You are on page 1of 17

pubs.acs.

org/jmc Article

Discovery and SAR Study of Boronic Acid-Based Selective PDE3B


Inhibitors from a Novel DNA-Encoded Library
Ann M. Rowley,* Gang Yao,* Logan Andrews, Aaron Bedermann, Ross Biddulph, Ryan Bingham,
Jennifer J. Brady, Rachel Buxton, Ted Cecconie, Rona Cooper, Adam Csakai, Enoch N. Gao,
Melissa C. Grenier-Davies, Meghan Lawler, Yiqian Lian, Justyna Macina, Colin Macphee,
Lisa Marcaurelle, John Martin, Patricia McCormick, Rekha Pindoria, Martin Rauch, Warren Rocque,
Yingnian Shen, Lisa M. Shewchuk, Michael Squire, Will Stebbeds, Westley Tear, Xin Wang, Paris Ward,
and Shouhua Xiao
Downloaded via VENATORX PHARMACEUTICALS INC on January 29, 2024 at 20:35:03 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Cite This: https://doi.org/10.1021/acs.jmedchem.3c01562 Read Online

ACCESS Metrics & More Article Recommendations *


sı Supporting Information

ABSTRACT: Human genetic evidence shows that PDE3B is


associated with metabolic and dyslipidemia phenotypes. A number
of PDE3 family selective inhibitors have been approved by the
FDA for various indications; however, given the undesirable
proarrhythmic effects in the heart, selectivity for PDE3B inhibition
over closely related family members (such as PDE3A; 48%
identity) is a critical consideration for development of PDE3B
therapeutics. Selectivity for PDE3B over PDE3A may be achieved
in a variety of ways, including properties intrinsic to the compound
or tissue-selective targeting. The high (>95%) active site homology
between PDE3A and B represents a massive obstacle for obtaining
selectivity at the active site; however, utilization of libraries with
high molecular diversity in high throughput screens may uncover
selective chemical matter. Herein, we employed a DNA-encoded library screen to identify PDE3B-selective inhibitors and identified
potent and selective boronic acid compounds bound at the active site.

■ INTRODUCTION
Cyclic nucleotide phosphodiesterases (PDEs) are a super-
while PDE3B is prevalently expressed in peripheral blood T-
cells, hepatocytes, and adipose tissue.1−4
Genome wide association studies from 23andMe and others
family of enzymes that catalyze the hydrolysis of the
have identified associations of PDE3B with lipid phenotypes. A
phosphodiester bonds of specific second messenger substrates rare stop-gain variant in the catalytic site of PDE3B (p.R783X,
such as 3′,5′-cyclic adenosine monophosphate (cAMP) and rs150090666) is associated with reduced risk of high
3′,5′-cyclic guanosine monophosphate (cGMP). They play a triglyceride, low HDL, high LDL, high cholesterol, as well
critical role in the indirect regulation of many intracellular obesity5 (Supporting Information, Figure S1). Literature
metabolism pathways. 11 broad families of PDE isoenzymes studies suggest that selective PDE3B inhibition would
(PDE1-11) have been reported thus far in the literature with effectively stimulate lipolysis and metabolism while avoiding
each PDE family consisting of distinct genes that in turn cardiovascular risks associated with PDE3A inhibition.6
generate more than 100 PDE isoforms. PDEs generally exist in Homology between PDE3A and PDE3B is high (identities:
the dimeric form, and each monomer is composed of three 48% positive substitutions: 67%) and increases significantly
distinct domains: a catalytic, an N-terminal, and a C-terminal (∼95% identity) within 15 Å in the active site pocket,
domain. The catalytic domain consists of approximately 350 presenting strong challenges in the development of PDE3
conserved amino acids and can selectively hydrolyze cyclic
nucleotides in different ways. Phosphodiesterase 3 (PDE3) Received: October 5, 2023
family members are dual specificity PDEs that hydrolyze both Revised: January 11, 2024
cAMP and cGMP with Km values in the submillimolar range Accepted: January 19, 2024
(Km cAMP = 0.2 mM, Km cGMP = 0.1 mM). There are two
PDE3 isoforms: PDE3A and PDE3B. PDE3A is mostly found
in cardiac tissue, platelets, and vascular smooth muscle cells,

© XXXX American Chemical Society https://doi.org/10.1021/acs.jmedchem.3c01562


A J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Figure 1. Known PDE3 inhibitors.7−10

Figure 2. Structure of the aminobenzoic acid-based DEL resulting in PDE3B-selective chemical series.

Figure 3. (a) Three-dimensional view of the DEL selection output. (b) Copy number vs cycle 3 BBs for the boronic acid series. Green: enriched for
PDE3B only; red: enriched for both PDE3A and PDE3B.

isozyme-selective inhibitors. Very few selective PDE3B technology has become a popular platform to generate novel
inhibitors have been reported to date, and those that have small molecule hits for various therapeutic targets due to its
exhibited low potency. Some known PDE3 inhibitors are high throughput and low cost nature. DELs can be prepared
shown in Figure 1.7−10 To identify novel PDE3B-selective using a split and pool synthesis strategy which allows very large
inhibitors, we screened the GSK collection of more than one libraries (>109 DEL molecules per library) to be generated in a
trillion DNA-encoded small molecules. few steps. Each DEL molecule contains a covalently attached
The DNA-encoded library (DEL) technology is an ultra- unique DNA tag that serves as an identifier for the small
high throughput screening technology which is capable of molecule. Affinity selections of a pool of DELs against the
screening millions to trillions of DNA-encoded molecules in a immobilized target proteins, PCR amplification, and sequenc-
single tube via affinity selection. In recent years, the DEL ing of the DNA tags of the enriched DEL molecules afford
B https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Table 1. Biochemical Data for Off-DNA Compounds, Varying Three Different Vectors

a
All compounds were tested in the assays n ≥ 2. Potency values quoted are mean of n’s. bPotency measurements for RF-MS PDE3B and PDE3A
listed in Table S3. cOn a single test occasion, the compound was inactive (pIC50 < 4.0).

putative binders with rich structure−activity data (on-DNA shown in Figure 2, was capped with 1178 amine building
SAR). Due to the facileness of affinity selection, multiple blocks (BBs) (carboxylic acids, amino acids, and sulfonyl
selection conditions can be performed in parallel enabling chlorides). Half of the pool of DEL molecules were further
further triage of hits based on desired selectivities or binding oxidized with N-hydroxypiperidine in cycle 4 to give the
modes (ortho or allosteric binders etc.).11 The DEL corresponding phenol derivatives, resulting in a total of 3.8
technology represents a unique approach to finding a selective million unique DNA-encoded compounds (Figure 2).
binder for PDE3B that has not been profiled previously and The initial off-DNA boronic acid molecules from one of the
therefore we sought to employ this technique to try to find PDE3B-selective clusters were designed based on the most
molecules with high selectivity. enriched disynthon combination of cycle 2 BB 3-borono-5-

■ RESULTS AND DISCUSSION


In an effort to identify novel selective PDE3B small molecule
nitrobenzoic acid and cycle 3 BB 2-methoxy-3,5-dimethylben-
zoic acid (Figure 3a). Compounds 1 and 2 exhibited a pIC50
value of 6.7 and 6.5, respectively, when tested against full-
inhibitors, we performed a parallel selection of both human length (FL) PDE3B (M1-E1112)/PDE3A(M1-Q1141) in the
His-tagged PDE3B (511-1112) and human His-tagged PDE3A IMAP assay; they also showed ∼3-fold selectivity for PDE3B
(511-1141) against 1.9 trillion DNA-encoded small molecules over PDE3A (Table 1). Substitution of the boronic acid
from the GSK collection (106 libraries in pool). DEL moiety with a hydroxy group resulted in a significant loss of
molecules that were enriched against PDE3B were clustered activity (Table 1, compound 3, IMAP PDE3B pIC50 = 4.4),
and analyzed. Clusters with >50% of members showing indicating the critical importance of the boronic acid group for
PDE3B specific enrichment over PDE3A were prioritized for maintaining PDE3B inhibition. This result was consistent with
further evaluation. Representative compounds from these the selection results, in which neither the corresponding
clusters were designed and synthesized without their DNA phenol nor the desboronic acid compounds from the DEL
tag, to confirm potency and selectivity in immobilized metal were enriched. Further analysis of the on-DNA SAR of the
ion affinity for phosphochemical (IMAP) and/or RapidFire boronic acid series revealed that certain cycle 3 BBs might
mass spectrometry (RF-MS) activity assays.12 Several chemical confer greater PDE3B/A IMAP selectivity. When examining a
series were identified to be PDE3B inhibitors but only one plot of the enriched boronic acid DEL molecules (copy
series from a four-cycle library, defined by a boronic acid number) versus cycle 3 BBs, it was observed that certain BB3
functional group, showed PDE3B selectivity over PDE3A. As derived series had a higher percentage of PDE3B-specific
shown in Figure 2, this particular four-cycle boronic acid members than those derived from 2-methoxy-3,5-dimethyl-
containing DEL was constructed utilizing 810 amines or amino benzoic acid (Figure 3b). Therefore, we targeted a compound
acids at cycle 1. In cycle 2, two 3-nitrobenzoic acid derivatives derived from one of the more selective cycle 3 BBs: 4,7-
were installed via amide bond formation and then dimethoxyquinoline-2-carboxylic acid and synthesized com-
subsequently reduced and then in cycle 3, the aniline, as pound 4, Table 1. Compound 4 exhibited moderate PDE3B
C https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Scheme 1. Representative Synthesis of Boronic Acid Analogues Exemplified by Compound 5

Scheme 2. Representative Synthesis of Boronic Acid Analogues in Table 2

activity (IMAP pIC50 = 6.5) and much improved PDE3B/A highlighting the importance of this moiety. Less drastic
IMAP selectivity (>30-fold). truncations of 4 to small polar groups such as the primary
The on-DNA SAR of cycle 1 BBs illustrated in the Spotfire amide (7), methyl ester (9), and methyl amide (10) all showed
cube view (Figure 3a) indicated that broad variation at this restoration of some of the PDE3B activity (IMAP pIC50 = 5.7
position was tolerated. Indeed, both the (R)- and (S)-2-amino- for all three). Encouragingly, these compounds all showed no
2-cyclohexylacetic acid-derived hits (compounds 4 and 5 activity against PDE3A, improving upon the PDE3B/A
IMAP PDE3B pIC50 of 6.5 and 7.1, respectively) were active selectivity (>51, >52, and >44-fold IMAP selectivity for 7, 9,
PDE3B inhibitors with the R-isomer (4), showing greater and 10, respectively). This reduction in PDE3A activity was
PDE3B/A IMAP selectivity than the S-isomer (5) (37-fold vs confirmed by the orthogonal RF-MS assay (Table S1), with
12-fold). This finding indicated that the cycle 1 portion of the only 7 showing any measurable activity (RF-MS PDE3A pIC50
molecule may be amenable to further optimization to improve = 4.1). Interestingly, the carboxylic acid analogue 8 showed
the PDE3B/A IMAP selectivity. In addition to the PDE3B similar PDE3B activity to compounds 7, 9, and 10 but
IMAP assay, the off-DNA hits were further evaluated in a RF- displayed higher inhibition of PDE3A (IMAP pIC50 = 5.1),
MS assay using the native substrate cAMP. Both compounds 4 resulting in a significant reduction in the PDE3B/A selectivity
and 5 showed PDE3B/A RF-MS selectivity with the R-isomer (3-fold selective). Swapping a primary methyl amine to a
(4) displaying better PDE3B/A selectivity compared to the S- secondary dimethyl amine, as exemplified by analogues 10 and
isomer (5) (78-fold vs 26-fold), in agreement with the IMAP 11, resulted in a significant loss in PDE3B/A selectivity (>44
assay data. vs 23-fold, respectively). Further increasing the size of the
A representative synthesis of the boronic acid analogues amide group substituent (12, IMAP pIC50 = 6.0) modestly
described above is illustrated in Scheme 1. Starting with the improved the activity but resulted in a decrease in PDE3B/A
(S)-2-amino-2-cyclohexyl-N-methylacetamide, coupling to 3- selectivity (15-fold selective). N-methylation of the amide
(N-Boc-amino)-5-carboxyphenylboronic acid under HATU- adjacent to the isoquinoline ring (R2�Me, compound 13)
mediated conditions afforded the corresponding amide eliminated PDE3B activity. Reducing the size of the α-
intermediate. Removal of the Boc protecting group with HCl substituent and replacing the methyl amide with a primary
and reaction with the requisite carboxylic acids under TCFH- amide on compounds 4 and 5 resulted in 14 and 15. Again the
mediated coupling conditions provided the desired compound (S)-enantiomer (15) showed higher PDE3B potency than the
4 and its related analogues. (R)-enantiomer 14 (IMAP pIC50 = 6.5 vs 5.7 for 15 and 14,
A representative synthesis of the boronic acid analogues respectively), although both showed significant reduction in
described above is illustrated in Scheme 2. Starting with 1- selectivity compared to compounds 7, 9, and 10. This
methyl 3-amino-5-boronobenzoate, coupling to the requisite selectivity was regained when the primary amide in 14 was
isoquinoline carboxylic acid under TCFH-mediated coupling replaced with a methoxymethyl (17). Compound 17
conditions provided the intermediate in good yield. Following demonstrated submicromolar activity (IMAP pIC50 = 6.3)
hydrolysis of the methyl ester, subsequent HATU-mediated against PDE3B and high PDE3B/A selectivity (>190-fold
coupling was amenable to a range of amines allowing for rapid selective IMAP, 50-fold selective RF-MS). Substitution at the
SAR exploration. The IMAP and RF-MS results are shown in α-position appears critical for the selectivity, with 18 (>300-
Table 2. fold selective) showing significant PDE3B/A selectivity
Complete removal of the amide group resulted in a compared to its β-substituted matched pair 19 (14-fold
significant loss of activity (IMAP pIC50 = 5.0 vs 6.5, selective). Cyclization of the ether reduced the PDE3B/A
compounds 6 and 4, respectively) and selectivity (1.4-fold vs selectivity (20 and 21). Increasing the steric bulk of the amide
37-fold IMAP PDE3B/A selectivity for 6 and 4, respectively) substituent and adding a chiral center (R1), from an isopentane
D https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Table 2. Potency and Selectivity SAR Table Examining Variation of the Left-Hand Portion of the Molecule While Maintaining
the Quinoline Moiety

a
All compounds were tested in the assays n ≥ 2. Potency values quoted are mean of n’s. bPotency measurements for RF-MS PDE3B and PDE3A
listed in Table S4. cCompound does not contain carbonyl attachment. dOn a singled test occasion, the compound was inactive (pIC50 < 4.0). eOn a
twoe test occasion, the compound was inactive (pIC50 < 4.0). fOn a threef test occasion, the compound was inactive (pIC50 < 4.0).

Scheme 3. Representative Synthesis of Right-Hand SAR Analogues

E https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Table 3. Potency and Selectivity Assay Results for Right-Hand SAR Analogues

a
All compounds were tested in the assays n ≥ 2. Potency values quoted are mean of n’s. bPotency measurements for RF-MS PDE3B and PDE3A
listed in Table S5. cOn a singlec test occasion, the compound was inactive (pIC50 < 4.0). dOn a twod test occasion, the compound was inactive
(pIC50 < 4.0). eOn a threee test occasion, the compound was inactive (pIC50 < 4.0). fOn a single test occasion, the compound was highly active
(pIC50 > 8.77).

F https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Figure 4. (a) X-ray crystal structure of boronic compound 23 binding to PDE3B (PDB 8SYC). (b) Overlay of crystal structure of 23 in PDE3B
(green protein, green VDM surface, and orange ligand, PDB 8SYC) with an aligned crystal structure of DNMDP in PDE3A (purple protein, purple
VDW surface, and cyan ligand PDB 7KWE). Structures superposed with MOE2020.0901. (c) Docking of ICOS compound (IMAP PDE3B pIC50
7.16, IMAP PDE3A pIC50 6.05, cyan ligand) (structure shown in Figure 1)8 to the cocrystal structure of Table 2, compound 23 with PDE3B (gray
protein, gray VDM surface, and orange ligand, PDB 8SYC).

to a 1-methoxy-3-methylbutan-2-yl, improved the PDE3B/A PDE3B activity with compounds 35 and 47 being the most
selectivity, as demonstrated by the comparison of compounds potent. Compared to compound 5, both showed ∼10-fold
22 and 17 (IMAP pIC50 = 14 vs >190-fold selective, improvement in potency, albeit at the expense of PDE3B/A
respectively). Compounds 23−26 derived from reactions selectivity (0.74-fold selective for both 35 and 47 vs 12-fold
with secondary amines (namely, N-methylbenzyl amine, 3- selective for compound 5). Both the 4- and 7-methoxy groups
cyclopropoxyazetidine, pyrrolidine, and 2,3-dihydro-1H- on the quinoline ring appeared to be required for selectivity.
pyrrolo[3,4-c]pyridine, respectively) demonstrated comparable Removing the 7-methoxy group on the quinoline ring resulted
PDE3B activity (IMAP pIC50 ranging from 6.1 to 6.7) to in a significant loss of selectivity (1.6-fold selective for 32 vs 12
compound 18 but with reduced PDE3B/A selectivity. Overall, for 5). Moving the 7-methoxy group to the 6 position (33)
compound 18 demonstrated the best combination of IMAP resulted in both lower activity and complete loss of selectivity.
PDE3B activity and PDE3B/A selectivity (IMAP pIC50 = 6.5, Removing both the methoxy groups on the quinoline (37)
>300-fold selective). improved the PDE3A activity more than PDE3B activity and
With the left-hand amide portion of the molecule optimized, again resulted in a complete loss of selectivity. Moving or
we turned our attention to the quinoline moiety with the goal removing the nitrogen on the quinoline resulted in a significant
of further improving potency and PDE3B/A selectivity. A loss of PDE3B activity (38, 39, 44−46 vs 37), highlighting the
representative synthesis of these analogs is exemplified in importance of this nitrogen atom. Interestingly, other 6, 6-
Scheme 3. Monoaryl systems generally gave lower activity and nitrogen containing heterocyclic ring systems 40−43 and the
minimal selectivity (Table 3, compounds 2, 28−31) with the quinoxaline 47 were connected at a different position of the
trisubstituted phenyl derivates showing the greatest potency ring compared to the quinolines and still maintained PDE3B
(Table 3, compounds 2 and 29). In contrast to the quinoline activity even without the nitrogen at the 1 position. This
derivatives (Table 3, compounds 4 and 5), no appreciable observation implies that a different binding pose may be
selectivity difference was observed between the (R)- and the operative here. Overall, all the 6,6 fused ring systems explored
(S)-enantiomers (Table 3, compounds 29 vs 2). Analogues the 4,7-dimethoxyquinoline system and still provided the best
containing biaryl 6,6 fused ring systems showed variable PDE3B/A selectivity (up to >300 fold, IMAP). While we were
G https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

able to improve the PDE3B activity with other 6,6 fused ring PDE3A can also be displaced in the same way as in PDE3B,
systems, no improvement in PDE3B/A selectivity was giving rise to questions that may only be answered by the
observed. The SAR around both the left-hand amide and the cocrystal structure of compound 23 with PDE3A which we
quinoline region of the molecule implies that both parts of the were unable to obtain. This hypothesis could explain why both
molecule have a synergistic effect in the high PDE3B/A 4- and 7-methoxy groups on the quinoline were required for
selectivity observed for compound 18. higher PDE3B/A selectivity.
To gain structural insights, several efforts were undertaken The difference between PDE3A and PDE3B at the solvent-
to obtain cocrystallographic structures. We were successful in exposed region where the left-hand amide moiety resides is still
crystallizing compound 23 with human PDE3B (Figure 4a). more subtle. Some side chain movement (i.e., Thr855 in
The structure confirmed that the boronic acid of compound 23 PDE3A and Thr829 in PDE3B) could be responsible for the
binds at the active site of PDE3B. As shown in Figure 4a, the improved selectivity for some compounds over others (e.g., 23
boronic acid group occupied the metal subdomain and formed vs 24) but the structural impact of this part of the ligand
potential key interactions with the two magnesium metal ions molecules are less obvious from the crystal structure. It is
as well as a network of water molecules. In addition, the conceivable that in the FL enzyme, polypeptide chains in the
boronic acid is also shown to form likely hydrogen bond (HB) noncatalytic domain, which have lower homology between
interactions with the two conserved aspartic acids (i.e., Asp822 PDE3A and PDE3B, interact with the binding site in this
and Asp937). These extensive metal-chelating interactions solvent-exposed region and thus are contributing to the
serve as an anchor point for the binding of compound 23 to specificity. Overall, both the SAR and structural information
PDE3B, consistent with the observation that the boronic acid suggested that cooperative interactions between the left-hand
group is essential for activity. The dimethoxyquinoline sits in amide and the quinoline region of the boronic acids with the
the hydrophobic site between the “hydrophobic clamp” formed enzyme are required to achieve high PDE3B/A selectivity.
by Phe991 and aliphatic amino acid Ile955. In contrast to Conceivably one way to maximize this cooperation is to
cAMP or cGMP, compound 23 is not within the typical HB rigidify the ligand molecule which will make it easier to control
distance to the highly conserved Gln988. Instead, the side the relative geometry of the substituents on the two ends of the
chain of Gln988 flipped out and moved away to accommodate molecule and systematically explore the two binding sites. In
the 4-methoxy group of the quinoline moiety. The 7-methoxy addition to improving PDE3B/A selectivity, preorganization of
group extended into a small pocket between Try736, Trp951, the compound into the bound conformation could also
and Gly940, which has not been observed in other PDE3B increase the binding affinity by decreasing ligand strain in
inhibitors such as the aryl dihydropyridazinones.8 A trapped the bound conformation or an entropic effect. Further
water molecule was observed at the bottom of the pocket chemistry efforts would be required to test this hypothesis
which potentially could be replaced with a polar group to which is beyond the scope of this paper.
increase the potency of the PDE3B inhibitor. The active In order to gain insights into how an active site binder could
binding conformation has the quinoline and center phenyl ring exhibit selectivity, we computationally docked the ICOS
coplanar, which may be a result of the intramolecular HB compound (Figure 1),7 which showed ∼12× PDE3B/A
between the amide hydrogen and the nitrogen on the selectivity (PDE3B/3A pIC50 = 7.15/6.05) in our IMAP
quinoline ring system. This would explain the complete loss assay (Supporting Information) to the cocrystal structure of
of activity of compound 13 as methylation of the amide compound 23, with PDE3B (Figure 4b, right). In this docking
nitrogen would result in the loss of this intramolecular HB and model, the 7-methoxy group of compound 23 occupied the
make the coplanar conformation highly energetically unfavor- same binding pocket as the hydroxy ethyl group of the ICOS
able. Losing the nitrogen in the quinoline ring likely had the compound.8 In contrast, the nonselective PDE3B inhibitor
same effect (44, 45, and 46). Finally, the left-hand amide DNMDP (Figure 1)13 does not occupy this pocket. This
group is seen occupying the hydrophobic cavity at the entrance observation further suggests that binding in this region of the
of the PDE3B catalytic site, consistent with the DEL protein may play a role in driving PDE3B/PBE3A selectivity.
information wherein this position was attached to the DNA Finally, it was of interest to us to explore the phenotypic
tag and assumed to be solvent exposed. activity of our top selective compound (Table 3, compound
To understand the origin of the selectivity of the boronic 18) as well as our top active compound (Table 3, compound
acid compounds, we overlaid the crystal structure of Table 1, 35). Compound 18 showed selectivity far superior to
compound 23 in PDE3B with an aligned crystal structure of previously characterized compounds Merck 8a8 (reported:
DNMDP (Figure 1) in PDE3A (Figure 4c).13 In contrast to pIC50 = 10, selectivity 6) and ICOS7 (IMAP PDE3B pIC50 =
the PDE3A inhibitor DNMDP (Figure 1) and other reported 7.16, selectivity = 12). In order to determine how potency of
nonselective PDE3B inhibitors, 23 does not appear to form these compounds translated from a biochemical assay to a
any polar interactions with the highly conserved Gln988. The cellular assay context, we investigated the compounds in
4-methoxy group of the quinoline moiety instead displaces the adipocyte lipolysis cellular assay. The adipocyte lipolysis assay
Gln988 and occupies the small pocket made available by the examines PDE-mediated hydrolysis of cAMP to 5′-AMP,
displacement of Gln988. The major polar interactions between which results in decreased lipolysis. In this assay, inhibition of
23 and PDE3B are derived from the boronic acid HB PDE activity leads to a reduction in lipolysis and increase in
interactions in the metal-binding site instead. Close examina- glycerol levels. PDE3B expression is significantly enriched in
tion of the hydrophobic binding site indicates the pocket adipocytes compared to PDE3A, allowing us to interrogate
between Try736, Trp951, and Gly940 are smaller in PDE3A PDE3B activity in a cellular context. Upon testing compound
than PDE3B due to the movement of the side chains (i.e., 18, at a single 10 μM dose, we saw limited but significant
Gly953 in PDE3A and Gly940 in PDE3B) and therefore steric activity, whereas Merck 8a8 demonstrated a larger response,
clash may occur with the 7-methoxy group of the quinoline consistent with its higher biochemical potency (Supporting
moiety in PDE3A but not in PDE3B. However, Gln988 in Information, Figure S2). Lack of demonstration of substantial
H https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

cellular activity for compound 18 could be due to differences Following the hydrolysis of the FAM-cAMP substrate by PDE3, the
in the bioactive conformation of the compound, inadequate FAM-AMP product is generated. When the product is bound to the
cell penetration, or lack of biochemical potency for PDE3B IMAP nanoparticle (and Tb is also bound), there is a high signal in
compared to Merck8a. Compound 35 demonstrated a the 520 nm channel, and the ratio of 520:490 nm channels is also
high. When PDE3B activity is inhibited, no product is formed, and a
response in the adipocyte assay similar to that of Merck 8a8 decrease in the signal at 520 nm and the ratio 520:490 nm are
and ICOS,7 which is an indication that a more biochemically observed as there is no longer any FAM-AMP in close proximity to
potent compound is needed for a more significant cellular the Tb-donor.
response. A TR-FRET IMAP assay (Molecular Devices R8160) was used to
Importantly, our rank ordering of potency held over other measure the inhibition of cAMP hydrolysis by PDE3A or PDE3B.
compounds profiled simultaneously, suggesting that the Test compounds were serially diluted 1:4 in DMSO as an 11-point
activity is not an off-target effect. While rather preliminary, curve with a top final assay concentration of 100 μM (40 nL/well in a
this data led us to hypothesize that upon further investigation 384 well white low volume assay plate (Greiner Bio-One, 782075).
of this series, perhaps one could improve potency while Columns 6 and 18 contained DMSO or 1 mM Trequinsin (10 μM
final assay concentration), respectively, and served as the high
maintaining selectivity, enough to show meaningful selectivity (uninhibited) and low (inhibited) controls, respectively. Assays were
data in the adipocyte lipolysis cellular assay. performed using either FL PDE3B (1.6 nM) with 130 nM FAM-

■ CONCLUSIONS
Employing a screen of PDE3B with greater diversity of
cAMP substrate (Molecular Devices, R7506) and a 120 min reaction
time or FL PDE3A (1.6 nM) with 50 nM FAM-cAMP substrate and a
45 min reaction time. Enzymes and substrates were prepared in assay
chemical matter than previous efforts outlined in the buffer [50 mM Tris−HCl pH7.5 (Gibco), 8.3 mM MgCl2 (Sigma),
literature,7−9 such as the DELs used here, demonstrates that 1.7 mM ethylene glycol-bis(2-aminoethyl ether)-N,N,N′,N′-tetra
acetic acid (EGTA) (Sigma), 0.05% BSA (Sigma), and 0.01%
the PDE3B active site remains a viable target. We identified a Pluronic F127 (Sigma)].
small molecule bound in the active site capable of achieving Following the defined reaction time, the IMAP quench reagent was
selectivity for PDE3A/B, where activity was mediated by a dispensed to the plate and allowed to equilibrate for a minimum of 3
boronic acid group. Activity of the compound was confirmed h, with assay plates covered to protect from light, prior to reading on
in cells, but selectivity was not as apparent as in the the PHERAstar plate reader (BMG Labtech Ltd.). Plates were read at
biochemical assays. We encountered challenges with engineer- an excitation of 337 nm and a dual emission of 520 and 490 nm.
ing better biochemical potency at the expense of selectivity. In RapidFire MS/MS Activity Assay. To determine compound
addition, we were unable to engineer away the boronic acid inhibitory potencies for FL PDE3B and 3A, a biochemical assay
group, without compromising compound potency. The measuring the formation of 5′-AMP product by the RapidFire-MS/
MS system was applied. 200 nL of test compound stocks (in 100%
boronic acid is, in general, a challenge for development of an DMSO) were prestamped into 384-well polypropylene microtiter
orally bioavailable drug. plates (781280, Greiner Bio One) after being serial diluted 1:3 (one
Despite these challenges, PDE3B remains a compelling volume stock adds into 2 volumes of 100% DMSO) from top
therapeutic target, supported by human genetics. The concentration of 10 mM for a 11-step concentration series. A 10 μL of
development of oral small molecule therapeutics for PDE3B total assay volume in an assay buffer consisted of 50 mM Tris−HCl,
may represent an attractive alternative to antibody therapy, pH 7.5, 8.3 mM MgCl2, 1.7 mM EGTA (BM-151, Boston
such as that used for blockage of PCSK9 to achieve an BioProducts), supplemented with 0.1 mg/mL bovine serum albumin
improved lipid profile with cardiovascular benefit. Our data (BSA) (A7030, Sigma), 0.0018% (V/V) ultrapure Tween-20 (359T-
show that the active site should not be ruled out as a path for A,G-Biosciences), and 2% (V/V) DMSO, freshly prepared 5.0 μL of
substrate solution of adenosine 3′,5′-cyclic monophosphate (3′5′-
obtaining selectivity. Minimization of on-target toxicity in the
cyclic AMP) (A6885, Sigma) at 2× apparent Km level, or 450 nM and
heart may also be achieved by selective tissue targeting. For 270 nM, for FL PDE3B (GST-Tev-10H-PDE3B (M1-E1112)-FLAG)
example, potent but nonselective PDE3A/B compounds could and 3A (GST-Tev-10H-PDE3A (M1-Q1141)-FLAG), respectively)
be delivered to fat deposits by engineering lipid nanoparticles and then 5.0 μL of enzyme solution (2.4 and 0.54 nM, for FL PDE3B
functionalized to target white adipose vasculature, resulting in and 3A, respectively) were added. The enzyme reaction was allowed
the subsequent uptake by the surrounding adipose tissue and to proceed at room temperature (22 °C) for 50 min. The reaction was
initiation of lipolysis. However, at this time, the technology for quenched with the addition of 40 μL of quench solution containing
selective adipose tissue-targeting requires further maturation 0.13% (V/V) trifluoracetic acid (TFA) (302031, Sigma) and 0.1 μM
15
before it can be used to robustly deliver small molecule N5-Mono-AMP (900382, Aldrich). As 0 or 100% inhibition
payloads in the clinic. We look forward to this research controls, on each assay plate, columns 6 and 18 contain only
informing future efforts in these areas to further explore the DMSO and column 18 with 40 μL of preadded quenching solution.
After being centrifuged at 2000 rpm for >10 min, the quenched
impact of a PDE3B novel therapeutic. reaction mixture was analyzed on a RapidFire/MS/MS system by

■ EXPERIMENTAL SECTION
IMAP TR-FRET Activity Assay. This protocol describes a TR-
detecting the formation of a 5′-AMP product. With eluent A of 0.1%
(V/V) TFA in 100% (V/V) water and eluent B of 0.5% (V/V) TFA
in 50% (V/V)/50% (V/V) acetonitrile/water, the mixture was first
FRET-based IMAP assay to measure inhibition of cAMP hydrolysis desalted by a graphite-C Hypercarb type D (C18) (G9203-80106,
by PDE3. The immobilized metal ion affinity for phosphochemical Agilent) separation matrix cartridge on a RapidFire 300 LC system
(IMAP) TR-FRET assay is a homogeneous, antibody-free, and (Agilent Technologies) and then analyzed by a coupled Agilent
sensitive assay provided in kit form by Molecular Devices. The PDE 6495C triple quadruple mass spectrometer (Agilent Technologies)
reaction is performed using a fluorescent labeled substrate (FAM- with electrospray ionization (ESI) source in the multireaction
cAMP). IMAP assays are based on the binding of phosphate to monitoring mode. For the ESI source, the curtain gas (GF) is 20
immobilized metal coordination complexes on nanoparticles. A L/min; NEB gas 25 psi; ion spray voltage (CAP) 4000 V; spray
terbium (Tb) donor enables a fluorescent energy transfer to occur temperature (ST) 400 °C; spray flow (SF) 12 L/min; NOZ 500 V;
when FAM-AMP product is present�FAM-AMP and Tb donor both HP RF 150 V; LP RF 60 V; and Delta EMV 200 V14
bind to the nanoparticle, bringing the Tb donor in close proximity to Fragments of enzyme reaction product 5′-mono-AMP, internal
the fluorescein acceptor on the product and enabling FRET. standard (IS) 15N5-5′-AMP, and substrate 3′-5′ cAMP were

I https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

monitored by the transition of 348.1 m/z → 136.1 m/z (Q1 → Q3), (1) Flash column chromatography was performed using a
353.1 m/z → 141.1 m/z, and 330.1 m/z → 136.1 m/z, respectively, Teledyne Isco CombiFlash RF Flash chromatography system
and with 50 ms dwell time, and using an instrument-specific fragment with SNAP silica cartridges.
voltage (FV), collision energy (CE) + 20 V, and collision cell exit (2) Prep HPLC was performed using a Agilent 1290 Infinity II
potential (CAV) + 5 V. Calibration curves (12 points 1:2 serial HPLC/MS, with an Agilent 1290 Infinity II Mass Directed
diluted from top concentration up to 1.0 μM) were set up using a 5′- Automated Purification System, while scanning at wavelengths
AMP product standard (01930, Fluka) dissolved in the same assay 215 and 254 nm. A liquid-phase method was used on a Water
buffer. Data were acquired and processed with Analyst for Windows Xselect C18 column (100 mm × 19 mm, 5 μm packing
(version 1.6.2, build 8489). The mass spectrometry detection peak diameter, 40 mL/min flow rate). Gradient elution at ambient
areas for both product and substrate fragments were normalized using temperature was used with the mobile phases as (A) H2O
the peak area of the internal standard. Product concentrations were containing 0.1% (v/v) formic acid and (B) acetonitrile
determined with normalized peak area from product calibration containing 0.1% (v/v) formic acid.
curves. On each assay plate, column 6 contains only DMSO and (3) Prep HPLC was performed using a Gilson Prep HPLC system.
column18 with predispensed TFA as high (zero inhibition) and low Analytes were detected scanning for specific wavelengths
(100% inhibition) controls, respectively. Data were fit to the following between 200 and 400 nm. A liquid-phase method was used on
equation (as described for the IMAP assays) to determine IC50. In the a Water Xselect CSH C18 column (100 mm × 19 mm, 5 μm
case of IC50s ≫ [Enzyme], a simple four parameter fit was used, with packing diameter, 40 mL/min flow rate). Gradient elution at
data normalized to high and low controls, where y is the % of ambient temperature was used with the mobile phases as (A)
normalized enzyme activity (or inhibition), x is the concentration of H2O containing 0.1% (v/v) formic acid and (B) acetonitrile
inhibitor, and s is the Hill slope factor. The mean values and standard containing 0.1% (v/v) formic acid.
deviation of pIC50 values from at least 2−3 replicates were reported. (4) Teledyne ISCO AccqPrep HP150 equipped with a CSH
General Chemistry Experimental. Solvents and reagents were XSELECT 30 × 75 Prep HPLC column (30 mm, 75 mm, 5
purchased from commercial suppliers and used as received. Reactions μm). The liquid-phase method used gradient elution with
were monitored by liquid chromatography−mass spectrometry (LC− mobile phases as (A) water containing 0.1% v/v formic acid
MS). The LC−MS analysis was performed on one of the following: and (B) acetonitrile containing 0.1% v/v formic acid.
(1) Thermo Scientific LC−MS instrument equipped with a The purity of the synthesized compounds was determined by LC-
Kinetex XB-C18 column (2.1 × 50 mm, 1.7 μm packing MS analysis. All compounds for biological testing were more than
diameter) and a Thermo Scientific LTQ XL linear ion trap 95% pure unless otherwise stated.
mass spectrometer (positive ion). Analytes were detected as a General Procedure for TCFH Amide Couplings. A 4 mL vial
summed UV wavelength of 210−400 nm. The liquid-phase was charged with a magnetic stir bar and carboxylic acid (0.165
method with 0.6 mL/min flow rate used gradient elution with mmoL, 1.1 equiv). To this vial, a 0.6 mL aliquot of a 0.25 M stock
the mobiles phases as (A) water containing 0.1% volume/ solution of (S)-3-borono-5-((1-cyclohexyl-2-(methylamino)-2-
volume (v/v) formic acid and (B) acetonitrile containing 0.1% oxoethyl)carbamoyl)benzenaminium chloride (55 mg, 0.15 mmol)
v/v formic acid. in NMP was added. Next, a 0.2 mL aliquot of a 2.625 M stock
(2) Thermo Scientific LC−MS instrument equipped with a solution of N-ethyl-N-isopropan-2-amine (68 mg, 0.525 mmol, 3.5
Kinetex C8 column (2.1 × 30 mm, 1.7 μm packing diameter) equiv) in NMP was added. Lastly, a 0.2 mL aliquot of a 0.9 M stock
and a Thermo Scientific LCQ fleet ion trap mass spectrometer solution of N-(chloro(dimethylamino)methylene)-N-methylmethana-
(positive ion). Analytes were detected as a summed UV minium hexafluorophosphate(V) (51 mg, 0.18 mmol, 1.2 equiv) in
wavelength of 200−400 nm. The liquid-phase method with NMP was added. The reaction mixture was then allowed to stir at
0.65 mL/min flow rate used gradient elution with the mobiles room temperature for 18 h. Compounds were purified via reverse-
phases as (A) water containing 0.1% v/v formic acid and (B) phase chromatography to afford the desired amide products.
acetonitrile containing 0.1% v/v formic acid and 0.1% v/v General Procedure for HATU Amide Couplings. A 4 mL vial
water. was charged with a magnetic stir bar and carboxylic acid (0.087 mmol,
(3) Waters Acquity UPLC instrument equipped with a CSH C19 1 equiv). To this vial was added a solution of HATU (0.095 mmol,
column (2.1 × 300 mm, 1.8 μm packing diameter) and a 0.2 M in DMF, 1.09 equiv) and DIPEA 0.379 mmol, 4.33 equiv). The
Waters electrospray ionization mass spectrometer (positive resulting mixture was allowed to stir at room temperature for 5 min.
and negative ion). The liquid-phase method used gradient After this time, a solution of amine (0.094 mmol, 1.08 equiv) was
elution with the mobiles phases as (A) water containing 0.1% added, and the reaction was allowed to stir overnight. After this time,
v/v formic acid and (B) acetonitrile containing 0.1% v/v the reaction was concentrated to dryness. Compounds were purified
formic acid. via reverse-phase chromatography to afford the desired amide
products.
(4) Waters Acquity UPLC instrument equipped with a CSH C18
General Procedure for DMTMM Amide Coupling. To a 4 mL
column (2.1 × 30 mm, 1.7 μm packing diameter) and a Waters
vial charged with a magnetic stir bar and amine (0.125 mmol, 1.25
electrospray ionization mass spectrometer (positive and
equiv) was added sodium 3-borono-5-(4,7-dimethoxyquinoline-2-
negative ion). The liquid-phase method used gradient elution
carboxamido)benzoate (compound 8, 176 mM stock solution in
with the mobile phases as (A) water containing 0.1% v/v
NMP, 0.100 mmol, 1.0 equiv), DMTMM (687 mM stock solution in
formic acid and (B) acetonitrile containing 0.1% v/v formic
NMP, 0.120 mmol, 1.2 equiv), and N-methylmorpholine (1.5 M
acid or (A) 95:5 water containing 0.1% NH4OH/acetonitrile
solution in NMP). The reaction mixture was allowed to stir at room
(pH = 9.4) and (B) acetonitrile.
temperature for 18 h. Compounds were purified by reverse-phase
The NMR spectra were recorded at ambient temperature (unless chromatography to afford the desired amide products.
otherwise stated) using standard pulse methods on any of the tert-Butyl (1-(Ethylcarbamoyl)azepan-3-yl)carbamate (Inter-
following spectrometers and signal frequencies: Bruker Ascend 400 mediate A). To a vial charged with a magnetic stir bar and tert-
(1H = 400 MHz), Bruker AVANCE 400 (1H = 400 MHz), or Bruker butyl azepan-3-ylcarbamate (610.0 mg, 2.84 mmol) was added DCM
DPX400 (1H = 400 MHz). Chemical shifts are referenced to the (6.1 mL). The solution was chilled in an ice bath, then to the solution
residual solvent peak and are reported in parts per million. Coupling was added ethyl isocyanate (225 μL, 2.84 mmol, 1 equiv) dropwise.
constants are quoted to the nearest 0.1 Hz, and multiplicities are given The reaction mixture was allowed to stir at room temperature for 20
by the following abbreviations and combinations thereof: s (singlet), h. The product was purified by normal-phase chromatography to
d (doublet), t (triplet), dd (doublet of doublets), dt (doublet of afford the desired product (595 mg, 73% yield). LC−MS m/z: 186.07
triplets), dquin (doublet of quintets), m (multiplet), and br (broad). [M + H-Boc]+. 1H NMR (methanol-d4, 400 MHz): δ 6.6−6.8 (m,
The purifications were performed on one of the following: 1H), 3.4−3.8 (m, 3H), 3.1−3.3 (m, 3H), 3.0−3.1 (m, 1H), 1.7−2.0

J https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

(m, 3H), 1.6−1.7 (m, 1H), 1.47 (s, 9H), 1.3−1.4 (m, 2H), 1.17 (t, Compound 2 was prepared according to the HATU amide coupling
3H, J = 1.0 Hz). procedure. LC−MS m/z: 496.25 [M + H]+. 1H NMR (400 MHz,
(3-((tert-Butoxycarbonyl)amino)-5-((1-(ethylcarbamoyl)azepan- MeOD-d4): δ ppm 8.18−7.84 (m, 3H), 7.42 (s, 1H), 7.22 (s, 1H),
3-yl)carbamoyl)phenyl)boronic Acid (Intermediate B). To a vial 4.35 (d, 1H, J = 8.3 Hz), 3.81 (s, 3H), 2.76 (s, 3H), 2.33 (s, 6H),
charged with a magnetic stir bar and tert-butyl (1-(ethylcarbamoyl)- 1.90−1.67 (m, 6H), 1.37−1.06 (m, 5H).
azepan-3-yl)carbamate (Intermediate A, 36 mg, 126 μmol) was N-Ethyl-3-(3-hydroxy-5-(2-methoxy-3,5-dimethylbenzamido)-
added DCM (630 μL). The solution was chilled in an ice bath, then benzamido)azepane-1-carboxamide (3). To a vial charged with a
to the solution was added TFA (19 μL, 252 μmol, 2 equiv) dropwise. magnetic stir bar and tert-butyl (3-((1-(ethylcarbamoyl)azepan-3-
The reaction mixture was allowed to stir at room temperature for 18 yl)carbamoyl)-5-hydroxyphenyl)carbamate (Intermediate C, 214 mg,
h, then concentrated to afford the crude oil. The crude product was 0.509 mmol) was added DCM (4 mL) followed by TFA (0.78 mL,
dissolved in DMF (630 μL) and used directly. To a separate glass vial 10.2 mmol, 20 equiv). The reaction mixture was allowed to stir at
charged with 3-borono-5-((tert-butoxycarbonyl)amino)benzoic acid room temperature for 2 h, then concentrated to afford the crude
(35 mg, 126 μmol, 1 equiv) and HATU (48 mg, 126 μmol, 1 equiv) product, which was dissolved in DMF (2 mL) and used directly. To
was added DMF (630 μL) followed by DIPEA (66 μL, 378 μmol, 3 the solution was added 2-methoxy-3,5-dimethylbenzoic acid (70 mg,
equiv). The premix solution was allowed to react at room temperature 0.388 mmol, 1 equiv) followed by DIPEA (0.27 mL, 1.55 mmol, 4
for 15 min, then the intermediate amine (126 μmol, 1 equiv) was equiv) and HATU (148 mg, 0.388 mmol, 1.3 equiv). The reaction
added. The reaction mixture was allowed to stir at room temperature mixture was allowed to stir at room temperature for 3 h, then
for 2 h, then concentrated to afford the crude product. A portion of concentrated to afford the crude product. The crude product was
the crude product was purified by reverse-phase chromatography to purified by reverse-phase chromatography to afford the desired
afford the desired product. LC−MS m/z: 449.18 [M + H]+. 1H NMR product (65 mg, 35% yield). LC−MS m/z: 483.13 [M + H]+. 1H
(methanol-d4, 400 MHz): δ ppm 7.82 (s, 1H), 7.73 (br s, 1H), 7.68 NMR DMSO-d6: δ ppm 10.2−10.3 (s, 1H), 9.6−9.8 (s, 1H), 8.4−8.5
(br s, 1H), 7.58 (br s, 1H), 3.87 (br s, 1H), 3.50 (br dd, 2H, J = 3.1, (d, 1H), 7.5−7.6 (m, 2H), 7.1−7.2 (s, 2H), 6.9−7.0 (s, 1H), 6.6−6.7
14.9 Hz), 3.2−3.3 (m, 1H), 3.2−3.2 (m, 3H), 1.9−2.0 (m, 1H), 1.7− (s, 1H), 3.9−4.0 (br s, 1H), 3.7−3.8 (s, 3H), 3.4−3.6 (m, 8H), 3.0−
1.8 (m, 2H), 1.4−1.7 (m, 2H), 1.43 (s, 9H), 1.2−1.4 (m, 1H), 1.09 3.3 (m, 4H), 2.2−2.3 (d, 6H), 1.7−1.9 (m, 3H), 1.4−1.7 (m, 2H),
(t, 3H, J = 7.2 Hz). 1.2−1.4 (m, 1H), 1−1.1 (t, 3H).
(3-Amino-5-((1-(ethylcarbamoyl)azepan-3-yl)carbamoyl)- 2,2,2-Trifluoroacetic Acid Compound with (R)-(3-((1-Cyclohexyl-
phenyl)boronic Acid (Intermediate C). To a vial charged with a 2-(methylamino)-2-oxoethyl)carbamoyl)-5-(4,7-dimethoxyquino-
magnetic stir bar and tert-butyl (1-(ethylcarbamoyl)azepan-3-yl)- line-2-carboxamido)phenyl)boronic Acid (1:1) (4). To a solution of
carbamate (Intermediate A, 386 mg, 1.35 mmol) was added DCM (R)-(3-((tert-butoxycarbonyl)amino)-5-((1-cyclohexyl-2-(methylami-
(6.8 mL). The solution was chilled in an ice bath, then to the solution no)-2-oxoethyl)carbamoyl)phenyl)boronic acid (28 mg, 0.065 mmol)
was added TFA (313 μL, 4.07 mmol, 3 equiv) dropwise. The reaction in dioxane (1 mL) was added 4 N HCl in dioxane (1 mL). The
mixture was allowed to stir at room temperature for 18 h, then reaction mixture was stirred at room temperature for 18 h, diluted
concentrated to afford the crude product as a yellow oil. The crude with ether (10 mL). The reaction mixture was centrifuged, the
product was dissolved in DMF (6.8 mL) and used directly. To a supernatant was decanted, and the solid was dried and used for the
separate glass vial charged with 3-((tert-butoxycarbonyl)amino)-5- next step directly. The above prepared amine was dissolved in a
hydroxybenzoic acid (343 mg, 1.35 mmol, 1 equiv) and HATU (515 mixture of acetonitrile (2 mL)/DMF (0.5 mL), and 4,7-dimethox-
mg, 1.35 mmol, 1 equiv) was added DMF (6.8 mL) followed by yquinoline-2-carboxylic acid (19.59 mg, 0.084 mmol), 1-methyl-1H-
DIPEA (708 μL, 4.07 mmol, 3 equiv). The premix solution was imidazole (26.5 mg, 0.323 mmol), and TCFH (46.5 mg, 0.084 mmol)
allowed to react at room temperature for 15 min, then the were added successively. The reaction mixture was stirred at room
intermediate amine (1.35 mmol, 1 equiv) was added. The reaction temperature for 18 h, concentrated to dryness, and the crude was
mixture was allowed to stir at room temperature for 2 h, then purified by reverse-phase chromatography to afford the desired
concentrated to afford the crude product. A portion of the crude product (17 mg, 38% yield). LC−MS m/z: 549.2 [M + H]. 1H NMR
product was purified by reverse-phase chromatography to afford the DMSO-d6: δ ppm 10.76 (s, 1H), 8.41 (s, 1H), 8.37 (s, 1H), 7.98−
desired product. LC−MS m/z: 421.15 [M + H]+. 1H NMR 8.20 (m, 4H), 7.62 (s, 1H), 7.59 (d, J = 2.5 Hz, 1H), 7.34 (dd, J =
(methanol-d4, 400 MHz): δ ppm 7.1−7.2 (m, 1H), 7.0−7.1 (m, 9.19, 2.5 Hz, 1H), 4.31−4.38 (m, 1H), 4.16 (s, 3H), 3.97 (s, 3H),
1H), 6.7−6.8 (m, 1H), 3.8−3.9 (m, 1H), 3.3−3.6 (m, 4H), 3.0−3.2 2.62 (d, J = 4 Hz, 3H), 1.67−1.82 (m, 4H), 1.54−1.67 (m, 2H),
(m, 4H), 2.9−3.0 (m, 1H), 1.5−2.0 (m, 5H), 1.4−1.4 (m, 2H), 1.3− 1.13−1.25 (m, 3H), 0.96−1.12 (m, 2H).
1.4 (m, 9H), 1.2−1.3 (m, 2H), 1.0−1.1 (m, 3H). 2,2,2-Trifluoroacetic Acid Compound with (S)-(3-((1-Cyclohexyl-
(3-((1-(Ethylcarbamoyl)azepan-3-yl)carbamoyl)-5-(2-methoxy- 2-(methylamino)-2-oxoethyl)carbamoyl)-5-(4,7-dimethoxyquino-
3,5-dimethylbenzamido)phenyl)boronic Acid (1). To a vial charged line-2-carboxamido)phenyl)boronic Acid (1:1) (5). To a solution of
with a magnetic stir bar and (3-((tert-butoxycarbonyl)amino)-5-((1- (S)-(3-((tert-butoxycarbonyl)amino)-5-((1-cyclohexyl-2-(methylami-
(ethylcarbamoyl)azepan-3-yl)carbamoyl)phenyl)boronic acid (Inter- no)-2-oxoethyl)carbamoyl)phenyl)boronic acid (28 mg, 0.065 mmol)
mediate B, 170 mg, 0.379 mmol) was added DCM (3 mL) followed in dioxane (1 mL) was added 4 N HCl in dioxane (1 mL). The
by TFA (0.58 mL, 7.58 mmol, 20 equiv). The reaction mixture was reaction mixture was stirred at room temperature for 18 h, diluted
allowed to stir at room temperature for 2 h, then concentrated to with ether (10 mL). The reaction mixture was centrifuged, the
afford the crude product, which was dissolved in DMF (2 mL) and supernatant was decanted, and the solid was dried and used for the
used directly. To the solution was added 2-methoxy-3,5-dimethyl- next step directly. The above prepared amine was dissolved in a
benzoic acid (50 mg, 0.277 mmol, 1 equiv) followed by DIPEA (0.19 mixture of acetonitrile (2 mL)/DMF (0.5 mL), and 4,7-dimethox-
mL, 1.11 mmol, 4 equiv) and HATU (106 mg, 0.277 mmol, 1.4 yquinoline-2-carboxylic acid (19.59 mg, 0.084 mmol), 1-methyl-1H-
equiv). The reaction mixture was allowed to stir at room temperature imidazole (26.5 mg, 0.323 mmol), and TCFH (46.5 mg, 0.084 mmol)
for 3 h, then concentrated to afford the crude product. The crude were added successively. The reaction mixture was stirred at room
product was purified by reverse-phase chromatography to afford the temperature for 18 h, concentrated to dryness, and the crude was
desired product (83 mg, 59% yield). LC−MS m/z: 511.18 [M + H]+. purified by reverse-phase chromatography to afford the desired
1
H NMR DMSO-d6: δ ppm 10.2−10.3 (s, 1H), 8.4−8.5 (d, 1H), product (21 mg, 47% yield). LC−MS m/z: 549.3 [M + H]+. 1H NMR
8.1−8.2 (d, 3H), 7.9−8.0 (s, 1H), 7.2−7.3 (d, 2H), 6.6−6.7 (br s, (400 MHz, DMSO-d6): δ ppm 10.76 (s, 1H), 8.41 (s, 1H), 8.37 (s,
1H), 3.9−4.0 (br s, 1H), 3.7−3.8 (s, 3H), 3.5−3.7 (m, 7H), 3.1−3.2 1H), 8.13 (d, J = 9.13 Hz, 1H), 8.10−8.05 (m, 3H), 7.62 (s, 1H),
(m, 4H), 2.2−2.3 (d, 6H), 1.7−1.9 (m, 3H), 1.5−1.6 (m, 2H), 1.2− 7.58 (d, J = 2.38 Hz, 1H), 7.34, (dd, J = 9.26, 1.5 Hz, 1H), 4.31−4.38
1.4 (m, 1H), 1.0−1.1 (t, 3H). (m, 1H), 4.16 (s, 3H), 3.97 (s, 3H), 2.62 (d, J = 4.5 Hz, 3H), 1.67−
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- 1.82 (m, 4H), 1.54−1.67 (m, 2H), 1.13−1.27 (m, 3H), 0.96−1.13
5-(2-methoxy-3,5-dimethylbenzamido)phenyl)boronic Acid (2). (2H).

K https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

(3-(4,7-Dimethoxyquinoline-2-carboxamido)phenyl)boronic (3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-
Acid (6). In a vial, (3-aminophenyl)boronic acid (18 mg, 0.13 mmol) (methylcarbamoyl)phenyl)boronic Acid (10). Compound 10 was
was stirred in ACN (1 mL) and DMF (0.25 mL). To the solution, prepared according to the general procedure for DMTMM amide
4,7-dimethoxyquinoline-2-carboxylic acid (30 mg, 0.13 mmol) was coupling. LC−MS m/z: 410.28 [M + H]+. 1H NMR (400 MHz,
added followed by N-methylimidazole (52 μL, 0.64 mmol), then MeOD-d4): δ ppm 8.31−8.41 (m, 1H) 8.18−8.25 (m, 1H) 8.09−8.17
TCFH (54 mg, 0.19 mmol). The reaction was stirred at RT for 1 h, (m, 1H) 7.82−8.01 (m, 1H) 7.62−7.67 (m, 1H) 7.51−7.56 (m, 1H)
then volatiles were removed in vacuo. The resulting mixture was 7.23−7.29 (m, 1H) 4.16 (s, 3H) 3.94−4.05 (m, 3H) 2.90−3.00 (m,
purified via reverse-phase chromatography to afford the desired 3H).
product (28 mg, 58%). LC−MS m/z: [353.2]+. 1H NMR (400 MHz, (3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-
MeOD-d4): δ ppm 8.14−8.22 (m, 1H) 8.04−8.10 (m, 1H) 7.89−7.98 (dimethylcarbamoyl)phenyl)boronic Acid (11). In a vial, 3-borono-
(m, 1H) 7.68−7.74 (m, 1H) 7.53−7.60 (m, 1H) 7.38−7.47 (m, 2H) 5-(4,7-dimethoxyquinoline-2-carboxamido)benzoic acid (100 mg,
7.25−7.33 (m, 1H) 4.20 (s, 3H) 4.01 (s, 3H). 0.25 mmol), 40% dimethylamine in water (64 μL, 0.51 mmol), and
(3-Carbamoyl-5-(4,7-dimethoxyquinoline-2-carboxamido)- HATU (140 mg, 0.38 mmol) were stirred in DCM (2 mL) with DMF
phenyl)boronic Acid (7). Compound 7 was prepared according to the (0.5 mL), and DIPEA (176 μL, 1.010 mmol) was added. The reaction
general procedure for DMTMM amide coupling (hydrochloride salt was stirred at RT for 19 h. Volatiles were removed in vacuo, then the
of amine used). LC−MS m/z: 396.31 [M + H]+. 1H NMR (400 subsequent reaction mixture was taken up in DCM and saturated
MHz, MeOD-d4): δ ppm 8.37−8.44 (m, 1H) 8.24−8.32 (m, 1H) sodium bicarbonate. The layers were separated, and the aqueous layer
8.14−8.22 (m, 1H) 7.90−8.09 (m, 1H) 7.66−7.75 (m, 1H) 7.53− was extracted the DCM three times. The organic layers were
7.58 (m, 1H) 7.25−7.32 (m, 1H) 4.19 (s, 3H) 3.94−4.07 (m, 3H). combined, and volatiles were removed in vacuo. The precipitate was
Sodium 3-Borono-5-(4,7-dimethoxyquinoline-2-carboxamido)- then purified via reverse-phase chromatography to afford the desired
benzoate (8). To a 250 mL round-bottomed flask charged with a product (36 mg, 32%). LC−MS m/z: 424.3 [M + H]+. 1H NMR (400
magnetic stir bar, 4,7-dimethyoxyquinoline-2-carboxylic acid (2.00 g, MHz, MeOD-d4): δ ppm 8.30−8.36 (m, 1H) 8.11−8.16 (m, 1H)
8.58 mmol, 1.0 equiv) and BOP (4.16 g, 9.43 mmol, 1.1 equiv) were 8.05−8.09 (m, 1H) 7.97−8.01 (m, 1H) 7.63−7.68 (m, 1H) 7.51−
added NMP (20 mL). To the solution was added DIPEA (4.49 mL, 7.57 (m, 1H) 7.43−7.50 (m, 1H) 4.38 (s, 3H) 4.02−4.11 (m, 3H)
25.7 mmol, 3.0 equiv), and the solution was allowed to stir at room 3.04−3.18 (m, 6H).
temperature for 10 min. Separately, to a 40 mL reaction vial charged (3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-
with (3-amino-5-(methoxycarbonyl)phenyl)boronic acid (1.67 g, 8.58 (isopropylcarbamoyl)phenyl)boronic Acid (12). In a vial, 4,7-
mmol, 1.0 equiv) was added NMP (10 mL), and the solution was dimethoxyquinoline-2-carboxylic acid (53 mg, 0.23 mmol) and
sonicated to aid dissolution. Following the 10 min preactivation, the HATU (130 mg, 0.34 mmol) were stirred in DMF (1.1 mL), and
amine solution was added to the reaction mixture via a syringe over 5 DIPEA (160 μL, 0.90 mmol) was added. The reaction was stirred at
min, and the resulting reaction mixture was allowed to react at room RT for 15 min, then (3-amino-5-(isopropylcarbamoyl)phenyl)boronic
temperature for 1 h. The reaction mixture was then slowly diluted acid (50 mg, 0.225 mmol) was added in DMF (1.1 mL), and the
with half-saturated aqueous ammonium chloride solution (250 mL), reaction was stirred at RT for 1 h. All volatiles were removed in vacuo.
and the resulting suspension was stirred at room temperature for 10 The reaction mixture was then purified via reverse-phase chromatog-
min. The suspended solids were isolated via filtration, washed with raphy to afford the desired product. LC−MS m/z: 438.3 [M + H]+.
half-saturated sodium chloride, and dried. The crude solid was 1
H NMR (400 MHz, MeOD-d4): δ ppm 8.28−8.35 (m, 1H) 8.21−
dispersed into a bilayer of ethyl acetate (150 mL) and water (75 mL), 8.26 (m, 1H) 8.13−8.18 (m, 1H) 7.81−7.87 (m, 1H) 7.64−7.69 (m,
and the resulting suspended solid was isolated via filtration and 1H) 7.53−7.57 (m, 1H) 7.25−7.30 (m, 1H) 4.21−4.30 (m, 1H) 4.17
washed with ethyl acetate and dried to afford the acid intermediate as (s, 3H) 4.00 (s, 3H) 1.29 (d, J = 6.6 Hz, 6H).
a light tan solid (2.63 g, 6.41 mmol, 74.8% yield, LC−MS m/z 411.2 (3-(4,7-Dimethoxy-N-methylquinoline-2-carboxamido)-5-
[M + H]+). The crude product was transferred to a 250 mL round- (isopropylcarbamoyl)phenyl)boronic Acid (13). A vial containing N-
bottomed flask and dissolved in acetonitrile (60 mL) and water (20 (3-bromo-5-(isopropylcarbamoyl)phenyl)-4,7-dimethoxy-N-methyl-
mL). The solution was treated with 6 N aqueous sodium hydroxide quinoline-2-carboxamide (35 mg, 0.072 mmol), Pd(dppf)Cl2 (5 mg,
(4.29 mL, 25.7 mmol, 4.0 equiv), and the reaction mixture was 0.006 mmol), B2Pin2 (20 mg, 0.079 mmol), potassium acetate (21
allowed to react at room temperature for 1.5 h. The solution was mg, 0.214 mmol), 1,4-dioxane (0.75 mL), and water (0.010 mL) was
further diluted with 40 mL water, and the reaction mixture was sparged with nitrogen gas for 1 min and heated at 80 °C for 2 h. After
allowed to continue to react at room temperature overnight. The this time, the reaction was allowed to cool to room temperature. LC−
mixture was concentrated under reduced pressure to remove the MS shows two desired products (in the boronic acid form as well as
majority of acetonitrile, then treated with 1 N aqueous hydrochloric the boronic ester form). The reaction was concentrated to dryness
acid to adjust the pH to 6. The suspended solid was isolated via and then purified via reverse-phase chromatography to afford the
filtration and triturated with 2-propanol and dried to afford the desired product (5 mg, 15%). LC−MS m/z: 452.25 [M + H]+. 1H
desired product (2.35 g, 5.34 mmol, 62.3%). LC−MS m/z: 397.2 [M NMR (400 MHz, MeOD-d4): δ ppm 8.1−8.3 (s, 1H), 7.8−8.0 (m,
+ H]+. 1H NMR (400 MHz, DMSO-d6): δ ppm 10.6−10.8 (m, 1H), 1H), 7.6−7.8 (m, 2H), 7.3−7.5 (s, 1H), 7.0−7.2 (s, 2H), 6.7−6.9 (s,
9.3−9.9 (m, 2H), 8.5 (s, 1H), 8.3 (s, 1H), 8.2 (s, 1H), 8.1 (d, J = 9.26 1H), 4.0−4.2 (br s, 1H), 3.8−4.0 (m, 6H), 3.5−3.7 (s, 3H), 1.1−1.4
Hz, 1H), 7.6−7.7 (m, 2H), 7.3 (dd, J = 9.13, 2.13 Hz, 1H) 4.2 (s, 3H) (m, 8H).
4.0 (s, 3H). (R)-(3-((1-Amino-3-methyl-1-oxobutan-2-yl)carbamoyl)-5-(4,7-
(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5- dimethoxyquinoline-2-carboxamido)phenyl)boronic Acid (14).
(methoxycarbonyl)phenyl)boronic Acid (9). In a vial 4,7-dimethox- Compound 14 was prepared according to the general procedure for
yquinoline-2-carboxylic acid (500 mg, 2.1 mmol) was stirred in NMP DMTMM amide coupling. LC−MS m/z: 495.24 [M + H]+. 1H NMR
(24 mL) with BOP (1.04 g, 2.4 mmol) and DIPEA (1.1 mL, 6.4 (400 MHz, MeOD-d4): δ ppm 8.34−8.44 (m, 1H) 8.22−8.31 (m,
mmol), the reaction was stirred for 5 min, then (3-amino-5- 1H) 8.12−8.18 (m, 1H) 7.85−8.06 (m, 1H) 7.64−7.69 (m, 1H)
(methoxycarbonyl)phenyl)boronic acid (420 mg, 2.1 mmol) was 7.53−7.58 (m, 1H) 7.23−7.30 (m, 1H) 4.38−4.49 (m, 1H) 4.17 (s,
added in NMP (12 mL). The reaction was stirred for 1H, then half- 3H) 3.95−4.05 (m, 3H) 2.15−2.30 (m, 1H) 0.94−1.18 (m, 6H).
saturated ammonium chloride (75 mL) was added slowly, and the (S)-(3-((1-Amino-3-methyl-1-oxobutan-2-yl)carbamoyl)-5-(4,7-
reaction was filtered. The precipitate was then purified via reverse- dimethoxyquinoline-2-carboxamido)phenyl)boronic Acid (15).
phase chromatography to afford the desired product (15.2 mg, 1.7%). Compound 15 was prepared according to the general procedure for
LC−MS m/z: 411.2 [M + H]+. 1H NMR (400 MHz, MeOD-d4): δ DMTMM amide coupling. LC−MS m/z: 495.2 [M + H]+. 1H NMR
ppm 8.59−8.65 (m, 1H) 8.29−8.35 (m, 1H) 8.17−8.22 (m, 1H) (700 MHz, DMSO-d6): δ ppm 10.76 (s, 1H) 8.44 (s, 1H) 8.38 (d, J =
8.07−8.13 (m, 1H) 7.69−7.76 (m, 1H) 7.56−7.60 (m,1H) 7.26−7.36 1.29 Hz, 1H) 8.25 (s, 2H) 8.06 (s,1H) 8.12 (d, J = 9.46 Hz, 1H) 7.91
(m, 1H) 4.18−4.24 (m, 3H) 4.02 (s, 3H) 3.91−3.97 (m, 3H). (d, J = 9.04 Hz, 1H) 7.61 (s, 1H) 7.58 (d, J = 2.58 Hz, 1H) 7.53 (s,

L https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

1H) 7.33 (dd, J = 9.04, 2.58 Hz, 1H) 7.15 (s, 1H) 4.34−4.39 (m, 1H) phenylmethanamine (65 μL, 0.51 mmol), HATU (140 mg, 0.38
4.15 (s, 3H) 3.97 (s, 3H) 2.08−2.15 (m, 1H) 0.95 (dd, J = 6.45, 5.16 mmol), and DIPEA (180 mL, 1.0 mmol) were added, and the
Hz, 6H). reaction was stirred for an additional 2 h. All volatiles were removed
(R)-(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-((1-(methyla- in vacuo, then taken up in brine and DCM, the aqueous layer was
mino)-1-oxopropan-2-yl)carbamoyl)phenyl)boronic Acid (16). extracted with DCM three times, then the organic layers were
Compound 16 was prepared according to the general procedure for combined, and all volatiles were removed in vacuo. The reaction
DMTMM amide coupling. LC−MS m/z: 481.24 [M + H]+. 1H NMR mixture was then purified via reverse-phase chromatography to afford
(400 MHz, MeOD-d4): δ ppm 8.33−8.41 (m, 1H) 8.21−8.31 (m, the desired product (64 mg, 49%). LC−MS m/z: 500.3 [M + H]+. 1H
1H) 8.11−8.18 (m, 1H) 7.88−8.08 (m, 1H) 7.62−7.69 (m, 1H) NMR (400 MHz, methanol-d4): δ ppm 8.1−8.3 (m, 3H) 7.7−7.8 (m,
7.50−7.56 (m, 1H) 7.22−7.31 (m, 1H) 4.51−4.63 (m, 1H) 4.17 (s, 1H) 7.5−7.6 (m, 2H) 7.2−7.5 (m, 6H) 4.6−4.8 (m, 2H) 4.3 (br s,
3H) 3.95−4.06 (m, 3H) 2.74−2.81 (m, 3H) 1.43−1.56 (m, 3H). 3H) 4.0−4.1 (m, 3H) 2.9−3.0 (m, 3H). 94% pure by LCMS.
(R)-(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-((1-methoxy- (3-(Benzyl(methyl)carbamoyl)-5-(4,7-dimethoxyquinoline-2-
3-methylbutan-2-yl)carbamoyl)phenyl)boronic Acid (17). Com- carboxamido)phenyl)boronic Acid (23). In a vial, 3-borono-5-(4,7-
pound 17 was prepared according to the general procedure for dimethoxyquinoline-2-carboxamido)benzoic acid (100 mg, 0.25
DMTMM amide coupling. LC−MS m/z: 496.29 [M + H]+. 1H NMR mmol), N-methyl-1-phenylmethanamine (65 μL, 0.51 mmol), and
(400 MHz, MeOD-d4): δ ppm 8.30−8.36 (m, 1H) 8.19−8.26 (m, HATU (140 mg, 0.38 mmol) were stirred in DCM (2.0 mL) with
1H) 8.12−8.17 (m, 1H) 7.82−8.04 (m, 1H) 7.63−7.69 (m, 1H) DMF (0.5 mL), and DIPEA (180 μL, 1.0 mmol) was added. The
7.51−7.57 (m, 1H) 7.24−7.30 (m, 1H) 4.17 (s, 3H) 4.04−4.12 (m, reaction was stirred at RT for 16 h, then additional N-methyl-1-
1H) 3.96−4.03 (m, 3H) 3.53−3.63 (m, 2H) 3.37−3.42 (m, 3H) phenylmethanamine (65 μL, 0.51 mmol), HATU (140 mg, 0.38
1.95−2.07 (m, 1H) 0.98−1.08 (m, 6H). mmol), and DIPEA (180 mL, 1.0 mmol) were added, and the
(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-((1-methoxypro- reaction was stirred for an additional 2 h. All volatiles were removed
pan-2-yl)carbamoyl)phenyl)boronic Acid (18). Compound 18 was in vacuo, then taken up in brine and DCM, the aqueous layer was
prepared according to the general procedure for DMTMM amide
extracted with DCM three times, then the organic layers were
coupling. LC−MS m/z: 468.27 [M + H]+. 1H NMR (400 MHz,
combined, and all volatiles were removed in vacuo. The reaction
MeOD-d4): δ ppm 8.28−8.34 (m, 1H) 8.12−8.27 (m, 2H) 7.80−8.03
mixture was then purified via reverse-phase chromatography to afford
(m, 1H) 7.63−7.68 (m, 1H) 7.52−7.57 (m, 1H) 7.24−7.30 (m, 1H)
the desired product (64 mg, 49%). LC−MS m/z: 500.3 [M + H]+. 1H
4.30−4.43 (m, 1H) 4.17 (s, 3H) 3.95−4.07 (m, 3H) 3.50−3.57 (m,
(400 MHz, MeOD-d4): δ ppm 8.08−8.27 (m, 3H) 7.74−7.83 (m,
1H) 3.39−3.49 (m, 4H) 1.22−1.35 (m, 3H).
1H) 7.47−7.62 (m, 2H) 7.19−7.45 (m, 6H) 4.59−4.83 (m, 2H) 4.26
(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-((2-
methoxypropyl)carbamoyl)phenyl)boronic Acid (19). Compound (br s, 3H) 3.97−4.07 (m, 3H) 2.94−3.10 (m, 3H). 94% pure by LC−
19 was prepared according to the general procedure for DMTMM MS.
amide coupling. LC−MS m/z: 468.27 [M + H]+. 1H NMR (400 (3-(3-Cyclopropoxyazetidine-1-carbonyl)-5-(4,7-dimethoxyqui-
MHz, MeOD-d4): δ ppm 8.32−8.38 (m, 1H) 8.12−8.28 (m, 2H) noline-2-carboxamido)phenyl)boronic Acid (24). Compound 24
7.81−8.04 (m, 1H) 7.63−7.68 (m, 1H) 7.51−7.56 (m, 1H) 7.23− was prepared according to the general procedure for DMTMM amide
7.30 (m, 1H) 4.17 (s, 3H) 3.96−4.05 (m, 3H) 3.57−3.67 (m, 1H) coupling. LC−MS m/z: 492.2 [M + H]+. 1H NMR1H NMR (700
3.39−3.52 (m, 5H) 1.18−1.26 (m, 3H). MHz, DMSO-d6): δ ppm 10.73−10.86 (m,7.33, 1H) 4.49−4.55 (m,
(S)-(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-((tetrahydro- 1H), 4.43−4.49 (m, 1H), 4.27−4.32 (m, 1H), 4.23−4.27 (m, 1H),
2H-pyran-3-yl)carbamoyl)phenyl)boronic Acid (20). Compound 20 4.14 (s, 3H) 3.97 (s, 3H) 3.88−3.93 (m, 1H) 3.36 (dt, J = 6.02, 3.01
was prepared according to the general procedure for DMTMM amide Hz, 1H) 0.41−0.60 (m, 4H).
coupling. LC−MS m/z: 480.34 [M + H]+. 1H NMR (400 MHz, (3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-(pyrrolidine-1-
MeOD-d4): δ ppm 8.28−8.36 (m, 1H) 8.19−8.27 (m, 1H) 8.06−8.17 carbonyl)phenyl)boronic Acid (25). Compound 25 was prepared
(m, 1H) 7.80−8.01 (m, 1H) 7.62−7.67 (m, 1H) 7.49−7.57 (m, 1H) according to the general procedure for DMTMM amide. LC−MS m/
7.20−7.32 (m, 1H) 4.17 (s, 3H) 4.04−4.12 (m, 1H) 3.93−4.02 (m, z: 514.3 [M + H]+. 1H NMR (700 MHz, DMSO-d6): δ ppm 10.71−
4H) 3.80−3.89 (m, 1H) 3.45−3.56 (m, 1H) 3.34−3.42 (m, 1H) 10.84 (m, 1H) 8.19 (m, 2H) 8.19−8.28 (m, 3H) 8.10−8.19 (m, 2H)
1.66−2.12 (m, 4H). 93% pure by LCMS. 7.63 (s, 1H) 7.54−7.61 (m, 2H) 7.44 (m, 6H) 7.18−7.44 (m, 6H)
(R)-(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5-((tetrahydro- 4.50−4.76 (m, 2H) 4.14 (s, 3H) 3.96 (s, 3H) 3.18−3.26 (m, 2H)
2H-pyran-3-yl)carbamoyl)phenyl)boronic Acid (21). Compound 21 1.02−1.18 (m, 3H). 93% pure by LC−MS.
was prepared according to the general procedure for DMTMM amide (3-(2,3-Dihydro-1H-pyrrolo[3,4-c]pyridine-2-carbonyl)-5-(4,7-di-
coupling. LC−MS m/z: 480.34 [M + H]+. 1H NMR (400 MHz, methoxyquinoline-2-carboxamido)phenyl)boronic Acid (26). Com-
MeOD-d4): δ ppm 8.28−8.34 (m, 1H) 8.21−8.27 (m, 1H) 8.12−8.18 pound 26 was prepared according to the general procedure for
(m, 1H) 7.80−8.00 (m, 1H) 7.63−7.68 (m, 1H) 7.51−7.57 (m, 1H) DMTMM amide. LC−MS m/z: 499.2 [M + H]+. 1H NMR (700
7.23−7.30 (m, 1H) 4.17 (s, 3H) 4.04−4.12 (m, 1H) 3.94−4.02 (m, MHz, DMSO-d6): δ ppm 10.82 (d, J = 2.58 Hz, 1H) 8.55−8.69 (m,
4H) 3.80−3.88 (m, 1H) 3.45−3.55 (m, 1H) 3.34−3.43 (m, 1H) 1H) 8.51 (dd, J = 17.85, 4.95 Hz, 1H) 8.34 (d, J = 1.29 Hz, 1H) 8.28
1.66−2.13 (m, 4H). (br d, J = 8.17 Hz, 1H) 8.26 (br s, 2H) 8.12 (d, J = 9.03 Hz, 1H) 7.81
(3-(4,7-Dimethoxyquinoline-2-carboxamido)-5- (d, J = 1.29 Hz, 1H) 7.59 (s, 1H) 7.57 (d, J = 2.15 Hz, 1H) 7.38−7.53
(isobutylcarbamoyl)phenyl)boronic Acid (22). Compound 22 was (m, 1H) 7.33 (dd, J = 9.04, 2.58 Hz, 1H) 4.88−5.01 (m, 4H) 4.14 (s,
prepared according to the general procedure for DMTMM amide 3H) 3.96 (s, 3H). 91% pure by LC−MS.
coupling. LC−MS m/z: 452.2 [M + H]+. 1H NMR (600 MHz, (R)-(3-((tert-Butoxycarbonyl)amino)-5-((1-cyclohexyl-2-(methyl-
DMSO-d6): δ ppm 10.72 (s, 1H) 8.40 (t, J = 5.87 Hz, 1H) 8.38 (t, J = amino)-2-oxoethyl)carbamoyl)phenyl)boronic Acid (Intermediate
2.02 Hz, 1H) 8.31−8.33 (m, 1H) 8.20 (s, 2H) 8.12 (d, J = 9.17 Hz, D). To a glass vial charged with 3-borono-5-((tert-butoxycarbonyl)-
1H) 8.00 (s, 1H) 7.60 (s, 1H)7.57 (d, J = 2.57 Hz, 1H) 7.33 (dd, J = amino)benzoic acid (528 mg, 1.88 mmol, 1 equiv) and HATU (786
9.17, 2.57 Hz, 1H) 4.15 (s, 3H) 3.97 (s, 3H) 3.07−3.14 (m, 2H) 1.88 mg, 2.07 mmol, 1.1 equiv) was added DMF (4.7 mL) followed by
(dquin, J = 13.48, 6.81, 6.81, 6.81, 6.81 Hz, 1H) 0.92 (d, J = 6.60 Hz, DIPEA (982 μL, 5.64 mmol, 3 equiv). The premix solution was
6H). allowed to react at room temperature for 15 min, then a solution of
(3-(Benzyl(methyl)carbamoyl)-5-(4,7-dimethoxyquinoline-2- (R)-2-amino-2-cyclohexyl-N-methylacetamide (320 mg, 1.88 mmol, 1
carboxamido)phenyl)boronic Acid (23). In a vial, 3-borono-5-(4,7- equiv) in DMF (4.7 mL) was added. The reaction mixture was
dimethoxyquinoline-2-carboxamido)benzoic acid (100 mg, 0.25 allowed to stir at room temperature for 2 h, then concentrated to
mmol), N-methyl-1-phenylmethanamine (65 μL, 0.51 mmol), and afford the crude product. The crude oil was purified by reverse-phase
HATU (140 mg, 0.38 mmol) were stirred in DCM (2.0 mL) with chromatography to afford the desired product. LC−MS m/z: 434.09
DMF (0.5 mL), and DIPEA (180 μL, 1.0 mmol) was added. The [M + H]+. 1H NMR (MeOD-d4, 400 MHz): δ ppm 7.81 (s, 1H), 7.71
reaction was stirred at RT for 16 h, then additional N-methyl-1- (br s, 1H), 7.63 (br s, 1H), 4.2−4.3 (m, 1H), 2.6−2.7 (m, 3H), 1.5−

M https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

1.8 (m, 8H), 1.4−1.5 (m, 1H), 1.43 (s, 9H), 1.34 (s, 1H), 1.0−1.2 washed with brine, dried over magnesium sulfate, filtered, and
(m, 7H). volatiles removed in vacuo. The reaction mixture purified via reverse-
(R)-(3-(6-Chloro-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-8- phase chromatography to afford the desired product (8.6 mg, 12%).
carboxamido)-5-((1-cyclohexyl-2-(methylamino)-2-oxoethyl)- LC−MS m/z: 469.3 [M + H]+. 1H NMR (400 MHz, MeOD-d4): δ
carbamoyl)phenyl)boronic Acid (27). To a solution of (R)-(3- ppm 1.04−1.17 (m, 2H) 1.20−1.38 (m, 4H) 1.63−1.73 (m, 2H)
amino-5-((1-cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- 1.74−1.82 (m, 2H) 1.83−1.92 (m, 2H) 2.77 (s, 3H) 4.07 (s, 3H)
phenyl)boronic acid (59 mg, 0.18 mmol), in DCM (0.4 mL) and 4.34 (d, J = 8.51 Hz, 1H) 7.35 (d, J = 4.88 Hz, 1H) 7.87 (s, 1H) 7.96
ACN (0.4 mL), 6-chloro-3-oxo-3,4-dihydro-2H-benzo[b][1,4]- (s, 1H) 8.20 (s, 1H) 8.28 (s, 1H) 8.60 (d, J = 5.63 Hz, 1H).
oxazine-8-carboxylic acid (40 mg, 0.18 mmol) and 1-methyl-1H- (R)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
imidazole (98 μL, 1.2 mmol) were added, followed by TCFH (99 mg, 5-(5-methoxy-2-methylpyrimidine-4-carboxamido)phenyl)boronic
0.351 mmol). The reaction was stirred at RT for 1 h, then all volatiles Acid, 2,2,2-Trifluoroacetic Acid Compound (1:1) (31). To a solution
were removed in vacuo. The reaction mixture was purified via reverse- of (R)-(3-((tert-butoxycarbonyl)amino)-5-((1-cyclohexyl-2-(methyla-
phase chromatography to afford the desired product (64 mg, 49%). mino)-2-oxoethyl)carbamoyl)phenyl)boronic acid (28 mg, 0.065
LC−MS m/z: 543.2 [M + H]+. 1H NMR (400 MHz, methanol-d4): δ mmol) in dioxane (1 mL) was added 4 N HCl in dioxane (1 mL).
ppm 8.1−8.3 (m, 2H), 8.1 (s, 1H), 7.8 (s, 1H), 7.5 (d, J = 2.25 Hz, The reaction mixture was stirred at room temperature for 18 h, then
1H), 7.1 (d, J = 2.38 Hz, 1H), 4.8 (s, 2H), 4.3−4.4 (m, 1H), 2.8 (d, J diluted with ether (10 mL). The reaction mixture was centrifuged, the
= 4.00 Hz, 3H), 1.8−1.9 (m, 2H), 1.7−1.8 (m, 2H), 1.6−1.7 (m, supernatant was decanted, and the resulting solid was dried to afford
2H), 1.2−1.4 (m, 5H), 1.0−1.2 (m, 3H). the crude intermediate. The intermediate was dissolved in a mixture
(R)-(3-(6-Chloro-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-8- of acetonitrile (2 mL)/DMF (0.5 mL), and 5-methoxy-2-methylpyr-
carboxamido)-5-((1-cyclohexyl-2-(methylamino)-2-oxoethyl)- imidine-4-carboxylic acid (16.30 mg, 0.097 mmol), 1-methyl-1H-
carbamoyl)phenyl)boronic Acid (27). To a solution of (R)-(3- imidazole (26.5 mg, 0.323 mmol), and TCFH (53.6 mg, 0.097 mmol)
amino-5-((1-cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- were added successively. The reaction mixture was stirred at room
phenyl)boronic acid (59 mg, 0.18 mmol), in DCM (0.4 mL) and temperature for 18 h, concentrated to dryness, and the crude was
ACN (0.4 mL), 6-chloro-3-oxo-3,4-dihydro-2H-benzo[b][1,4]- purified by reverse-phase chromatography to afford the desired
oxazine-8-carboxylic acid (40 mg, 0.18 mmol) and 1-methyl-1H- product (18 mg, 44% yield). LC−MS m/z: 484.17 [M + H]+. 1H
imidazole (98 μL, 1.2 mmol) were added, followed by TCFH (99 mg, NMR (400 MHz, DMSO-d6): δ ppm 10.63 (s, 1H), 8.71 (s, 1H),
0.351 mmol). The reaction was stirred at RT for 1 h, then all volatiles 8.17−8.13 (m, 2H), 8.11−7.97 (m, 3H), 4.4−4.18 (m, 1H), 3.94 (s,
were removed in vacuo. The reaction mixture was purified via reverse- 3H), 2.62 (s, 3H), 2.6 (d, J = 8.0 Hz, 3H), 1.80−1.65 (m, 4H), 1.65−
phase chromatography to afford the desired product (64 mg, 49%). 1.51 (m, 2H), 1.24−1.10 (m, 3H), 1.10−0.95 (m, 2H).
LC−MS m/z: 543.2 [M + H]+. 1H NMR (400 MHz, MeOD-d4): δ (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
ppm 1.00−1.19 (m, 3H) 1.22−1.38 (m, 5H) 1.63−1.73 (m, 2H) 5-(4-methoxyquinoline-2-carboxamido)phenyl)boronic Acid (32).
1.75−1.82 (m, 2H) 1.83−1.91 (m, 2H) 2.76 (d, J = 4.00 Hz, 3H) Compound 32 was prepared according to the general procedure for
4.30−4.38 (m, 1H) 4.80 (s, 2H) 7.08 (d, J = 2.38 Hz, 1H) 7.45 (d, J TCFH couplings. LC−MS m/z: 519.3 [M + H]+. 1H NMR (400
= 2.25 Hz, 1H) 7.84 (s, 1H) 8.05 (s, 1H) 8.12−8.28 (m, 2H). 100% MHz, DMSO-d6): δ ppm 10.77 (s, 1H) 8.41 (t, 1H, J = 1.9 Hz) 8.38
pure by LC−MS. (s, 1H) 8.25 (d, 2H, J = 7.3 Hz) 8.21 (d, 1H, J = 8.2 Hz) 8.08−8.03
(3-(((R)-1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- (m, 3H) 7.91 (t, 1H, J = 7.9 Hz) 7.74−7.71 (m, 2H) 4.35 (t, 1H, J =
5-(2-(2,6-dimethylphenoxy)propanamido)phenyl)boronic Acid 8.4 Hz) 4.19 (s, 3H) 2.62 (d, 3H, J = 2.0 Hz) 1.80−1.75 (m, 2H)
(28). To a solution of (R)-(3-amino-5-((1-cyclohexyl-2-(methylami- 1.74−1.69 (m, 2H) 1.64−1.56 (m, 2H) 1.22−1.11 (m, 3H) 1.11−
no)-2-oxoethyl)carbamoyl)phenyl)boronic acid (30 mg, 0.09 mmol), 0.98 (m, 2H). 87% pure by LCMS.
in DCM (0.2 mL) and ACN (0.2 mL), and 2-(2,6-dimethylphenoxy)- (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
propanoic acid (17.5 mg, 0.090 mmol) and 1-methyl-1H-imidazole 5-(4,6-dimethoxyquinoline-2-carboxamido)phenyl)boronic Acid
(50 μL, 0.6 mmol) were added, followed by TCFH (51 mg, 0.18 (33). Compound 33 was prepared according to the general procedure
mmol). The reaction was stirred at RT for 2 h, then water was added. for TCFH couplings. LC−MS m/z: 549.3 [M + H]+. 1H NMR (400
The aqueous layer was extracted with DCM three times, the organic MHz, DMSO-d6): δ ppm 10.70 (s, 1H) 8.40 (s, 1H) 8.37 (s, 1H)
layers were combined, washed with brine, the dried over magnesium 8.23 (s, 2H) 8.12 (d, 2H, J = 9.0 Hz) 8.07−8.05 (m, 2H) 8.03 (d, 1H,
sulfate, filtered, and volatiles removed in vacuo. The reaction mixture J = 8.6 Hz) 7.70 (s, 1H) 7.55−7.53 (m, 1H) 7.50 (d, 1H, J = 3.0 Hz)
was purified via reverse-phase chromatography to afford the desired 4.35−4.33 (m, 1H) 4.18 (s, 3H) 3.95 (s, 3H) 2.63−2.62 (m, 3H)
product (11 mg, 25%). LC−MS m/z: 510.3 [M + H1H NMR (400 2.53−2.52 (m, 3H) 1.80−1.75 (m, 2H) 1.73−1.69 (m, 2H) 1.17−
MHz, MeOD-d4): δ ppm 1.04−1.15 (m, 2H) 1.17−1.36 (m, 5H) 1.14 (m, 3H).
1.55 (d, J = 6.63 Hz, 3H) 1.64−1.71 (m, 2H) 1.73−1.82 (m, 2H) (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
1.82−1.91 (m, 2H) 2.32 (s, 6H) 2.75 (s, 3H) 3.21 (dd, J = 14.76, 5-(6-methoxyquinoline-2-carboxamido)phenyl)boronic Acid (34).
7.25 Hz, 1H) 4.33 (d, J = 8.38 Hz, 1H) 4.55 (dd, J = 13.38, 6.63 Hz, Compound 34 was prepared according to the general procedure for
1H) 6.94 (t, J = 7.50 Hz, 1H) 7.04 (d, J = 7.63 Hz, 2H) 7.83 (s, 1H) TCFH couplings. LC−MS m/z: 519.3 [M + H]+. 1H NMR (400
8.07 (d, J = 14.13 Hz, 2H). MHz, DMSO-d6): δ ppm 10.7 (s, 1H) 8.5 (d, J = 8.2 Hz, 1H) 8.40
(R)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- (m, 2H) 8.2 (m, 3H) 8.2 (d, J = 9.5 Hz, 1H) 8.0−8.1 (m, 3H) 7.5−
5-(2-methoxy-3,5-dimethylbenzamido)phenyl)boronic Acid (29). 7.6 (m, 1H) 7.5 (d, J = 3.0 Hz, 1H) 4.0 (s, 3H) 2.6 (m, 3H) 2.5 (m,
Compound 29 was prepared according to the HATU amide coupling 2H) 1.8 (m, 2H) 1.7 (m, 2H) 1.5−1.6 (m, 3H) 1.1−1.2 (m, 3H).
procedure to give the desired product (7.5 mg, 17%). LC−MS m/z: (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
496.25 [M + H]+. 1H NMR (400 MHz, MeOD-d4): δ ppm 8.18−7.84 5-(4-methylquinoline-2-carboxamido)phenyl)boronic Acid (35).
(m, 3H), 7.42 (s, 1H), 7.22 (s, 1H), 4.35 (d, 1H, J = 8.3 Hz), 3.81 (s, Compound 35 was prepared according to the general procedure for
3H), 2.76 (s, 3H), 2.33 (s, 6H), 1.90−1.67 (m, 6H), 1.37−1.06 (m, TCFH couplings. LC−MS m/z: 503.3 [M + H]+. 1H NMR (400
5H). MHz, DMSO-d6): δ ppm 10.77 (s, 1H) 8.40 (d, 2H, J = 10.9 Hz)
(R)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- 8.27 (d, 1H, J = 7.8 Hz) 8.25−8.23 (m, 3H) 8.14 (s, 1H) 8.08−8.03
5-(4-methoxypicolinamido)phenyl)boronic Acid (30). To a solution (m, 3H) 7.94−7.91 (m, 1H) 7.79 (t, 1H, J = 7.8 Hz) 4.34 (t, 1H, J =
of (R)-(3-amino-5-((1-cyclohexyl-2-(methylamino)-2-oxoethyl)- 8.4 Hz) 2.84 (d, 3H, J = 0.9 Hz) 2.63−2.62 (m, 3H) 2.53−2.52 (m,
carbamoyl)phenyl)boronic acid (50 mg, 0.15 mmol), 4-methoxypi- 1H) 1.80−1.75 (m, 2H) 1.74−1.69 (m, 2H) 1.61−1.57 (m, 1H)
colinic acid (23 mg, 0.15 mmol), 1-methyl-1H-imidazole (84 μL, 1.1 1.22−1.11 (m, 3H) 1.09−0.98 (m, 2H).
mmol) in DCM (0.4 mL), and acetonitrile (0.4 mL) was added (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
TCFH (84 mg, 0.300 mmol). The reaction was stirred for 2 h at room 5-(5-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrido[2,3-d]pyrimidine-
temperature, then water was added, and the aqueous layer was 7-carboxamido)phenyl)boronic Acid (36). Compound 36 was
extracted with DCM three times. The organic layers were combined, prepared according to the general procedure for TCFH couplings.

N https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

LC−MS m/z: 537.3 [M + H]+. 1H NMR (400 MHz, DMSO-d6): δ 8.75 (m, 1H) 8.65 (d, 1H, J = 8.1 Hz) 8.47−8.34 (m, 3H) 8.10−8.02
ppm 11.74 (s, 1H) 11.44 (s, 1H) 10.32 (s, 1H) 8.20 (s, 2H) 8.12 (d, (m, 3H) 7.83−7.76 (m, 1H) 4.36−4.29 (m, 1H) 2.64−2.61 (m, 3H)
1H, J = 2.2 Hz) 8.09 (s, 1H) 8.06−8.03 (m, 1H) 8.00 (s, 1H) 7.96 (d, 1.82−1.76 (m, 2H) 1.75−1.69 (m, 2H) 1.67−1.54 (m, 2H) 1.24−
1H, J = 8.6 Hz) 7.14 (s, 1H) 4.31 (t, 1H, J = 8.6 Hz) 2.61 (d, 3H, J = 1.12 (m, 3H) 1.11−0.95 (m, 2H). 87% pure by LCMS.
4.7 Hz) 2.54 (s, 3H) 1.77−1.74 (m, 2H) 1.71−1.68 (m, 2H) 1.63− (S)-(3-(2-Naphthamido)-5-((1-cyclohexyl-2-(methylamino)-2-
1.53 (m, 2H) 1.20−1.11 (m, 3H) 1.08−0.94 (m, 2H). oxoethyl)carbamoyl)phenyl)boronic Acid (44). Compound 44 was
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- prepared according to the general procedure for TCFH couplings.
5-(quinoline-2-carboxamido)phenyl)boronic Acid (37). Compound LC−MS m/z: 488.2 [M + H]+. 1H NMR (400 MHz, DMSO-d6): δ
37 was prepared according to the general procedure for TCFH ppm 10.5 (s, 1H) 8.6 (s, 1H) 8.3 (s, 2H) 8.2 (s, 2H) 8.1 (d, J = 9.9
couplings. LC−MS m/z: 489.2 [M + H]+. 1H NMR (400 MHz, Hz, 1H) 8.0−8.1 (m, 5H) 8.0 (d, J = 8.6 Hz, 1H) 7.6−7.7 (m, 2H)
DMSO-d6): δ ppm 10.81 (s, 1H) 8.66 (d, 1H, J = 8.6 Hz) 8.41 (d, 4.3 (t, J = 8.4 Hz, 1H) 2.6 (m, 3H) 1.7−1.8 (m, 4H) 1.6 (m, 2H)
2H, J = 2.6 Hz) 8.28 (s, 1H) 8.27 (d, 1H, J = 3.0 Hz) 8.24 (s, 2H) 1.1−1.2 (m, 3H) 1.0−1.1 (m, 2H).
8.14 (d, 1H, J = 7.8 Hz) 8.08−8.04 (m, 3H) 7.94 (t, 1H, J = 7.9 Hz) (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
7.79−7.77 (m, 1H) 4.35 (t, 1H, J = 8.4 Hz) 3.41 (s, 1H) 2.53−2.52 5-(6-methoxy-2-naphthamido)phenyl)boronic Acid (45). Com-
(m, 3H) 1.80−1.75 (m, 2H) 1.69−1.73 (m, 2H) 1.63−1.47 (m, 2H) pound 45 was prepared according to the general procedure for
1.21−1.16 (m, 2H) 1.16−1.15 (s, 2H). TCFH couplings. LC−MS m/z: 518.3 [M + H]+. 1H NMR (400
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- MHz, DMSO-d6): δ ppm 10.42 (s, 1H) 8.57 (s, 1H) 8.30−8.28 (m,
5-(isoquinoline-3-carboxamido)phenyl)boronic Acid (38). Com- 2H) 8.21 (s, 2H) 8.08−8.02 (m, 3H) 7.98 (t, 2H, J = 9.7 Hz) 7.94 (d,
pound 38 was prepared according to the general procedure for 1H, J = 8.6 Hz) 7.43 (d, 1H, J = 2.6 Hz) 7.29−7.27 (m, 1H) 4.34 (t,
TCFH couplings. LC−MS m/z: 489.2 [M + H]+. 1H NMR (400 1H, J = 8.6 Hz) 3.9 (s, 3H) 2.63−2.60 (m, 3H) 1.79−1.75 (m, 2H)
MHz, DMSO-d6): δ ppm 10.77 (s, 1H) 9.49 (s, 1H) 8.74−8.71 (m, 1.72−1.69 (m, 2H) 1.63−1.57 (m, 2H) 1.21−1.11 (m, 3H) 1.08−
1H), 8.39 (d, 2H, J = 9.5 Hz) 8.33 (d, 1H, J = 7.7 Hz) 8.28 (d, 2H, J 0.98 (m, 2H).
= 7.7 Hz) 8.07−8.02 (m, 3H) 7.95−7.92 (m, 1H) 7.89−7.86 (m, 1H) (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
4.34 (t, 1H, J = 8.4 Hz) 2.63−2.61 (m, 3H) 2.53−2.52 (m, 1H) 5-(7-methoxy-2-naphthamido)phenyl)boronic Acid (46). Com-
1.80−1.76 (m, 2H) 1.74−1.69 (m, 3H) 1.64−1.57 (m, 2H) 1.22− pound 46 was prepared according to the general procedure for
TCFH couplings. LC−MS m/z: 518.3 [M + H]+. 1H NMR (400
1.11 (m, 3H).
MHz, DMSO-d6): δ ppm 10.47 (s, 1H) 8.52 (s, 1H) 8.30 (d, 2H, J =
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
5-(quinoline-3-carboxamido)phenyl)boronic Acid (39). Compound 0.9 Hz) 8.21 (s, 2H) 8.08−8.05 (m, 1H) 8.04 (s, 1H) 7.99−7.97 (m,
39 was prepared according to the general procedure for TCFH 2H) 7.94 (d, 1H, J = 8.6 Hz) 7.91−7.89 (m, 1H) 7.47 (d, 1H, J = 2.6
couplings. LC−MS m/z: 489.2 [M + H]+. 1H NMR (400 MHz, Hz) 7.31−7.29 (m, 1H) 4.34 (t, 1H, J = 8.4 Hz) 3.92 (s, 3H) 2.63−
DMSO-d6): δ ppm 10.70 (s, 1H) 9.40 (d, 1H, J = 2.2 Hz) 9.02 (d, 2.61 (m, 3H) 2.53−2.52 (m, 1H) 1.79−1.70 (m, 4H) 1.61−1.56 (m,
1H, J = 2.2 Hz) 8.31 (d, 2H, J = 6.0 Hz) 8.25−8.21 (m, 1H) 8.21− 1H) 1.22−1.10 (m, 3H) 1.08−0.97 (m, 2H).
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
8.12 (m, 3H) 8.08−8.06 (m, 2H) 8.01 (d, 1H, J = 9.0 Hz) 7.94−7.90 5-(7-fluoroquinoxaline-5-carboxamido)phenyl)boronic Acid (47).
(m, 1H) 7.75 (t, 1H, J = 7.6 Hz) 4.35 (t, 1H, J = 8.4 Hz) 2.62 (d, 3H, Compound 47 was prepared according to the general procedure for
J = 4.8 Hz) 1.80−1.69 (m, 4H) 1.65−1.55 (m, 2H) 1.22−1.11 (m, TCFH couplings. LC−MS m/z: 508.3 [M + H]+. 1H NMR (400
3H) 1.10−0.98 (m, 2H). MHz, DMSO-d6): δ ppm 11.59 (s, 1H) 9.14 (d, 1H, J = 1.7 Hz) 9.12
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- (d, 1H, J = 1.7 Hz) 8.32 (s, 1H) 8.31 (d, 1H, J = 9.3 Hz) 8.26 (s, 2H)
5-(isoquinoline-1-carboxamido)phenyl)boronic Acid (40). Com-
8.22 (d, 1H, J = 1.3 Hz) 8.16−8.13 (m, 1H) 8.07−8.04 (m, 3H) 4.33
pound 40 was prepared according to the general procedure for (t, 1H, J = 8.4 Hz) 2.62−2.61 (m, 3H) 1.79−1.74 (m, 2H) 1.73−1.67
TCFH couplings. LC−MS m/z: 489.2 [M + H]+. 1H NMR (400 (m, 2H) 1.63−1.56 (m, 2H) 1.21−1.11 (m, 3H) 1.09−0.97 (m, 2H).
MHz, DMSO-d6): δ ppm 10.86 (s, 1H) 8.87 (d, 1H, J = 8.7 Hz) 8.64 (S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)-
(d, 1H, J = 5.6 Hz) 8.36 (s, 1H) 8.32 (t, 1H, J = 1.9 Hz) 8.23 (s, 2H) 5-(8-methoxy-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-
8.13−8.08 (m, 2H) 8.07−8.03 (m, 3H) 7.89−7.86 (t, 1H, J = 7.8 Hz) carboxamido)phenyl)boronic Acid (48). To a vial containing 8-
7.80−7.77 (m, 1H) 4.33 (t, 1H, J = 8.4 Hz) 2.62 (d, 3H, J = 4.7 Hz) methoxy-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-carboxylic
1.80−1.75 (m, 2H) 1.73−1.69 (m, 2H) 1.65−1.55 (m, 2H) 1.22− acid (72 mg, 0.323 mmol) and HATU (123 mg, 0.324 mmol) were
1.10 (m, 3H) 1.09−0.98 (m, 2H). added DMF (2 mL) and DIPEA (0.154 mL, 0.882 mmol). The
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- reaction was allowed to stir at room temperature for 5 min. After this
5-(1,5-naphthyridine-2-carboxamido)phenyl)boronic Acid (41). time, a solution of (S)-(3-amino-5-((1-cyclohexyl-2-(methylamino)-
Compound 41 was prepared according to the general procedure for 2-oxoethyl)carbamoyl)phenyl)boronic acid (98 mg, 0.294 mmol) in
TCFH couplings. LC−MS m/z: 490.2 [M + H]+. 1H NMR (400 DMF (2 mL) was added, and the reaction was allowed to stir at room
MHz, DMSO-d6): δ ppm 10.85 (s, 1H) 9.16 (s, 1H) 8.69 (d, 1H, J = temperature for 18 h. The reaction was concentrated to dryness, and
8.6 Hz) 8.65 (d, 1H, J = 8.2 Hz) 8.49 (d, 1H, J = 8.6 Hz) 8.40 (s, 2H) the compound was isolated via reverse-phase chromatography to
8.33−8.19 (m, 2H) 8.08 (s, 1H) 8.07 (d, 1H, J = 6.0 Hz) 8.04 (d, 1H, afford the desired product (93 mg, 59%). LC−MS m/z: 539.17 [M +
J = 8.6 Hz) 7.97−7.95 (m, 1H) 4.35 (t, 1H, J = 8.6 Hz) 2.63−2.62 H]+. 1H NMR (400 MHz, DMSO-d6): δ ppm 10.87 (s, 1H,
(m, 3H) 1.80−1.75 (m, 2H) 1.73−1.69 (m, 2H) 1.63−1.57 (m, 2H) exchangeable), 10.22 (s, 1H, exchangeable), 8.26−8.19 (m, 3H),
1.22−1.11 (m, 3H) 1.09−0.98 (m, 2H). 8.08−7.96 (m, 3H), 7.3−7.4 (s, 1H), 7.2−7.1 (s, 1H), 4.63 (s, 3H),
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- 4.3−4.4 (m, 1H), 3.89 (m, 4H), 2.61−2.60 (m, 2H), 1.81−1.51 (m,
5-(1,7-naphthyridine-2-carboxamido)phenyl)boronic Acid (42). 5H), 1.29−0.94 (m, 5H). One exchangeable proton not observed,
Compound 42 was prepared according to the general procedure for likely under solvent peak.
TCFH couplings. LC−MS m/z: 490.3 [M + H]+. 1H NMR (400
MHz, DMSO-d6): δ ppm 10.90 (s, 1H) 9.63 (s, 1H) 8.76 (d, 1H, J =
5.6 Hz) 8.73 (d, 1H, J = 8.6 Hz) 8.48 (d, 1H, J = 8.2 Hz) 8.41 (d, 2H,
J = 0.9 Hz) 8.25 (s, 2H) 8.09−8.06 (m, 3H) 8.04 (d, 1H, J = 9.0 Hz)

*
ASSOCIATED CONTENT
sı Supporting Information

4.35 (s, 1H) 2.53−2.52 (m, 3H) 1.82−1.75 (m, 2H) 1.75−1.68 (m, The Supporting Information is available free of charge at
2H) 1.65−1.55 (m, 2H) 1.16−1.14 (m, 3H) 1.10−0.97 (m, 2H). https://pubs.acs.org/doi/10.1021/acs.jmedchem.3c01562.
(S)-(3-((1-Cyclohexyl-2-(methylamino)-2-oxoethyl)carbamoyl)- Experimental details, data for compound character-
5-(1,8-naphthyridine-2-carboxamido)phenyl)boronic Acid (43).
Compound 43 was prepared according to the general procedure for ization, protein preparation, and in vitro data and
TCFH couplings. LC−MS m/z: 490.2 [M + H]+. 1H NMR (400 crystallography parameters (PDF)
MHz, DMSO-d6): δ ppm 10.86 (s, 1H) 9.28−9.26 (m, 1H) 8.81− Formula strings (CSV)
O https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

Accession Codes Martin Rauch − GSK, Collegeville, Pennsylvania 19426,


PDB ID Codes: 8SYC. United States
Warren Rocque − GSK, Collegeville, Pennsylvania 19426,

■ AUTHOR INFORMATION
Corresponding Authors
United States
Yingnian Shen − GSK, Collegeville, Pennsylvania 19426,
United States
Ann M. Rowley − GSK, Collegeville, Pennsylvania 19426, Lisa M. Shewchuk − GSK, Collegeville, Pennsylvania 19426,
United States; orcid.org/0000-0002-3857-9447; United States
Email: Ann.X.Rowley@gsk.com Michael Squire − GSK, Collegeville, Pennsylvania 19426,
Gang Yao − GSK, Encoded Library Technologies, NCE United States
Molecular Discovery, Cambridge, Massachusetts 02140, Will Stebbeds − GSK Medicines Research Centre, Stevenage
United States; Email: Gang.X.Yao@gsk.com SG1 2NY, U.K.
Westley Tear − GSK, Encoded Library Technologies, NCE
Authors Molecular Discovery, Cambridge, Massachusetts 02140,
Logan Andrews − 23andMe Inc, Therapeutics, South San United States
Francisco, California 94080, United States; Present Xin Wang − 23andMe Inc, Therapeutics, South San Francisco,
Address: Animol Discovery, One Boston Place, Suite California 94080, United States; orcid.org/0000-0001-
3930, 201 Washington St., Boston, MA 02108, United 7242-357X
States; orcid.org/0000-0002-5092-4530 Paris Ward − GSK, Collegeville, Pennsylvania 19426, United
Aaron Bedermann − GSK, Collegeville, Pennsylvania 19426, States
United States; Present Address: Janssen Pharmaceuticals, Shouhua Xiao − 23andMe Inc, Therapeutics, South San
1400 McKean Rd, Springhouse, PA 19477, United Francisco, California 94080, United States; Present
States.; orcid.org/0009-0003-3168-2083 Address: RA Capital Management, 200 Berkeley St., 18th
Ross Biddulph − GSK Medicines Research Centre, Stevenage Floor, Boston, MA 02116, United States.
SG1 2NY, U.K. Complete contact information is available at:
Ryan Bingham − GSK Medicines Research Centre, Stevenage https://pubs.acs.org/10.1021/acs.jmedchem.3c01562
SG1 2NY, U.K.
Jennifer J. Brady − 23andMe Inc, Therapeutics, South San Author Contributions
Francisco, California 94080, United States; orcid.org/ The manuscript was written through contributions of all
0009-0000-1794-0494 authors. These authors contributed equally: Logan Andrews,
Rachel Buxton − GSK Medicines Research Centre, Stevenage Aaron Bedermann, Ross Biddulph, Ryan Bingham, Jennifer J.
SG1 2NY, U.K. Brady, Rachel Buxton, Ted Cecconie, Rona Cooper, Adam
Ted Cecconie − GSK, Collegeville, Pennsylvania 19426,
Csakai, Enoch N. Gao, Melissa C. Grenier-Davies, Meghan
United States
Lawler, Yiqian Lian, Justyna Macina, Colin Macphee, Lisa
Rona Cooper − GSK, Collegeville, Pennsylvania 19426,
United States Marcaurelle, John Martin, Patricia McCormick, Rekha
Adam Csakai − GSK, Encoded Library Technologies, NCE Pindoria, Martin Rauch, Warren Rocque, Yingnian Shen, Lisa
Molecular Discovery, Cambridge, Massachusetts 02140, M. Shewchuk, Michael Squire, Will Stebbeds, Westley Tear,
United States Xin Wang, Paris Ward, and Shouhua Xiao.
Enoch N. Gao − GSK, Collegeville, Pennsylvania 19426, Notes
United States The authors declare no competing financial interest.
Melissa C. Grenier-Davies − GSK, Encoded Library
Technologies, NCE Molecular Discovery, Cambridge,
Massachusetts 02140, United States
Meghan Lawler − GSK, Encoded Library Technologies, NCE
■ ABBREVIATIONS
BOP, benzotriazol-1-yloxytris(dimethylamino)phosphonium
Molecular Discovery, Cambridge, Massachusetts 02140, hexafluorophosphate; DIPEA, diisopropylethylamine;
United States; Present Address: Anagenex, 20 Maguire Rd. DMTMM, 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methyl-mor-
Suite 302, Lexington, MA 02421, United States. pholinium chloride; HATU, hexafluorophosphate azabenzo-
Yiqian Lian − GSK, Collegeville, Pennsylvania 19426, United triazole tetramethyl uronium; IMAP, immobilized metal
States; orcid.org/0000-0002-2163-0189 affinity for phosphochemicals; RF-MS, rapid fire mass
Justyna Macina − GSK Medicines Research Centre, Stevenage spectrometry; TCFH, chloro-N,N,N′,N′-tetramethylformami-
SG1 2NY, U.K. dinium hexafluorophosphate
Colin Macphee − GSK Medicines Research Centre, Stevenage
SG1 2NY, U.K.
Lisa Marcaurelle − GSK, Encoded Library Technologies, NCE
■ REFERENCES
(1) Jurgens, S. J.; Choi, S. H.; Morrill, V. N.; Chaffin, M.; Pirruccello,
Molecular Discovery, Cambridge, Massachusetts 02140, J. P.; Halford, J. L.; Weng, L. C.; Nauffal, V.; Roselli, C.; Hall, A. W.;
United States et al. Analysis of rare genetic variation underlying cardiometabolic
John Martin − GSK, Collegeville, Pennsylvania 19426, United diseases and traits among 200,000 individuals in the UK Biobank. Nat.
States Genet. 2022, 54, 240−250.
Patricia McCormick − GSK, Collegeville, Pennsylvania (2) Liu, H.; Maurice, D. H. Expression of cyclic GMP-inhibited
19426, United States phosphodiesterases 3A and 3B (PDE3A and PDE3B) in rat tissues:
Rekha Pindoria − GSK Medicines Research Centre, Stevenage differential subcellular localization and regulated expression by cyclic
SG1 2NY, U.K. AMP. Br. J. Pharmacol. 1998, 125 (7), 1501−1510.

P https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Article

(3) Baillie, G. S.; Tejeda, G. S.; Kelly, M. P. Therapeutic targeting of


3′,5′-cyclic nucleotide phosphodiesterases: inhibition and beyond.
Nat. Rev. Drug Discovery 2019, 18, 770−796.
(4) Lin, Y.-K.; Cheng, C.-C.; Huang, J.-H.; Chen, Y.-A.; Lu, Y.-Y.;
Chen, Y.-C.; Chen, S.-A.; Chen, Y.-J. Various subtypes of
phosphodiesterase inhibitors differentially regulate pulmonary vein
and sinoatrial node electrical activities. Exp. Ther. Med. 2020, 19,
2773−2782.
(5) Deaton, A. M.; Dubey, A.; Ward, L. D.; Dornbos, P.; Flannick, J.;
Yee, E.; Ticau, S.; Noetzli, L.; Parker, M. M.; Hoffing, R. A.; et al.
Rare loss of function variants in the hepatokine gene INHBE protect
from abdominal obesity. Nat. Commun. 2022, 13, 4319.
(6) Chung, Y. W.; Lagranha, C.; Chen, Y.; Sun, J.; Tong, G.;
Hockman, S. C.; Ahmad, F.; Esfahani, S. G.; Bae, D. H.; Polidovitch,
N.; et al. Targeted disruption of PDE3B, but not PDE3A, protects
murine heart from ischemia/reperfusion injury. Proc. Natl. Acad. Sci.
U.S.A. 2015, 112 (17), No. E2253.
(7) Snyder, P. B.; et al. Selective pde3b inhibitors and use of the
same in therapy. WO 2002070469 A2, September 9, 2002.
(8) Edmondson, S. D.; Mastracchio, A.; He, J.; Chung, C. C.;
Forrest, M. J.; Hofsess, S.; MacIntyre, E.; Metzger, J.; O’Connor, N.;
Patel, K.; Tong, X.; Tota, M. R.; Van der Ploeg, L. H. T.; Varnerin, J.
P.; Fisher, M. H.; Wyvratt, M. J.; Weber, A. E.; Parmee, E. R. Benzyl
Vinylogous Amide Substituted Aryldihydropyridazinones and Aryldi-
methylpyrazolones as Potent and Selective PDE3B Inhibitors. Bioorg.
Med. Chem. Lett. 2003, 13, 3983−3987.
(9) (a) Scapin, G.; Patel, S. B.; Chung, C.; Varnerin, J. P.;
Edmondson, S. D.; Mastracchio, A.; Parmee, E. R.; Singh, S. B.;
Becker, J. W.; Van der Ploeg, L. H. T.; Tota, M. R. Crystal Structure
of Human Phosphodiesterase 3B: Atomic Basis for Substrate and
Inhibitor Specificity. Biochemistry 2004, 43, 6091−6100. (b) Sudo, T.;
Tachibana, K.; Toga, K.; Tochizawa, S.; Inoue, Y.; Kimura, Y.;
Hidaka, H. Potent effects of novel anti-platelet aggregatory
cilostamide analogues on recombinant cyclic nucleotide phospho-
diesterase isozyme activity. Biochem. Pharmacol. 2000, 59 (4), 347−
356.
(10) Yan, B.; Ding, Z.; Zhang, W.; Cai, G.; Han, H.; Ma, Y.; Cao, Y.;
Wang, J.; Chen, S.; Ai, Y. Multiple PDE3A modulators act as
molecular glues promoting PDE3A-SLFN12 interaction and induce
SLFN12 dephosphorylation and cell death. Cell Chem. Biol. 2022, 29,
958−969.e5.
(11) Chen, Q.; Hall, J.; Foley, T. L.; Wan, J.; Li, Y.; Israel, D. I. A
method for estimating binding affinity from primary DEL selection
data. Biochem. Biophys. Res. Commun. 2020, 533 (2), 249−255.
(12) Sportsman, R. IMAP assays for protein kinases. Drug Discovery
Ser. 2006, 5, 85−96.
(13) Garvie, C. W.; Wu, X.; Papanastasiou, M.; Lee, S.; Fuller, J.;
Schnitzler, G. R.; Horner, S. W.; Baker, A.; Zhang, T.; Mullahoo, J. P.;
et al. Structure of PDE3A-SLFN12 complex reveals requirements for
activation of SLFN12 RNase. Nat. Commun. 2021, 12, 4375.
(14) Rendina, A. R.; Pietrak, B.; Smallwood, A.; Zhao, H.; Qi, H.;
Quinn, C.; Adams, N. D.; Concha, N.; Duraiswami, C.; Thrall, S. H.;
Sweitzer, S.; Schwartz, B. Mutant IDH1 enhances the production of 2-
hydroxyglutarate due to its kinetic mechanism. Biochemistry 2013, 52,
4563−4577.

Q https://doi.org/10.1021/acs.jmedchem.3c01562
J. Med. Chem. XXXX, XXX, XXX−XXX

You might also like