Acs - Jmedchem.1c02001 Ispinesib As An Effective Warhead For The Design of Autophagosome-Tethering Chimeras Discovery of Potent Degraders of Nicotinamide Phosphoribosyltransferase (NAMPT)

You might also like

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

pubs.acs.

org/jmc Article

Ispinesib as an Effective Warhead for the Design of


Autophagosome-Tethering Chimeras: Discovery of Potent
Degraders of Nicotinamide Phosphoribosyltransferase (NAMPT)
Guoqiang Dong,*,∥ Ying Wu,∥ Junfei Cheng,∥ Long Chen, Rui Liu, Yu Ding, Shanchao Wu, Junhui Ma,
and Chunquan Sheng*
Cite This: J. Med. Chem. 2022, 65, 7619−7628 Read Online
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ACCESS Metrics & More Article Recommendations *


sı Supporting Information

ABSTRACT: Autophagosome-tethering compounds (ATTECs) are an emerging new


Downloaded via NATL TAIWAN UNIV on July 3, 2022 at 15:18:00 (UTC).

technology in targeted protein degradation. However, effective tools and successful


examples for autophagosome-tethering chimeras are still rather limited. Herein, ATTEC
ispinesib was identified for the first time to be an effective warhead to design
autophagosome-tethering chimeras. As a conceptual validation study, the first generation
of autophagic degraders of nicotinamide phosphoribosyltransferase (NAMPT) were
developed by connecting the NAMPT inhibitor and LC3-binding ispinesib through a
flexible linker. In particular, compound A3 significantly induced the degradation of
NAMPT through the autophagy-lysosomal pathway, leading to excellent cellular antitumor
potency. Ispinesib may have broad applications in the design of potent autophagosome-
tethering chimeras.

■ INTRODUCTION
Over the past 2 decades, targeted protein degradation
and engulfs cellular cargos (e.g., proteins and organelles) to form
the double-membrane autophagosome,14 followed by the fusion
exemplified by proteolysis-targeting chimeras (PROTACs) has with lysosomes to generate the autolysosome, resulting in the
aroused great attention and represents an emerging new degradation of cellular cargoes.15,16
technology in the field of drug discovery.1−3 PROTACs are While UPS-dependent PROTACs are limited to degrade the
heterobifunctional chimeras consisting of a warhead binding to soluble intracellular proteins, the ALP pathway can effectively
the protein of interest (POI), a flexible linker, and an E3 ligase remove a broader range of cellular biomolecules (e.g., protein
aggregates and other nonprotein molecules). Therefore,
ligand.4,5 Based on the ubiquitin proteasome system (UPS),
exploiting the power of ALP to degrade target proteins may
PROTACs induce the POI−PROTAC−E3 ternary complex
greatly enrich the targeted protein degradation techniques and
formation by hijacking E3 ligase to recruit POI, leading to its
open a new avenue for drug discovery. Recently, autophagy-
ubiquitination and subsequent proteasomal degradation.4
targeting chimeras (AUTACs)17 and autophagosome-tethering
Recently, a large number of PROTAC molecules have been
compounds (ATTECs)18 have been developed based on
successfully designed to degrade a variety of drug targets.6−8
macroautophagy by hijacking autophagosomes to degrade
More than 10 PROTACs (e.g., ARV-110 and ARV-471) have
bulky cytosolic cargos. Among them, ATTECs act by binding
progressed into clinical trials.9 However, the PROTAC
directly and simultaneously to the mutant HTT protein
technology has several inherent obstacles and limitations,10
(mHTT) and LC3.18 Because the LC3 is attached to the
such as E3 ligase-related drug resistance11 and limited scope of
phagophore membrane and plays a central role in recruiting the
protein targets (only soluble proteins in the cytoplasm).10
specific cargos into the autophagosome (Figure 1A),19 ATTECs
The autophagy-lysosomal pathway (ALP) is another major
have the capacity to drive mHTT directly to the autophagosome
cellular degradation system in eukaryotic cells, which is primarily
for subsequent autophagic degradation. As a proof-of-concept
responsible for the clearance of dispensable proteins, protein
aggregates, droplets, and even defective or surplus organ-
elles.12,13 ALP can be divided into three major subtypes: Received: November 19, 2021
macroautophagy, microautophagy, and chaperone-mediated Published: May 19, 2022
autophagy (CMA).13 Macroautophagy is the most widely
studied form, which is initiated by the formation of a cup-
shaped double-membraned structure known as phagophore
(Figure 1A). During the process, phagophore further expands

© 2022 American Chemical Society https://doi.org/10.1021/acs.jmedchem.1c02001


7619 J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Figure 1. Design of NAMPT degraders via autophagy. (A) Overview of the macroautophagy process. (B) Binding mode of MS2 (green) with NAMPT
and the design strategy of the autophagy-based NAMPT degraders (PDB: 2GVJ). (C) Schematic illustration of the POI degradation via autophagy.

study, Lu et al. identified four ATTECs (i.e., 10O5, ispinesib, potent inhibitor of kinesin spindle protein (KSP) with an IC50
AN1, and AN2, Figure S1 in the Supporting Information). Based value of 0.5 nM.21,22 KSP is a kinesin motor protein, and it plays
on these molecular glues, they further developed chimeric an important role in the formation of a bipolar mitotic spindle
autophagy-tethering compounds for the autophagic degradation and cell cycle progression through mitosis.22 It has been shown
of lipid droplets (LDs).20 These molecules were made of LC3- that abnormal expression of KSP is correlated with a variety of
binding molecules (10O5 and AN2) and LD-binding probes
human cancers. Using ispinesib as a warhead, autophagy-based
connected by flexible linkers, which was similar to that of
degraders of nicotinamide phosphoribosyltransferase
PROTACs. Despite the progress made in this field, the design
tools for chimeric autophagy-tethering degraders are still rather (NAMPT) were successfully identified, which showed excellent
limited. Moreover, whether such compounds could effectively degradation potency as well as antitumor activity. This study
degrade therapeutic targets still remains unknown. To answer demonstrated that autophagosome-tethering chimeras could be
these questions, herein, ispinesib was used for the first time to an effective strategy for targeted protein degradation of drug
design chimeric autophagy-tethering degraders. Ispinesib is a targets and discovery of novel therapeutic agents.
7620 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Figure 2. Colocalization experiment of LC3-B protein probes (green) and lysosomes (red). (A) Design of the fluorescent probe for LC3-B by
connecting the FITC group to the amino group of ispinesib. (B) Schematic of the theory for FP assays and the affinity measurement by fluorescence
anisotropy with LC3-B and P1. Error bars represent a standard deviation of the measurements performed in triplicate. (C) Confocal microscopy
images of Hela cells treated by our probe P1 (green) and lysosome probe (red).

■ RESULTS AND DISCUSSION


Design and Synthesis of NAMPT-Degrading ATTECs.
NAMPT inhibitors and identified compound 1 (MS2, Figure
1B) as a potent NAMPT inhibitor (IC50 = 85 nM).29 Docking
We choose NAMPT for ATTEC design because it was a highly studies showed that the terminal piperazine group of compound
challenging target for the development of antitumor agents. 1 extended out of the active site of NAMPT without direct
NAMPT is the rate-limiting enzyme of nicotinamide adenine interaction with NAMPT (Figure 1B), offering a favorable site
dinucleotide (NAD), which plays a crucial role in multiple for the extension of the linker and LC3 ligand.
cellular processes.23−26 However, clinical development of Then, the key step for the degrader design is to select an
NAMPT inhibitors (i.e., FK866 and CHS828, Figure S2 in the effective LC3 warhead. Among the reported LC3-binding
Supporting Information) failed due to limited efficacy and dose- molecules, indolone LC3 ligands 10O5 and AN1 exist in the
dependent toxicity.27,28 We envisioned that autophagy-based mixture of cis-configuration (active form) and trans-config-
NAMPT degradation might offer a novel strategy for antitumor uration (inactive form), which significantly decreased the
drug discovery. degrading potency. Due to its stability and verified config-
Similar to the molecular design of PROTAC, the autophagy- uration, herein, we explored the feasibility of ispinesib for the
based degrader can also be divided into three parts: a ligand of design of autophagosome-tethering chimeras. To further verify
POI, a flexible linker, and an LC3 ligand (Figure 1B), which is the direct binding between ispinesib warhead and LC3 protein,
supposed to bind to both NAMPT and LC3. As a result, POI we measured the binding affinity through the isothermal
and the LC3 protein could be brought closer, and then, the POI titration calorimetry (ITC) assay and the KD was 11.8 μM,
is labeled, recruited, and fused into autophagosome. The target indicating that ispinesib could bind to LC3 effectively (Figure S9
protein will be degraded via lysosome-mediated autophagy in the Supporting Information). Currently, the binding mode of
(Figure 1C). Previously, our group screened and optimized ispinesib with LC3 is still unknown. Thus, it is important to
7621 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Scheme 1. Synthetic Route of Target Compounds A1−A5a

a
Reagents and conditions: (a) bromide-substituted linkers, DIPEA, DCM, 60 °C, 2 h, yield 60−80%; (b) i. CF3COOH, DCM or LiOH, THF/
MeOH/H2O; ii. compound 2, HATU, DIPEA, DMF, rt, 2 h, yield 36−56%.

determine a suitable site to attach the linker without interfering described previously.31 As shown in Table 1, all the target
the critical interactions with LC3. compounds showed good inhibitory activities with the IC50
To validate the rationality of the structural modification
position of ispinesib and its ability to be engulfed into Table 1. Biological Evaluation of NAMPT Autophagy
autophagosome, the first small-molecule fluorescent probe Degraders
(P1) for LC3 protein was designed and synthesized by
connecting the fluorophore FITC to the amino group of
ispinesib (compound 2) directly through a thiourea linker
(Figure 2A, Scheme S1 in the Supporting Information).
Fluorescence polarization (FP) technology was used to measure
the binding affinity between probe P1 and LC3.30 As shown in
Figure 2B, probe P1 demonstrated nanomolar affinity with a KD
value of 175 nM, indicating that the attachment point for linker
conjugation would improve the binding affinity to LC3 protein.
The colocalization experiment was conducted to verify the
distribution of probe P1 in HeLa cells, which were treated with
the probe for 2 h, followed by staining with red lysosome probes
for another 1.5 h. As shown in Figure 2C, the green fluorescent
probes were colocalized with the red lysosomal probe,
suggesting that the conjugate binding with LC3 could be
engulfed into autophagosome.
On the basis of the above analysis, a series of NAMPT
autophagy degraders were rationally designed and synthesized
by connecting NAMPT inhibitor 1 and ispinesib with different
lengths of alkyl or poly ethylene glycol (PEG) linkers. The target
compounds were synthesized according to Scheme 1. NAMPT
inhibitor 1 was prepared according to the literature protocols.29
Subsequently, intermediates 3a−h were obtained through
substitution reactions between compound 1 and bromide-
substituted linkers under the alkaline condition. After removing
the protective group, the intermediates underwent amide values ranging from 10 to 34 nM, suggesting the good binding
condensation with compound 2 to afford target compounds affinity between the target compounds and NAMPT. Then, we
A1−7. investigated whether target compounds could degrade NAMPT
Evaluation of ATTEC-Induced NAMPT Degradation. in human ovarian cancer A2780 cells, in which NAMPT is
Initially, the NAMPT inhibitory activities of all the designed overexpressed. The results indicated that all the compounds
NAMPT autophagy degraders were determined by the assays were able to trigger NAMPT degradation after treatment for 48
7622 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Figure 3. Characterization of potent NAMPT autophagy degraders. (A) Expression level of NAMPT in A2780 cells treated with target compounds for
48 h. (B) Cell growth inhibition activity of compound A3 and MS2. Error bars represent a standard deviation of the measurements performed in
triplicate.

h (Figure 3A). The degradation efficiency increased gradually Compound A3 also significantly inhibited the viability of
along with the linker length. When the length was 5−7 atoms HCT116 cells, which were highly dependent on the NAD
(compounds A6, A1 and A2), they showed relatively weak salvage pathway for survival. In particular, the IC50 value was 46
efficiency with the maximum degradation rates in the range of nM for compound A3 against A2780 cells, which was
1−9% at 0.1 μM. As the length was extended to 8-atom significantly more potent than NAMPT inhibitor MS2 (IC50 =
(compound A3), it showed the best degradation activity. 490 nM). Moreover, compound A3 was stable under the
Compound A3 induced the NAMPT degradation in a experimental condition and did not degrade into free drugs
concentration- and time-dependent manner (Figure S3 in the (Figure S7 in the Supporting Information).
Supporting Information) with a maximal degradation rate of Investigation of the Mechanism of Action of NAMPT
91% at 3 μM. When the linker length was further extended to 10- Autophagic Degrader A3. The underlying mechanism for the
atoms (compound A5), the degradation activity was obviously NAMPT-degrading effect of compound A3 was further studied.
decreased (maximum degradation rate: 33% at about 0.1 μM). First, direct target engagement was performed in the cell lysate
In addition, surface plasmon resonance (SPR) experiments were by cellular thermal shift assay (CETSA) to investigate whether
performed to further verify the binding affinity between the designed compounds bound to NAMPT directly. As shown
compound A3 and LC3 protein, with a KD value of 933 nM in Figure S8, treatment with compound A3 enhanced the
(Figure S4 in the Supporting Information). Ispinesib was a well- stability of NAMPT proteins compared to the blank control,
known kinesin spindle protein (KSP) inhibitor. Therefore, we indicating that it was capable of stabilizing NAMPT in cells. In
further evaluated the selectivity of compound A3 by monitoring addition, the CETSA assay also revealed that compound A3 was
the degradation of KSP. As shown in Figure S5, compound A3 able to bind with KSP, which might partly contribute to the
had no effect on KSP degradation, indicating the selective cytotoxicity of compound A3. Thus, as an ATTEC warhead,
NAMPT degrading activity. To evaluate whether the NAMPT ispinesib remained to be further optimized to reduce the binding
degradation could led to antitumor potency, the cellular activity to KSP. In order to investigate whether the target
antiproliferative activity of compound A3 was assayed for compounds caused the NAMPT degradation via lysosomal-
several cell lines (Figure 3B), including A2780, MBA-MB-231 mediated autophagy, different autophagy inhibitors (ammo-
(NAMPT overexpression), HCT116 (NAMPT in normal), and nium chloride, chloroquine, wortmannin, 3-methyladenine, and
A549 (NAMPT low expression, Figure S6 in the Supporting LY294002)19,32−35 were added to evaluate their effects on
Information). Interestingly, the excellent inhibitory activities reversing the expression level of NAMPT. Among them,
were observed in cell lines with overexpressed NAMPT. ammonium chloride (NH4Cl) and chloroquine can block the
7623 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Figure 4. Compound A3 induced NAMPT degradation via autophagy. (A) A2780 cells were pretreated with MS2, FK866, NH4Cl, chloroquine,
wortmannin, 3-methyladenine or LY294002, followed by treatment with A3 for an additional 48 h. (B) Compound A3 could not reduce NAMPT
levels in Atg7-knockdown cells. Left panel: relative Atg7 levels in A2780 cells and Atg7-knockdown A2780 cells. Right panel: NAMPT levels in A2780
cells and Atg7-knockdown A2780 cells treated with A3 at 500 nM concentration.

function of lysosomes by acidifying the environment of degraders.36 Although compound A3 did not show better
lysosomes and then impede the fusion process of autophagy degradation activity than NAMPT PROTACs, this work proved
and lysosome. Wortmannin, 3-methyladenine and LY294002 that NAMPT could also be effectively degraded by the ALP
are able to block or interfere with the formation of system, providing an alternative strategy for targeted degrada-
autophagosome, thus inhibiting autophagy.19 As shown in tion of NAMPT. Moreover, we identified ispinesib as an
Figure 4A, the total protein level of NAMPT was reversed upon effective degradation tag and provided a useful tool for ALP-
the addition of autophagy inhibitors, indicating that compound based targeted protein degradation. However, it should be noted
A3 induced NAMPT degradation through lysosomal-mediated that ispinesib is a potent KSP inhibitor and further optimization
autophagy. Meanwhile, the coculture of NAMPT inhibitor MS2 is required to improve the affinity and selectivity toward LC3
or FK866 also led to the recuperation of NAMPT expression, protein. The application of LC3 binder ispinesib to degrade
indicating that the binding mode of compound A3 with more drug targets and structural optimization of ispinesib
NAMPT was similar to that of FK866 and MS2. LC3 ligand warhead is currently in progress.


ispinesib was added to compete with compound A3 for binding
with LC3 protein. However, the A2780 cells were all dead after EXPERIMENTAL SECTION
pretreating with ispinesib because of high toxicity. As an General Methods. All the reagents and solvents were obtained
alternative, probe P1 was used in the competitive experiment commercially and used without further purification. 1H NMR and 13C
and the result revealed that the degrading activity was reversed NMR spectra were collected using Bruker AVANCE300 or
due to the blockage of LC3 binding (Figure S10 in the AVANCE600 spectrometers (Leipzig, Bruker Company, Germany)
Supporting Information). Atg7 is essential to lysosome- using tetramethylsilane (TMS) as an interior label and dimethyl
mediated autophagy.13 When Atg7 was knocked down through sulfoxide (DMSO)-d6 as solvents. Chemical shifts (δ) are expressed in
infection via lentivirus in A2780 cells, compound A3 was unable parts per million (ppm). The mass spectra were measured on an Esquire
to induce NAMPT degradation (Figure 4B), indicating that the 3000 LC-MS mass spectrometer. Thin-layer chromatography analysis
was performed with GF254 silica gel plates to detect reactions (Haiyang
degradation relied on the pathway of lysosome-mediated
Chemical, Qingdao, China), and visualization of reactants occurred
autophagy. under 254 nm UV light. Silica gel column chromatography was carried

■ CONCLUSIONS
In summary, ispinesib was identified to be an effective warhead
out with Silica Gel 60 G (Haiyang Chemical, Qingdao, China). High-
performance liquid chromatography analyses were performed on an
Agilent Technologies 1206, instrument to determine purity, and water
and methanol were used as the mobile phase. The final compounds
to design autophagosome-tethering chimeras. Using ispinesib as exhibited the purity greater than 95%.
the LC3 ligand, a series of novel NAMPT autophagic degraders tert-Butyl 7-(4-((4-(3-(pyridin-3-ylmethyl)thioureido)-
were designed and synthesized. Compound A3 effectively phenyl)sulfonyl)piperazin-1-yl)heptanoate (3a). Compound 1
degraded NAMPT, leading to excellent cellular antitumor (500 mg, 1.3 mmol) and tert-butyl bromoacetate (338 mg, 1.3 mmol)
potency. Mechanism studies confirmed that compound A3 were dissolved in dry dichloromethane (DCM) (20 mL), and then,
N,N-diisopropylethylamine (DIPEA) (335 mg, 2.6 mmol) was slowly
caused the NAMPT degradation via lysosomal-mediated added dropwise. The mixture was stirred at 60 °C for 2 h. After that, the
autophagy. PROTAC has been emerging as a promising reaction solvent was evaporated under reduced pressure and the residue
approach in the field of drug discovery and development. was purified by silica gel column chromatography (DCM/MeOH =
Recently, using PROTAC technology, we also designed and 100/1) to obtain a white solid intermediate 3a (512 mg, yield 68%). 1H
identified several NAMPT PROTACs as potent NAMPT NMR (DMSO-d6, 600 MHz) δ: 10.17 (s, 1H), 8.66 (s, 1H), 8.59 (s,

7624 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

1H), 8.49 (d, J = 4.7 Hz, 1H), 7.82 (d, J = 8.6 Hz, 2H), 7.78 (d, J = 7.9 J = 4.8, 1.3 Hz, 1H), 8.22 (d, J = 8.6 Hz, 1H), 7.81 (d, J = 8.6 Hz, 2H),
Hz, 1H), 7.67 (d, J = 8.6 Hz, 2H), 7.40 (dd, J = 7.8, 4.9 Hz, 1H), 4.81 (d, 7.75−7.78 (m, 2H), 7.63−7.67 (m, 3H), 7.34−7.38 (m, 3H), 7.28−
J = 5.5 Hz, 2H), 2.87 (s, 4H), 2.41 (s, 4H), 2.24 (s, 2H), 1.43−1.47 (m, 7.33 (m, 2H), 7.18−7.26 (m, 6H), 8.87 (d, J = 16.4 Hz, 1H), 5.53 (d, J =
2H), 1.38 (s, 9H), 131−1.37 (m, 2H), and 1.17−1.27 (m, 6H). 10.4 Hz, 1H), 4.80 (d, J = 5.5 Hz, 2H), 3.17−3.26 (m, 2H), 2.66−2.75
Preparation methods of target compounds 3b−g were the same as (m, 1H), 2.34−2.50 (m, 4H), 2.31 (s, 3H), 2.16−2.27 (m, 2H), 1.68−
that of target compound 3a. 1.79 (m, 2H), 1.21−1.35 (m, 9H), 1.05−1.17 (m, 4H), 0.89 (d, J = 6.8
tert-Butyl 8-(4-((4-(3-(pyridin-3-ylmethyl)thioureido)- Hz, 3H), 0.79−0.86 (m, 2H), and 0.47 (d, J = 6.2 Hz, 3H); 13C NMR
phenyl)sulfonyl)piperazin-1-yl)octanoate (3b). White solid, (DMSO-d6, 150 MHz) δ: 185.98, 177.24, 176.85, 166.33, 160.45,
yield 53%. 1H NMR (DMSO-d6, 600 MHz) δ: 10.14 (s, 1H), 8.64 (s, 154.11, 153.40, 152.37, 149.25, 144.70, 143.88, 141.87, 140.51, 139.37,
1H), 8.58 (d, J = 2.0 Hz, 1H), 8.48 (dd, J = 4.8, 1.4 Hz, 1H), 7.76−7.83 138.96, 134.09, 133.87, 133.62, 133.22, 132.65, 131.86, 131.60, 131.07,
(m, 3H), 7.66 (d, J = 8.74 Hz, 2H), 7.39 (dd, J = 7.9, 4.8 Hz, 1H), 4.80 128.66, 126.76, 124.27, 99.99, 64.21, 66.75, 50.95, 50.39, 49.92, 47.67,
(d, J = 5.6 Hz, 2H), 2.86 (s, 4H), 2.40 (s, 4H), 2.23 (s, 2H), 2.14 (t, J = 40.92, 40.43, 35.39, 34.16, 33.71, 33.57, 31.65, 30.24, 26.11, 24.71, and
7.3 Hz, 2H), 1.41−1.48 (m, 2H), 1.38 (s, 9H), 1.30−1.36 (m, 2H), and 23.36; HRMS (ESI, positive) m/z calcd for C54H65ClN9O5S2 [M +
1.18−1.23 (m, 6H). High-resolution mass spectrometry (HRMS) (ESI, H]+: 1018.4233; found, 1018.4260.
negative) m/z calcd for C29H43N5O4S2 [M − H]+, 588.2684; found, Preparation methods of target compounds A2-A7 were the same as
588.2701. that of target compound A1.
Ethyl 9-(4-((4-(3-(pyridin-3-ylmethyl)thioureido)phenyl)- N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
sulfonyl)piperazin-1-yl)nonanoate (3c). White solid, yield 55%. 2-methylpropyl)-4-methyl-N-(3-(8-(4-((4-(3-(pyridin-3-
1
H NMR (DMSO-d6, 600 MHz) δ: 10.11 (s, 1H), 8.59−8.66 (m, 1H), ylmethyl)thioureido)phenyl)sulfonyl)piperazin-1-yl)-
8.57 (d, J = 1.78 Hz, 1H), 8.47 (dd, J = 1.4, 15.4 Hz, 1H), 7.72−7.83 (m, octanamido)propyl)benzamide (A2). White solid, yield 52%. 1H
3H), 7.60−7.69 (m, 2H), 7.38 (dd, J = 7.8, 4.8 Hz, 1H), 4.79 (d, J = 5.5 NMR (DMSO-d6, 600 MHz) δ: 10.13 (s, 1H), 8.39−8.72 (m, 3H),
Hz, 2H), 3.98−4.11 (m, 2H), 3.17 (d, J = 5.3 Hz, 1H), 2.85 (s, 4H), 8.23 (d, J = 8.2 Hz, 1H), 7.72−7.88 (m, 4H), 7.59−7.70 (m, 3H),
2.39 (s, 4H), 2.16−2.23 (m, 4H), 1.27−1.51 (m, 4H), and 1.08−1.27 7.14−7.43 (m, 11H), 5.88 (d, J = 16.2 Hz, 1H), 5.54 (d, J = 9.8 Hz, 1H),
(m, 10H). 5.06 (d, J = 16.5 Hz, 1H), 4.80 (s, 2H), 3.19−3.28 (m, 2H), 2.86 (s,
Ethyl 10-(4-((4-(3-(pyridin-3-ylmethyl)thioureido)phenyl)- 4H), 2.67−2.76 (m, 1H), 2.39 (s, 4H), 2.32 (s, 3H), 2.22 (s, 2H),
sulfonyl)piperazin-1-yl)decanoate (3d). White solid, yield 58%. 1.67−1.81 (m, 2H), 1.37−1.20 (m, 7H), 1.15 (s, 4H), 1.09 (s, 2H),
1
H NMR (DMSO-d6, 600 MHz) δ: 8.89 (s, 1H), 7.77 (d, J = 8.2 Hz, 0.82−0.94 (m, 4H), and 0.47 (d, J = 4.6 Hz, 3H); HRMS (ESI,
1H), 7.71 (d, J = 7.8 Hz, 1H), 7.42−7.47 (m, 5H), 7.33−7.39 (m, 2H), positive) m/z calcd for C55H67ClN9O5S2 [M + H]+, 1032.4390; found,
5.02 (q, J = 6.75.5 Hz, 1H), 4.71 (t, J = 9.2 Hz, 1H), 4.50 (s, 1H), 4.07 1032.4368.
(s, 1H), 3.84 (d, J = 11.4 Hz, 1H), 3.68 (dd, J = 11.4, 3.6 Hz, 1H), 2.49 N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
(s, 4H), 2.27−2.34 (m, 1H), 1.95−2.00 (m, 1H), 1.53 (d, J = 7.0 Hz, 2-methylpropyl)-4-methyl-N-(3-(9-(4-((4-(3-(pyridin-3-
3H), and 1.13−1.18 (m, 11H). HRMS (ESI, negative) m/z calcd for ylmethyl)thioureido)phenyl)sulfonyl)piperazin-1-yl)-
C29H43N5O4S2 [M − H]+, 588.2684; found, 588.2673. nonanamido)propyl)benzamide (A3). White solid, yield 43%. 1H
tert-Butyl 11-(4-((4-(3-(pyridin-3-ylmethyl)thioureido)- NMR (DMSO-d6, 600 MHz) δ: 10.17 (s, 1H), 8.38−8.77 (m, 3H),
phenyl)sulfonyl)piperazin-1-yl)undecanoate (3e). White solid, 8.23 (s, 1H), 7.72−7.95 (m, 4H), 7.66 (s, 3H), 7.06−7.52 (m, 12H),
yield 65%. 1H NMR (DMSO-d6, 600 MHz) δ: 10.13 (s, 1H), 8.62 (s, 5.88 (d, J = 12.0 Hz, 1H), 5.55 (s, 1H), 5.06 (d, J = 14.0 Hz, 1H), 4.80
1H), 8.57 (d, J = 1.7 Hz, 1H), 8.47 (dd, J = 4.8, 1.6 Hz, 1H), 7.79 (d, J = (s, 2H), 3.24 (s, 4H), 2.58−3.05 (m, 6H), 2.07−2.47 (m, 10H), 1.75 (s,
8.8 Hz, 2H), 7.74−7.78 (m, 1H), 7.64 (d, J = 8.8 Hz, 2H), 7.36−7.39 2H), 1.01−1.50 (m, 15H), 0.90 (s, 5H), and 0.48 (s, 3H); 13C NMR
(m, 1H), 4.79 (d, J = 5.4 Hz, 2H), 3.56 (s, 3H), 2.85 (s, 4H), 2.39 (s, (DMSO-d6, 150 MHz) δ: 181.23, 172.45, 172.02, 161.56, 155.72,
4H), 2.26 (t, J = 7.4 Hz, 2H), 2.22 (t, J = 7.2 Hz, 2H), 1.44−1.52 (m, 149.42, 148.67, 147.64, 144.46, 139.92, 139.09, 137.16, 135.72, 134.61,
2H), and 1.29−1.35 (m, 2H). 134.26, 129.32, 129.11, 128.85, 128.44, 127.87, 127.12, 126.88, 126.33,
tert-Butyl 3-(2-(4-((4-(3-(pyridin-3-ylmethyl)thioureido)- 123.86, 121.93, 119.55, 59.45, 57.72, 52.12, 46.38, 45.64, 45.19, 42.90,
phenyl)sulfonyl)piperazin-1-yl)ethoxy)propanoate (3f). White 40.93, 36.17, 35.72, 30.69, 29.24, 29.15, 29.07, 28.81, 27.22, 26.59,
solid, yield 70%. 1H NMR (DMSO-d6, 600 MHz) δ: 10.12 (s, 1H), 8.62 25.53, 21.36, 19.95, and 18.63; HRMS (ESI, positive) m/z calcd for
(t, J = 5.7 Hz, 1H), 8.55 (d, J = 1.2 Hz, 1H), 8.46 (d, J = 3.5 Hz, 1H), C56H67ClN9O5S2 [M − H]−, 1044.4401; found, 1045.4424.
7.56−7.82 (m, 5H), 7.36 (dd, J = 7.9, 5.0 Hz, 1H), 4.77 (d, J = 5.7 Hz, N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2H), 3.48 (t, J = 6.4 Hz, 2H), 3.39 (t, J = 5.3 Hz, 2H), 2.81 (s, 4H), 2-methylpropyl)-4-methyl-N-(3-(10-(4-((4-(3-(pyridin-3-
2.38−2.47 (m, 6H), 2.34 (t, J = 5.9 Hz, 2H), and 1.33 (s, 9H). HRMS ylmethyl)thioureido)phenyl)sulfonyl)piperazin-1-yl)-
(ESI, positive) m/z calcd for C26H37N5O5S2 [M + H]+, 564.2309; decanamido)propyl)benzamide (A4). White solid, yield 42%. 1H
found, 564.2329. NMR (DMSO-d6, 600 MHz) δ: 10.15 (s, 1H), 8.61−8.71 (m, 1H),
tert-Butyl 3-(2-(2-(2-(4-((4-(3-(pyridin-3-ylmethyl)- 8.56 (s, 1H), 8.46 (dd, J = 4.7 Hz, 1H), 8.22 (d, J = 8.5 Hz, 1H), 7.71−
thioureido)phenyl)sulfonyl)piperazin-1-yl)ethoxy)ethoxy)- 7.84 (m, 3H), 7.56−7.70 (m, 3H), 7.11−7.45 (m, 11H), 5.87 (d, J =
ethoxy)propanoate (3g). White solid, yield 65%. 1H NMR (DMSO- 16.1 Hz, 1H), 5.53 (d, J = 10.0 Hz, 1H), 5.05 (d, J = 16.7 Hz, 1H), 4.79
d6, 600 MHz) δ: 10.19 (s, 1H), 8.69 (s, 1H), 8.57 (s, 1H), 8.47 (d, J = (s, 2H), 3.10−3.27 (m, 4H), 2.84 (s, 3H), 2.54 (s, 2H), 2.34−2.43 (m,
4.1 Hz, 1H), 7.72−7.89 (m, 3H), 7.66 (d, J = 8.8 Hz, 2H), 7.37 (dd, J = 4H), 2.32 (s, 3H), 2.14−2.24 (m, 2H), 1.95−2.09 (m, 1H), 1.66−1.83
7.6, 4.9 Hz, 1H), 4.79 (d, J = 5.3 Hz, 2H), 3.56 (t, J = 6.1 Hz, 2H), 3.42− (m, 2H), 1.00−1.40 (m, 14H), 0.73−0.97 (m, 5H), and 0.46 (d, J = 5.5
3.50 (m, 10H), 2.73−3.01 (m, 4H), 2.55−2.72 (m, 4H), 2.40 (t, J = 6.4 Hz, 3H); 13C NMR (DMSO-d6, 150 MHz) δ: 181.23, 172.46, 172.07,
Hz, 2H), 1.38 (s, 9H), and 1.22−1.26 (m, 2H). 161.56, 155.71, 149.38, 148.64, 147.63, 144.43, 139.92, 139.10, 137.13,
N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)- 135.73, 134.61, 134.23, 130.08, 129.51, 129.32, 129.11, 128.83, 128.44,
2-methylpropyl)-4-methyl-N-(3-(7-(4-((4-(3-(pyridin-3- 127.87, 127.11, 126.85, 126.32, 123.87, 121.97, 119.53, 59.45, 57.72,
ylmethyl)thioureido)phenyl)sulfonyl)piperazin-1-yl)- 52.11, 46.36, 45.63, 45.17, 42.89, 40.88, 36.17, 35.72, 31.71, 30.67,
heptanamido)propyl)benzamide (A1). Intermediate 3a (519 mg, 29.45, 29.29, 29.14, 29.05, 28.80, 27.21, 27.04, 26.56, 25.53, 22.51,
1.0 mmol), ispinesib (516 mg, 1.0 mmol), HATU (570 mg, 1.5 mmol), 21.34, 19.945, 18.61, and 14.37; HRMS (ESI, positive) m/z calcd for
and DIPEA (194 mg, 1.5 mmol) were dissolved in dry dimethylforma- C57H69ClN9O5S2 [M − H]+, 1058.4557; found, 1058.4539.
mide (DMF) (10 mL), and then, the mixture was stirred at room N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
temperature for 1 h. After that, the mixture was poured into iced water 2-methylpropyl)-4-methyl-N-(3-(11-(4-((4-(3-(pyridin-3-
(30 mL). A large amount of white solid was precipitated and then ylmethyl)thioureido)phenyl)sulfonyl)piperazin-1-yl)-
filtrated under vacuum. The precipitant was dried at 50 °C to constant undecanamido)propyl)benzamide (A5). White solid, yield 39%.
weight and then purified via column chromatography to obtain the 1
H NMR (DMSO-d6, 600 MHz) δ: 10.21 (s, 1H), 8.69 (s, 1H), 8.58 (d,
target compound A1 (360 mg, yield 36%). 1H NMR (DMSO-d6, 600 J = 1.8 Hz, 1H), 8.49 (dd, J = 4.6, 1.4 Hz, 1H), 8.23 (d, J = 8.6 Hz, 1H),
MHz) δ: 10.14 (s, 1H), 8.63 (s, 1H), 8.57 (d, J = 1.7 Hz, 1H), 8.47 (dd, 7.82 (d, J = 8.7 Hz, 2H), 7.75−7.79 (m, 2H), 7.63−7.67 (m, 3H),

7625 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

7.20−7.40 (m, 11H), 5.88 (d, J = 16.2 Hz, 1H), 5.54 (d, J = 10.5 Hz, NAMPT Enzymatic Inhibition Assay. All of the enzymatic
1H), 5.06 (d, J = 16.2 Hz, 1H), 4.80 (d, J = 5.6 Hz, 2H), 3.24 (t, J = 8.1 reactions were conducted at 30 °C for 90 min in a 50 μL mixture
Hz, 2H), 2.86 (s, 4H), 2.67−2.76 (m, 1H), 2.39 (s, 4H), 2.33 (s, 3H), containing 50 mM Tris HCl (pH 8.0), 12.5 mM MgCl2, 2 mM ATP, 1
2.20−2.24 (m, 2H), 1.96−2.01 (m, 1H), 1.72−1.81 (m, 3H), 1.46− mM DTT, 0.02% bovine serum albumin (BSA), 0.4 mM phosphor-
1.55 (m, 2H), 1.27−1.34 (m, 6H), 1.23−1.26 (m, 6H), 1.09−1.13 (m, ibosyl pyrophosphate, 20 μM nicotinamide, 30 μg/mL of alcohol
2H), 0.90 (d, J = 6.8 Hz, 3H), and 0.47 (d, J = 6.2 Hz, 3H); 13C NMR dehydrogenase, 1.5% alcohol, 0.01% Tween 20, 10 μg/mL NAMPT,
(DMSO-d6, 150 MHz) δ: 181.23, 172.45, 172.03, 161.56, 155.71, and the test compounds. Fluorescence intensity was measured at an
149.39, 148.65, 147.63, 144.48, 139.91, 139.09, 137.14, 135.71, 134.59, excitation of 360 nm and an emission of 460 nm, and then, the
134.25, 130.09, 129.31, 129.09, 128.83, 128.43, 127.87, 127.12, 126.86, fluorescent intensity data were analyzed using Graphpad Prism.
126.32, 123.86, 121.88, 119.54, 59.44, 57.67, 52.07, 46.34, 45.63, 45.14, Fluorescence Polarization Measurements. KD values were
42.89, 36.17, 35.72, 31.71, 30.84, 30.68, 29.52, 29.36, 29.30, 29.15, measured using the fluorescence anisotropy assay with fluorescein-
29.07, 28.80, 27.20, 27.00, 26.56, 25.54, 22.52, 21.34, 19.93, 18.61, and labeled ispinesib as the ligand in black flat-bottom half-area 96-well
14.37; HRMS (ESI, positive) m/z calcd for C58H73ClN9O5S2 [M + H]+, plates (Corning #3650). The fluorescein-labeled ispinesib was diluted
1074.4859; found, 1074.4846. to 50 nM with N-(2-hydroxyethyl)piperazine-N’-ethanesulfonic acid
N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)- (HEPES) buffer. A twofold serial dilution of LC3 protein in HEPES
2-methylpropyl)-4-methyl-N-(3-(3-(2-(4-((4-(3-(pyridin-3- buffer (100 μL) was added to a 200 μL of solution of fluorescent probe
ylmethyl)thioureido)phenyl)sulfonyl)piperazin-1-yl)ethoxy)- (final concentration at 25 nM). The fluorescence anisotropy was
propanamido)propyl)benzamide (A6). White solid, yield 40%. 1H measured on BioTek Synergy H2 with the 485 nm excitation and 535
NMR (DMSO-d6, 600 MHz) δ: 10.15 (s, 1H), 8.60−8.72 (m, 1H), nm emission filters after 30 min incubation at room temperature. The
8.57 (d, J = 1.6 Hz, 1H), 8.47 (dd, J = 3.4, 1.4 Hz, 1H), 8.22 (d, J = 8.6 KD values were determined by fitting the displacement curves to the
Hz, 1H), 7.74−7.83 (m, 4H), 7.59−7.66 (m, 3H), 7.19−7.40 (m, following equation using Mathamatica 9 (Wolfram Research Inc.)
12H), 5.87 (d, J = 16.3 Hz, 1H), 5.52 (d, J = 10.3 Hz, 1H), 5.04 (d, J =
16.3 Hz, 1H), 4.79 (d, J = 5.3 Hz, 2H), 3.19−3.27 (m, 2H), 2.80 (m, Q × F × AB + (1 − F ) × AF
4H), 2.35−2.47 (m, 8H), 2.31 (s, 4H), 1.93−2.01 (m, 2H), 1.25−1.33 A=
1 − (1 − Q ) × F
(m, 2H), 0.88 (d, J = 6.5 Hz, 3H), 0.75−0.87 (m, 2H), and 0.46 (d, J =
6.2 Hz, 3H); HRMS (ESI, positive) m/z calcd for C52H59ClN9O6S2 [M
− H]−, 1004.3724; found, 1004.3693. F=
N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-methylpropyl)-4-methyl-N-(12-oxo-1-(4-((4-(3-(pyridin-3- (KD + LST + RT − (KD + LST + RT)2 − 4 × LST × RT )
ylmethyl)thioureido)phenyl)sulfonyl)piperazin-1-yl)-3,6,9-tri- 2 × LST
oxa-13-azahexadecan-16-yl)benzamide (A7). White solid, yield
51%. 1H NMR (600 MHz, DMSO-d6) δ: 10.16 (s, 1H), 8.66 (s, 1H), In the equations, A is the measured fluorescence polarization value,
8.58 (s, 1H), 8.48 (d, J = 4.5 Hz, 1H), 8.23 (d, J = 8.7 Hz, 1H), 7.74− Q is the ratio of the fluorescence intensity of the probe in the bound and
7.86 (m, 4H), 7.60−7.71 (m, 3H), 7.18−7.43 (m, 11H), 5.89 (d, J = free status, AB and F represent the anisotropies of bound and free probe,
16.1 Hz, 1H), 5.54 (d, J = 10.0 Hz, 1H), 5.06 (d, J = 15.6 Hz, 1H), 4.79 respectively, KD is the equilibrium dissociation constant of the
(s, 2H), 3.37−3.48 (m, 11H), 3.21−3.28 (m, 2H), 2.85 (s, 4H), 2.70− fluorescent probe, LST is the concentration of the probe, and RT is
2.77 (m, 1H), 2.41−2.49 (m, 6H), 2.34 (s, 3H), 1.98−2.06 (m, 2H), the receptor protein concentration.
1.24−1.31 (m, 3H), 0.90 (d, J = 6.5 Hz, 3H), 0.81−0.88 (m, 2H), and Western Blot Analysis. A2780 cells were seeded in six-well plates
0.48 (d, J = 6.3 Hz, 3H); 13C NMR (150 MHz, DMSO-d6) δ: 185.98, at a density of 1 × 106/well. After incubation at 37 °C with 5% CO2
177.20, 174.66, 166.34, 160.45, 154.18, 153.43, 152.41, 149.20, 144.72, overnight, compounds at different concentrations were added. After 48
143.91, 141.92, 140.48, 139.37, 138.99, 134.12, 133.88, 133.63, 133.24, h, the medium was discarded. Cells were washed with phosphate-
132.64, 131.94, 131.64, 131.11, 128.63, 126.70, 124.31, 74.87, 74.82, buffered saline (PBS) three times and then lysed on ice with RIPA cell
74.69, 73.46, 74.84, 64.20, 61.76, 57.13, 51.13, 49.96, 47.53, 41.13, lysis buffer for 30 min. Cell lysates were collected and centrifuged at 1.2
41.01, 33.57, 26.11, 24.70, and 23.38; HRMS (ESI, positive) m/z calcd × 105 g at 4 °C for 15 min. The supernatants were quantified via the
for C56H69ClN9O8S2 [M + H]+, 1094.4394; found, 1094.4362. bicinconinic acid assay kit and then degenerated at 100 °C. Then, they
(R)-N-(1-(3-Benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2- were analyzed at an equal amount of protein via sodium dodecyl
yl)-2-methylpropyl)-N-(3-(3-(3′,6′-dihydroxy-3-oxo-3H-spiro- sulfate−polyacrylamide gel electrophoresis (SDS−PAGE). Subse-
[isobenzofuran-1,9′-xanthen]-5-yl)thioureido)propyl)-4- quently, they were transferred to the polyvinylidene fluoride membrane
methylbenzamide (P1). The intermediate 4 (0.1 g, 0.2 mmol) and 5 and then blocked with 5% BSA in Tris-buffered saline and Tween 20 at
(75 mg, 0.2 mmol) were dissolved in anhydrous DMF (10 mL). Then, room temperature for 2 h. Then, they were incubated with primary
tetraethylammonium (40 mg, 0.4 mmol) was added and the mixture antibodies at 4 °C overnight separately. After being washed with PBS
was reacted at room temperature. After 2 h, the reaction solution was three times every 10 min, they were incubated with secondary
poured into water (100 mL) and extracted with EA (40 mL × 3). The antibodies at room temperature for 1 h. After being washed with PBS
organic phases were collected and concentrated under reduced pressure another three times every 10 min, the membranes were read by Odyssey
to give the crude product. Purification was applied with C18 column Infrared Imaging.
[H2O (0.1% TFA): MeOH = 20: 80] to afford the target compound P1 Cell Proliferation Assay. Cells were seeded at a density of 5 × 103
as a yellow solid (74 mg, yield 41%). 1H NMR (600 MHz, DMSO-d6) δ cells per well. After being incubated overnight, they were treated with
10.11 (s, 2H), 9.68 (s, 1H), 8.21 (d, 1H, J = 8.5 Hz), 8.05 (s, 1H), 7.78 DMSO, MS2, or A3 as indicated for 72 h. The culture medium was
(s, 1H), 7.70 (s, 1H), 7.57−7.60 (m, 2H), 7.35−7.37 (m, 2H), 7.20− discarded, and 10% CCK8 containing culture medium was added. After
7.31 (m, 7H), 7.13 (d, 1H, J = 8.0 Hz), 6.67 (d, 2H, J = 2.4 Hz), 6.55− being incubated at 37 °C, the OD values at a wavelength of 450 nm were
6.59 (m, 4H), 5.89 (d, 1H, J = 16.0 Hz), 5.57 (d, 1H, J = 10.4 Hz), 5.09 read. The cell proliferation rates versus concentration of compounds
(d, 1H, J = 16.5 Hz), 3.36−3.37 (m, 2H), 3.00 (s, 2H), 2.76−2.80 (m, was analyzed using Graphpad Prism.
1H), 2.24 (s, 3H), 1.51−1.53 (m, 1H), 1.04−1.11 (m, 1H), 0.92 (d, Cell Thermal Shift Assay. A2780 cells were seeded at a density of 1
3H, J = 6.6 Hz), and 0.49 (d, 3H, J = 6.6 Hz). 13C NMR (150 MHz, × 107/well and then incubated at 37 °C with 5% CO2 overnight. Then,
DMSO-d6) δ 180.63, 172.45, 168.95, 161.61, 159.95, 155.67, 152.35, compounds at the indicated concentration were added. After 4 h,
147.70, 141.63, 139.94, 139.19, 137.18, 134.19, 129.39, 129.11, 128.46, culture medium was discarded and the cells were collected and lysed
127.87, 127.23, 126.88, 126.40, 124.39, 119.64, 112.99, 110.22, 102.72, under treatment in liquid nitrogen for 5 min (three times). After being
59.546, 45.676, 42.602, 41.417, 29.725, 28.884, 21.321, 20.001, and centrifuged at 1.2 × 105 g at 4 °C for 15 min, the supernatants were
18.64. HRMS (ESI): m/z calcd for C51H44ClN5O7S, 906.4421; found, divided into four parts and heated at 48 , 52, 55, and 58 °C, respectively.
906.2742 [M + H]+. Subsequently, they were centrifuged at 1.2 × 105 g at 4 °C for 15 min.

7626 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

The supernatants were degenerated at 100 °C and then analyzed via Pharmacy, 920th Hospital of Joint Logistics Support Force,
SDS-PAGE to detect the expression level of NAMPT protein. Kunming 650032, China
Knockdown of Atg7 in A2780 Cells. Cells were seeded into 24- Junfei Cheng − School of Pharmacy, Second Military Medical
well plates at a density of 1.5×105/well. After being incubated at 37 °C University, Shanghai 200433, China
with 5% CO2 overnight, 250 μL of medium containing shRNA and 2
μg/mL polybrene was replaced for culture medium. After 4 h, another
Long Chen − School of Pharmacy, Second Military Medical
250 μL of culture medium was added. After 24 h, the medium was University, Shanghai 200433, China
discarded and replaced by fresh culture medium. Cells were collected Rui Liu − School of Life Sciences, Fudan University, Shanghai
after another 48 h for analysis of the expression level of Atg7. 200433, China
Molecular Docking. The crystal structure of NAMPT in complex Yu Ding − School of Life Sciences, Fudan University, Shanghai
with its inhibitor was obtained from the Protein Data Bank (PDB: 200433, China
2GVJ37) and prepared for docking using the protein preparation tool in Shanchao Wu − School of Pharmacy, Second Military Medical
Discovery Studio 3.0.38 During this process, the ligands and waters were University, Shanghai 200433, China
removed and hydrogens were added to the structure. Staged Junhui Ma − School of Pharmacy, Second Military Medical
minimization was performed with default setting. The docking studies
were carried out using GOLD 5.0.39 The binding site was defined as
University, Shanghai 200433, China
whole residues within a 15 Å radius subset encompassing the ligand. Complete contact information is available at:
Conformations were generated by genetic algorithm and scored using https://pubs.acs.org/10.1021/acs.jmedchem.1c02001
GoldScore as fitness function. The best conformation was chosen to
analyze the ligand−protein interaction. The image representing the Author Contributions
best pose was prepared using PyMOL. ∥
Isothermal Titration Calorimetry. The thermodynamic param- G.D., Y.W. and J.C. made equal contributions to this work.
eters of the binding of ispinesib to recombinant LC3B were determined Notes
by ITC using VP-ITC or ITC200 calorimetry (MicroCal VP-ITC, The authors declare no competing financial interest.


Malvern). Since the solubility of ispinesib in the ITC buffer is relatively
low, we performed the titration by injecting a 12 μl of aliquot of LC3B
protein sample (1500 μM) into the cell containing 100 μM ispinesib ACKNOWLEDGMENTS
(the ITC buffer includes 20 mM HEPES-HCl, pH 7.3, 1% DMSO, and This work was supported by the National Natural Science
100 mM NaCl) at a time interval of 120 s to ensure that the titration Foundation of China (grant 82030105 to C.S.), the National
peak returned to baseline. Altogether, 13 aliquots were titrated in each Key Research and Development Program of China (grant
individual experiment. The dissociation constant KD, stoichiometry of
binding (n), and the binding enthalpy ΔH were calculated using 2020YFA0509100 to C.S.), and Shanghai Rising-Star Program
MicroCal Origin 7.0 software with a one-site binding model. (20QA1411700 to G.D.).

■sı
ASSOCIATED CONTENT
* Supporting Information
■ ABBREVIATIONS
ALP, autophagy-lysosomal pathway; ATTEC, autophagosome-
The Supporting Information is available free of charge at tethering compound; AUTACs, autophagy-targeting chimeras;
https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c02001. CMA, chaperone-mediated autophagy; DIPEA, N,N-diisopro-
Chemical structures of ATTECs; chemical structures of pylethylamine; LDs, lipid droplets; Mhtt, mutant HTT protein;
NAMPT inhibitors; NAMPT levels in A2780 cells treated NAD, nicotinamide adenine dinucleotide; NAMPT, nicotina-
with compound A3 at 1 μM concentration for the mide phosphoribosyltransferase; PEG, poly(ethylene glycol);
indicated time points; affinity measurement by SPR with PROTACs, proteolysis targeting chimeras
LC3-B and A3; KSP levels in A2780 cells treated with
compound A3; cell growth inhibition of compound A3;
stability of A3; CETSA of NAMPT with compound A3;
■ REFERENCES
(1) Paiva, S.-L.; Crews, C. M. Targeted protein degradation: elements
validation of the interaction between LC3-B and ispinesib of PROTAC design. Curr. Opin. Chem. Biol. 2019, 50, 111−119.
by ITC at 25 °C; A2780 cells pretreated with ispinesib, (2) Cully, M. PROTAC moves in on lymphoma. Nat. Rev. Drug
followed by treatment with A3 for an additional 48 h; Discovery 2019, 18, 584.
synthetic route of probe P1; and 1H- and 13C NMR (3) Burslem, G. M.; Crews, C. M. Proteolysis-Targeting Chimeras as
spectra of target compounds (PDF) Therapeutics and Tools for Biological Discovery. Cell 2020, 181, 102−
SMILES molecular formula strings (CSV) 114.
(4) Toure, M.; Crews, C. M. Small-Molecule PROTACS: New
Molecular docking of compound MS2 with NAMPT Approaches to Protein Degradation. Angew. Chem., Int. Ed. 2016, 55,
(PDB)


1966−1973.
(5) Cheng, J.; Li, Y.; Wang, X.; Dong, G.; Sheng, C. Discovery of
AUTHOR INFORMATION Novel PDEdelta Degraders for the Treatment of KRAS Mutant
Corresponding Authors Colorectal Cancer. J. Med. Chem. 2020, 63, 7892−7905.
(6) Schapira, M.; Calabrese, M. F.; Bullock, A. N.; Crews, C. M.
Guoqiang Dong − School of Pharmacy, Second Military
Targeted protein degradation: expanding the toolbox. Nat. Rev. Drug
Medical University, Shanghai 200433, China; orcid.org/ Discovery 2019, 18, 949−963.
0000-0003-1708-7803; Email: gdong@smmu.edu.cn (7) Bond, M. J.; Crews, C. M. Proteolysis targeting chimeras
Chunquan Sheng − School of Pharmacy, Second Military (PROTACs) come of age: entering the third decade of targeted protein
Medical University, Shanghai 200433, China; orcid.org/ degradation. RSC Chem. Biol. 2021, 2, 725−742.
0000-0001-9489-804X; Email: shengcq@smmu.edu.cn (8) Sun, X.; Gao, H.; Yang, Y.; He, M.; Wu, Y.; Song, Y.; Tong, Y.; Rao,
Y. PROTACs: great opportunities for academia and industry. Signal
Authors Transduction Targeted Ther. 2019, 4, 64.
Ying Wu − School of Pharmacy, Second Military Medical (9) Mullard, A. Targeted protein degraders crowd into the clinic. Nat.
University, Shanghai 200433, China; Department of Rev. Drug Discovery 2021, 20, 247−250.

7627 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

(10) Clift, D.; McEwan, W. A.; Labzin, L. I.; Konieczny, V.; Mogessie, (29) Xu, T.-Y.; Zhang, S.-L.; Dong, G.-Q.; Liu, X.-Z.; Wang, X.; Lv, X.-
B.; James, L. C.; Schuh, M. A Method for the Acute and Rapid Q.; Qian, Q.-J.; Zhang, R.-Y.; Sheng, C.-Q.; Miao, C.-Y. Discovery and
Degradation of Endogenous Proteins. Cell 2017, 171, 1692−1706. characterization of novel small-molecule inhibitors targeting nicotina-
(11) Zhang, L.; Riley-Gillis, B.; Vijay, P.; Shen, Y. Acquired Resistance mide phosphoribosyltransferase. Sci. Rep. 2015, 5, 10043.
to BET-PROTACs (Proteolysis-Targeting Chimeras) Caused by (30) Du, Y. Fluorescence polarization assay to quantify protein-
Genomic Alterations in Core Components of E3 Ligase Complexes. protein interactions in an HTS format. Methods Mol. Biol. 2015, 1278,
Mol. Cancer Ther. 2019, 18, 1302−1311. 529−544.
(12) Balchin, D.; Hayer-Hartl, M.; Hartl, F. U. In vivo aspects of (31) Dong, G.; Chen, W.; Wang, X.; Yang, X.; Xu, T.; Wang, P.;
protein folding and quality control. Science 2016, 353, aac4354. Zhang, W.; Rao, Y.; Miao, C.; Sheng, C. Small Molecule Inhibitors
(13) Whitmarsh-Everiss, T.; Laraia, L. Small molecule probes for Simultaneously Targeting Cancer Metabolism and Epigenetics:
targeting autophagy. Nat. Chem. Biol. 2021, 17, 653−664. Discovery of Novel Nicotinamide Phosphoribosyltransferase
(14) Zhao, Y. G.; Codogno, P.; Zhang, H. Machinery, regulation and (NAMPT) and Histone Deacetylase (HDAC) Dual Inhibitors. J.
pathophysiological implications of autophagosome maturation. Nat. Med. Chem. 2017, 60, 7965−7983.
Rev. Mol. Cell Biol. 2021, 22, 733−750. (32) Vakifahmetoglu-Norberg, H.; Xia, H.-g.; Yuan, J. Pharmacologic
(15) Glick, D.; Barth, S.; Macleod, K. F. Autophagy: cellular and agents targeting autophagy. J. Clin. Invest. 2015, 125, 5−13.
molecular mechanisms. J. Pathol. 2010, 221, 3−12. (33) Verbaanderd, C.; Maes, H.; Schaaf, M. B.; Sukhatme, V. P.;
(16) Levy, J. M. M.; Towers, C. G.; Thorburn, A. Targeting autophagy Pantziarka, P.; Sukhatme, V.; Agostinis, P.; Bouche, G. Repurposing
in cancer. Nat. Rev. Cancer 2017, 17, 528−542. Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as
(17) Takahashi, D.; Moriyama, J.; Nakamura, T.; Miki, E.; Takahashi, anti-cancer agents. Ecancermedicalscience 2017, 11, 781.
E.; Sato, A.; Akaike, T.; Itto-Nakama, K.; Arimoto, H. AUTACs: cargo- (34) Wu, Y.; Wang, X.; Guo, H.; Zhang, B.; Zhang, X.-B.; Shi, Z.-J.; Yu,
specific degraders using selective autophagy. Mol. Cell 2019, 76, 797− L. Synthesis and screening of 3-MA derivatives for autophagy
inhibitors. Autophagy 2013, 9, 595−603.
810.
(35) Gharbi, S. I.; Zvelebil, M. J.; Shuttleworth, S. J.; Hancox, T.;
(18) Li, Z.; Wang, C.; Wang, Z.; Zhu, C.; Li, J.; Sha, T.; Ma, L.; Gao,
Saghir, N.; Timms, J. F.; Waterfield, M. D. Exploring the specificity of
C.; Yang, Y.; Sun, Y.; Wang, J.; Sun, X.; Lu, C.; Difiglia, M.; Mei, Y.;
the PI3K family inhibitor LY294002. Biochem. J. 2007, 404, 15−21.
Ding, C.; Luo, S.; Dang, Y.; Ding, Y.; Fei, Y.; Lu, B. Allele-selective
(36) Wu, Y.; Pu, C.; Fu, Y.; Dong, G.; Huang, M.; Sheng, C. NAMPT-
lowering of mutant HTT protein by HTT-LC3 linker compounds. targeting PROTAC promotes antitumor immunity via suppressing
Nature 2019, 575, 203−209. myeloid-derived suppressor cell expansion. Acta Pharm. Sin. B 2021
(19) Kocak, M.; Ezazi Erdi, S.; Jorba, G.; Maestro, I.; Farres, J.; Kirkin, doi:10.1016/j.apsb.2021.12.017, DOI: 10.1016/j.apsb.2021.12.017.
V.; Martinez, A.; Pless, O. Targeting autophagy in disease: established (37) Khan, J. A.; Tao, X.; Tong, L. Molecular basis for the inhibition of
and new strategies. Autophagy 2021, 18, 473. human NMPRTase, a novel target for anticancer agents. Nat. Struct.
(20) Fu, Y.; Chen, N.; Wang, Z.; Luo, S.; Ding, Y.; Lu, B. Degradation Mol. Biol. 2006, 13, 582−588.
of lipid droplets by chimeric autophagy-tethering compounds. Cell Res. (38) Sampath, D.; Zabka, T. S.; Misner, D. L.; O’Brien, T.; Dragovich,
2021, 31, 965−979. P. S. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT)
(21) Carol, H.; Lock, R.; Houghton, P. J.; Morton, C. L.; Kolb, E. A.; as a therapeutic strategy in cancer. Pharmacol. Ther. 2015, 151, 16−31.
Gorlick, R.; Reynolds, C. P.; Maris, J. M.; Keir, S. T.; Billups, C. A.; (39) Jones, G.; Willett, P.; Glen, R. C.; Leach, A. R.; Taylor, R.
Smith, M. A. Initial testing (stage 1) of the kinesin spindle protein Development and validation of a genetic algorithm for flexible docking.
inhibitor ispinesib by the pediatric preclinical testing program. Pediatr. J. Mol. Biol. 1997, 267, 727−748.
Blood Cancer 2009, 53, 1255−1263.
(22) Purcell, J. W.; Davis, J.; Reddy, M.; Martin, S.; Samayoa, K.; Vo,
H.; Thomsen, K.; Bean, P.; Kuo, W. L.; Ziyad, S.; Billig, J.; Feiler, H. S.;
Gray, J. W.; Wood, K. W.; Cases, S. Activity of the kinesin spindle
protein inhibitor ispinesib (SB-715992) in models of breast cancer.
Clin. Cancer Res. 2010, 16, 566−576.
(23) Grolla, A. A.; Travelli, C.; Genazzani, A. A.; Sethi, J. K.
Extracellular nicotinamide phosphoribosyltransferase, a new cancer
metabokine. Br. J. Pharmacol. 2016, 173, 2182−2194.
(24) Chen, H.; Wang, S.; Zhang, H.; Nice, E. C.; Huang, C.
Nicotinamide phosphoribosyltransferase (Nampt) in carcinogenesis:
new clinical opportunities. Expert Rev. Anticancer Ther. 2016, 16, 827−
838.
(25) Garten, A.; Schuster, S.; Penke, M.; Gorski, T.; de Giorgis, T.;
Kiess, W. Physiological and pathophysiological roles of NAMPT and
NAD metabolism. Nat. Rev. Endocrinol. 2015, 11, 535−546.
(26) Guo, Q.; Han, N.; Shi, L.; Yang, L.; Zhang, X.; Zhou, Y.; Yu, S.;
Zhang, M. NAMPT: A potential prognostic and therapeutic biomarker
in patients with glioblastoma. Oncol. Rep. 2019, 42, 963−972.
(27) Galli, U.; Travelli, C.; Massarotti, A.; Fakhfouri, G.; Rahimian, R.;
Tron, G. C.; Genazzani, A. A. Medicinal chemistry of nicotinamide
phosphoribosyltransferase (NAMPT) inhibitors. J. Med. Chem. 2013,
56, 6279−6296.
(28) Korotchkina, L.; Kazyulkin, D.; Komarov, P. G.; Polinsky, A.;
Andrianova, E. L.; Joshi, S.; Gupta, M.; Vujcic, S.; Kononov, E.;
Toshkov, I.; Tian, Y.; Krasnov, P.; Chernov, M. V.; Veith, J.; Antoch, M.
P.; Middlemiss, S.; Somers, K.; Lock, R. B.; Norris, M. D.; Henderson,
M. J.; Haber, M.; Chernova, O. B.; Gudkov, A. V. OT-82, a novel
anticancer drug candidate that targets the strong dependence of
hematological malignancies on NAD biosynthesis. Leukemia 2020, 34,
1828−1839.

7628 https://doi.org/10.1021/acs.jmedchem.1c02001
J. Med. Chem. 2022, 65, 7619−7628

You might also like