sedic, S. Kostrun, S.K. Pavelic,
[84] I. Mohammed, I.R. Kummetha, G. Singh, N. Sharova, G. Lichinchi, J. Dang, M. Bajic, S. Raic-Malic, Design, synthesis and biological evaluation of novel
M. Stevenson, T.M. Rana, 1,2,3-Triazoles as amide bioisosteres: discovery of a benzimidazole amidines as potent multi-target inhibitors for the treatment of non-
new class of potent HIV-1 vif antagonists, J. Med. Chem. 59 (2016) 7677–7682. small cell lung cancer, Eur. J. Med. Chem. 143 (2018) 1616–1634.
[85] G. Wu, W.A. Zalloum, M.E. Meuser, L. Jing, D. Kang, C.-H. Chen, Y. Tian, [104] A.M. Macan, N. Perin, S. Jakopec, M. Mioc, M.R. Stojkovic, M. Kralj, M. Hranjec,
F. Zhang, S. Cocklin, K.-H. Lee, X. Liu, P. Zhan, Discovery of phenylalanine S. Raic-Malic, Synthesis, antiproliferative activity and DNA/RNA-binding
derivatives as potent HIV-1 capsid inhibitors from click chemistry-based properties of mono- and bis-(1,2,3-triazolyl)-appended benzimidazo[1,2-a]
compound library, Eur. J. Med. Chem. 158 (2018) 478–492. quinoline derivatives, Eur. J. Med. Chem. 185 (2020), 111845.
[86] Z. Zhou, T. Liu, G. Wu, D. Kang, Z. Fu, Z. Wang, E. De Clercq, C. Pannecouque, [105] A.M. Macan, A. Harej, I. Cazin, M. Klobucar, V. Stepanic, K. Pavelic, S.K. Pavelic,
P. Zhan, X. Liu, Targeting the hydrophobic channel of NNIBP: discovery of novel D. Schols, R. Snoeck, G. Andrei, S. Raic-Malic, Antitumor and antiviral activities
1,2,3-triazole-derived diarylpyrimidines as novel HIV-1 NNRTIs with high of 4-substituted 1,2,3-triazolyl-2,3-dibenzyl-L-ascorbic acid derivatives, Eur. J.
potency against wild-type and K103N mutant virus, Org. Biomol. Chem. 17 Med. Chem. 184 (2019), 111739.
(2019) 3202–3217. [106] T.G. Kraljevic, A. Harej, M. Sedic, S.K. Pavelic, V. Stepanic, D. Drenjancevic,
[87] A.V. Aralov, G.V. Proskurin, A.A. Orlov, L.I. Kozlovskaya, A.A. Chistov, S. J. Talapko, S. Raic-Malic, Synthesis, in vitro anticancer and antibacterial activities
V. Kutyakov, G.G. Karganova, V.A. Palyulin, D.I. Osolodkin, V.A. Korshun, and in silico studies of new 4-substituted 1,2,3-triazole-coumarin hybrids, Eur. J.
Perylenyltriazoles inhibit reproduction of enveloped viruses, Eur. J. Med. Chem. Med. Chem. 124 (2016) 794–808.
138 (2017) 293–299. [107] S.P. Shaik, V.L. Nayak, F. Sultana, A.V.S. Rao, A.B. Shaik, K.S. Babu, A. Kamal,
[88] A. Musa, H.S. Abulkhair, A. Aljuhani, N. Rezki, M.A. Abdelgawad, K. Shalaby, A. Design and synthesis of imidazo[2,1-b]thiazole linked triazole conjugates:
H. El-Ghorab, M.R. Aouad, Phenylpyrazolone-1,2,3-triazole hybrids as potent microtubule-destabilizing agents, Eur. J. Med. Chem. 126 (2017) 36–51.
antiviral agents with promising SARS-CoV-2 main protease inhibition potential, [108] V.G. Reddy, S.R. Bonam, T.S. Reddy, R. Akunuri, V.G.M. Naidu, V.L. Nayak, S.
Pharmaceuticals 16 (2023) 463. K. Bhargava, H.M.S. Kumar, P. Srihari, A. Kamal, 4β-amidotriazole linked
[89] T. Ni, F. Xie, Y. Hao, L. Li, S. Zhu, H. Wu, X. Chi, L. Yan, Y. Jiang, D. Zhang, podophyllotoxin congeners: DNA topoisomerase-IIα inhibition and potential
Discovery of novel orally bioavailable triazoles with potent and broad-spectrum anticancer agents for prostate cancer, Eur. J. Med. Chem. 144 (2018) 595–611.
antifungal activity in vitro and in vivo, J. Med. Chem. 65 (2022) 16665–16678. [109] Z. Zuo, X. Liu, X. Qian, T. Zeng, N. Sang, H. Liu, Y. Zhou, L. Tao, X. Zhou, N. Su,
[90] L. Goswami, L. Gupta, S. Paul, M. Vermani, P. Vijayaraghavan, A.K. Bhattacharya, Y. Yu, Q. Chen, Y. Luo, Y. Zhao, Bifunctional naphtho[2,3-d][1,2,3]triazole-4,9-
Design and synthesis of eugenol/isoeugenol glycoconjugates and other analogues dione compounds exhibit antitumor effects in vitro and in vivo by inhibiting
as antifungal agents against Aspergillus fumigatus, RSC Med. Chem. 13 (2022) dihydroorotate dehydrogenase and inducing reactive oxygen species production,
955–962. J. Med. Chem. 63 (2020) 7633–7652.
[91] Z. Jiang, J. Gu, C. Wang, S. Wang, N. Liu, Y. Jiang, G. Dong, Y. Wang, Y. Liu, [110] V. Janganati, J. Ponder, M. Balasubramaniam, P. Bhat-Nakshatri, E.E. Bar,
J. Yao, Z. Miao, W. Zhang, C. Sheng, Design, synthesis and antifungal activity of H. Nakshatri, C.T. Jordan, P.A. Crooks, MMB triazole analogs are potent NF-κB
novel triazole derivatives containing substituted 1,2,3-triazole-piperdine side inhibitors and anti-cancer agents against both hematological and solid tumor
chains, Eur. J. Med. Chem. 82 (2014) 490–497. cells, Eur. J. Med. Chem. 157 (2018) 562–581.
[92] N. Fu, S. Wang, Y. Zhang, C. Zhang, D. Yang, L. Weng, B. Zhao, L. Wang, Efficient [111] W. Chen, X. Chen, D. Li, X. Wang, G. Long, Z. Jiang, Q. You, X. Guo, Discovery of
click chemistry towards fatty acids containing 1,2,3-triazole: design and synthesis a potent MLL1 and WDR5 protein-protein interaction inhibitor with in vivo
as potential antifungal drugs for Candida albicans, Eur. J. Med. Chem. 136 (2017) antitumor activity, Eur. J. Med. Chem. 223 (2021), 113677.
596–602. [112] W. Hou, Z. Luo, G. Zhang, D. Cao, D. Li, H. Ruan, B.H. Ruan, L. Su, H. Xu, Click
[93] T. Vijai Kumar Reddy, A. Jyotsna, B.L.A. Prabhavathi Devi, R.B.N. Prasad, chemistry-based synthesis and anticancer activity evaluation of novel C-14 1,2,3-
Y. Poornachandra, C. Ganesh Kumar, Design, synthesis and in vitro biological triazole dehydroabietic acid hybrids, Eur. J. Med. Chem. 138 (2017) 1042–1052.
evaluation of short-chain C12-sphinganine and its 1,2,3-triazole analogs as

16

You might also like