Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

Article

Cite This: J. Med. Chem. 2019, 62, 7160−7184 pubs.acs.org/jmc

Structure-Based Development of (1-(3′-


Mercaptopropanamido)methyl)boronic Acid Derived Broad-
Spectrum, Dual-Action Inhibitors of Metallo- and Serine-β-
lactamases
Yao-Ling Wang,†,§ Sha Liu,†,§ Zhu-Jun Yu,† Yuan Lei,† Meng-Yi Huang,† Yu-Hang Yan,† Qiang Ma,†
Yang Zheng,† Hui Deng,‡ Ying Sun,‡ Chengyong Wu,‡ Yamei Yu,‡ Qiang Chen,‡ Zhenling Wang,‡
Yong Wu,*,† and Guo-Bo Li*,†

See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for
Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy,
Sichuan University, Sichuan 610041, China
Downloaded via UNIV DE ALCALA on December 12, 2022 at 08:30:37 (UTC).


State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University,
Chengdu 610041, China
*
S Supporting Information

ABSTRACT: The emergence and spread of bacterial pathogens acquired metallo-β-lactamase (MBL) and serine-β-lactamase
(SBL) medicated β-lactam resistance gives rise to an urgent need for the development of new dual-action MBL/SBL inhibitors.
Application of a pharmacophore fusion strategy led to the identification of (2′S)-(1-(3′-mercapto-2′-methylpropanamido)-
methyl)boronic acid (MS01) as a new dual-action inhibitor, which manifests broad-spectrum inhibition to representative MBL/
SBL enzymes, including the widespread VIM-2 and KPC-2. Guided by the VIM-2:MS01 and KPC-2:MS01 complex structures,
further structural optimization yielded new, more potent dual-action inhibitors. Selectivity studies indicated that the inhibitors
had no apparent inhibition to human angiotensin-converting enzyme-2 and showed selectivity across serine hydrolyases in E.
coli and human HEK293T cells labeled by the activity-based probe TAMRA-FP. Moreover, the inhibitors displayed potentiation
of meropenem efficacy against MBL- or SBL-positive clinical isolates without apparent cytotoxicity. This work will aid efforts to
develop new types of clinically useful dual-action inhibitors targeting MBL/SBL enzymes.

■ INTRODUCTION
The β-lactam antibiotics are the most widely used class of
β-lactamases (MBLs; ambler class B), which use one or two
active site zinc ions to activate a nucleophilic water molecule
broad-spectrum antibacterial agents; however, they have been and thereby cleave the β-lactam ring (Figure 1A).14 MBLs are
plagued by the ongoing problems of resistance.1−5 Resistance further divided into subclasses B1, B2, B3, and somewhere B4,
to β-lactam antibiotics is mainly due to the production of β- primarily by difference in active site zinc coordination
lactamases, which can be classified into two categories features.15,16 The B1 MBLs (e.g., NDM, VIM, and IMP) and
according to their hydrolytic mechanisms.6−8 The first B3 MBLs (e.g., GOB, AIM, SMB, and L1) are catalyticlly
category is serine-β-lactamases (SBLs; ambler classes A, C, active with two zinc ions and show a broad substrate profile,
and D), which employ an active site nucleophilic serine to while the monozinc B2 MBLs, such as CphA and Sfh-I, have a
destroy the β-lactam ring (Figure 1A). Clinically important
SBL families include KPC, TEM, SHV, CTX-M, GES, SME, Received: May 5, 2019
AmpC, OXA, and others.4,9−13 The second category is metallo- Published: July 3, 2019

© 2019 American Chemical Society 7160 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Figure 1. (A) Mechanisms for metallo- (MBL) and serine-β-lactamase (SBL) catalyzed hydrolysis of carbapenems. (B) Dual-action MBL/SBL
inhibitors.

narrow specificity profile, solely hydrolyzing carbapenems. experimental assays, we recently identified aromatic bi-
Among them, the dizinc B1 MBLs, such as NDM-1, are of sphosphonates and rosmarinic acid as dual-action MBL/SBL
widespread clinical significance.14,17−20 inhibitors,31,32 which are only active against some types of
Several SBL inhibitors have been used in combination with a MBL and SBL enzymes. Here, we report structure-based
β-lactam partner to overcome resistance, such as, for example, development of (1-(3′-mercaptopropanamido)methyl)boronic
clavulanate/amoxicillin, tazobactam/piperacillin, sulbactam/ acid derived new broad-spectrum, dual-action MBL/SBL
ampicillin, and avibactam/ceftazidime.2,4 Recently vaborbac- inhibitors.
tam, a boron-heterocyclic SBL inhibitor, was approved for use Considering the structural and mechanistic difference
with meropenem for treating complicated urinary tract between MBL and SBL enzymes, we proposed a pharmaco-
infections and pyelonephritis.21,22 By contrast, there are no phore fusion strategy to obtain dual-action MBL/SBL
equivalent inhibitors for MBLs, which can hydrolyze almost all inhibitors. Application of this strategy led to the identification
β-lactam antibiotics including carbapenems (i.e., “last resort” of the compound (2′S)-(1-(3′-mercapto-2′-
antibiotics) as well as the β-lactam SBL inhibitors (e.g., methylpropanamido)methyl)boronic acid (MS01, Figure
clavulanate, tazobactam, and sulbactam).18,19,23 More concern- 1B), which manifested broad-spectrum inhibition to all classes
ing is the continuous emergence and spread of multidrug- of representative MBL and SBL enzymes, including subclass
resistant pathogens acquired MBL and SBL genes,24,25 leading B1 VIM-2, subclass B1 NDM-1, subclass B2 Sfh-I, subclass B3
to an urgent need for the development of broad-spectrum, GOB-18, class A KPC-2, class A TEM-1, class C AmpC, and
dual-action MBL/SBL inhibitors to overcome MBL or MBL/ class D OXA-48 enzymes; with the exception of TEM-1 and
SBL-mediated resistance. AmpC, these enzymes are carbapenemases that are able to
Until recently, there have been a few reports of dual-action inactivate carbapenems and other β-lactams.11,33 Crystallo-
MBL/SBL inhibitors (Figure 1B). Cyclic boronates are the graphic analyses reveal insights into the dual-action inhibition
most desirable dual-action inhibitors, which can potently modes of MS01 with VIM-2 and KPC-2, providing important
inhibit all classes of MBL and SBL enzymes via a mechanism basis for structural optimization. The subsequent multitarget
involving mimicking of their common natural tetrahedral structure−activity relationship (SAR) studies identified new
intermediates as revealed by crystallographic analyses.26,27 The (1-(3′-mercaptopropanamido)methyl)boronic acid derivatives
benzo[b]thiophen-2-ylboronic acids28,29 and cyclobutanones30 as broad-spectrum, dual-action inhibitors. Some of them were
(Figure 1B) inhibit multiple MBL and SBL enzymes by an observed by crystallographic analyses to bind with VIM-2 and
analogous manner to cyclic boronates (i.e., mimicking the KPC-2 via unexpected binding modes. The inhibitors were
tetrahedral intermediates). Using combined computational and further evaluated for their selectivity to human angiotensin-
7161 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Scheme 1. Synthesis of Compounds MS01−MS04a

a
(a) CH2Br2, n-BuLi, THF, −78 °C, 2 h; CH3SO2OH, 0 °C, 1 h; pinacol, 0 °C, 1 h; (b) LiHMDS, THF, −78 °C to rt, overnight; (c) HCl/1,4-
dioxane, Et2O, −60 °C to rt, 3 h; (d) TBTU, DIPEA, DMF, −10 °C, 2 h; (e) 2-methylpropylboronic acid, HCl (aq), CH3OH/hexane (1:1), rt
overnight.

Table 1. Inhibitory Potency (Ki, μM) of MS01−MS04 to the Representative MBL and SBL Enzymesa

a
The method for measuring IC50/Ki values is described in Experimental Section, and the dose−response curves of compounds inhibiting the
selected enzymes are given in Supporting Information Figure S2.

converting enzyme (ACE)-2 and to the serine hydrolyases in E. structures of MBL enzymes in complex with small-molecule
coli and human HEK293T cells by activity-based protein inhibitors,21,34 we observed that most of MBL inhibitors
profiling using TAMRA-FP, as well as their efficacy to involve direct active site metal chelation via metal-binding
potentiate meropenem activity against MBL- or SBL-positive pharmacophores (MBPs),35 such as (S)-3-mercapto-2-methyl-
clinical isolates. propanamido,36−38 5-aminothiazole-4-carboxylic acid,39 (Z)-2-

■ RESULTS AND DISCUSSION


Design of (2′S)-(1-(3′-Mercapto-2′-
mercapto-3-phenylacrylic acid,40 2-(mercaptomethyl)benzoic
acid,41 and phthalic acid.42 Comparatively, most of SBL
inhibitors are positioned to covalently bind with the active site
methylpropanamido)methyl)boronic Acid (MS01) as a nucleophilic serine, e.g., via the boronic acid group.29,43−47
Dual-Action MBL/SBL Inhibitor. By analyses of crystal With the aim to obtain dual-action inhibitors targeting MBL
7162 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Figure 2. Crystallographic analyses reveal compound MS01 binding modes to VIM-2 MBL and KPC-2 SBL. (A) The mode of MS01 binding as
defined by the mFo − DFc electron density maps observed in the VIM-2:MS01 structure (PDB code 6J8R), indicating that MS01 fits well with the
electron density and chelates to and bridges with the two active site zinc ions. (B) Mode of MS01 binding as defined by the mFo − DFc electron
density maps observed in the KPC-2:MS01 structure (PDB code 6J8Q), indicating that MS01 fits well with the electron density and forms a
covalent bond with the residue Ser70. (C) View from the VIM-2:MS01 structure (PDB code 6J8R) reveals that the inhibitor binds to form
hydrophobic and electrostatic interactions with residues on the L3 and L10 loops (e.g., hydrogen-bonding interactions with Asn233 and Arg228 and
hydrophobic interactions with Phe61). (D) View from the KPC-2:MS01 structure (PDB code 6J8Q) reveals that the inhibitor binds to make
hydrogen-bonding interactions with the residues Asn132, Glu166, Asn170, and Thr237 in the active site.

and SBL enzymes, we came up with a strategy of integrating in their kinetic parameters (Figure S1 and Table S1),
the key pharmacophores of MBL and SBL inhibitors. As an indicating that FC5 was a suitable substrate for these enzymes.
initial attempt, we designed and synthesized the compound Consequently, we determined IC50 values of MS01−MS04
(2′S)-(1-(3′-mercapto-2′-methylpropanamido)methyl)boronic with all the representative MBL and SBL enzymes using FC5.
acid (MS01, Figure 1B and Scheme 1). The (2′S)-3′- Given the differences in the enzyme kinetics, we calculated the
mercapto-2′-methylpropanamido moeity of MS01, as a key inhibition constant (Ki) values from the corresponding IC50
motif of captopril (an old drug used in clinical for more than values using the Cheng−Prusoff equation49 to compare the
30 years), may have potential biocompatibility and achieve inhibitory potency for the compounds (for details see the
broad-spectrum inhibition to multiple MBL enzymes, partic- Experimental Section).
ularly B1 MBL enzymes (e.g., NDM-1 and VIM-2), whereas As anticipated, MS01 displayed inhibitory activity against all
the α-aminoboronic acid moiety of MS01 is expected to target of the tested MBL and SBL enzymes (Table 1 and Figure S2).
the catalytic serine residue of SBL enzymes. Meanwhile, we For the subclass B1 MBLs, MS01 showed Ki values of 0.76 and
synthesized the non-thiol analogues MS02 and MS03 (Table 1 67.13 μM to the widespread VIM-2 and NDM-1, respectively,
and Scheme 1), to test whether thiol is essential for MBL comparable to L-captopril, a well-known MBL inhibitor (Table
inhibition, and the non-boronic acid compound MS04 (Table 1), while the non-thiol compounds MS02 and MS03, bearing a
1 and Scheme 1), bearing a carboxylic acid group instead of boronic acid group, had low inhibition to VIM-2 and NDM-1
boronic acid, to examine the necessity of the boronic acid (Table 1), suggesting the importance of the thiol group (rather
group for SBL inhibition. than boronic acid) for B1 MBL inhibition. Another compound
To investigate the MBL/SBL inhibitory activity of MS01− MS04, which contains a thiol group but no boronic acid,
MS04, we selected a representative panel of clinically relevant displayed potent inhibition to these two B1 MBLs as MS01
MBLs, including VIM-2 (subclass B1), NDM-1 (subclass B1), (Table 1), probably owing to the contributions of both the
Sfh-I (subclass B2), and GOB-18 (subclass B3), and SBLs, thiol and carboxylate group, similar to previously reported
including KPC-2 (class A), TEM-1 (class A), AmpC (class C), inhibitors.36−38 Notably, MS01 can also inhibit subclass B2
and OXA-48 (class D). All of these recombinant enzymes were Sfh-I and B3 GOB-18 with Ki values of 7.34 and 2.23 μM,
expressed and purified from E. coli DH5α and BL21 cultures; respectively (Table 1). The non-thiol compound MS02
the details are described in the Experimental Section. Since exhibited comparable inhibition as MS01 to Sfh-I and GOB-
FC5 has been demonstrated as a robust fluorescent substrate 18; conversely, the thiol compounds MS04 and L-captopril did
for the dizinc B1 MBLs (e.g., VIM-2 and NDM-1),48 we here not show marked inhibition against Sfh-I and GOB-18 (Table
examined whether FC5 could be used as a substrate for activity 1), indicating that the thiol group is likely not essential for
test for other classes of β-lactamases. The enzyme kinetic inhibiting these B2/B3 enzymes.
analyses revealed that all of the tested β-lactamases exhibited For the tested SBLs, MS01 manifested Ki values of 2.70,
substantial hydrolytic activity toward FC5 resulting in 40.97, 6.46, and 70.19 μM, respectively, to class A SBL KPC-2
measurable fluorescent signals, albeit with apparent difference and TEM-1, class C AmpC, and class D OXA-48 (Table 1 and
7163 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Table 2. Inhibitory Potency (Ki, μM) of MS05−MS12, with Modifications of the (S)-3′-Mercapto-2′-methylpropanamido
Moiety, to the MBL and SBL Enzymesa

a
The dose−response curves of compounds inhibiting the selected enzymes are given in Supporting Information Figure S8.

Figure S2). Similarly, the boronic acid compounds MS02 and in the asymmetric unit (ASU) (Table S3). In the VIM-2:MS01
MS03 displayed inhibitory activity against these SBL enzymes, structure, the observed clear Fo − Fc electron density adjacent
whereas structurally similar non-boronic acid compound MS04 to the two zinc ions allow unambiguous modeling of MS01
had no or weak SBL inhibition, suggesting that the boronic (Figures 2A and S3A). KPC-2:MS01 crystallized in space
acid group is likely important for SBL inhibition. These SAR group P321 with four molecules in the ASU, different from
results demonstrated feasibility of integrating the key those previously reported45,51,52 (Table S4); MS01 could also
pharmacophores of MBL and SBL inhibitors to develop be well-modeled into clearly defined electron density in the
broad-spectrum, dual-action inhibitors. active site of KPC-2 (Figures 2B and S3B).
X-ray Structure Determination of the VIM-2:MS01 The VIM-2:MS01 structure reveals that MS01 works via a
and KPC-2:MS01 Complexes. We then conducted crystallo- thiol-participating metal chelating mode of inhibition, similar
graphic analyses for the VIM-2:MS01 and KPC-2:MS01 to that observed for previously reported thiol-based MBL
complexes to investigate the dual inhibition modes of MS01. inhibitors.36−38,53 In VIM-2:MS01, the thiol binds to displace
Details of crystallization and structure determinations are the zinc-bridging nucleophilic hydroxide that is observed in the
described in Experimental Section and Table S2. Cocrystalliza- native VIM-2 active site (PDB code 4BZ3),36 and chelates
tion experiments yielded high-resolution crystal structures of with the two active site zinc ions (2.2−2.3 Å) (Figure 2C),
the VIM-2:MS01 (PDB code 6J8R) and KPC-2:MS01 (PDB demonstrating the importance of the thiol for VIM-2
code 6J8Q) complexes. VIM-2:MS01 crystallized in the inhibition, consistent with the SAR data shown in Table 1.
previously described space group P21221,50 with one molecule The amidocarbonyl group of MS01 is positioned to make
7164 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Scheme 2. Synthetic Routes for 1-Substituted (1-(3′-Mercaptopropanamido)methyl)boronic Acid Derivatives MS05 and
MS13−MS31a

a
(a) Nis(pinacolato)diboron, CuBr, NaOtBu, DMF, Ar, 80 °C, 10 h; (b) (+)-pinanediol or (−)-pinanediol, Et2O, rt, overnight; (c) CH2Cl2, n-
BuLi, ZnCl2, THF, −100 °C to rt, overnight; (d) LiHMDS, THF, −78 °C to t., overnight; HCl/1,4-dioxane, Et2O, −60 °C to rt, 3 h; (e) TBTU,
DIPEA, DMF, −10 °C, 2 h; (f) 2-methylpropylboronic acid, HCl (aq), CH3OH/hexane (1:1), rt overnight. (+)-Pinanediol was used as chiral
auxiliary reagent to generate final (1R)-enantiomers, while (−)-pinanediol was used as chiral auxiliary reagent to generate final (1S)-enantiomers.

hydrogen-bonding interactions with the residue Asn233 (3.0 Å) hydrolytic pathway.43 Moreover, MS01 binding is further
on the L10 loop of VIM-2 (Figure 2C), a conserved residue stabilized by multiple hydrogen-bonding interactions with the
observed in active site of subclass B1 MBLs (e.g., NDM-1 catalytically important residues Ser70 (2.8 Å), Thr237 (2.8 Å),
Asn233, IMP-1 Asn233, and BcII Asn233),36 and its boronic acid Asn170 (2.7 Å), Glu166 (2.7 Å), and Asn132 (2.9 Å) (Figure 2D).
form hydrogen-bonds with Arg228 (3.5 Å) (Figure 2C), which Superimposing structures of KPC-2 with TEM-1 (PDB code
is structurally equivalent to the catalytic important positively 1M40),57 AmpC (PDB code 1FSY),58 and OXA-48 (PDB
charged residue on L10 loop of B1 MBLs, for example, NDM- code 5FAQ)59 revealed that despite differences in their active
1 Lys211, IMP-1 Lys224, and BcII Lys224.36 This may indicate sites, MS01 may have a similar binding mode with these SBL
that MS01 has a similar inhibition mode to subclass B1 MBLs, enzymes (Figure S5).
including NDM-1. By contrast, for B2 MBL Shf-I and B3 MBL The VIM-2:MS01 and KPC-2:MS01 structures not only
GOB-18, MS01 appears to bind via boronic acid (rather than reveal the substrate-competitive, dual-action MBL/SBL
thiol)-involving zinc ion contacts as predicted by molecular inhibition modes but also provide important basis for further
docking analyses (Figure S4), similar to that observed in structural optimization. On superimposing VIM-2:MS01 with
crystal structures of B2 Sfh-I:L-CS319 (a thiol containing other crystal structures of VIM-2 complexed with 3-
compound) (PDB code 5EW0)53 and B2 CphA:D-captopril mercaptopropanamido-containing inhibitors,36−38 we observed
(PDB code 2QDS);54 this distinct inhibition mode of MS01 to that compared to other inhibitors, MS01 only makes weak
B2/B3 MBLs may explain the differences in the SAR trends hydrophobic interactions with Phe61 and Tyr67 on the flexible
(e.g., MS01 vs MS02 and MS01 vs MS04; see Table 1). L3 loop that is important for inhibitor capture as previously
The KPC-2:MS01 structure reveals continuous electron described36−38,50 (Figure S6). Further comparison of KPC-
density connecting the boron atom of MS01 with the terminal 2:MS01 and reported KPC-2 structures45,56,60 revealed that
hydroxyl group of the catalytic Ser70 (Figures 2B and S3B), MS01 did not occupy the subpocket around the residues
demonstrating that MS01 binds with KPC-2 via formation of a Trp105, Ser130, and Thr235 (Figure S7). These results suggested
covalent adduct between its boronic acid and Ser70 (Figure that introduction of a substituent at the 1-position of MS01
2D), consistent with the SAR results in Table 1; this inhibition might be able to improve the potency to VIM-2 and KPC-2 as
mode is similar to that of previously reported boronic acid- well as other MBL/SBL enzymes.
based SBL inhibitors.22,46,47,55,56 The KPC-2:MS01 structure Structure-Based Optimization of (1-(3′-
represents a covalent inhibition mode (i.e., the formed Mercaptopropanamido)methyl)boronic Acid Deriva-
tetrahedral boron atom) that mimics the tetrahedral transition tives. On the basis of the VIM-2:MS01 and KPC-2:MS01
state of substrate binding, which is transiently present on the structures, we performed fragment-growing analyses on the 1-
7165 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

position using the LEADOPT program, a tool for structure-


based lead optimization.61 The results suggested that the
introduced fragments in (R)-configuration might be better for
binding with VIM-2 and KPC-2. According to the synthetic
accessibility, we synthesized a compound MS05, bearing a
benzyl group at 1-position in (R)-enantiomer (Table 2), via
the route shown in Scheme 2. MS05 displayed potent
inhibition to VIM-2 (Ki = 0.36 μM), comparable to MS01
(Ki = 0.76 μM), while it showed slightly less potent inhibition
to KPC-2 (Ki = 6.85 μM) as compared with MS01 (Ki = 2.70
μM) (Table 2 and Figure S8). MS05 also manifested
inhibition to subclass B1 MBL NDM-1, B2 Sfh-I, B3 GOB-
18, and class A SBL TEM-1, class C AmpC, and class D OXA-
48, with Ki values of 3.88, 3.48, 9.03, 15.41, 7.87, and 25.83
μM, respectively; by comparison with MS01, MS05 had
slightly improved potency to NDM-1, TEM-1, and OXA-48
(Table 2 and Figure S8).
We obtained a crystal structure of KPC-2:MS05 (PDB code
6JN3) to 2.2 Å resolution (Tables S2 and S4), in which MS05
could be confidently fitted with the electron density maps in
the KPC-2 active site (Figure S9). Similar to MS01, MS05 was
observed to form a covalent bond with the catalytic residue
Ser70 and make multiple hydrogen bonds with the residues
Ser70 (3.0 Å), Thr237 (2.7 Å), Asn170 (2.6 Å), Glu166 (2.7 Å),
and Asn132 (3.1 Å) (Figures 2B and 3A). The introduced
benzyl group of MS05 bound to the subpocket located
between the L3 and L5 loops and made edge-to-face aromatic
interactions with Trp105 (Figure 3A). By superimposing the
structures of KPC-2:MS05 and KPC-2:MS01, we observed
that the binding of MS05 resulted in substantial rotation of the
side chain of Trp105 to accommodate the introduced benzyl
group, providing evidence for flexibility in the conformation of
Trp105 and suggesting its important role in substrate/inhibitor
capture (Figure 3B and Figure S7).45,56,60
Exploring the Modifications of (2′S)-3′-Mercapto-2′-
methylpropanamide Motif. We then explored modifications
of the (2′S)-3′-mercapto-2′-methylpropanamido moiety, with
fixing the benzyl group at 1-position (Table 2), to examine
whether it is possible to improve the inhibitory activity. The
replacement of the 2′-methyl group of (2′S)-3′-mercapto-2′- Figure 3. Crystallographic analyses reveal MS05 binding mode to
methylpropanamido with relatively larger chemical groups, KPC-2. (A) View from the KPC-2:MS05 structure (PDB code 6JN3)
including the benzyl (MS06), 1,3-dioxoisoindolin-2-yl reveals that the inhibitor binds to form a covalent bond with the
(MS07), and benzylamino (MS08) groups (Scheme 3), had catalytic residue Ser70, makes hydrogen-bonding interactions with the
residues Ser70, Thr237, Asn170, Glu166, and Asn132, and makes edge-to-
no improvement in the inhibitory potency to the tested MBL
face aromatic interactions with Trp105. (B) Comparison of KPC-
and SBL enzymes as compared with MS05 and even led to a 2:MS05 and KPC-2:MS01 (PDB code 6J8Q) reveals evidence for the
decrease in ligand efficiency (Table 2 and Figure S8). In important role of Trp105 in substrate/inhibitor capture.45,56,60
contrast, the removal of the 2′-methyl group (MS09) appeared
to improve the inhibitory activity for subclass B2 Sfh-I (Ki = optimal moiety to achieve broad-spectrum inhibition to the
0.89 μM), B3 GOB-18 (Ki = 1.64 μM) and class C AmpC (Ki representative MBL and SBL enzymes.
= 1.14 μM) but reduce the potency to subclass B1 VIM-2 (Ki Substitutions at 1-Position Improve the Dual-Action
= 4.04 μM) and NDM-1 (Ki = 190.53 μM) (Table 2 and MBL/SBL Inhibitory Activity. With fixing the optimal (2′S)-
Figure S8), compared with MS05. Similarly, MS10, with 2′- 3′-mercapto-2′-methylpropanamide group, we further explored
mercaptoacetamide instead of (2′S)-3′-mercapto-2′-methyl- the influences of substitutions and enantiomers at 1-position to
propanamide, exhibited decreased activity to VIM-2 (Ki = 3.34 the dual-action MBL/SBL inhibition. Compounds MS13−
μM) and NDM-1 (Ki = 165 μM), and comparable activity to MS31 were synthesized via the routes shown in Scheme 2, and
other tested MBL and SBL enzymes as MS05 and MS09 their SARs with the representative MBLs and SBLs are given in
(Table 2 and Figure S8). The non-thiol analogues MS11 and Table 3. Compared with MS05, the (1S)-enantiomer MS13,
MS12 displayed low inhibitory activity to VIM-2 and NDM-1 with a benzyl group at 1-position, showed comparable
and also had decreased inhibitory activity to the tested SBL inhibition to subclass B1 VIM-2 (Ki = 1.04 μM) and NDM-
enzymes (Table 2), similar to that observed for MS02 and 1 (Ki = 16.86 μM), B2 Sfh-I (Ki = 1.00 μM), and B3 GOB-18
MS03 (Table 1). These results indicated that the (2′S)-3′- (Ki = 4.38 μM) (Table 3). In contrast, for the tested SBLs,
mercapto-2′-methylpropanamide moiety was a relatively MS13 showed decreased potency to class A KPC-2/TEM-1
7166 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Scheme 3. Synthesis of MS06−MS12 and MS32−MS33 Modified at (S)-3′-Mercapto-2′-methylpropanamide Motifa

a
(a) TBTU, DIPEA, DMF, −10 °C, 2 h; (b) 2-methylpropylboronic acid, HCl (aq), CH3OH/hexane (1:1), rt overnight.

and class D OXA-48 (Table 3), probably due to the benzyl S10). To investigate the binding mode of MS19 with VIM-2, a
group in (1S)-configuration making certain influences for the high-quality crystal structure of the VIM-2:MS19 complex to
formation of a covalent bond between the boronic acid and 1.60 Å resolution was obtained (Tables S2 and S3). MS19 was
active site catalytic Ser70. The enantiomer pairs MS14 vs well-fitted with the electron density maps found in the VIM-2
MS15, MS16 vs MS17, and MS18 vs MS19 were observed to active site (Figure S11).
have similar inhibition profiles to the tested SBL enzymes as The VIM-2:MS19 structure revealed that similar to MS01,
MS05 and MS13, e.g., the (1R)-enantiomers MS14, MS16, the (2′S)-3′-mercapto-2′-methylpropanamido moiety of MS19
and MS18 showing comparable or slightly better (<5-fold) was positioned to chelate with the two zinc ions and form
potency to KPC-2, TEM-1, and AmpC than the corresponding hydrogen-bonding interactions with Asn233 (3.0 Å) that is a
(1S)-enantiomers MS15, MS17, and MS19 (Table 3 and conserved residue on the L10 loop in subclass B1 MBLs;36,50
Figure S10). Notably, the (1R)-enantiomer MS18, bearing a the boronic acid made hydrogen bonds with the residue Arg228
(benzofuran-3-yl)methyl substituent at 1-position, displayed (2.8 Å) (Figure 4A) that is structurally equivalent to B1 MBLs,
broad-spectrum inhibition to all of the tested MBL/SBL e.g., NDM-1 Lys211, IMP-1 Lys224, and BcII Lys224, which are
enzymes with the exception of class D OXA-48; in particular, it important to recognize β-lactam 2-C carboxylic acid.36,50
manifested nanomolar potent inhibition to subclass B1 VIM-2 Unexpectedly, the structure revealed an unusual binding mode
(Ki = 0.44 μM), B2 Sfh-I (Ki = 0.26 μM), B3 GOB-18 (Ki = of the benzofuran moiety of MS19, i.e., which was positioned
0.13 μM), class A KPC-2 (Ki = 0.61 μM), and class C AmpC to make orthophoric face-to-face π−π stacking interactions
(Ki = 0.11 μM) (Table 3 and Figure S10). Compared with with residue Phe61 on the VIM-2 L3 loop (Figure 4A), partly
KPC-2, TEM-1, and AmpC, OXA-48 bears a distinct active explaining why MS19 had very potent inhibition to VIM-2 (Ki
site,57−59 e.g., with several hydrophic residues (Val120, Leu158, = 0.07 μM). Notably, superimposition of the VIM-2:MS19 and
Ile102, and Trp105) and the lack of a structurally equivalent VIM-2:MS01 structures revealed that the L3 loop of VIM-2
residue to KPC-2/TEM-1 Asn132 and AmpC Asn152 (Figure was positioned away from the active zinc ions (2.8 and 0.8 Å
S5) to form hydrogen bonds with the amido-carbonyl of MS18 movements of the C-α atoms of Phe61 and Tyr67 respectively,
(see subsequent crystallographic analyses for MS22 and Figure 4B) for MS19 binding to VIM-2, providing further
MS23), partly explaining why MS18 does not potently inhibit evidence for the proposal that the L3 loop contributes to the
OXA-48. More interestingly, the (1S)-enantiomer MS19 highly efficient nature of MBL catalysis by capturing the
inhibited the subclass B1 MBLs VIM-2 (Ki = 0.07 μM) and potential substrates and delivering them to the zinc ions for
NDM-1 (Ki = 6.06 μM) more potently (>6-fold) than the hydrolysis.50
(1R)-enantiomer MS18 (with Ki values of 0.44 μM and 37.95 Further comparison of the enantiomeric pairs, with the
μM to VIM-2 and NDM-1, respectively) (Table 3 and Figure phenyl (MS20 vs MS21), para-fluorophenyl (MS22 vs MS23),
7167 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Table 3. Inhibitory Potency (Ki, μM) of 1-Substituted (1-(3′-Mercaptopropanamido)methyl)boronic Acid Derivatives MS13−
MS31 and MS32−MS33 against the Selected MBL and SBL Enzymesa

7168 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Table 3. continued

a
The dose−response curves of compounds inhibiting the selected enzymes are given in Supporting Information Figure S10.

para-methylphenyl (MS24 vs MS25), and naphthyl (MS26 vs compounds were well-modeled into the observed electron
MS27) moiety at 1-position (Table 3), revealed that the (1R)- density maps in the active site (Figures S12 and S13).
enantiomeric compounds MS20, MS22, and MS24 had The structures revealed that both MS22 and MS23 bound to
comparable inhibition as the corresponding (1S)-enantiomeric form a covalent bond with the catalytic residue Ser70 and make
compounds MS21, MS23, and MS25 to the tested B1/B2/B3 multiple hydrogen bonds with the active site residues Ser70,
MBL enzymes; (1R)-enantiomer MS26, with a rigid substitent Thr237, Asn170, Glu166, and Asn132 (Figure 5A−C), similar to
of naphthyl, inhibited subclass B1 VIM-2 and NDM-1 more that observed for MS01 and MS05 (Figures 2D and 3A).
potently than (1S)-enantiomer MS27, while MS26 had Similar to MS05, MS22 and MS23 were positioned to occupy
comparable potency to B2 Sfh-I and B3 GOB-18 as MS27 the subpocket between the L3 and L5 loops and made
(Table 3 and Figure S10). By comparison, for the SBL hydrophobic interactions with the flexible residue Trp105
enzymes KPC-2, TEM-1, and AmpC, these (1R)-enantiomeric (Figure 5A,B). Notably, in the KPC-2:MS22 structure, the
compounds generally displayed more potent inhibitory amido group adopts a trans-conformation and makes hydro-
activities compared with their corresponding (1S)-enantio- gen-bonding interactions with Thr237 (2.9 Å) and Asn132 (3.0
meric compounds, similar to that observed for other Å) (Figure 5A), same as that observed in the structure of (1R)-
enantiomeric pairs in Table 3. Consequently, we conducted enantiomer MS05 complexed with KPC-2 (Figures 3A and
crystallographic analyses for KPC-2 in complex with the (1R)- S14). By contrast, the amido group of the (1S)-enantiomer
enantiomer MS22 and (1S)-enantiomer MS23, with the aim to MS23 was unexpectedly observed to bind in a cis-conformation
investigate the difference in their binding modes. The crystal and only form hydrogen bonds with Asn132 (2.9 and 3.5 Å)
structures of KPC-2:MS22 (PDB code 6JN4) and KPC- (Figure 5B). These differences may, in part, explain why (1R)-
2:MS23 (PDB code 6JN5) were obtained and solved to 1.90 enantiomers inhibit the SBL enzymes more potently than (1S)-
and 1.97 Å resolution, respectively (Table S4); both enantiomers. Additionally, these structures provided important
7169 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

structural basis for developing chiral α-aminoboronic acid-


based SBL inhibitors.
We then compared the MBL/SBL inhibitory activity for
compounds MS28−MS31, which bear small moieties at 1-
position (Table 3). We observed that for the tested MBL
enzymes, the (1R)-enantiomeric compounds MS28 and MS30
have comparable activity as the corresponding (1S)-enantio-
meric compounds MS29 and MS31, with the exception of
MS29; MS29 has relatively weak inhibition (5-fold lower) to
subclass B2 Shf-I and B3 GOB-18. By comparison with MS18,
compounds MS28−MS31 mostly had less potent inhibition to
the SBL enzymes. Interestingly, to class A KPC-2/TEM-1 and
class C AmpC, the (1R)-enantiomer MS28 was more potent
than (1S)-enantiomer MS29, whereas (1R)-enantiomer MS30
had less inhibitory activity than (1S)-enantiomer MS31 (Table
3 and Figure S10). Comparison of the dual-action inhibition
potencies of MS32/MS33 with MS19 further demonstrated
the importance of the (2′S)-3′-mercapto-2′-methylpropanami-
do group for achieving broad-spectrum MBL inhibition,
particularly for the subclass B1 VIM-2 and NDM-1.
The overall SAR studies of various thiol- and/or boronic
acid-containing compounds with the representative MBL/SBL
enzymes (Table 1−3 and Figure S15) revealed the substantial
influences of the thiol moiety and substitutions/enantiomers at
1-position on the dual-action MBL/SBL inhibition. To
subclass B1 MBLs, the (2′S)-3′-mercapto-2′-methylpropana-
mide moiety was demonstrated to be an optimal MBP for
targeting VIM-2 and NDM-1, similar to that previously
reported,36−38 while the inhibitory activities were synthetically
affected by the 1-position substituents and enantiomeric
conformations (Table 3). By contrast, for subclass B2 Shf-I
and B3 GOB-18, no significant differences were observed
between the tested enantiomer pairs (Table 3), implying that
the inhibitors may have different inhibition modes to subclass
Figure 4. Crystallographic analyses reveal an unexpected mode of B1 and B2/B3 MBLs. To class A KPC-2/TEM-1 and class C
MS19 binding to VIM-2. (A) Close-up view from the VIM-2:MS19 AmpC, the (1R)-enantiomeric compounds, in most cases,
structure (PDB code 6JN6) reveals that the inhibitor binds to form displayed better activities than the corresponding (1S)-
hydrophobic and electrostatic interactions with residues on the L3 enantiomeric compounds (Table 3 and Figure S15), possibly
and L10 loops (e.g., hydrogen-bonding interactions with Asn233 and
Arg228 and, unexpectedly, π−π stacking interactions with Phe61). (B)
owing to the difference in their amino binding conformations
Superimposition of VIM-2:MS19 and VIM-2:MS01 (PDB code as observed by crystallographic analyses (Figure 5). Yet, almost
6J8R) reveals evidence for flexibility in the conformation of Phe61 and all of the tested compounds had no or weak inhibition to the
Tyr67 on the L3 loop, suggesting they may play important roles in structurally different class D OXA-48 (Figure S5). Notably, the
capturing substrates and delivering them to the zinc ions for crystallographic analyses revealed that the flexible loops in the
hydrolysis.50 active sites of MBL and SBL enzymes played important roles in

Figure 5. Crystallographic analyses reveal similar binding modes of (1R)-enantiomer MS22 and (1S)-enantiomer MS23 with KPC-2. (A) Close-up
view from a structure of MS22 complexed with KPC-2 (PDB code 6JN4). (B) Close-up view from a structure of MS23 complexed with KPC-2
(PDB code 6JN5). (C) Superimposition of KPC-2:MS22 and KPC-2:MS23 reveals MS22 and MS23 bind in similar modes involving covalent
bonding and hydrogen-bonding interactions. Note that unlike MS22, MS23 is bound with cis-amide conformation.

7170 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Figure 6. In vitro selectivity profile of the representative inhibitors across serine hydrolases in E. coli cells (A, B) and human 293T cells (C, D) as
determined by competitive activity-based protein profiling using broad-spectrum probe TAMRA-FP. (A) E. coli cells or (C) human 293T cells were
lysed and treated with inhibitors MS01, MS05, MS06, MS18, MS19, MS20, and MS21 at 50 μM for 30 min, followed by the treatment of activity-
based probe TAMRA-FP (750 nM, 20 min). The heat-maps (B) and (D) showing the quantification of data of gels in (A) and (C), respectively;
data represent average values, n = 3 per group (Table S8).

capturing various inhibitors, which may be one of the critical inhibition of 63.2% and 44.6% to hACE-2 at 100 μM and 50
aspects to understand the relatively complicated multitarget μM, respectively (Table S7 and Figure S17).
SAR data. More importantly, the SAR studies identified new We also examined the selectivity of the representative
more potent dual-action inhibitors compared with the starting inhibitors MS01, MS05, MS06, MS13, MS18, MS19, MS20,
compound MS01. For example, MS18 manifested potent and MS21 across serine hydrolases in E. coli cells and human
nanmolar inhibition to several tested MBL and SBL enzymes, HEK293T cells by competitive activity-based protein profiling
including the clinically widespread carbapenemases subclass B1 using TAMRA-FP, since they bear an α-aminoboronic acid
VIM-2 (Ki = 0.44 μM) and class A KPC-2 (Ki = 0.61 μM); moiety. TAMRA-FP is a well-characterized broad-spectrum
MS18 was observed to be a reversible inhibitor to both VIM-2 serine hydrolase activity-based probe, which provides selectiv-
and KPC-2, which had a relatively slow dissociation rate for ity profile assays across a broad array of serine hydrolases. As
KPC-2 compared with other tested inhibitors, including shown in Figure 6, the tested inhibitors have weak inhibitory
tazobactam (Figure S16). activity against most of the labeled serine hydrolases in E. coli
Selectivity to Metalloenzyme ACE-2 and Serine and HEK293T cells. Particularly, MS01 showed high
Hydrolyases. Since these dual-action inhibitors have the selectivity with no significant off-targets (enzyme inhibition
(2′S)-3′-mercapto-2′-methylpropanamido moiety, which is a are all below 50%, Table S8) identified in both E. coli and
key scaffold of captopril, an angiotensin-converting enzyme HEK293T cell lysates (Figure 6 and Table S8). Of note, the
(ACE) inhibitor, we tested the dual-action inhibitors MS01, control compound AN2728, an FDA-approved boronate-
MS05, MS06, and MS13−MS33 to examine their selectivity to containing drug, exhibited less selectivity compared with
the commercially available human ACE-2 (hACE-2) enzyme. most of the tested inhibitors with several off-targets (Figure
With the exception of MS06, all these compounds have no 6 and Table S8). These results indicate that the dual-action
apparent inhibitory activity to hACE-2 at the concentration of inhibitors are selective among serine hydrolases in E. coli and
100 μM (Figure S17 and Table S7); MS06 displayed HEK293T cells labeled by TAMRA-FP.
7171 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

Table 4. Susceptibility Testing Data of Clinical Isolates Harboring NDM-1, KPC-2, or AmpC to Meropenem alone and in
Combination with the Inhibitors MS01, MS05, MS13, MS18, and MS19 at 50 μM
meropenem MIC (μg·mL−1)
bacterial strain characterized β-lactamase DMSO MS01 MS05 MS13 MS18 MS19 L-cap Tazo
E. coli BAA-2452 blaNDM‑1 32 16 16 32 32 16 8 16
K. pneumoniae 13249 blaNDM‑1 64 64 64 64 64 32 32 64
E. coli BAA-2340 blaKPC‑2 4 0.5 2 2 1 2 4 2
E. coli 11119 blaKPC‑2 16 4 16 16 8 8 16 8
K. pneumoniae BAA-1705 blaKPC‑2 16 8 16 16 8 16 16 16
K. pneumoniae 5846 blaKPC‑2 16 4 8 16 8 8 8 8
K. pneumoniae C660 blaAmpC 32 8 16 16 8 8 32 16

Inhibitors Potentiate Meropenem Activity against cupric bromide (CuBr), sodium tert-butoxide (NaOtBu) in
Clinical Isolates Harboring MBL or SBL Enzymes. We DMF to obtain boronic acid pinacol esters 9a−d,65 which were
then tested the activity of the inhibitors MS01, MS05, MS13, then subjected to transesterification reaction with
MS18, and MS19 (L-captopril and tazobactam as control (1S,2S,3R,5S)-(+)-2,3-pinanediol ((+)-pinanediol) in diethyl
compounds) against seven multidrug-resistant Gram-negative ether (Et2O) to generate boronic acid (+)-pinanediol esters
clinical isolates: E. coli BAA-2452 (blaNDM‑1), K. pneumoniae 11a−d. Compounds 11e−j were produced by esterification
13249 (blaNDM‑1), E. coli BAA-2340 (blaKPC‑2), E. coli 11119 reaction from diverse boronic acid 10e−j with (+)-pinanediol
(blaKPC‑2), K. pneumoniae BAA-1705 (blaKPC‑2), K. pneumoniae in Et2O. Subsequent Matteson reaction of 11a−j with
5846 (blaKPC‑2), and K. pneumoniae C660 (blaAmpC). The dichloromethane (CH2Cl2) and n-butyllithium (n-BuLi) in
minimum inhibitory concentration (MIC) values of merope- THF at −100 °C afforded 12a−j with (S)-configuration,66−68
nem against these bacterial strains were determined with or followed by SN2 reaction with LiHMDS in THF at −78 °C,
without the inhibitors. The results showed that MS01, MS05, generating 13a−j with inverted (R)-configuration (obtained as
MS13, MS18, and MS19 were able to potentiate the efficacy of hydrochloride salts).69,70 Next, 13a−j were reacted with (S)-
meropenem against the clinically relevant E. coli and K. (3-acetylsulfanyl-2-methyl)propionic acid to form 14a−j,
pneumonia strains, particularly harboring KPC-2 and AmpC, as which were then subjected to deprotection reaction to yield
evidenced by the substantial reductions of the MIC values (≤8 (1R)-1-substituted (1-(3′-mercaptopropanamido)methyl)-
μg·ml−1), which are better than or comparable to L-captopril boronic acid derivatives (including MS05, MS14, MS16,
and tazobactam (Table 4); these compounds did not cause MS18, MS20, MS22, MS24, MS26, MS28, and MS30).
marked inhibition of bacterial growth at 64 μg·mL−1, indicating Followed the same synthetic routes as described above, the
that they exhibited synergistic antibacterial effects when used corresponding (1S)-enantiomers (including MS13, MS15,
along with meropenem. Besides, none of these inhibitors MS17, MS19, MS21, MS23, MS25, MS27, MS29, and
showed cytotoxicity in human HEK293T and MCF-7 cells MS31) were obtained using (1R,2R,3S,5R)-(−)-2,3-pinanediol
when treated at concentrations up to 100 μM (Table S9 and ((−)-pinanediol) instead of (+)-pinanediol.
Figure S18).


The intermediate (R)-(1-amino-2-phenylethyl)boronic acid
(1S,2S,3R,5S)-(+)-2,3-pinanediol ester 13a was subjected to a
CHEMISTRY coupling reaction with various carboxylic acids to provide
As shown in Scheme 1A, triisopropyl borate (1) was prestirred 15a−g, followed by the deprotection reactions to give MS06−
with dibromomethane and n-butyllithium (n-BuLi) in MS12 (Scheme 3A). Similarly, (S)-(1-amino-2-(3′-
tetrahydrofuran (THF) at −78 °C for 2 h, and the mixture benzofuran)ethyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pinane-
was then reacted with methanesulfonic acid and pinacol at 0 diol ester 13d′ reacted with (3-acetylsulfanyl)propionic acid or
°C to afford compound 2.62 A nucleophilic substitution (2-acetylsulfanyl)acetic acid to form 16a or 16b, which then
reaction was then carried out from bromide 2 which was deprotected to give MS32 or MS33 (Scheme 3B).


stirred in THF with lithium bis(trimethylsilyl)amide
(LiHMDS) at −78 °C to obtain bis-silylamine intermediate CONCLUSION
3. The amine hydrochloride 4 was synthesized by deprotection
from 3 using 4 N hydrochloride/1,4-dioxane solution and Overall, the results revealed the potential of a pharmacophore
diethyl ether at −60 °C.63 Compound 4 was then subjected to fusion strategy targeting the key active site features of MBL
a coupling reaction with diverse carboxylic acid using O- and SBL enzymes for the identification of dual-action
(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoro- inhibitors. The exquisite fusion of (2′S)-3′-mercapto-2′-
borate (TBTU), N,N-diisopropylethylamine (DIPEA), and methylpropanamido with acyclic α-aminoboronic acid led to
N,N-dimethylformamide (DMF) at −10 °C to generate 5a− the identification of new dual-action MBL/SBL inhibitors. The
c, 64 followed by the deprotection reaction using 2- crystallographic analyses demonstrated the hypothetical MBL/
methylpropylboronic acid and 3 N hydrochloric acid solution SBL inhibition modes, i.e., (2′S)-3′-mercapto-2′-methylpropa-
in methanol and hexane to give MS01−MS03. Similarly, namido positioned to chelate with active site zinc ions of at
treatment of glycine ethyl ester hydrochloride (6) with (S)-(3- least subclass B1 MBL enzymes, and α-aminoboronic acid
acetylsulfanyl-2-methyl)propionic acid in DMF gave 7, which covalently bound with the catalytic residue Ser70 of SBL
was then subjected to deprotection reaction to yield MS04 enzymes. These results may prompt a fresh start for using
(Scheme 1B). pharmacophore fusion to obtain new dual-action inhibitor
As outlined in Scheme 2, various bromides 8a−d were chemotypes targeting MBL and SBL enzymes as well as other
carried out with borylation reaction by bis(pinacolato)diboron, structurally and mechanistically distinct enzymes.
7172 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

The structure-guided optimization studies yielded inter- with water followed by brine. The organic layer was collected, dried
preted multitarget structure−activity relationships and resulted over Na2SO4, and concentrated under reduced pressure. The resulting
in new more potent, broad-spectrum, dual-action MBL/SBL residue was purified by silica gel chromatography with dichloro-
inhibitors, e.g., MS18. The crystal structures revealed insights methane/methanol combinations as eluent, giving rise to the amide
compounds.
into the binding modes of these inhibitors with VIM-2 and (1-Bromomethyl)boronic Acid Pinacol Ester (2). To a solution of
KPC-2, providing important structural basis for developing triisopropyl borate (10 g, 55 mmol) and dibromomethane (4.2 mL,
dual-action MBL/SBL inhibitors. The structural results also 60 mmol) in dried THF (80 mL) at −78 °C under argon, n-BuLi (20
provided evidence for the flexibility of the loops near active mL, 50 mmol; 2.5 N in hexane) was added dropwise, and the mixture
sites of MBL and SBL enzymes, suggesting their important was stirred at the same temperature for 2 h. The reaction mixture was
roles in substrate/inhibitor capture, which hence need to be then warmed to 0 °C, methanesulfonic acid (3.2 mL, 50 mmol) was
considered in inhibitor design and development. added, and the mixture was stirred at room temperature for 1 h.
Selectivity studies indicated that the inhibitors had no Subsequently, the obtained mixture was cooled to 0 °C, pinacol (5.9
apparent inhibition to human ACE-2 and showed selectivity g, 50 mmol) was added, and the mixture was stirred at room
temperature for another 1 h. The solvents were evaporated under
across serine hydrolyases in E. coli and HEK293T cells labeled
reduced pressure from the reaction mixture and then the obtained
by TAMRA-FP. The inhibitors could reduce MICs of residue was distilled under reduced pressure (62−63 °C, 10 mmHg),
meropenem against MBL- or SBL-positive K. pneumoniae obtaining the product 2 7.5 g in 68% yield as colorless oil. 1H NMR
and E. coli clinical isolates without apparent cytotoxicity, (400 MHz, CDCl3): δ 1.29 (s, 12H), 2.59 (s, 2H).
indicating that they are suitable for further structural (Bis(trimethylsilyl)aminomethyl)boronic Acid Pinacol Ester (3).
optimization, possibly along with membrane permeability To a solution of (1-bromomethyl)boronic acid pinacol ester (2, 4.0 g,
and intracellular accumulation, to obtain clinically useful 18.1 mmol) in dried THF (36 mL) was added LiHMDS (22.6 mL,
dual-action MBL/SBL inhibitors. 22.6 mmol; 1 N in THF) dropwise under argon atmosphere at −78


°C. The reaction mixture was allowed to warm to room temperature
and stirred overnight. The resulting solution was concentrated under
EXPERIMENTAL SECTION reduced pressure and reconstituted in hexane (50 mL) and filtered
Chemistry. Unless otherwise noted, all reactions were carried out through Celite. The organic layer was dried with Na2SO4 and
under anhydrous conditions, argon atmosphere, and dry solvents. evaporated to give crude N-bis(trimethylsilyl)amine product 3 as
Reactions were monitored by thin-layer chromatography (TLC) and light-brown oil. The product was used for the next step without
were visualized using UV light or phosphomolybdic acid (10% in purification.
ethanol). TLC characterization was performed with precoated silica (1-Aminomethyl)boronic Acid Pinacol Ester (4). The crude light-
gel GF254 (0.2 mm). The purification of products by silica gel brown oil (3, 9 mmol) was dissolved in diethyl ether (25 mL) and
column chromatography was carried out on silica gel (100−200 cooled down to −60 °C. To the mixture was added 4 N HCl/1,4-
mesh). All organic extracts were dried over MgSO4, and solvents were dioxane solution (6.75 mL, 27 mmol), and the reaction was allowed
removed with a rotary evaporator, followed by high vacuum oil pump to stir for 3 h. The hydrochloride salt crashed out of solution and was
pressure. NMR spectra were taken on a Varian INOVA 400 or 600 filtered. The final product was washed three times with cold diethyl
MHz (Varian, Palo Alto, CA, USA). Chemical shifts were expressed in ether, resulting in a white solid with 1.5 g in 88% yield. 1H NMR (400
parts per million (δ, ppm), with tetramethylsilane (TMS) functioning MHz, DMSO-d6): δ 1.24 (s, 12H), 2.36 (q, J = 7.6 Hz, 2H), 7.83 (br,
as the internal standard, and coupling constants (J) were expressed in 3H).
Hz. The following abbreviations were used to identify the multi- (S)-(1-(3′-Acetylsulfanyl-2′-methylpropanamido)methyl)boronic
plicities: s = singlet, d = doublet, dd = doublet of doublets, dt = Acid Pinacol Ester (5a). Followed general method using (1-
doublet of triplets, t = triplet, q = quartet, m = multiplet, br = broad, aminomethyl)boronic acid pinacol ester (4) as amine and (S)-(3-
and all combinations thereof can be explained by their integral parts. acetylsulfanyl-2-methyl)propionic acid as carboxylic acid. Colorless
High resolution mass spectroscopy (HRMS) data of the products oil, 212 mg in 74% yield. 1H NMR (400 MHz, CDCl3): δ 1.23 (d,
were collected on a SCIEX X500 QTOF mass spectrometer 3H, J = 6.8 Hz), 1.27 (s, 12H), 2.10 (s, 3H), 2.22 (s, 2H), 2.62 (m,
instrument. High performance liquid chromatography (HPLC) 1H), 2.78 (m, 1H), 3.00−3.15 (m, 1H), 7.66 (br, 1H).
analyses were run on a Agilent 1200 HPLC analytical system (1-(n-Butyramido)methyl)boronic Acid Pinacol Ester (5b).
(monitoring at a wavelength of 210 or 254 nm) with a Amethyst C18- Followed general method using (1-aminomethyl)boronic acid pinacol
H column (4.6 mm × 250 mm, 5 μm) and mixtures of methanol and ester (4) as amine and n-butyric acid as carboxylic acid. Colorless oil,
H2O containing 0.03% triethylamine and 0.05% trifluoacetic acid as 96 mg in 42% yield. 1H NMR (400 MHz, CDCl3): δ 0.86 (t, 3H, J =
eluent. All final tested compounds were >95% pure by HPLC 7.2 Hz), 1.23 (s, 12H), 1.53−1.63 (m, 2H), 2.18−2.24 (m, 2H), 2.29
analyses. Commercial reagents were from Best-reagent or Astatech (s, 2H).
Chemical Technology Co, Ltd. All reagents were used without further (1-(n-Propanamido)methyl)boronic Acid Pinacol Ester (5c).
purification. Followed general method using (1-aminomethyl)boronic acid pinacol
Synthesis. Boronic Acid Pinanediol Ester. The key inter- ester (4) as amine and n-propionic acid as carboxylic acid. Colorless
mediate (R)-(1-amino-2-phenylethyl)boronic acid (1S,2S,3R,5S)- oil, 98 mg in 46% yield. 1H NMR (400 MHz, CDCl3): δ 1.11 (t, 3H, J
(+)-2,3-pinanediol ester (13a) was synthesized as described in = 7.2 Hz), 1.23 (s, 12H), 2.34 (q, 2H, J = 7.2 Hz), 2.64 (s, 2H).
Scheme 2 from benzyl bromide (8a) as starting material. 1- (S)-2-(3′-Acetylsulfanyl-2′-methylpropanamido)acetic Acid Ethyl
Substituted amine intermediates were generated by similar synthetic Ester (7). Followed general method using glycine ethyl ester
method from corresponding starting material using either hydrochloride (6) as amine and (S)-(3-acetylsulfanyl-2-methyl)-
(1S,2S,3R,5S)-(+)-2,3-pinanediol ester or (1R,2R,3S,5R)-(−)-2,3- propionic acid as carboxylic acid. Colorless oil, 150 mg in 61%
pinanediol ester as chiral auxiliary reagent. The products were yield. 1H NMR (400 MHz, CDCl3): δ 1.21−1.31 (m, 6H), 2.33 (s,
obtained as hydrochloride salts.67,68 3H), 2.47−2.56 (m, 1H), 2.95 (dd, 1H, J = 13.6, 6.4 Hz), 3.11 (dd,
General Method of Coupling Reaction for Synthesis of 1H, J = 13.6, 7.6 Hz), 4.00−4.05 (m, 2H), 4.21 (q, 2H, J = 7.2 Hz),
Amide Compounds (5a−c, 7, 14a−j, 14a′−j′, 15a−g, 16a, 6.20 (s, 1H).
16b). TBTU (385 mg, 1.5 mmol) was added into a solution of amine (1-Phenylmethyl)boronic Acid Pinacol Ester (9a). To a solution of
(1 mmol), carboxylic acid (1 mmol), and DIPEA (387 mg, 3 mmol) benzyl bromide (8a, 2.0 g, 11.69 mmol) in degassed DMF (40 mL)
in DMF (10 mL) at −10 °C. The reaction solution was stirred at the were added bis(pinacolato)diboron (4.45 g, 17.54 mmol), CuBr (340
same temperature until the TLC indicates complete disappearance of mg, 2.34 mmol), and NaOtBu (1.69 g, 17.54 mmol). The solution
amine. Dilute the reaction mass with ethyl acetate (30 mL), and wash was stirred under argon atmosphere at 80 °C for 10 h, and the

7173 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

contents of the flask were cooled to room temperature. The reaction boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (13a) as amine
mixture was filtered out, and the organic layer was extracted with ethyl and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as carboxylic acid.
acetate (50 mL) and washed with water (20 mL × 2) followed by White solid, 226 mg in 51% yield. 1H NMR (600 MHz, CDCl3): δ
brine (20 mL × 2). The organic layer was collected, dried over 0.86 (s, 3H), 1.18 (d, 3H, J = 7.2 Hz), 1.28 (s, 3H), 1.36 (d, 1H, J =
Na2SO4, and concentrated under reduced pressure. The resulting 6.0 Hz), 1.38 (s, 3H), 1.83−1.88 (m, 2H), 2.01 (t, 1H, J = 5.4 Hz),
residue was purified by silica gel chromatography with hexane/ethyl 2.11−2.15 (m, 1H), 2.30 (s, 3H), 2.34 (m, 1H), 2.47 (q, 1H, J = 6.6
acetate (40:1) combinations as eluent, giving rise to the product 9a Hz), 2.74−2.78 (m, 1H), 2.95−3.00 (m 2H), 3.03−3.07 (m, 2H),
1.78 g in 70% yield as colorless oil. 1H NMR (400 MHz, CDCl3): δ 4.29 (d, 1H, J = 7.8 Hz), 6.35 (s, 1H), 7.19−7.24 (m, 3H), 7.26−7.33
1.23 (s, 12H), 2.29 (s, 2H), 7.11 (t, 1H, J = 7.2 Hz), 7.16−7.19 (m, (m, 2H). 13C NMR (100 MHz, CDCl3): δ 16.98, 23.91, 26.29, 27.07,
2H), 7.21−7.25 (m, 2H). 28.68, 30.45, 31.64, 36.01, 37.03, 37.89, 38.36, 39.65, 51.89, 76.78,
(1-Phenylmethyl)boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol 84.09, 126.69, 128.44 (2C), 128.75 (2C), 140.17, 177.31, 195.02.
Ester (11a). To a solution of (1-phenylmethyl)boronic acid pinacol [(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2-
ester (9a, 1.0 g, 4.58 mmol) in diethyl ether (20 mL) was added phenylethyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol Ester
(1S,2S,3R,5S)-(+)-2,3-pinanediol ester (859 mg, 5.04 mmol). The (14a′). Followed general method using (S)-(1-amino-2-phenylethyl)-
reaction mixture was stirred at room temperature overnight and then boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester (13a′) as amine
washed with water (20 mL × 2) followed by brine (20 mL × 2). The and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as carboxylic acid.
organic layer was collected, dried over Na2SO4, and concentrated White solid, 275 mg in 62% yield. 1H NMR (600 MHz, CDCl3): δ
under reduced pressure. The resulting residue was purified by silica 0.83 (s, 3H), 1.16 (d, 3H, J = 6.6 Hz), 1.25 (s, 3H), 1.33 (d, 1H, J =
gel chromatography with hexane/ethyl acetate (50:1) combinations as 6.0 Hz), 1.35 (s, 3H), 1.80−1.86 (m, 2H), 1.98 (t, 1H, J = 5.4 Hz),
eluent, giving rise to the product 11a 1.13 g in 91% yield as colorless 2.08−2.12 (m, 1H), 2.28 (s, 3H), 2.29−2.32 (m, 1H), 2.44 (q, 1H, J
oil. 1H NMR (400 MHz, CDCl3): δ 0.82 (s, 3H), 1.06 (d, 1H, J = = 6.6 Hz), 2.74 (dd, 1H, J = 13.8, 10.8 Hz), 2.96 (dd, 2H, J = 13.8, 5.4
10.8 Hz), 1.27 (s, 3H), 1.38 (s, 3H), 1.79−1.84 (m, 1H), 1.85−1.90 Hz), 3.02 (dd, 2H, J = 13.8, 7.8 Hz), 4.26 (d, 1H, J = 7.8 Hz), 6.33 (s,
(m, 1H), 2.04 (t, 1H, J = 5.6 Hz), 2.14−2.21 (m, 1H), 2.26−2.33 (m, 1H), 7.16−7.20 (m, 3H), 7.25−7.28 (m, 2H). 13C NMR (150 MHz,
1H), 2.34 (s, 2H), 4.27 (dd, 1H, J = 8.8, 2.0 Hz), 7.09−7.14 (m, 1H), CDCl3): δ 16.65, 24.06, 26.38, 27.19, 28.79, 30.47, 32.04, 36.12,
7.18−7.21 (m, 2H), 7.22−7.26 (m, 2H). 37.17, 38.03, 38.69, 39.80, 51.87, 76.93, 84.22, 126.16, 128.43 (2C),
(S)-(1-Chloro-2-phenylethyl)boronic Acid (1S,2S,3R,5S)-(+)-2,3- 128.88 (2C), 140.23, 176.96, 195.59.
Pinanediol Ester (12a). In a three-neck flask equipped with [(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2-(4′-
thermometer, a solution of CH2Cl2 (0.8 mL) in freshly distilled fluorophenyl)ethyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol
THF (10 mL) was cooled to −100 °C (in ethanol/liquid nitrogen Ester (14b). Followed general method using (R)-(1-amino-2-(4′-
bath) under argon atmosphere. n-BuLi (2.5 mL, 6.25 mmol; 2.5 N in fluorophenyl)ethyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol
hexane) was added via syringe by running it down the cold wall of the ester (13b) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic
reaction flask over a period of 20 min. White precipitate of LiCHCl2 acid as carboxylic acid. White solid, 118 mg in 25% yield. 1H NMR
was formed, and after 20 min of stirring, cold solution of 1- (400 MHz, CDCl3): δ 0.83 (s, 3H), 1.20 (d, 3H, J = 6.8 Hz), 1.24 (m,
(phenylmethyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester 4H), 1.35 (s, 3H), 1.78−1.88 (m, 2H), 1.98 (t, 1H, J = 5.2 Hz),
(11a, 1.13 g, 4.18 mmol) in 5 mL of dry THF was added via syringe 2.06−2.11 (m, 1H), 2.28 (m, 4H), 2.43−2.50 (m, 1H), 2.68−2.75
by running it down the cold wall of the reaction flask over a period of (m, 1H), 2.89−2.96 (m, 2H), 3.01−3.07 (m, 2H), 4.25 (d, 1H, J =
15 min. After 20 min of stirring at −100 °C, ZnCl2 solution (4.2 mL, 8.4 Hz), 6.41 (s, 1H), 6.93−6.98 (m, 2H), 7.12−7.17 (m, 2H). 13C
4.2 mmol; 1.0 N in THF) was added via syringe by running it down NMR (100 MHz, CDCl3): δ 16.06, 23.12, 25.38, 26.24, 27.87, 29.55,
the cold wall of the reaction flask over a period of 10 min. The 30.92, 35.08, 35.33, 37.12, 37.87, 38.82, 50.85, 76.11, 83.48, 114.15,
solution was stirred overnight, and the temperature was allowed to 114.36, 129.34, 129.41, 134.78, 159.31, 176.21, 194.87.
rise to room temperature. The reaction mixture was evaporated and [(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2-(4′-
redissolved in hexane. The organic phase was washed with satd fluorophenyl)ethyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol
NH4Cl (20 mL × 2), water (20 mL), and brine (20 mL), then dried Ester (14b′). Followed general method using (S)-(1-amino-2-(4′-
and concentrated to give about 70% crude product as colorless oil. fluorophenyl)ethyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol
The product was used for the next step without purification. 1H NMR ester (13b′) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic
(400 MHz, CDCl3): δ 0.83 (s, 3H), 1.06 (d, 1H, J = 11.2 Hz), 1.28 acid as carboxylic acid. White solid, 106 mg in 23% yield. 1H NMR
(s, 3H), 1.34 (s, 3H), 1.83−1.91 (m, 2H), 2.06 (t, 1H, J = 5.6 Hz), (400 MHz, CDCl3): δ 0.85 (s, 3H), 1.19 (d, 3H, J = 6.8 Hz), 1.27 (m,
2.14−2.22 (m, 1H), 2.30−2.36 (m, 1H), 3.10 (dd, 1H, J = 14.0, 8.8 4H), 1.37 (s, 3H), 1.80−1.88 (m, 2H), 1.98−2.02 (m, 1H), 2.10−
Hz), 3.21 (dd, 1H, J = 14.0, 7.6 Hz), 3.66 (t, 1H, J = 8.0 Hz), 4.35 (d, 2.16 (m, 1H), 2.31 (m, 4H), 2.41−2.50 (m, 1H), 2.72−2.78 (m, 1H),
1H, J = 9.2 Hz), 7.20−7.25 (m, 2H), 7.26−7.30 (m, 3H). 2.93−2.99 (m, 2H), 3.02−3.09 (m, 2H), 4.28 (d, 1H, J = 8.4 Hz),
(R)-(1-Amino-2-phenylethyl)boronic Acid (1S,2S,3R,5S)-(+)-2,3- 6.20 (s, 1H), 6.95−7.00 (m, 2H), 7.15−7.19 (m, 2H). 13C NMR (100
Pinanediol Ester (13a). To a solution of crude (S)-1-chloro-2- MHz, CDCl3): δ 16.79, 24.14, 26.44, 27.26, 28.83, 30.60, 32.20,
(phenylethyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester 36.09, 36.40, 38.15, 39.07, 39.84, 51.87, 77.14, 84.62, 115.19, 115.40,
(12a, 4.0 mmol) in dried THF (10 mL) was added LiHMDS (4.0 130.37, 130.45, 135.82, 160.35, 176.99, 195.79.
mL, 4 mmol; 1 N in THF) dropwise under argon atmosphere at −78 [(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2-(4′-
°C. The reaction mixture was stirred overnight and allowed to warm methoxyphenyl)ethyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinane-
to room temperature. The resulting solution was concentrated under diol Ester (14c). Followed general method using (R)-(1-amino-2-
reduced pressure and reconstituted in hexane (25 mL) and filtered (4′-methoxyphenyl)ethyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinane-
through Celite. The organic layer was dried with Na2SO4 and diol ester (13c) as amine and (S)-(3-acetylsulfanyl-2-methyl)-
evaporated to give crude N-bis(trimethylsilyl)amine product as light- propionic acid as carboxylic acid. White solid, 238 mg in 50% yield.
brown oil. The crude light-brown oil was dissolved in diethyl ether
1
H NMR (400 MHz, CDCl3): δ 0.86 (s, 3H), 1.21 (d, 3H, J = 7.2
(15 mL) and cooled down to −60 °C. To the mixture was added 4 N Hz), 1.28 (s, 3H), 1.33 (d, 1H, J = 10.4 Hz), 1.38 (s, 3H), 1.82−1.90
HCl/1,4-dioxane solution (3.0 mL), and the reaction was allowed to (m, 2H), 2.01 (t, 1H, J = 5.6 Hz), 2.10−2.16 (m, 1H), 2.29 (s, 3H),
stir for 3 h. The hydrochloride salt crashed out of solution and was 2.31−2.36 (m, 1H), 2.43−2.49 (m, 1H), 2.70 (d, 1H, J = 14.4, 10.4
filtered. The final product was washed three times with cold diethyl Hz), 2.89−2.99 (m, 2H), 3.02−3.08 (m, 2H), 3.79 (s, 3H), 4.28 (d,
ether, resulting in white solid with 811 mg in 68% yield as 1H, J = 8.8 Hz), 6.17 (s, 1H), 6,84 (d, 2H, J = 8.8 Hz), 7.12 (d, 2H, J
hydrochloride salt. = 8.8 Hz). 13C NMR (100 MHz, CDCl3): δ 16.14, 23.13, 25.41,
[(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2- 26.26, 27.87, 29.55, 30.96, 35.11, 35.20, 37.13, 38.03, 38.84, 50.87,
phenylethyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol Ester 54.25, 76.10, 83.49, 112.98 (2C), 128.87 (2C), 131.10, 157.11,
(14a). Followed general method using (R)-(1-amino-2-phenylethyl)- 175.82, 194.78.

7174 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

[(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2-(4′- (dd, 1H, J = 14.0, 6.0 Hz), 3.15 (dd, 1H, J = 14.0, 7.6 Hz), 4.01 (s,
methoxyphenyl)ethyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinane- 1H), 4.16 (d, 1H, J = 7.6 Hz), 7.15−7.18 (m, 3H), 7.25−7.30 (m,
diol Ester (14c′). Followed general method using (S)-(1-amino-2- 2H). 13C NMR (150 MHz, CDCl3): δ 16.80, 24.09, 26.43, 27.23,
(4′-methoxyphenyl)ethyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pina- 28.76, 30.61, 31.78, 36.20, 38.07, 38.49, 39.81, 52.02, 76.94, 77.23,
nediol ester (13c′) as amine and (S)-(3-acetylsulfanyl-2-methyl)- 84.31, 126.01, 126.21 (2C), 128.29 (2C), 140.53, 178.17, 195.92.
propionic acid as carboxylic acid. White solid, 171 mg in 36% yield. [(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1-(4′-
1
H NMR (400 MHz, CDCl3): δ 0.86 (s, 3H), 1.19 (d, 3H, J = 6.8 fluorophenyl)methyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol
Hz), 1.28 (s, 3H), 1.34 (d, 1H, J = 10.4 Hz), 1.38 (s, 3H), 1.82−1.90 Ester (14f). Followed general method using (R)-(1-amino-1-(4′-
(m, 2H), 2.01 (t, 1H, J = 5.6 Hz), 2.11−2.17 (m, 1H), 2.30−2.37 (m, fluorophenyl)methyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol
4H), 2.43−2.48 (m, 1H), 2.68−2.74 (m, 1H), 2.91−3.06 (m, 4H), ester (13f) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic
3.79 (s, 3H), 4.29 (d, 1H, J = 8.8 Hz), 6.17 (s, 1H), 6,84 (d, 2H, J = acid as carboxylic acid. White solid, 268 mg in 60% yield. 1H NMR
8.8 Hz), 7.14 (d, 2H, J = 8.8 Hz). 13C NMR (100 MHz, CDCl3): δ (400 MHz, CDCl3): δ 0.80 (s, 3H), 1.23 (s, 3H), 1.27 (d, 3H, J = 4.4
16.79, 24.15, 26.47, 27.28, 28.86, 30.59, 32.17, 36.17, 36.29, 38.15, Hz), 1.32 (s, 3H), 1.51 (d, 1H, J = 14.0 Hz), 1.77 (m, 1H), 1.89−1.99
38.99, 39.88, 51.93, 55.27, 77.09, 84.45, 113.98 (2C), 129.91 (2C), (m, 2H), 2.01−2.06 (m, 1H), 2.21 (m, 1H), 2.32 (s, 3H), 2.69 (q,
132.20, 158.13, 176.83, 195.70. 1H, J = 6.8 Hz), 2.99−3.04 (m, 1H), 3.11−3.17 (m, 1H), 3.99 (s,
[(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2-(3′- 1H), 4.16 (d, 1H, J = 7.2 Hz), 6.94−6.98 (m, 2H), 7.10−7.14 (m,
benzofuran)ethyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol 2H), 7.31 (br, 1H). 13C NMR (100 MHz, CDCl3): δ 17.11, 24.10,
Ester (14d). Followed general method using (R)-(1-amino-2-(3′- 26.43, 27.23, 28.82, 30.61, 31.88, 36.22, 38.09, 38.72, 39.80, 52.03,
benzofuran)ethyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester 69.59, 77.27, 84.37, 115.00 (2C), 127.77 (2C), 136.37, 162.58,
(13d) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as 178.03, 196.07.
carboxylic acid. White solid, 392 mg in 81% yield. 1H NMR (400 [(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1-(4′-
MHz, CDCl3): δ 0.83 (s, 3H), 1.19 (d, 3H, J = 7.2 Hz), 1.25 (m, 4H), fluorophenyl)methyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol
1.33 (s, 3H), 1.78−1.87 (m, 2H), 1.96−2.00 (m, 1H), 2.08−2.15 (m, Ester (14f′). Followed general method using (S)-(1-amino-1-(4′-
1H), 2.22 (s, 3H), 2.29−2.36 (m, 1H), 2.46−2.55 (m, 1H), 2.84− fluorophenyl)methyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol
2.87 (m, 1H), 2.96 (m, 1H), 2.99−3.07 (m, 2H), 3.14−3.17 (m, 1H), ester (13f′) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic
4.25 (d, 1H, J = 7.2 Hz), 6.83 (s, 1H), 7.21 (t, 1H, J = 7.6 Hz), 7.26 acid as carboxylic acid. White solid, 333 mg in 74% yield. 1H NMR
(t, 1H, J = 7.6 Hz), 7.40−7.49 (m, 2H), 7.59 (d, 1H, J = 7.6 Hz). 13C (400 MHz, CDCl3): δ 0.79 (s, 3H), 1.23 (s, 3H), 1.28 (d, 3H, J = 8.0
NMR (150 MHz, CDCl3): δ 16.75, 24.10, 25.26, 26.49, 27.26, 28.84, Hz), 1.31 (s, 3H), 1.48 (d, 1H, J = 14.0 Hz), 1.76 (m, 1H), 1.90−2.03
30.49, 32.06, 36.25, 38.10, 38.80, 38.91, 39.88, 51.97, 77.04, 84.37, (m, 3H), 2.20 (m, 1H), 2.34 (s, 3H), 2.68 (q, 1H, J = 6.8 Hz), 3.03−
111.47, 118.45, 119.78, 122.43, 124.32, 127.96, 141.97, 155.41, 3.09 (m, 1H), 3.11−3.17 (m, 1H), 3.95 (s, 1H), 4.16 (d, 1H, J = 8.0
177.28, 195.73. Hz), 6.97 (t, 2H, J = 8.4 Hz), 7.11−7.15 (m, 2H), 7.49 (br, 1H). 13C
[(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2-(3′- NMR (150 MHz, CDCl3): δ 16.86, 24.04, 26.38, 27.18, 28.77, 30.57,
benzofuran)ethyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol 31.96, 36.20, 38.02, 38.54, 39.75, 52.02, 68.99, 76.96, 84.24, 114.96,
Ester (14d′). Followed general method using (S)-(1-amino-2-(3′- 115.10, 127.71, 127.76, 136.32, 160.48, 178.02, 195.83.
benzofuran)ethyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol [(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1-(4′-
ester (13d′) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic methylphenyl)methyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinane-
acid as carboxylic acid. White solid, 365 mg in 75% yield. 1H NMR diol Ester (14g). Followed general method using (R)-(1-amino-1-
(400 MHz, CDCl3): δ 0.85 (s, 3H), 1.18 (d, 3H, J = 6.8 Hz), 1.26 (m, (4′-methylphenyl)methyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinane-
4H), 1.36 (s, 3H), 1.81−1.90 (m, 2H), 2.00 (t, 1H, J = 5.2 Hz), diol ester (13g) as amine and (S)-(3-acetylsulfanyl-2-methyl)-
2.12−2.18 (m, 1H), 2.25 (s, 3H), 2.28−2.37 (m, 1H), 2.44−2.50 (m, propionic acid as carboxylic acid. White solid, 235 mg in 53% yield.
1H), 2.88−2.92 (m, 1H), 2.96−3.05 (m, 3H), 3.11−3.18 (m, 1H),
1
H NMR (400 MHz, CDCl3): δ 0.79 (s, 3H), 1.24 (s, 3H), 1.26 (d,
4.28 (d, 1H, J = 8.8 Hz), 6.56 (s, 1H), 7.22 (t, 1H, J = 7.6 Hz), 7.28 3H, J = 6.0 Hz), 1.31 (s, 3H), 1.54 (d, 1H, J = 14.0 Hz), 1.74−1.80
(t, 1H, J = 7.6 Hz), 7.46 (d, 1H, J = 7.6 Hz), 7.49 (s, 1H), 7.59 (d, (m, 1H), 1.87−1.98 (m, 2H), 1.99−2.08 (m, 1H), 2.17−2.23 (m,
1H, J = 7.6 Hz). 13C NMR (100 MHz, CDCl3): δ 16.81, 24.15, 25.28, 1H), 2.29 (s, 3H), 2.31 (s, 3H), 2.62−2.68 (m, 1H), 2.98−3.05 (m,
26.52, 27.30, 28.85, 30.51, 32.11, 36.25, 38.16, 38.60, 38.94, 39.93, 1H), 3.08−3.16 (m, 1H), 3.97 (s, 1H), 4.16 (d, 1H, J = 8.4 Hz),
52.01, 77.15, 84.49, 111.51, 118.48, 119.83, 122.46, 124.35, 128.03, 7.04−7.11 (m, 4H), 7.22 (br, 1H). 13C NMR (100 MHz, CDCl3): δ
142.03, 155.47, 177.19, 195.71. 17.14, 21.05, 24.10, 26.42, 27.25, 28.83, 30.59, 31.86, 36.20, 38.05,
[(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1- 38.70, 39.82, 52.01, 69.57, 77.03, 84.25, 126.27 (2C), 129.03 (2C),
phenylmethyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol Ester 135.46, 137.48, 177.70, 196.04.
(14e). Followed general method using (R)-(1-amino-1- [(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1-(4′-
phenylmethyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester methylphenyl)methyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinane-
(13e) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as diol Ester (14g′). Followed general method using (S)-(1-amino-1-
carboxylic acid. White solid, 144 mg in 33% yield. 1H NMR (600 (4′-methylphenyl)methyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pina-
MHz, CDCl3): δ 0.81 (s, 3H), 1.21 (d, 1H, J = 10.2 Hz), 1.25 (s, nediol ester (13g′) as amine and (S)-(3-acetylsulfanyl-2-methyl)-
3H), 1.34−1.36 (m, 6H), 1.57 (d, 1H, J = 9.2 Hz), 1.79 (br, 1H), propionic acid as carboxylic acid. White solid, 317 mg in 71% yield.
2.03−2.10 (m, 2H), 2.19−2.23 (m, 1H), 2.34 (s, 3H), 2.68−2.71 (m, 1
H NMR (400 MHz, CDCl3): δ 0.80 (s, 3H), 1.23 (s, 3H), 1.27 (d,
1H), 3.04 (dd, 1H, J = 13.8, 6.0 Hz), 3.18 (dd, 1H, J = 13.8, 7.2 Hz), 3H, J = 6.4 Hz), 1.34 (s, 3H), 1.56 (d, 1H, J = 14.0 Hz), 1.75−1.80
4.11 (s, 1H), 4.22 (d, 1H, J = 8.4 Hz), 6.84 (br, 1H), 7.18−7.21 (m, (m, 1H), 1.92−2.08 (m, 3H), 2.15−2.20 (m, 4H), 2.30 (s, 3H),
3H), 7.29−7.32 (m, 2H). 13C NMR (100 MHz, CDCl3): δ 17.00, 2.62−2.69 (m, 1H), 3.02−3.10 (m, 1H), 3.11−3.18 (m, 1H), 4.00 (s,
24.08, 26.39, 27.21, 28.64, 30.63, 31.78, 36.06, 38.14, 38.50, 39.80, 1H), 4.19 (d, 1H, J = 8.4 Hz), 6.94 (br, 1H), 7.08−7.15 (m, 4H). 13C
51.87, 76.88, 77.23, 84.99, 126.21, 126.31 (2C), 128.40 (2C), 140.16, NMR (100 MHz, CDCl3): δ 16.89, 21.06, 24.10, 26.42, 27.24, 28.78,
178.57, 196.16. 30.63, 32.14, 36.16, 38.07, 38.92, 39.81, 51.99, 69.55, 77.17, 84.45,
[(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1- 126.39 (2C), 129.08 (2C), 135.58, 137.40, 177.38, 195.97.
phenylmethyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol Ester [(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1-(2′-
(14e′). Followed general method using (S)-(1-amino-1- naphthyl)methyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol
phenylmethyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester Ester (14h). Followed general method using (R)-(1-amino-1-(2′-
(13e′) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as naphthyl)methyl)boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester
carboxylic acid. White solid, 126 mg in 29% yield. 1H NMR (400 (13h) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as
MHz, CDCl3): δ 0.79 (s, 3H), 1.18−1.22 (m, 4H), 1.20 (d, 3H, J = carboxylic acid. White solid, 389 mg in 81% yield. 1H NMR (400
7.2 Hz), 1.31 (s, 3H), 1.47−1.52 (m, 1H), 1.75 (br, 1H), 1.93−2.04 MHz, CDCl3): δ 0.77 (s, 3H), 1.21 (s, 3H), 1.27 (d, 3H, J = 10.0
(m, 2H), 2.13−2.20 (m, 1H), 2.34 (s, 3H), 2.65−2.74 (m, 1H), 3.07 Hz), 1.32 (s, 3H), 1.48 (d, 1H, J = 14.0 Hz), 1.72 (br, 1H), 1.88−1.92

7175 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

(m, 1H), 1.95−1.98 (m, 1H), 2.00−2.05 (m, 1H), 2.10−2.17 (m, and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as carboxylic acid.
1H), 2.32 (s, 3H), 2.69−2.78 (m, 1H), 3.02−3.07 (m, 1H), 3.17 (dd, White solid, 279 mg in 68% yield. 1H NMR (600 MHz, CDCl3): δ
1H, J = 13.6, 7.6 Hz), 4.17 (d, 1H, J = 8.4 Hz), 4.19 (s, 1H), 7.27− 0.84 (s, 3H), 0.90−0.94 (m, 6H), 1.22 (d, 3H, J = 6.6 Hz), 1.26 (s,
7.34 (m, 2H), 7.37−7.46 (m, 2H), 7.57 (s, 1H), 7.76−7.79 (m, 3H). 3H), 1.31 (d, 1H, J = 10.2 Hz), 1.37 (s, 3H), 1.44 (t, 2H, J = 7.6 Hz),
13
C NMR (150 MHz, CDCl3): δ 17.13, 24.05, 26.43, 27.21, 28.82, 1.62−1.67 (m, 1H), 1.80−1.83 (m, 1H), 1.87 (m, 1H), 1.98−2.02
30.59, 31.78, 36.15, 38.03, 38.50, 39.80, 52.03, 76.93, 77.12, 84.24, (m, 1H), 2.12−2.15 (m, 1H), 2.32 (m, 4H), 2.47−2.51 (m, 1H),
123.86, 125.12, 125.40, 125.83, 127.57, 127.58, 127.89, 132.16, 2.95−3.00 (m, 2H), 3.05−3.09 (m, 1H), 4.26 (d, 1H, J = 8.4 Hz),
133.45, 138.32, 178.32, 196.03. 6.25 (s, 1H). 13C NMR (100 MHz, CDCl3): δ 16.99, 22.09, 23.16,
[(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-1-(2′- 24.15, 25.69, 26.42, 27.25, 28.79, 30.61, 32.35, 36.03, 38.16, 38.78,
naphthyl)methyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol 39.53, 39.81, 40.34, 51.77, 77.24, 84.68, 175.95, 196.28.
Ester (14h′). Followed general method using (S)-(1-amino-1-(2′- [( 1R )-1- (3′ -Acetylsulfanyl-2 ′-benzylpropanamido)-2-
naphthyl)methyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol phenylethyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol Ester
ester (13h′) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic (15a). Followed general method using (R)-(1-amino-2-phenylethyl)-
acid as carboxylic acid. White solid, 317 mg in 66% yield. 1H NMR boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (13a) as amine
(400 MHz, CDCl3): δ 0.75 (s, 3H), 1.20 (s, 3H), 1.24 (d, 3H, J = and (3-acetylsulfanyl-2-benzyl)propionic acid as carboxylic acid.
10.0 Hz), 1.30 (s, 3H), 1.48 (d, 1H, J = 14.0 Hz), 1.71 (br, 1H), 1.95 White solid, 214 mg in 41% yield. 1H NMR (600 MHz, CDCl3): δ
(t, 1H, J = 5.2 Hz), 1.98−2.05 (m, 1H), 2.08−2.16 (m, 2H), 2.31 (s, 0.88 (s, 3H), 1.27−1.32 (m, 4H), 1.40 (s, 3H), 1.84−1.94 (m, 2H),
3H), 2.66−2.72 (m, 1H), 3.02 (dd, 1H, J = 13.6, 6.0 Hz), 3.14 (dd, 2.03 (br, 1H), 2.13−2.16 (m, 1H), 2.31−2.37 (m, 4H), 2.44−2.52
1H, J = 13.6, 7.6 Hz), 4.12−4.15 (m, 2H), 7.30 (d, 1H, J = 8.4 Hz), (m, 1H), 2.60−2.65 (m, 1H), 2.84−3.07 (m, 4H), 3.09−3.16 (m,
7.36−7.45 (m, 3H), 7.54 (s, 1H), 7.72−7.78 (m, 3H). 13C NMR (150 2H), 4.34 (m, 1H), 6.10 (s, 1H), 6.87 (d, 1H, J = 7.2 Hz), 7.05 (d,
MHz, CDCl3): δ 16.12, 23.04, 25.42, 26.20, 27.81, 29.58, 30.77, 1H, J = 7.2 Hz), 7.17−7.23 (m, 5H), 7.24−7.28 (m, 2H), 7.33 (t, 1H,
35.14, 37.02, 37.49, 38.79, 51.02, 75.92, 76.11, 83.23, 122.85, 124.11, J = 7.2 Hz). 13C NMR (100 MHz, CDCl3): δ 24.13, 26.41, 27.24,
124.39, 124.82, 126.56, 126.57, 126.88, 131.15, 132.44, 137.31, 28.77, 30.60, 30.82, 35.98, 37.11, 37.78, 38.16, 39.83, 46.78, 51.78,
177.31, 195.02. 77.23, 84.86, 126.20, 126.81, 128.38, 128.47, 128.62, 128.77, 128.92,
[(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2- 128.98, 129.06, 129.12, 137.85, 139.88, 175.88, 195.65.
methylpropyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol Ester [(R)-1-(2′-Acetylsulfanylacetamido)-2-phenylethyl]boronic Acid
(14i). Followed general method using (R)-(1-amino-2-methylpropyl)- (1S,2S,3R,5S)-(+)-2,3-Pinanediol Ester (15e). Followed general
boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (13i) as amine method using (R)-(1-amino-2-phenylethyl)boronic acid
and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as carboxylic acid. (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (13a) as amine and (2-
White solid, 238 mg in 60% yield. 1H NMR (400 MHz, CDCl3): δ acetylsulfanyl)acetic acid as carboxylic acid. White solid, 154 mg in
0.85 (s, 3H), 0.97 (dd, 6H, J = 6.4, 4.4 Hz), 1.24 (d, 3H, J = 6.8 Hz), 37% yield. 1H NMR (400 MHz, CDCl3): δ 0.84 (s, 3H), 1.27 (s, 3H),
1.27−1.30 (m, 4H), 1.40 (s, 3H), 1.82−1.91 (m, 2H), 1.92−2.00 (m, 1.33 (d, 1H, J = 10.0 Hz), 1.37 (s, 3H), 1.81−1.93 (m, 2H), 1.98−
1H), 2.02 (t, 1H, J = 5.6 Hz), 2.13−2.21 (m, 1H), 2.29−2.38 (m, 2.05 (m, 1H), 2.10−2.20 (m, 1H), 2.25−2.46 (m, 4H), 2.75−2.82
4H), 2.47−2.57 (m, 1H), 2.94−3.03 (m, 2H), 3.09 (dd, 1H, J = 13.6, (m, 1H), 2.90−3.00 (m, 1H), 3.17−3.22 (m, 1H), 3.44−3.55 (m,
7.6 Hz), 4.30 (d, 1H, J = 8.4 Hz), 6.23 (s, 1H). 13C NMR (100 MHz, 2H), 4.30 (t, 1H, J = 9.2 Hz), 6.38 (s, 1H), 7.12−7.36 (m, 5H). 13C
CDCl3): δ 17.42, 19.92, 20.32, 24.13, 26.42, 27.16, 28.83, 29.90, NMR (100 MHz, CDCl3): δ 23.07, 25.29, 26.14, 27.58, 29.14, 30.34,
30.64, 32.39, 35.83, 38.14, 39.70, 40.14, 51.52, 77.42, 84.95, 175.76, 34.63, 35.71, 37.16, 38.60, 50.49, 76.67, 84.55, 125.33, 127.46 (2C),
196.32. 128.05 (2C), 138.58, 168.54, 193.85.
[(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-2- [(S)-1-(3′-Acetylsulfanylpropanamido)-2-(3′-benzofuran)ethyl]-
methylpropyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol Ester boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol Ester (16a). Followed
(14i′). Followed general method using (S)-(1-amino-2-
general method using (S)-(1-amino-2-(3′-benzofuran)ethyl)boronic
methylpropyl)boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester
acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester (13d′) as amine and (3-
(13i′) as amine and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as
acetylsulfanyl)propionic acid as carboxylic acid. White solid, 244 mg
carboxylic acid. White solid, 255 mg in 64% yield. 1H NMR (400
in 52% yield. 1H NMR (400 MHz, DMSO-d6): δ 0.80 (s, 3H), 1.20
MHz, CDCl3): δ 0.85 (s, 3H), 0.97 (d, 6H, J = 6.8 Hz), 1.24 (d, 3H, J
= 6.8 Hz), 1.28 (s, 3H), 1.30 (d, 1H, J = 11.6 Hz), 1.40 (s, 3H), (s, 3H), 1.23 (s, 3H), 1.27 (d, 1H, J = 10.4 Hz), 1.59−1.64 (m, 1H),
1.83−1.91 (m, 2H), 1.92−1.97 (m, 1H), 2.02 (t, 1H, J = 5.6 Hz), 1.74 (br, 1H), 1.80 (t, 1H, J = 5.6 Hz), 1.90−1.97 (m, 1H), 2.13−
2.13−2.20 (m, 1H), 2.29−2.38 (m, 4H), 2.48−2.57 (m, 1H), 2.90 (t, 2.21 (m, 1H), 2.31 (s, 3H), 2.54 (t, 2H, J = 6.8 Hz), 2.61−2.69 (m,
1H, J = 5.6 Hz), 3.00 (dd, 1H, J = 13.6, 6.4 Hz), 3.09 (dd, 1H, J = 1H), 2.78−2.82 (m, 2H), 3.01 (t, 2H, J = 6.8 Hz), 4.05 (d, 1H, J = 8.0
13.6, 7.6 Hz), 4.30 (d, 1H, J = 8.4 Hz), 6.29 (s, 1H). 13C NMR (100 Hz), 7.22−7.31 (m, 2H), 7.55 (dd, 2H, J = 12.4, 8.0 Hz), 7.76 (s,
MHz, CDCl3): δ 17.14, 19.92, 20.27, 24.08, 26.40, 27.12, 28.75, 1H), 9.33 (s, 1H).
29.78, 30.57, 32.38, 35.81, 38.08, 39.67, 39.88, 51.52, 77.30, 84.77, [(S)-1-(2′-Acetylsulfanylacetamido)-2-(3′-benzofuran)ethyl]-
boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol Ester (16b). Followed
175.74, 196.17.
general method using (S)-(1-amino-2-(3′-benzofuran)ethyl)boronic
[(1R)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-3-
methylbutyl]boronic Acid (1S,2S,3R,5S)-(+)-2,3-Pinanediol Ester acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester (13d′) as amine and (2-
(14j). Followed general method using (R)-(1-amino-3-methylbutyl)- acetylsulfanyl)acetic acid as carboxylic acid. White solid, 168 mg in
boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (13j) as amine 37% yield. 1H NMR (400 MHz, DMSO-d6): δ 0.78 (s, 3H), 1.15 (d,
and (S)-(3-acetylsulfanyl-2-methyl)propionic acid as carboxylic acid. 1H, J = 10.4 Hz), 1.20 (s, 3H), 1.21 (s, 3H), 1.58−1.62 (m, 1H), 1.73
White solid, 292 mg in 71% yield. 1H NMR (400 MHz, CDCl3): δ (br, 1H), 1.81 (t, 1H, J = 5.6 Hz), 1.88−1.97 (m, 1H), 2.13−2.22 (m,
0.83 (s, 3H), 0.91 (d, 6H, J = 6.0 Hz), 1.22 (d, 3H, J = 6.8 Hz), 1.26 1H), 2.36 (s, 3H), 2.68−2.84 (m, 2H), 2.92 (m, 1H), 3.69 (s, 2H),
(s, 3H), 1.31 (d, 1H, J = 10.8 Hz), 1.38 (s, 3H), 1.42 (t, 2H, J = 7.6 4.11 (d, 1H, J = 8.0 Hz), 7.22−7.31 (m, 2H), 7.53 (d, 1H, J = 8.0
Hz), 1.60−1.69 (m, 1H), 1.80−1.87 (m, 2H), 2.00 (t, 1H, J = 5.6 Hz), 7.59 (d, 1H, J = 7.6 Hz), 7.74 (s, 1H), 9.07 (s, 1H).
Hz), 2.10−2.17 (m, 1H), 2.30 (m, 4H), 2.47−2.54 (m, 1H), 2.92− General Method of Hydrolysis Reaction for Synthesis of
3.02 (m, 2H), 3.04−3.12 (m, 1H), 4.23 (d, 1H, J = 7.6 Hz), 6.39 (s, MS01−MS33. 3 N aqueous HCl solution (0.2 mL) was added into a
1H). 13C NMR (100 MHz, CDCl3): δ 17.16, 22.11, 23.13, 24.13, solution of protected amide (1 equiv), 2-methylpropylboronic acid (4
25.68, 26.37, 27.23, 28.81, 30.60, 32.18, 36.01, 38.15, 38.72, 39.44, equiv) in methanol/hexane (1:1, 10 mL) at 0 °C. The reaction
39.80, 40.33, 51.75, 77.13, 84.64, 176.19, 196.24. solution was stirred at the same temperature until the TLC indicates
[(1S)-1-((2′S)-(3′-Acetylsulfanyl-2′-methylpropanamido))-3- complete disappearance of amide. The reaction mixture was extracted
methylbutyl]boronic Acid (1R,2R,3S,5R)-(−)-2,3-Pinanediol Ester with hexane (5 mL) thrice, and the aqueous methanol layer was
(14j′). Followed general method using (S)-(1-amino-3-methylbutyl)- concentrated at temperature below 30 °C. The residue was treated
boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester (13j′) as amine with ice and basified with 2 N aqueous NaOH solution and extracted

7176 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

with dichloromethane 5 times. The aqueous layer was then acidified general method using [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-(1″,3″-
with 3 N aqueous HCl solution and extracted with dichloromethane dioxoisoindolin-2″-yl)propanamido))-2-phenylethyl]boronic acid
until the TLC indicates complete disappearance of product in (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (15b) as amide. White solid.
aqueous layer. The dichloromethane layer was collected, dried over HPLC purity: 96.2%. 1H NMR (400 MHz, CDCl3): δ 1.61 (t, 1H, J =
Na2SO4, and concentrated under reduced pressure to give a solid 8.8 Hz), 2.75−2.83 (m, 2H), 2.97−3.03 (m, 1H), 3.06−3.13 (m, 1H),
residue, which was purified by flash chromatography on high 3.22−3.27 (m, 1H), 3.59 (t, 1H, J = 6.8 Hz), 6.59 (br, 1H), 7.21−
performance silica gel to obtain the final compounds MS01−MS33. 7.34 (m, 5H), 7.85−7.97 (m, 4H). 13C NMR (100 MHz, CDCl3): δ
(S)-(1-(3′-Mercapto-2′-methylpropanamido)methyl)boronic 27.81, 35.94, 63.93, 77.44, 123.11, 123.26, 125.95, 128.44 (2C),
Acid (MS01). Followed general method using (S)-(1-(3′-acetylsul- 129.17 (2C), 132.14 (2C), 132.41 (2C), 141.07 166.46, 166.67,
fanyl-2′-methylpropanamido)methyl)boronic acid pinacol ester (5a) 177.42. MS (ESI+): 381.1 [M + H − H2O]+. HRMS (ESI) m/z:
as amide. White solid. HPLC purity: 96.2%. 1H NMR (400 MHz, calcd for C19H19BN2O5S [M + Na]+, 421.1000, found, 421.1011.
CDCl3): δ 1.25 (d, 3H, J = 6.8 Hz), 1.64 (br, 1H), 2.31 (s, 2H), 2.60 [(1R)-1-((2′S)-(3′-Mercapto-2′-benzylaminopropanamido))-2-
(m, 1H), 2.76 (m, 1H), 2.98−3.14 (m, 1H), 6.37 (br, 1H). 13C NMR phenylethyl]boronic Acid (MS08). Followed general method using
(100 MHz, CDCl3): δ 17.04, 27.73, 31.95, 41.49, 178.34. MS (ESI+): [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-benzylaminopropanamido))-2-
148.1 [M + H − H2O]+. phenylethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester
(1-(n-Butyramido)methyl)boronic Acid (MS02). Followed general (15c) as amide. White solid. HPLC purity: 95.4%. 1H NMR (400
method using (1-(n-butyramido)methyl)boronic acid pinacol ester MHz, CDCl3): δ 1.56 (t, 1H, J = 8.4 Hz), 2.51−2.60 (m, 2H), 2.95−
(5b) as amide. White solid. HPLC purity: 98.0%. 1H NMR (400 3.00 (m, 1H), 3.31−3.37 (m, 1H), 3.53−3.57 (m, 1H), 3.91 (t, 1H, J
MHz, CDCl3): δ 0.84 (t, 3H, J = 7.2 Hz), 1.54 (m, 2H), 2.14 (t, 2H, J = 6.8 Hz), 4.68 (dd, 2H, J = 43.6, 16.6 Hz), 6.76 (br, 2H), 7.11−7.22
= 7.2 Hz), 2.36 (s, 2H), 7.80 (br, 1H). 13C NMR (100 MHz, CDCl3): (m, 5H), 7.26−7.39 (m, 5H). 13C NMR (100 MHz, CDCl3): δ 28.68,
δ 13.65, 18.65, 36.82, 41.28, 176.80. MS (ESI+): 128.1 [M + H − 35.56, 52.41, 57.68, 76.21, 125.10, 125.34, 127.33, 127.37, 127.43,
H2O]+. HRMS (ESI) m/z: calcd for C5H12BNO3[M + H − H2O]+, 127.51, 127.98, 128.05, 128.09, 128.24, 139.67, 139.78, 171.19. MS
128.0877, found, 128.0873; calcd for C5H12BNO3 [M + Na]+, (ESI+): 341.2 [M + H − H2O]+. HRMS (ESI) m/z: calcd for
168.0802, found, 168.0807. C18H23BN2O3S [M + H − H2O]+, 341.1490, found, 341.1496; calcd
(1-(n-Propanamido)methyl)boronic Acid (MS03). Followed gen- for C18H23BN2O3S [M + H]+, 359.1595, found, 359.1606.
eral method using (1-(n-propanamido)methyl)boronic acid pinacol [(R)-1-(3′-Mercaptopropanamido)-2-phenylethyl]boronic Acid
ester (5c) as amide. White solid. HPLC purity: 97.8%. 1H NMR (400 (MS09). Followed general method using [(R)-1-(3′-acetylsulfanyl-
MHz, CDCl3): δ 1.16 (t, 3H, J = 7.2 Hz), 2.41 (q, 2H, J = 7.2 Hz), propanamido)-2-phenylethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pi-
2.68 (s, 2H). 13C NMR (100 MHz, CDCl3): δ 11.15, 21.35, 26.41, nanediol ester (15d) as amide. White solid. HPLC purity: 96.1%. 1H
179.41. MS (ESI+): 114.1 [M + H − H2O]+. HRMS (ESI) m/z: NMR (400 MHz, CDCl3): δ 1.66 (t, 1H, J = 8.0 Hz), 2.54 (t, 2H, J =
calcd for C4H10BNO3 [M + H − H2O]+, 114.0721, found, 114.0718. 6.8 Hz), 2.72−2.79 (m, 3H), 2.99 (dd, 1H, J = 14.4, 5.2 Hz), 3.05−
(S)-2-(3′-Mercapto-2′-methylpropanamido)acetic Acid (MS04). 3.14 (m, 1H), 6.80 (br, 1H), 7.18−7.25 (m, 3H), 7.25−7.31 (m, 2H).
Followed general method using (S)-2-(3′-acetylsulfanyl-2′-
13
C NMR (100 MHz, CDCl3): δ 22.06, 29.69, 36.57, 77.22, 126.09,
methylpropanamido)acetic acid ethyl ester (7) as amide. White 128.42 (2C), 129.04 (2C), 140.77, 175.13. MS (ESI+): 236.1 [M + H
solid. HPLC purity: 95.4%. 1H NMR (400 MHz, DMSO-d6): δ 1.07 − H2O]+. HRMS (ESI) m/z: calcd for C11H16BNO3S [M + H −
(d, 3H, J = 6.8 Hz), 2.22 (t, 1H, J = 8.0 Hz), 2.40−2.49 (m, 2H), 2.66 H2O]+, 236.0911, found, 236.0912; calcd for C11H16BNO3S [M +
(dt, 1H, J = 12.4, 6.8 Hz), 3.74 (s, 2H), 8.46 (br, 1H), 8.86 (br, 1H). Na]+, 276.0836, found, 276.0833.
13
C NMR (100 MHz, DMSO-d6): δ 17.48, 27.91, 41.03, 43.61, [(R)-1-(2′-Mercaptoacetamido)-2-phenylethyl]boronic Acid
171.80, 174.83. MS (ESI+): 178.0 [M + H] +. HRMS (ESI) m/z: (MS10). Followed general method using [(R)-1-(2′-acetylsulfanyla-
calcd for C6H11NO3S [M + H]+, 178.0532, found, 178.0534; calcd for cetamido)-2-phenylethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinane-
C6H11NO3S [M + Na]+, 200.0352, found, 200.0354. diol ester (15e) as amide. White solid. HPLC purity: 95.4%. 1H NMR
[(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2- (400 MHz, CDCl3): δ 1.88 (br, 1H), 2.70−2.78 (m, 1H), 2.96 (dd,
phenylethyl]boronic Acid (MS05). Followed general method using 1H, J = 14.4, 5.2 Hz), 3.06−3.10 (m, 1H), 3.31 (d, 2H, J = 8.4 Hz),
[(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2- 7.19−7.22 (m, 3H), 7.27−7.32 (m, 2H), 7.37 (br, 1H). 13C NMR
phenylethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (100 MHz, CDCl3): δ 31.59, 36.56, 77.23, 126.12, 128.53 (2C),
(14a) as amide. White solid. HPLC purity: 95.8%. 1H NMR (400 128.97 (2C), 140.64, 174.02. MS (ESI+): 222.1 [M + H − H2O]+.
MHz, CDCl3): δ 1.19 (d, 3H, J = 6.8 Hz), 1.46 (t, 1H, J = 8.8 Hz), HRMS (ESI) m/z: calcd for C10H14BNO3S [M + H − H2O]+,
2.52−2.59 (m, 2H), 2.67−2.74 (m, 2H), 2.93 (dd, 1H, J = 14.4, 5.2 222.0755, found, 222.0749; calcd for C10H14BNO3S [M + Na]+,
Hz), 3.01−3.08 (m, 1H), 6.74 (br, 1H), 7.10−7.19 (m, 3H), 7.19− 262.0680, found, 262.0684.
7.24 (m, 2H). 13C NMR (100 MHz, CDCl3): δ 15.19, 26.40, 35.72, [(R)-1-(n-Butyramido)-2-phenylethyl]boronic Acid (MS11). Fol-
41.94, 76.22, 125.04, 127.47 (2C), 128.07 (2C), 139.76, 177.90. MS lowed general method using [(R)-1-(n-butyramido)-2-phenylethyl]-
(ESI+): 250.1 [M + H − H2O]+. HRMS (ESI) m/z: calcd for boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (15f) as amide.
C12H18BNO3S [M + H − H2O]+, 250.1068, found, 250.1066; calcd White solid. HPLC purity: 96.0%. 1H NMR (400 MHz, CDCl3): δ
for C12H18BNO3S [M + Na]+, 290.0993, found, 290.0993. 0.92 (t, 3H, J = 7.2 Hz), 1.62 (q, 2H, J = 7.2 Hz), 2.21 (t, 2H, J = 7.2
[(1R)-1-(3′-Mercapto-2′-benzylpropanamido)-2-phenylethyl]- Hz), 2.58−2.79 (m, 1H), 2.79−2.99 (m, 1H), 3.00−3.09 (m, 1H),
boronic Acid (MS06). Followed general method using [(1R)-1-(3′- 6.72 (br, 1H), 7.15−7.22 (m, 3H), 7.24−7.28 (m, 2H). 13C NMR
acetylsulfanyl-2′-benzylpropanamido)-2-phenylethyl]boronic acid (100 MHz, CDCl3): δ 13.67, 18.67, 34.89, 36.84, 77.29, 125.85,
(1S,2S,3R,5S)-(+)-2,3-pinanediol ester (15a) as amide. White solid. 128.37 (2C), 129.10 (2C), 141.23, 176.82. MS (ESI+): 218.1 [M + H
HPLC purity: 96.8%. 1H NMR (600 MHz, CDCl3): δ 1.70 (br, 1H), − H2O]+. HRMS (ESI) m/z: calcd for C12H18BNO3[M + H −
2.56−2.84 (m, 5H), 2.89−2.98 (m, 2H), 3.08−3.11 (m, 1H), 6.15 H2O]+, 218.1347, found, 218.1347; calcd for C12H18BNO3[M + Na]+,
(br, 1H), 6.85 (d, 1H, J = 7.2 Hz), 7.03 (d, 1H, J = 7.2 Hz), 7.15− 258.1272, found, 258.1268.
7.22 (m, 5H), 7.23−7.26 (m, 2H), 7.30−7.33 (m, 1H). 13C NMR [(R)-1-(n-Propanamido)-2-phenylethyl]boronic Acid (MS12).
(100 MHz, CDCl3): δ 28.96, 35.77, 37.10, 49.77, 77.22, 126.14, Followed general method using [(R)-1-(n-propanamido)-2-
126.69, 128.38, 128.44, 128.61, 128.88, 128.91, 128.96, 129.05, phenylethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester
129.51, 137.54, 139.86, 175.17. MS (ESI+): 326.1 [M + H − H2O]+. (15g) as amide. White solid. HPLC purity: 96.6%. 1H NMR (400
HRMS (ESI) m/z: calcd for C18H22BNO3S [M + H − H2O]+, MHz, CDCl3): δ 1.13 (t, 3H, J = 7.2 Hz), 2.32 (q, 2H, J = 7.2 Hz),
326.1381, found, 326.1377; calcd for C18H22BNO3S [M + Na]+, 3.08−3.15 (m, 1H), 3.19−3.26 (m, 1H), 3.67 (t, 1H, J = 8.0 Hz),
366.1306, found, 366.1299. 7.22−7.34 (m, 5H). 13C NMR (100 MHz, CDCl3): δ 10.78, 34.13,
[(1R)-1-((2′S)-(3′-Mercapto-2′-(1″,3″-dioxoisoindolin-2″-yl)- 39.86, 77.08, 125.19, 128.97 (2C), 129.90 (2C), 141.12, 176.82. MS
propanamido))-2-phenylethyl]boronic Acid (MS07). Followed (ESI+): 204.1 [M + H − H2O]+. HRMS (ESI) m/z: calcd for

7177 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

C11H16BNO3[M + H − H2O]+, 204.1190, found, 204.1148; calcd for [(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2-(3′-


C11H16BNO3[M + Na]+, 244.1115, found, 244.1061. benzofuran)ethyl]boronic Acid (MS18). Followed general method
[(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2- using [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2-
phenylethyl]boronic Acid (MS13). Followed general method using (3′-benzofuran)ethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol
[(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2- ester (14d) as amide. White solid. HPLC purity: 96.2%. 1H NMR
phenylethyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester (400 MHz, CDCl3): δ 1.17 (d, 3H, J = 7.2 Hz), 1.55 (t, 1H, J = 8.4
(14a′) as amide. White solid. HPLC purity: 95.6%. 1H NMR (400 Hz), 2.45−2.56 (m, 2H), 2.70−2.87 (m, 3H), 3.18−3.21 (m, 1H),
MHz, CDCl3): δ 1.20 (d, 3H, J = 6.8 Hz), 1.59 (t, 1H, J = 8.8 Hz), 7.18−7.24 (m, 2H), 7.43 (d, 1H, J = 8.0 Hz), 7.51 (s, 1H), 7.59 (d,
2.44−2.60 (m, 2H), 2.72−2.82 (m, 2H), 3.03 (dd, 1H, J = 14.0, 4.8 1H, J = 8.0 Hz). 13C NMR (150 MHz, CDCl3): δ 15.34, 23.67, 26.77,
Hz), 3.07−3.14 (m, 1H), 6.57 (br, 1H), 7.16−7.26 (m, 3H), 7.27− 40.84, 76.22, 110.32, 117.85, 119.03, 121.30, 123.14, 127.42, 141.00,
7.32 (m, 2H). 13C NMR (100 MHz, CDCl3): δ 16.47, 27.91, 36.83, 154.28, 176.93. MS (ESI+): 290.1 [M + H − H2O]+. HRMS (ESI)
41.99, 77.24, 125.95, 128.42 (2C), 129.14 (2C), 141.07, 177.52. MS m/z: calcd for C14H18BNO4S [M + H − H2O]+, 290.1017, found,
(ESI+): 250.1 [M + H − H2O]+. HRMS (ESI) m/z: calcd for 290.1021; calcd for C14H18BNO4S [M + Na]+, 330.0942, found,
C12H18BNO3S [M + H − H2O]+, 250.1068, found, 250.1069; calcd 330.0942.
for C12H18BNO3S [M + Na]+, 290.0993, found, 290.0992. [(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2-(3′-
[(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2-(4′- benzofuran)ethyl]boronic Acid (MS19). Followed general method
fluorophenyl)ethyl]boronic Acid (MS14). Followed general method using [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2-
using [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2- (3′-benzofuran)ethyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol
(4′-fluorophenyl)ethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester (14d′) as amide. White solid. HPLC purity: 96.1%. 1H NMR
ester (14b) as amide. HPLC purity: 97.4%. White solid. 1H NMR (600 MHz, CDCl3): δ 1.18 (d, 3H, J = 7.2 Hz), 1.56 (t, 1H, J = 8.4
(400 MHz, CDCl3): δ 1.20 (d, 3H, J = 6.8 Hz), 1.61 (t, 1H, J = 8.8 Hz), 2.44−2.49 (m, 1H), 2.53−2.58 (m, 1H), 2.71−2.77 (m, 1H),
Hz), 2.53−2.58 (m, 1H), 2.62−2.66 (m, 1H), 2.69−2.77 (m, 2H), 2.81−2.87 (m, 1H), 3.01−3.04 (m, 1H), 3.18−3.25 (m, 1H), 6.81 (s,
2.92 (dd, 1H, J = 14.0, 5.2 Hz), 3.01−3.09 (m, 1H), 6.84 (br, 1H), 1H), 7.20 (t, 1H, J = 7.2 Hz), 7.25 (t, 1H, J = 7.2 Hz), 7.43 (d, 1H, J
6.95 (t, 2H, J = 8.8 Hz), 7.16 (dd, 2H, J = 8.8, 5.6 Hz). 13C NMR = 7.8 Hz), 7.52 (s, 1H), 7.59 (d, 1H, J = 7.8 Hz). 13C NMR (100
(100 MHz, CDCl3): δ 16.15, 27.67, 35.98, 41.27, 76.21, 115.05, MHz, CDCl3): δ 16.38, 24.73, 27.83, 41.90, 77.24, 111.36, 118.90,
115.19, 130.33, 130.38, 136.25, 162.16, 178.03. MS (ESI+): 268.1 [M 120.08, 122.34, 124.18, 128.46, 142.04, 155.34, 177.94. MS (ESI+):
+ H − H2O]+. HRMS (ESI) m/z: calcd for C12H13BFNO3S [M + H 290.1 [M + H − H2O]+. HRMS (ESI) m/z: calcd for C14H18BNO4S
− H2O]+, 268.0973, found, 268.0974; calcd for C12H13BFNO3S [M + [M + H − H 2 O]+ , 290.1017, found, 290.1022; calcd for
Na]+, 308.0898, found, 308.0896. C14H18BNO4S [M + Na]+, 330.0942, found, 330.0940.
[(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2-(4′- [(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-
fluorophenyl)ethyl]boronic Acid (MS15). Followed general method phenylmethyl]boronic Acid (MS20). Followed general method using
using [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2- [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-1-
(4′-fluorophenyl)ethyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-pinane- phenylmethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester
diol ester (14b′) as amide. White solid. HPLC purity: 97.6%. 1H (14e) as amide. White solid. HPLC purity: 95.0%. 1H NMR (400
NMR (400 MHz, CDCl3): δ 1.15 (d, 3H, J = 6.8 Hz), 1.56 (t, 1H, J = MHz, CDCl3): δ 1.13 (d, 3H, J = 6.4 Hz), 1.59 (br, 1H), 2.48−2.70
8.8 Hz), 2.42−2.58 (m, 2H), 2.63−2.72 (m, 2H), 2.88 (dd, 1H, J = (m, 3H), 3.79 (s, 1H), 7.03−7.18 (m, 5H). 13C NMR (100 MHz,
14.0, 4.8 Hz), 2.97−3.06 (m, 1H), 6.64 (br, 1H), 6.90 (t, 2H, J = 8.8 CDCl3): δ16.23, 27.45, 42.94, 77.24, 126.02, 126.08, 128.53, 128.58,
Hz), 7.12 (dd, 2H, J = 8.8, 5.6 Hz). 13C NMR (100 MHz, CDCl3): 134.50, 138.07, 175.02. MS (ESI+): 236.1 [M + H − H2O]+. HRMS
δ15.40, 26.79, 34.94, 40.76, 76.21, 114.06, 114.27, 129.38, 129.46, (ESI) m/z: calcd for C11H16BNO3S [M + H − H2O]+, 236.0911,
135.34, 161.62, 176.97. MS (ESI+): 268.1 [M + H − H2O]+. HRMS found, 236.0912; calcd for C11H16BNO3S [M + Na]+, 276.0836,
(ESI) m/z: calcd for C12H13BFNO3S [M + H − H2O]+, 268.0973, found, 276.0840.
found, 268.0969; calcd for C12H13BFNO3S [M + Na]+, 308.0898, [(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-
found, 308.0893. phenylmethyl]boronic Acid (MS21). Followed general method using
[(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2-(4′- [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-1-
methoxyphenyl)ethyl]boronic Acid (MS16). Followed general phenylmethyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester
method using [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanami- (14e′) as amide. White solid. HPLC purity: 95.3%. 1H NMR (400
do))-2-(4′-methoxyphenyl)ethyl]boronic acid (1S,2S,3R,5S)-(+)-2,3- MHz, CDCl3): δ 1.19 (d, 3H, J = 5.2 Hz), 1.60 (br, 1H), 2.49−2.70
pinanediol ester (14c) as amide. White solid. HPLC purity: 96.0%. 1H (m, 3H), 3.87 (s, 1H), 7.08−7.17 (m, 5H). 13C NMR (100 MHz,
NMR (400 MHz, CDCl3): δ 1.21 (d, 3H, J = 6.8 Hz), 1.68 (br, 1H), CDCl3): δ 16.18, 27.41, 42.88, 77.19, 126.07 (2C), 128.54 (2C),
2.55−2.79 (m, 4H), 2.93 (dd, 1H, J = 14.0, 5.2 Hz), 3.06−3.09 (m, 134.47, 138.09, 174.25. MS (ESI+): 236.1 [M + H − H2O]+. HRMS
1H), 3.76 (s, 3H), 6.82 (d, 2H, J = 8.8 Hz), 7.16 (d, 2H, J = 8.8 Hz). (ESI) m/z: calcd for C11H16BNO3S [M + H − H2O]+, 236.0911,
13
C NMR (150 MHz, CDCl3): δ 16.16, 27.77, 35.83, 41.31, 55.18, found, 236.0908; calcd for C11H16BNO3S [M + Na]+, 276.0836,
77.20, 113.79 (2C), 129.98 (2C), 132.69, 157.86, 177.81. MS (ESI+): found, 276.0833.
280.1 [M + H − H2O]+. HRMS (ESI) m/z: calcd for C13H20BNO4S [(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-(4′-
[M + H − H 2O]+ , 280.1173, found, 280.1178; calcd for fluorophenyl)methyl]boronic Acid (MS22). Followed general
C13H20BNO4S [M + Na]+, 320.1098, found, 320.1098. method using [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanami-
[(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2-(4′- do))-1-(4′-fluorophenyl)methyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-
methoxyphenyl)ethyl]boronic Acid (MS17). Followed general pinanediol ester (14f) as amide. White solid. HPLC purity: 95.8%. 1H
method using [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanami- NMR (400 MHz, CDCl3): δ 1.24 (d, 3H, J = 6.8 Hz), 1.52 (t, 1H, J =
do))-2-(4′-methoxyphenyl)ethyl]boronic acid (1R,2R,3S,5R)- 8.4 Hz), 2.55−2.71 (m, 3H), 3.73 (d, 1H, J = 23.6 Hz), 6.77−6.83
(−)-2,3-pinanediol ester (14c′) as amide. White solid. HPLC purity: (m, 2H), 6.86−6.91 (m, 2H). 13C NMR (100 MHz, CDCl3): δ 16.23,
96.9%. 1H NMR (400 MHz, CDCl3): δ 1.21 (d, 3H, J = 6.8 Hz), 1.64 27.45, 42.93, 77.23, 114.48, 114.69, 127.11, 127.19, 136.91, 159.69,
(t, 1H, J = 8.8 Hz), 2.50−2.79 (m, 4H), 2.94 (dd, 1H, J = 14.0, 5.2 178.56. MS (ESI+): 254.1 [M + H − H2O]+. HRMS (ESI) m/z:
Hz), 3.05−3.15 (m, 1H), 3.77 (s, 3H), 6.82 (d, 2H, J = 8.8 Hz), 7.15 calcd for C11H15BFNO3S [M + H − H2O]+, 254.0817, found,
(d, 2H, J = 8.8 Hz). 13C NMR (100 MHz, CDCl3): δ 16.43, 27.87, 254.0819; calcd for C11H15BFNO3S [M + Na]+, 294.0742, found,
35.81, 41.84, 55.24, 77.26, 113.84 (2C), 130.04 (2C), 132.82, 157.91, 294.0740.
177.74. MS (ESI+): 280.1 [M + H − H2O]+. HRMS (ESI) m/z: [(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-(4′-
calcd for C13H20BNO4S [M + H − H2O]+, 280.1173, found, fluorophenyl)methyl]boronic Acid (MS23). Followed general
280.1174; calcd for C13H20BNO4S [M + Na]+, 320.1098, found, method using [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanami-
320.1094. do))-1-(4′-fluorophenyl)methyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-

7178 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

pinanediol ester (14f′) as amide. White solid. HPLC purity: 95.3%. 2.81 (m, 1H). 13C NMR (150 MHz, CDCl3): δ 16.44, 20.14, 20.67,
1
H NMR (400 MHz, DMSO-d6): δ 1.14 (d, 3H, J = 6.0 Hz), 2.28 (m, 27.72, 29.36, 41.70, 77.24, 177.91. MS (ESI+): 202.1 [M + H −
1H), 2.60−2.68 (m, 3H), 3.52 (s, 1H), 6.90−6.97 (m, 4H), 9.34 (br, H2O]+. HRMS (ESI) m/z: calcd for C8H18BNO3S [M + H − H2O]+,
1H). 13C NMR (100 MHz, DMSO-d6): δ 16.90, 27.41, 43.13, 77.29, 202.1068, found, 202.1066; calcd for C8H18BNO3S [M + Na]+,
114.36, 114.57, 127.99, 128.07, 138.93, 159.38, 178.45. MS (ESI+): 242.0993, found, 242.0987.
254.1 [M + H − H2O]+. HRMS (ESI) m/z: calcd for C11H15BFNO3S [(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2-
[M + H − H 2O]+ , 254.0817, found, 254.0819; calcd for methylpropyl]boronic Acid (MS29). Followed general method using
C11H15BFNO3S [M + Na]+, 294.0742, found, 294.0740. [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2-
[(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-(4′- methylpropyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester
methylphenyl)methyl]boronic Acid (MS24). Followed general (14i′) as amide. White solid. HPLC purity: 95.1%. 1H NMR (400
method using [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanami- MHz, CDCl3): δ 0.91 (d, 3H, J = 6.4 Hz), 0.94 (d, 3H, J = 6.4 Hz),
do))-1-(4′-methylphenyl)methyl]boronic acid (1S,2S,3R,5S)-(+)-2,3- 1.24 (d, 3H, J = 6.4 Hz), 1.66 (t, 1H, J = 8.4 Hz), 1.90−1.96 (m, 1H),
pinanediol ester (14g) as amide. White solid. HPLC purity: 95.7%. 2.57−2.65 (m, 3H), 2.76−2.82 (m, 1H), 7.05 (br, 1H). 13C NMR
1
H NMR (400 MHz, CDCl3): δ 1.15 (br, 3H), 1.53 (m, 1H), 2.28 (s, (150 MHz, CDCl3): δ 16.48, 20.24, 20.58, 27.80, 29.30, 41.99, 77.16,
3H), 2.52−2.65 (m, 3H), 3.77 (d, 1H, J = 22.8 Hz), 6.75−6.85 (m, 177.37. MS (ESI+): 202.1 [M + H − H2O]+. HRMS (ESI) m/z:
2H), 6.93−6.96 (m, 2H). 13C NMR (100 MHz, CDCl3): δ 16.24, calcd for C8H18BNO3S [M + H − H2O]+, 202.1068, found, 202.1065;
21.07, 27.64, 42.95, 77.23, 126.01, 126.08, 128.52, 128.58, 134.49, calcd for C8H18BNO3S [M + Na]+, 242.0993, found, 242.0984.
138.06, 175.02. MS (ESI+): 250.1 [M + H − H2O]+. HRMS (ESI) [(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-3-
m/z: calcd for C12H18BNO3S [M + H − H2O]+, 250.1068, found, methylbutyl]boronic Acid (MS30). Followed general method using
250.1069; calcd for C12H18BNO3S [M + Na]+, 290.0993, found, [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-3-
290.0989. methylbutyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester
[(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-(4′- (14j) as amide. White solid. HPLC purity: 97.2%. 1H NMR (400
methylphenyl)methyl]boronic Acid (MS25). Followed general MHz, CDCl3): δ 0.89 (d, 3H, J = 1.6 Hz), 0.91 (d, 3H, J = 1.6 Hz),
method using [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanami- 1.26 (d, 3H, J = 6.8 Hz), 1.32−1.39 (m, 1H), 1.47−1.54 (m, 1H),
do))-1-(4′-methylphenyl)methyl]boronic acid (1R,2R,3S,5R)- 1.60−1.70 (m, 2H), 2.58−2.66 (m, 2H), 2.78−2.84 (m, 1H), 2.92−
(−)-2,3-pinanediol ester (14g′) as amide. White solid. HPLC purity: 2.97 (m, 1H), 7.23 (br, 1H). 13C NMR (150 MHz, CDCl3): δ 16.32,
96.1%. 1H NMR (400 MHz, CDCl3): δ 1.15 (d, 3H, J = 4.4 Hz), 1.53 22.78, 23.08, 25.99, 27.87, 40.33, 41.69, 77.20, 177.25. MS (ESI+):
(m, 1H), 2.28 (s, 3H), 2.43−2.64 (m, 3H), 3.75 (d, 1H, J = 22.4 Hz), 216.1 [M + H − H2O]+. HRMS (ESI) m/z: calcd for C9H20BNO3S
6.78−6.85 (m, 2H), 6.92−6.95 (m, 2H). 13C NMR (100 MHz, [M + H − H2O]+, 216.1224, found, 216.1220; calcd for C9H20BNO3S
CDCl3): δ 16.54, 21.07, 27.52, 41.42, 77.23, 126.06 (2C), 128.54 [M + Na]+, 256.1149, found, 256.1142.
(2C), 134.46, 138.09, 174.24. MS (ESI+): 250.1 [M + H − H2O]+. [(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-3-
HRMS (ESI) m/z: calcd for C12H18BNO3S [M + H − H2O]+, methylbutyl]boronic Acid (MS31). Followed general method using
250.1068, found, 250.1064; calcd for C12H18BNO3S [M + Na]+, [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-3-
290.0993, found, 290.0991. methylbutyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol ester
[(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-(2′- (14j′) as amide. White solid. HPLC purity: 95.6%. 1H NMR (400
naphthyl)methyl]boronic Acid (MS26). Followed general method MHz, CDCl3): δ 0.88 (t, 6H, J = 6.8 Hz), 1.22 (d, 3H, J = 6.8 Hz),
using [(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-1- 1.31−1.41 (m, 1H), 1.44−1.53 (m, 1H), 1.56−1.72 (m, 2H), 2.54−
(2′-naphthyl)methyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol
2.69 (m, 2H), 2.76−2.83 (m, 1H), 2.92 (m, 1H), 7.27 (br, 1H). 13C
ester (14h) as amide. White solid. HPLC purity: 95.7%. 1H NMR
NMR (150 MHz, CDCl3): δ 16.31, 22.67, 23.11, 26.01, 27.83, 40.20,
(600 MHz, DMSO-d6): δ 1.06 (d, 3H, J = 6.6 Hz), 1.50 (t, 1H, J = 8.4
41.72, 77.21, 177.48. MS (ESI+): 216.1 [M + H − H2O]+. HRMS
Hz), 2.57−2.60 (m, 1H), 2.64−2.72 (m, 2H), 3.75 (d, 1H, J = 8.4
(ESI) m/z: calcd for C9H20BNO3S [M + H − H2O]+, 216.1224,
Hz), 7.00 (d, 1H, J = 7.8 Hz), 7.39−7.43 (m, 4H), 7.66 (d, 1H, J =
found, 216.1219; calcd for C9H20BNO3S [M + Na]+, 256.1149, found,
7.8 Hz), 7.75 (d, 1H, J = 7.8 Hz), 9.08 (br, 1H). 13C NMR (100
MHz, DMSO-d6): δ17.03, 27.50, 43.16, 77.38, 122.98, 125.03, 256.1140.
126.09, 127.11, 127.59, 127.83, 130.13, 131.69, 133.41, 141.02, [(S)-1-(3′-Mercaptopropanamido))-2-(3′-benzofuran)ethyl]-
boronic Acid (MS32). Followed general method using [(S)-1-(3′-
178.39. MS (ESI+): 286.1 [M + H − H2O]+. HRMS (ESI) m/z:
acetylsulfanylpropanamido)-2-(3′-benzofuran)ethyl]boronic acid
calcd for C15H18BNO3S [M + H − H2O]+, 286.1068, found,
(1R,2R,3S,5R)-(−)-2,3-pinanediol ester (16a) as amide. White
286.1043; calcd for C15H18BNO3S [M + Na]+, 326.0993, found,
solid. HPLC purity: 95.9%. 1H NMR (400 MHz, DMSO-d6): δ
326.0971.
1.56 (t, 1H, J = 8.4 Hz), 2.49 (t, 2H, J = 6.8 Hz), 2.71−2.79 (m, 3H),
[(1S)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-1-(2′-
naphthyl)methyl]boronic Acid (MS27). Followed general method 2.98−3.03 (m, 1H), 3.06−3.11 (m, 1H), 6.77 (br, 1H), 7.17−7.26
using [(1S)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-1- (m, 2H), 7.48−7.53 (m, 2H), 7.71 (s, 1H). 13C NMR (100 MHz,
(2′-naphthyl)methyl]boronic acid (1R,2R,3S,5R)-(−)-2,3-pinanediol DMSO-d6): δ 20.70, 29.79, 42.47, 77.30, 111.34, 118.88, 120.05,
ester (14h′) as amide. White solid. HPLC purity: 96.6%. 1H NMR 122.33, 124.16, 128.43, 142.04, 155.30, 177.94. MS (ESI+): 276.1 [M
(400 MHz, CDCl3): δ 1.10 (d, 3H, J = 6.8 Hz), 1.50 (t, 1H, J = 8.8 + H − H2O]+. HRMS (ESI) m/z: calcd for C13H16BNO4S [M + H −
Hz), 2.62−2.69 (m, 1H), 2.70−2.77 (m, 2H), 3.72 (d, 1H, J = 8.8 H2O]+, 276.0860, found, 276.0869; calcd for C13H16BNO4S [M +
Hz), 7.02 (d, 1H, J = 6.8 Hz), 7.37−7.44 (m, 4H), 7.58 (d, 1H, J = Na]+, 316.0785, found, 316.0787.
6.8 Hz), 7.67 (d, 1H, J = 6.8 Hz). 13C NMR (100 MHz, CDCl3): [(S)-1-(2′-Mercaptoacetamido))-2-(3′-benzofuran)ethyl]boronic
δ17.10, 27.33, 42.81, 77.11, 122.94, 125.03, 125.61, 126.11, 127.12, Acid (MS33). Followed general method using [(S)-1-(2′-
127.57, 127.85, 131.70, 133.45, 141.03, 178.32. MS (ESI+): 286.1 [M acetylsulfanylacetamido)-2-(3′-benzofuran)ethyl]boronic acid
+ H − H2O]+. HRMS (ESI) m/z: calcd for C15H18BNO3S [M + H − (1R,2R,3S,5R)-(−)-2,3-pinanediol ester (16b) as amide. White
H2O]+, 286.1068, found, 286.1071; calcd for C15H18BNO3S [M + solid. HPLC purity: 95.2%. 1H NMR (400 MHz, CDCl3): δ 1.60
Na]+, 326.0993, found, 326.0990. (t, 1H, J = 8.8 Hz), 2.84−2.92 (m, 1H), 2.96−3.09 (m, 2H), 3.47 (d,
[(1R)-1-((2′S)-(3′-Mercapto-2′-methylpropanamido))-2- 2H, J = 6.8 Hz), 6.74 (br, 1H), 7.30−7.39 (m, 2H), 7.60−7.67 (m,
methylpropyl]boronic Acid (MS28). Followed general method using 2H), 7.83 (s, 1H). 13C NMR (100 MHz, CDCl3): δ 25.26, 36.24,
[(1R)-1-((2′S)-(3′-acetylsulfanyl-2′-methylpropanamido))-2- 77.13, 111.41, 118.42, 119.81, 122.45, 124.43, 127.01, 142.01, 157.45,
methylpropyl]boronic acid (1S,2S,3R,5S)-(+)-2,3-pinanediol ester 177.17. MS (ESI+): 262.1 [M + H − H2O]+. HRMS (ESI) m/z:
(14i) as amide. White solid. HPLC purity: 95.4%. 1H NMR (400 calcd for C12H14BNO4S [M + H − H2O]+, 262.0704, found,
MHz, CDCl3): δ 0.91 (t, 6H, J = 7.2 Hz), 1.23 (d, 3H, J = 6.8 Hz), 262.0710; calcd for C12H14BNO4S [M + Na]+, 302.0629, found,
1.60−1.71 (m, 1H), 1.83−1.93 (m, 1H), 2.57−2.66 (m, 3H), 2.75− 302.0633.

7179 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

The NMR and HRMS spectra of MS01−MS33 are given in examined by the jump-dilution recovery method.71 Incubation of 20
Supporting Information. nM VIM-2 enzymes (the enzyme concentration equal to 100-fold of
Constructs, Protein Expression, and Purification. The NDM- that used in the activity assays) was with 9.6 μM MS01, 4.5 μM
1 (aa 1−270), Sfh-I (aa 3−234), GOB-18 (aa 1−290), KPC-2 (aa MS05, 5.6 μM MS18, 0.89 μM MS19, or 6.5 μM L-captopril (i.e., the
29−289), TEM-1 (aa 24−286), AmpC (aa 20−377), and OXA-48 concentration of inhibitor equal to 10-fold of their respective IC50
(aa 1−265) (protein sequences given in Table S5) were cloned into value to VIM-2) for 10 min at room temperature. Then, the samples
pET28a vectors for expression of N-terminally His6-Tagged proteins were rapidly diluted 100-fold by adding the substrate FC-5 containing
that can be cleaved by TEV protease; the cloned VIM-2 (aa 27−266) buffer, followed by measuring the enzyme activity. The reversibility of
plasmids were obtained from C. J. Schofield group in University of the KPC-2 inhibition by MS01, MS05, MS18, MS19, or tazobactam
Oxford. All the proteins were overexpressed in E. coli Transetta (DE3) was determined similarly as described above.
cells (Novagen) at 37 °C using LB medium supplemented with 50 μg· X-ray Crystallography. The crystal structures of VIM-2:MS01
mL−1 ampicillin and 50 μg·mL−1 chloramphenicol (for KPC-2, (PDB code 6J8R) and VIM-2:MS19 (PDB code 6JN6), KPC-2:MS01
additionally complemented with 200 mM sorbitol and 5 mM (PDB code 6J8Q), KPC-2:MS05 (PDB code 6JN3), KPC-2:MS22
betaine). When cells grew to an OD600 of 0.6, the temperature was (PDB code 6JN4), and KPC-2:MS23 (PDB code 6JN5) were
lowered to 20 °C (for VIM-2 and NDM-1), 16 °C (for Sfh-1, GOB- obtained by cocrystallization experiments. The purified VIM-2 and
18, KPC-2, AmpC, and OXA-48), or 27 °C (for TEM-1), and 0.5 mM KPC-2 proteins were freshly prepared to a concentration of 10 mg·
IPTG was then added to induce the protein expression for 18−20 h. mL−1 in the crystallization buffer (for VIM-2, 20 mM Tris-HCl, pH
Cells were harvested by centrifugation (15 min, 4000 rpm), 7.5, 200 mM NaCl, and 0.5 mM TCEP; for KPC-2, 20 mM Tris-HCl,
resuspended in lysis buffer A (20 mM Tris-HCl, pH 8.0, 250 mM pH 8.0, 300 mM NaCl) and were then added with 5 mM inhibitor.
NaCl) supplemented with EDTA-free protease inhibitor, and lysed by The protein/inhibitor mixtures were incubated for 1 h at 4 °C prior to
using an ultrahigh-pressure homogenizer (JNBIO). Cellular debris crystallization. The crystallization drops constituted a 1:1 ratio of
were removed by centrifugation of 15 000 rpm for 30 min, and the protein/inhibitor mixtures/reservoir solutions. The crystallization
supernatant were loaded onto an Ni-NTA column (Roche), followed conditions for VIM-2 are 23−30% PEG 3350, 0.2 M magnesium
by extensive washing with buffer B (20 mM Tris-HCl, pH 8.0, 250
formate, and for KPC-2, conditions are 32−35% PEG 8000, 0.1 M
mM NaCl, 5 mM imidazole) to remove nonspecifically binding
lithium sulfate, 0.05 M sodium acetate, pH 4.5. The VIM-2 and KPC-
proteins. The target proteins were eluted with buffer C (20 mM Tris-
2 protein crystals were cryoprotected using a solution of 19% (v/v)
HCl, pH 8.0, 250 mM NaCl, 250 mM imidazole). Fractions
glycerol and perfluoropolyether oil (from Aladdin), respectively, and
containing the purified enzymes were concentrated using Amicon
Ultra 10K (Millipore) and then desalted using a HiTrap desalting flash-cooled in liquid nitrogen. X-ray diffraction data were obtained at
column (GE Healthcare) into the assay buffer (see Table S6) for the Shanghai Synchrotron Radiation Facility (SSRF) BL19U1
enzyme kinetic analyses. beamline and processed using HKL2000. The molecular replacement
The His-Tags of VIM-2 and KPC-2 proteins were further removed and refinement were carried out as previously described.37,38,50
by TEV protease cleavage (1:100 w/w) at 4 °C overnight and Crystallization conditions are in Table S2, and data collection and
captured on Ni-NTA resin (Roche). Proteins were then loaded onto a refinement statistics are in Tables S3 and S4.
Superdex S75 column equilibrated in buffer D (20 mM Tris-HCl, pH ACE-2 Inhibition Assays. The ACE-2 proteins were purchased
7.5, 200 mM NaCl, 0.5 mM TCEP) for VIM-2 or buffer E (20 mM from Sigma-Aldrich (catalog no. SAE0064), and the substrate Mca-
Tris-HCl, pH 8.0, 300 mM NaCl) for KPC-2, and concentrated for Ala-Pro-Lys(Dnp)-OH (CAS no. 305336-82-7) was purchased from
crystallization. The protein concentrations were determined using a Leon Biological Technology (catalog no. P180529-3). The ACE-2
NanoDrop 2000 spectrophotometer (Thermo Scientic). All the enzymes (4 nM) were incubated with each compound (100 μM or 50
proteins were stored at −80 °C. μM with a final DMSO concentration ≤0.5%) for 10 min at 37 °C.
Enzyme Kinetic Assays. All determinations were performed at Then, the substrate Mca-Ala-Pro-Lys(Dnp)-OH (2 μM) was added to
room temperature (∼25 °C) by using a Thermo microplate reader start the reactions, and the fluorescence was recorded at λex of 328 nm
and 96-well flat-bottom black plates. All the enzymes and substrates and λem of 393 nm at 37 °C using a Thermo microplate reader. All
were dissolved in the assay buffers (Table S6). The activity for all the determinations were performed in triplicate.
purified enzymes was determined using the fluorescent substrate Serine Hydrolyase Inhibition Assays. Cell Culture and Protein
FC548 and monitored by following the variation in fluorescence at λex Preparation. Human embryonic kidney 293T cells (HEK293T) were
of 380 nm and λem of 460 nm. Ten different concentrations of FC5 cultured in Dulbecco’s modified Eagle medium (DMEM) supple-
were used to determine the kinetic parameters (KM and kcat), which mented with 10% fetal bovine serum (FBS), 100 U/mL penicillin, and
were obtained by fitting the initial velocity data to the Michaelis− 100 U/mL streptomycin at 37 °C in a humidified 5% CO2
Menten equation using GraphPad Prism software (La Jolla, CA). atmosphere. Cells were passaged every 2−3 days and harvested by
Compound IC50/Ki Determinations. All the compounds were suspending them in 10 mL of medium. The suspension was
freshly prepared in 100 mM DMSO stock solutions. The IC50 values centrifuged for 5 min at 1000 rpm, and the supernatant was removed.
were determined by preincubation of the appropriate amount of The cell pellet was flash-frozen in liquid nitrogen and stored at −80
enzymes with the desired compound (with 10 different concen- °C. Cell pellets were thawed on ice and resuspended in lysis buffer
trations in 3-fold dilution) in the assay buffer for 10 min, followed by (20 mM HEPES, pH 7.2, 1 mM MgCl2, 0.25 M sucrose, 2 mM DTT,
adding the substrate FC5 to initiate the reactions and monitoring the 25 U/mL Benzonase). The suspension was homogenized by pipet.
fluorescence at λex of 380 nm and λem of 460 nm. All determinations Protein concentration was determined by NanoDrop 2000
were tested in triplicate. The IC50/pIC50/se pIC50 values were spectrophotometer (Thermo Scientic). The protein fractions were
obtained from the plot of activity versus inhibitor concentration using diluted to a total protein concentration of 2 mg/mL and stored in
the GraphPad Prism software. All the IC50 curves are given in small aliquots at −80 °C.
Supporting Information Figures S2, S8, and S10. The Ki values were E. coli Transetta (DE3) cells (Novagen) were growing at 37 °C
calculated from the IC50 values using the Cheng−Prusoff equation:49 using LB medium as described above. Cells were harvested by
centrifugation (15 min, 4000 rpm), followed by resuspending in the
IC50
Ki = lysis buffer (20 mM Tris-HCl, pH 8.0, 250 mM NaCl) supplemented
[S]
1+ KM
with EDTA-free protease inhibitor, and then lysed using an ultrahigh-
pressure homogenizer (JNBIO). Cellular debris was removed by
where [S] is the FC5 concentration for activity test (Table S6) and centrifugation of the lysate at 15 000 rpm for 30 min, and the
KM is the Michaelis constant of the enzymes (Table S1). supernatant was resuspended in the lysis buffer (20 mM HEPES, pH
Jump Dilution Recovery Assays. The reversibility of the VIM-2 7.2, 1 mM MgCl2, 0.25 M sucrose, 2 mM DTT, 25 U/mL
inhibition by MS01, MS05, MS18, MS19, or L-captopril was Benzonase). The protein fractions were diluted to a total protein

7180 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

concentration of 5 mg/mL and stored in small aliquots at −80 °C for data for the intermediates that are not included in the
later use. main text; and NMR and HRMS spectra for the target
Selectivity Profile by Activity-Based Protein Profiling (ABPP). For compounds (PDF)
gel-based ABPP experiments, the extracted proteome from HEK293T
and E. coli cells was preincubated for 30 min with vehicle (DMSO) or Molecular formula strings and the MBL/SBL Ki data
inhibitor (50 μM) at rt, followed by the treatment with activity-based (CSV)
probe TAMRA-FP (750 nM, final concentration) (from Thermo Molecular docking model of GOB-18:MS01 (PDB)
Fischer Scientific) for 20 min at rt. The reactions were quenched with Molecular docking model of Sfh-I:MS01 (PDB)
5 μL of standard 5× SDS−PAGE loading buffer. The samples were
directly loaded and resolved on SDS−PAGE gel (12% acrylamide). Accession Codes
The gels were then scanned with a ChemiDoc MP system (Cy 3 The coordinates and structure factors of VIM-2:MS01 (PDB
settings, 605/50 filter) and analyzed using Image Lab 4.1. code 6J8R), VIM-2:MS19 (PDB code 6JN6), KPC-2:MS01
Microbiological Susceptibility Testing. The inhibitors MS01, (PDB code 6J8Q), KPC-2:MS05 (PDB code 6JN3), KPC-
MS18, and MS19 (L-captopril and tazobactam as control com- 2:MS22 (PDB code 6JN4), and KPC-2:MS23 (PDB code
pounds) were tested against seven clinically isolated bacterial strains 6JN5) complex structures have been deposited with the
producing NDM-1, KPC-2 or AmpC, including E. coli BAA-2452
Protein Data Bank. Authors will release the atomic coordinates
(blaNDM‑1), K. pneumoniae 13249 (blaNDM‑1), E. coli BAA-2340
and experimental data upon article publication.


(blaKPC‑2), E. coli 11119 (blaKPC‑2), K. pneumoniae BAA-1705
(blaKPC‑2), K. pneumoniae 5846 (blaKPC‑2), and K. pneumoniae C660
(blaAmpC). The MIC values of meropenem against the bacterial strains AUTHOR INFORMATION
were determined with or without the inhibitors by broth micro- Corresponding Authors
dilution according to the Clinical Laboratory Standards Institute *Y.W.: phone, +86-028-85503235; e-mail, wyong@scu.edu.cn.
(CLSI) guidelines.72 Meropenem and inhibitors were first dissolved in *G.-B.L.: phone, +86-135-5016-1826; e-mail, liguobo@scu.
DMSO and then diluted in cation-adjusted Mueller−Hinton II broth edu.cn.
(CAMHB). The 0.5 McFarland suspension of the bacterial strains in
0.85% NaCl was diluted in CAMHB broth. The inoculum were ORCID
seeded in 96-well microtiter plates to make the final concentration of Yong Wu: 0000-0003-0719-4963
5 × 105 CFU/mL and treated with meropenem (128−0.25 μg/mL in Guo-Bo Li: 0000-0002-4915-6677
2-fold dilution) and/or the inhibitors (50 μM). The microtiter plates
were incubated at 37 °C for 16−20 h, followed by visual evaluation of Author Contributions
§
the bacterial growth. Y.-L.W. and S.L. are co-first-authors. G.B.L. and Y.W.
Cytotoxicity Assays. HEK293 cells were plated at ∼5000 cells/ designed the project. Y.-L.W, M.-Y.H., Y.L., Q.M., and Y.Z.
well in 96-well plate 24 h prior to treatment with the inhibitors at a performed molecule design and chemical synthesies and
concentration of 100 μM and incubated for another 72 h. 20 μL of analyzed the NMR and mass spectra, guided by G.-B.L. and
MTT solution (5.0 mg/mL) was added to the medium and incubated Y.W.; S.L., Z.-J.Y., and Y.-H.Y. performed protein production
for another 4 h. Then, 150 μL of DMSO was added to each well after and kinetic experiments, guided by G.-B.L.; S.L., G.-B.L., C.W.,
removing MTT solution. The absorbance was read at 570 nm Y.Y., and Q.C. performed crystal experiments and structural
wavelength on an automatic microplate spectrophotometer.
refinements; S.L. and H.D. performed the selectivity assays;
Molecular Docking and LEADOPT Optimization. The
molecular docking simulations were carried out by using AutoDock Y.S. and Z.W. performed microbiological susceptibility testing;
Vina program.73 The crystal structures of Sfh-I (PDB code 3SD9)53 G.-B.L., Y.-L.W., and S.L. wrote the manuscript. All authors
and GOB-18 (PDB code 5K0W)74 were used as the protein read and approved the manuscript.
templates. The protein and ligand structures were prepared as Notes
previously described.32,75 The docking poses were viewed using the The authors declare no competing financial interest.


PyMOL program. The LEADOPT program61 was used to perform
fragment-growing analyses, which involves a fragment library ACKNOWLEDGMENTS
consisting of 6877 unique fragments. The binding conformations of
MS01 with VIM-2 (PDB code 6J8R) and KPC-2 (PDB code 6J8Q) This work was supported by the funds from the National
were kept unchanged, and the two hydrogens at the 1-position of Natural Science Foundation of China (Grants 81874291,
MS01 were set as the fragment growing points. According the binding 8177357, 81502989, and 21807076), the Sichuan Science and
site information, LEADOPT generated a number of potential Technology Program (Grant 2018HH0100), the Scientic
derivatives with predicted binding affinities. The top-ranked Research Foundation of Sichuan University (Grants
compounds were visually inspected and further considered with 2082604401098 and 20822041A4193), and the Fundamental
respect to the synthetic accessibility.


Research Funds for the Central Universities. The authors
thank the staff of BL19U1 beamline of the National Center for
ASSOCIATED CONTENT
Protein Science Shanghai at Shanghai Synchrotron Radiation
*
S Supporting Information
Facility for assistance during data collection.


The Supporting Information is available free of charge on the
ACS Publications website at DOI: 10.1021/acs.jmed- ABBREVIATIONS USED
chem.9b00735. MBL, metallo-β-lactamase; SBL, serine-β-lactamase; SAR,
Tables showing kinetic data for FC5 with MBL and SBL structure−activity relationship; MBP, metal-binding pharma-
enzymes, enzymatic assay conditions, crystallization cophore; ACE, angiotensin-converting enzyme; VIM-2, Verona
conditions, crystallographic data collection and refine- integron-encoded MBL-2; NDM-1, New Delhi MBL-1; KPC-
ment statistics; figures showing Michaelis−Menten plots 2, Klebsiella pneumoniae carbapenemase-2; OXA-48, oxacilli-
of enzymes catalyzed hydrolysis of FC5, IC50/Ki curves nase-48; TAMRA-FP, carboxytetramethylrhodamine fluoro-
of dual-action inhibitors, modes of inhibitor binding as phosphonate; IPTG, isopropyl β-D-1-thiogalactopyranoside;
defined by electron density maps, and structure EDTA, ethylenediaminetetraacetic acid; TCEP, tris(2-
comparison; synthetic methods and characterization carboxyethyl)phosphine; DMSO, dimethyl sulfoxide; DTT,
7181 DOI: 10.1021/acs.jmedchem.9b00735
J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

dithiothreitol; SDS−PAGE, sodium dodecyl sulfate−polyacry- (20) Khan, A. U.; Maryam, L.; Zarrilli, R. Structure, genetics and
lamide gel electrophoresis; MTT, 3-(4,5-dimethyl-2-thiazolyl)- worldwide spread of New Delhi metallo-β-lactamase (NDM): a threat
2,5-diphenyl-2H-tetrazolium bromide; CFU, colony-forming to public health. BMC Microbiol. 2017, 17, 101.
unit; aa, amino acids; PDB, Protein Data Bank; n-BuLi, n- (21) Docquier, J.-D.; Mangani, S. An update on β-lactamase
inhibitor discovery and development. Drug Resist. Updates 2018, 36,
butyllithium; THF, tetrahydrofuran; LiHMDS, lithium bis-
13−29.
(trimethylsilyl)amide; TBTU, O-(benzotriazol-1-yl)- (22) Hecker, S. J.; Reddy, K. R.; Totrov, M.; Hirst, G. C.;
N,N,N′,N′-tetramethyluronium tetrafluoroborate; DIPEA, Lomovskaya, O.; Griffith, D. C.; King, P.; Tsivkovski, R.; Sun, D.;
N,N-diisopropylethylamine; DMF, N,N-dimethylformamide; Sabet, M.; Tarazi, Z.; Clifton, M. C.; Atkins, K.; Raymond, A.; Potts,
NaOtBu, sodium tert-butoxide; Et2O, diethyl ether; CH2Cl2, K. T.; Abendroth, J.; Boyer, S. H.; Loutit, J. S.; Morgan, E. E.; Durso,
dichloromethane; HCl, hydrogen chloride; CuBr, cupric S.; Dudley, M. N. Discovery of a cyclic boronic acid β-lactamase
bromide; Na2SO4, sodium sulfate; TLC, thin-layer chromatog- inhibitor (RPX7009) with utility vs class A serine carbapenemases. J.
raphy; NaOH, sodium hydroxide Med. Chem. 2015, 58, 3682−3692.

■ REFERENCES
(1) Garber, K. A β-lactamase inhibitor revival provides new hope for
(23) Lisa, M.-N.; Palacios, A. R.; Aitha, M.; González, M. M.;
Moreno, D. M.; Crowder, M. W.; Bonomo, R. A.; Spencer, J.;
Tierney, D. L.; Llarrull, L. I.; Vila, A. J. A general reaction mechanism
for carbapenem hydrolysis by mononuclear and binuclear metallo-β-
old antibiotics. Nat. Rev. Drug Discovery 2015, 14, 445−447.
(2) Bush, K. Game changers: new β-lactamase inhibitor combina- lactamases. Nat. Commun. 2017, 8, 538−549.
tions targeting antibiotic resistance in Gram-negative bacteria. ACS (24) Potter, R. F.; D’Souza, A. W.; Dantas, G. The rapid spread of
Infect. Dis. 2018, 4, 84−87. carbapenem-resistant Enterobacteriaceae. Drug Resist. Updates 2016,
(3) Chaudhary, A. S. A review of global initiatives to fight antibiotic 29, 30−46.
resistance and recent antibiotics’ discovery. Acta Pharm. Sin. B 2016, (25) Bush, K. A resurgence of β-lactamase inhibitor combinations
6, 552−556. effective against multidrug-resistant Gram-negative pathogens. Int. J.
(4) Bush, K.; Bradford, P. A. Interplay between β-lactamases and Antimicrob. Agents 2015, 46, 483−493.
new β-lactamase inhibitors. Nat. Rev. Microbiol. 2019, 17, 295−306. (26) Brem, J.; Cain, R.; Cahill, S.; McDonough, M. A.; Clifton, I. J.;
(5) Bush, K.; Page, M. G. P. What we may expect from novel Jiménez-Castellanos, J.-C.; Avison, M. B.; Spencer, J.; Fishwick, C. W.
antibacterial agents in the pipeline with respect to resistance and G.; Schofield, C. J. Structural basis of metallo-β-lactamase, serine-β-
pharmacodynamic principles. J. Pharmacokinet. Pharmacodyn. 2017, lactamase and penicillin-binding protein inhibition by cyclic
44, 113−132. boronates. Nat. Commun. 2016, 7, 12406−12413.
(6) Bush, K.; Jacoby, G. A. Updated functional classification of β- (27) Cahill, S. T.; Cain, R.; Wang, D. Y.; Lohans, C. T.; Wareham,
lactamases. Antimicrob. Agents Chemother. 2010, 54, 969−976. D. W.; Oswin, H. P.; Mohammed, J.; Spencer, J.; Fishwick, C. W.;
(7) Crowder, M. W.; Spencer, J.; Vila, A. J. Metallo-β-lactamases: McDonough, M. A.; Schofield, C. J.; Brem, J. Cyclic boronates inhibit
novel weaponry for antibiotic resistance in bacteria. Acc. Chem. Res. all classes of β-lactamases. Antimicrob. Agents Chemother. 2017, 61,
2006, 39, 721−728. e02260-16.
(8) King, A. M.; Reid-Yu, S. A.; Wang, W.; King, D. T.; De Pascale, (28) Cendron, L.; Quotadamo, A.; Maso, L.; Bellio, P.; Montanari,
G.; Strynadka, N. C.; Walsh, T. R.; Coombes, B. K.; Wright, G. D. M.; Celenza, G.; Venturelli, A.; Costi, M. P.; Tondi, D. X-ray
Aspergillomarasmine A overcomes metallo-β-lactamase antibiotic crystallography deciphers the activity of broad-spectrum boronic acid
resistance. Nature 2014, 510, 503−506. β-lactamase inhibitors. ACS Med. Chem. Lett. 2019, 10, 650−655.
(9) Tondi, D.; Cross, S.; Venturelli, A.; Costi, M. P.; Cruciani, G.; (29) Santucci, M.; Spyrakis, F.; Cross, S.; Quotadamo, A.; Farina, D.;
Spyrakis, F. Decoding the structural basis for carbapenem hydrolysis Tondi, D.; De Luca, F.; Docquier, J. D.; Prieto, A. I.; Ibacache, C.;
by class A β-lactamases: fishing for a pharmacophore. Curr. Drug Blazquez, J.; Venturelli, A.; Cruciani, G.; Costi, M. P. Computational
Targets 2016, 17, 983−1005. and biological profile of boronic acids for the detection of bacterial
(10) Docquier, J.-D.; Mangani, S. Structure-function relationships of serine- and metallo-β-lactamases. Sci. Rep. 2017, 7, 17716.
class D carbapenemases. Curr. Drug Targets 2016, 17, 1061−1071. (30) Abboud, M. I.; Kosmopoulou, M.; Krismanich, A.; Johnson, J.
(11) Queenan, A. M.; Bush, K. Carbapenemases: the versatile β- W.; Hinchliffe, P.; Brem, J.; Claridge, T. D. W.; Spencer, J.; Schofield,
lactamases. Clin. Microbiol. Rev. 2007, 20, 440−458. C.; Dmitrienko, G. I. Cyclobutanone mimics of intermediates in
(12) Bradford, P. A. Extended-spectrum β-lactamases in the 21st metallo-β-lactamase catalysis. Chem. - Eur. J. 2018, 24, 5734−5737.
century: characterization, epidemiology, and detection of this (31) Zhang, C.; Pu, Y.-c.; Yu, Z.-J.; Wu, C.-y.; Brem, J.; McDonough,
important resistance threat. Clin. Microbiol. Rev. 2001, 14, 933−951. M. A.; Schofield, C. J.; Li, G.-B.; Wu, Y. Rh(iii)-catalyzed directed C-
(13) Naas, T.; Poirel, L.; Nordmann, P. Minor extended-spectrum β- H carbenoid coupling reveals aromatic bisphosphonates inhibiting
lactamases. Clin. Microbiol. Infect. 2008, 14, 42−52. metallo- and serine-β-lactamases. Org. Chem. Front. 2018, 5, 1288−
(14) Mojica, M. F.; Bonomo, R. A.; Fast, W. B1-metallo-β- 1292.
lactamases: Where do we stand? Curr. Drug Targets 2016, 17, (32) Yu, Z.-J.; Liu, S.; Zhou, S.; Li, H.; Yang, F.; Yang, L.-L.; Wu, Y.;
1029−1050. Guo, L.; Li, G.-B. Virtual target screening reveals rosmarinic acid and
(15) Vella, P.; Miraula, M.; Phelan, E.; Leung, E. W.; Ely, F.; Ollis, salvianolic acid A inhibiting metallo- and serine-β-lactamases. Bioorg.
D. L.; McGeary, R. P.; Schenk, G.; Mitic, N. Identification and Med. Chem. Lett. 2018, 28, 1037−1042.
characterization of an unusual metallo-β-lactamase from Serratia (33) Frere, J.-M.; Sauvage, E.; Kerff, F. From “an enzyme able to
proteamaculans. JBIC, J. Biol. Inorg. Chem. 2013, 18, 855−863. destroy penicillin” to carbapenemases: 70 years of β-lactamase
(16) Hou, C.-F. D.; Phelan, E. K.; Miraula, M.; Ollis, D. L.; Schenk, misbehaviour. Curr. Drug Targets 2016, 17, 974−982.
G.; Mitić, N. Unusual metallo-β-lactamases may constitute a new (34) Linciano, P.; Cendron, L.; Gianquinto, E.; Spyrakis, F.; Tondi,
subgroup in this family of enzymes. Am. J. Mol. Biol. 2014, 4, 11−15. D. Ten years with New Delhi Metallo-β-lactamase-1 (NDM-1): from
(17) Palzkill, T. Metallo-β-lactamase structure and function. Ann. N. structural insights to inhibitor design. ACS Infect. Dis. 2019, 5, 9−34.
Y. Acad. Sci. 2013, 1277, 91−104. (35) Cohen, S. M. A bioinorganic approach to fragment-based drug
(18) Raczynska, J. E.; Shabalin, I. G.; Minor, W.; Wlodawer, A.; discovery targeting metalloenzymes. Acc. Chem. Res. 2017, 50, 2007−
Jaskolski, M. A close look onto structural models and primary ligands 2016.
of metallo-β-lactamases. Drug Resist. Updates 2018, 40, 1−12. (36) Brem, J.; van Berkel, S. S.; Zollman, D.; Lee, S. Y.; Gileadi, O.;
(19) Ju, L.-C.; Cheng, Z.; Fast, W.; Bonomo, R. A.; Crowder, M. W. McHugh, P. J.; Walsh, T. R.; McDonough, M. A.; Schofield, C. J.
The continuing challenge of metallo-β-lactamase inhibition: mecha- Structural basis of metallo-β-lactamase inhibition by captopril
nism matters. Trends Pharmacol. Sci. 2018, 39, 635−647. stereoisomers. Antimicrob. Agents Chemother. 2016, 60, 142−150.

7182 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

(37) Li, G.-B.; Brem, J.; Lesniak, R.; Abboud, M. I.; Lohans, C. T.; R.; Bhagwat, S. S.; Patel, M. V.; van den Akker, F.; Bonomo, R. A.
Clifton, I. J.; Yang, S.-Y.; Jimenez-Castellanos, J.-C.; Avison, M. B.; Strategic approaches to overcome resistance against Gram-negative
Spencer, J.; McDonough, M. A.; Schofield, C. J. Crystallographic pathogens using β-lactamase inhibitors and β-lactam enhancers:
analyses of isoquinoline complexes reveal a new mode of metallo-β- activity of three novel diazabicyclooctanes WCK 5153, Zidebactam
lactamase inhibition. Chem. Commun. 2017, 53, 5806−5809. (WCK 5107), and WCK 4234. J. Med. Chem. 2018, 61, 4067−4086.
(38) Liu, S.; Jing, L.; Yu, Z.-J.; Wu, C.; Zheng, Y.; Zhang, E.; Chen, (52) Pemberton, O. A.; Zhang, X.; Chen, Y. Molecular basis of
Q.; Yu, Y.; Guo, L.; Wu, Y.; Li, G.-B. ((S)-3-Mercapto-2- substrate recognition and product release by the Klebsiella pneumo-
methylpropanamido)acetic acid derivatives as metallo-β-lactamase niae carbapenemase (KPC-2). J. Med. Chem. 2017, 60, 3525−3530.
inhibitors: synthesis, kinetic and crystallographic studies. Eur. J. Med. (53) Hinchliffe, P.; Gonzalez, M. M.; Mojica, M. F.; Gonzalez, J. M.;
Chem. 2018, 145, 649−660. Castillo, V.; Saiz, C.; Kosmopoulou, M.; Tooke, C. L.; Llarrull, L. I.;
(39) Leiris, S.; Coelho, A.; Castandet, J.; Bayet, M.; Lozano, C.; Mahler, G.; Bonomo, R. A.; Vila, A. J.; Spencer, J. Cross-class metallo-
Bougnon, J.; Bousquet, J.; Everett, M.; Lemonnier, M.; Sprynski, N.; β-lactamase inhibition by bisthiazolidines reveals multiple binding
Zalacain, M.; Pallin, T. D.; Cramp, M. C.; Jennings, N.; Raphy, G.; modes. Proc. Natl. Acad. Sci. U. S. A. 2016, 113, E3745−E3754.
Jones, M. W.; Pattipati, R.; Shankar, B.; Sivasubrahmanyam, R.; (54) Lienard, B. M. R.; Garau, G.; Horsfall, L.; Karsisiotis, A. I.;
Soodhagani, A. K.; Juventhala, R. R.; Pottabathini, N.; Pothukanuri, Damblon, C.; Lassaux, P.; Papamicael, C.; Roberts, G. C. K.; Galleni,
S.; Benvenuti, M.; Pozzi, C.; Mangani, S.; De Luca, F.; Cerboni, G.; M.; Dideberg, O.; Frere, J.-M.; Schofield, C. J. Structural basis for the
Docquier, J.-D.; Davies, D. T. SAR studies leading to the broad-spectrum inhibition of metallo-beta-lactamases by thiols. Org.
identification of a novel series of metallo-β-lactamase inhibitors for Biomol. Chem. 2008, 6, 2282−2294.
the treatment of carbapenem-resistant Enterobacteriaceae infections (55) Caselli, E.; Romagnoli, C.; Vahabi, R.; Taracila, M. A.;
that display efficacy in an animal infection model. ACS Infect. Dis. Bonomo, R. A.; Prati, F. Click chemistry in lead optimization of
2019, 5, 131−140. boronic acids as β-lactamase inhibitors. J. Med. Chem. 2015, 58,
(40) Brem, J.; van Berkel, S. S.; Aik, W.; Rydzik, A. M.; Avison, M. 5445−5458.
B.; Pettinati, I.; Umland, K.-D.; Kawamura, A.; Spencer, J.; Claridge, (56) Celenza, G.; Vicario, M.; Bellio, P.; Linciano, P.; Perilli, M.;
T. D.; McDonough, M. A.; Schofield, C. J. Rhodanine hydrolysis leads Oliver, A.; Blazquez, J.; Cendron, L.; Tondi, D. Phenylboronic acid
to potent thioenolate mediated metallo-β-lactamase inhibition. Nat. derivatives as validated leads active in clinical strains overexpressing
Chem. 2014, 6, 1084−1090. KPC-2: a step against bacterial resistance. ChemMedChem 2018, 13,
(41) Cain, R.; Brem, J.; Zollman, D.; McDonough, M. A.; Johnson, 713−724.
R. M.; Spencer, J.; Makena, A.; Abboud, M. I.; Cahill, S.; Lee, S. Y.; (57) Minasov, G.; Wang, X.; Shoichet, B. K. An ultrahigh resolution
McHugh, P. J.; Schofield, C. J.; Fishwick, C. W. G. In silico fragment- structure of TEM-1 β-lactamase suggests a role for Glu166 as the
based design identifies subfamily B1 metallo-β-lactamase inhibitors. J. general base in acylation. J. Am. Chem. Soc. 2002, 124, 5333−5340.
Med. Chem. 2018, 61, 1255−1260. (58) Caselli, E.; Powers, R. A.; Blasczcak, L. C.; Wu, C. Y. E.; Prati,
(42) Hiraiwa, Y.; Saito, J.; Watanabe, T.; Yamada, M.; Morinaka, A.;
F.; Shoichet, B. K. Energetic, structural, and antimicrobial analyses of
Fukushima, T.; Kudo, T. X-ray crystallographic analysis of IMP-1
β-lactam side chain recognition by β-lactamases. Chem. Biol. 2001, 8,
metallo-β-lactamase complexed with a 3-aminophthalic acid deriva-
17−31.
tive, structure-based drug design, and synthesis of 3,6-disubstituted
(59) King, A. M.; King, D. T.; French, S.; Brouillette, E.; Asli, A.;
phthalic acid derivative inhibitors. Bioorg. Med. Chem. Lett. 2014, 24,
Alexander, J. A. N.; Vuckovic, M.; Maiti, S. N.; Parr, T. R.; Brown, E.
4891−4894.
D.; Malouin, F.; Strynadka, N. C. J.; Wright, G. D. Structural and
(43) Krajnc, A.; Lang, P. A.; Panduwawala, T. D.; Brem, J.;
Schofield, C. J. Will morphing boron-based inhibitors beat the β- kinetic characterization of diazabicyclooctanes as dual inhibitors of
lactamases? Curr. Opin. Chem. Biol. 2019, 50, 101−110. both serine-β-lactamases and penicillin-binding proteins. ACS Chem.
(44) Zhou, J.; Stapleton, P.; Haider, S.; Healy, J. Boronic acid Biol. 2016, 11, 864−868.
inhibitors of the class A β-lactamase KPC-2. Bioorg. Med. Chem. 2018, (60) Ke, W.; Bethel, C. R.; Papp-Wallace, K. M.; Pagadala, S. R. R.;
26, 2921−2927. Nottingham, M.; Fernandez, D.; Buynak, J. D.; Bonomo, R. A.; van
(45) Nguyen, N. Q.; Krishnan, N. P.; Rojas, L. J.; Prati, F.; Caselli, den Akker, F. Crystal structures of KPC-2 β-lactamase in complex
E.; Romagnoli, C.; Bonomo, R. A.; van den Akker, F. Crystal with 3-nitrophenyl boronic acid and the penam sulfone PSR-3-226.
structures of KPC-2 and SHV-1 β-lactamases in complex with the Antimicrob. Agents Chemother. 2012, 56, 2713−2718.
boronic acid transition state analog S02030. Antimicrob. Agents (61) Li, G.-B.; Ji, S.; Yang, L.-L.; Zhang, R.-J.; Chen, K.; Zhong, L.;
Chemother. 2016, 60, 1760−1766. Ma, S.; Yang, S.-Y. LEADOPT: an automatic tool for structure-based
(46) Tondi, D.; Calò, S.; Shoichet, B. K.; Costi, M. P. Structural lead optimization, and its application in structural optimizations of
study of phenyl boronic acid derivatives as AmpC β-lactamase VEGFR2 and SYK inhibitors. Eur. J. Med. Chem. 2015, 93, 523−538.
inhibitors. Bioorg. Med. Chem. Lett. 2010, 20, 3416−3419. (62) Struth, F. R.; Hirschhäuser, C. A modular approach to the
(47) Tondi, D.; Venturelli, A.; Bonnet, R.; Pozzi, C.; Shoichet, B. K.; asymmetric synthesis of cytisine. Eur. J. Org. Chem. 2016, 2016, 958−
Costi, M. P. Targeting class A and C serine β-lactamases with a broad- 964.
spectrum boronic acid derivative. J. Med. Chem. 2014, 57, 5449−5458. (63) Putty, S.; Rai, A.; Jamindar, D.; Pagano, P.; Quinn, C. L.; Mima,
(48) van Berkel, S. S.; Brem, J.; Rydzik, A. M.; Salimraj, R.; Cain, R.; T.; Schweizer, H. P.; Gutheil, W. G. Characterization of d-boroAla as
Verma, A.; Owens, R. J.; Fishwick, C. W.; Spencer, J.; Schofield, C. J. a novel broad spectrum antibacterial agent targeting d-Ala-d-Ala
Assay platform for clinically relevant metallo-β-lactamases. J. Med. ligase. Chem. Biol. Drug Des. 2011, 78, 757−763.
Chem. 2013, 56, 6945−6953. (64) Beenen, M. A.; An, C.; Ellman, J. A. Asymmetric copper-
(49) Cheng, Y.-C.; Prusoff, W. H. Relationship between the catalyzed synthesis of α-amino boronate esters from N-tert-
inhibition constant (K1) and the concentration of inhibitor which butanesulfinyl aldimines. J. Am. Chem. Soc. 2008, 130, 6910−6911.
causes 50 percent inhibition (I50) of an enzymatic reaction. Biochem. (65) Ito, H.; Kubota, K. Copper(I)-catalyzed boryl substitution of
Pharmacol. 1973, 22, 3099−3108. unactivated alkyl halides. Org. Lett. 2012, 14, 890−893.
(50) Li, G.-B.; Abboud, M. I.; Brem, J.; Someya, H.; Lohans, C. T.; (66) Matteson, D. S.; Sadhu, K. M. Boronic ester homologation with
Yang, S.-Y.; Spencer, J.; Wareham, D. W.; McDonough, M. A.; 99% chiral selectivity and its use in syntheses of the insect
Schofield, C. J. NMR-filtered virtual screening leads to non-metal pheromones (3S,4S)-4-methyl-3-heptanol and exo-brevicomin. J.
chelating metallo-β-lactamase inhibitors. Chem. Sci. 2017, 8, 928−937. Am. Chem. Soc. 1983, 105, 2077−2078.
(51) Papp-Wallace, K. M.; Nguyen, N. Q.; Jacobs, M. R.; Bethel, C. (67) Matteson, D. S.; Sadhu, K. M.; Peterson, M. L. 99% Chirally
R.; Barnes, M. D.; Kumar, V.; Bajaksouzian, S.; Rudin, S. D.; Rather, selective synthesis via pinanediol boronic esters: insect pheromones,
P. N.; Bhavsar, S.; Ravikumar, T.; Deshpande, P. K.; Patil, V.; Yeole, diols, and an amino alcohol. J. Am. Chem. Soc. 1986, 108, 810−819.

7183 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184
Journal of Medicinal Chemistry Article

(68) Matteson, D. S.; Ray, R.; Rocks, R. R.; Tsai, D. J. S. Directed


chiral synthesis by way of .alpha.-chloro boronic esters. Organo-
metallics 1983, 2, 1536−1543.
(69) Zhu, Y.; Wu, G.; Zhu, X.; Ma, Y.; Zhao, X.; Li, Y.; Yuan, Y.;
Yang, J.; Yu, S.; Shao, F.; Lei, M. Synthesis, in vitro and in vivo
biological evaluation, and comprehensive understanding of structure−
activity relationships of dipeptidyl boronic acid proteasome inhibitors
constructed from β-amino acids. J. Med. Chem. 2010, 53, 8619−8626.
(70) Kostova, M. B.; Rosen, D. M.; Chen, Y.; Mease, R. C.;
Denmeade, S. R. Structural optimization, biological evaluation, and
application of peptidomimetic prostate specific antigen inhibitors. J.
Med. Chem. 2013, 56, 4224−4235.
(71) Copeland, R. A. Evaluation of Enzyme Inhibitors in Drug
Discovery: A Guide for Medicinal Chemists and Pharmacologists; John
Wiley & Sons, Inc.: Hoboken, NJ, 2013.
(72) Performance Standards for Antimicrobial Susceptibility Testing;
Document M100-S27; 27th Informational Supplement; Clinical and
Laboratory Standards Institute: Wayne, PA, 2017.
(73) Trott, O.; Olson, A. J. AutoDock Vina: improving the speed
and accuracy of docking with a new scoring function, efficient
optimization, and multithreading. J. Comput. Chem. 2009, 31, 455−
461.
(74) Morán-Barrio, J.; Lisa, M.-N.; Larrieux, N.; Drusin, S. I.; Viale,
A. M.; Moreno, D. M.; Buschiazzo, A.; Vila, A. J. Crystal structure of
the metallo-β-lactamase GOB in the periplasmic dizinc form reveals
an unusual metal site. Antimicrob. Agents Chemother. 2016, 60, 6013−
6022.
(75) Li, G.-B.; Yu, Z.-J.; Liu, S.; Huang, L.-Y.; Yang, L.-L.; Lohans,
C. T.; Yang, S.-Y. IFPTarget: a customized virtual target identification
method based on protein−ligand interaction fingerprinting analyses. J.
Chem. Inf. Model. 2017, 57, 1640−1651.

7184 DOI: 10.1021/acs.jmedchem.9b00735


J. Med. Chem. 2019, 62, 7160−7184

You might also like