DOI:
10.1039/D5MD00393H
(Research Article)
RSC Med. Chem., 2025, Advance Article
Identification of novel N-benzyloxy-amino acid hydroxamates as inhibitors of the virulence factor LasB from Pseudomonas aeruginosa†
Received
6th May 2025
, Accepted 19th July 2025
First published on 13th August 2025
Abstract
The emergence of multidrug-resistant pathogens, particularly Pseudomonas aeruginosa, represents a global health concern. Among its major virulence factors, elastase B (LasB), a zinc-dependent metalloprotease, plays a pivotal role in host tissue degradation, immune evasion, and biofilm formation. Targeting LasB with selective inhibitors offers a promising therapeutic strategy to mitigate bacterial virulence while minimizing selective pressure for resistance development. In this study, a series of N-benzyloxy amino acid derivatives were designed, synthesized, and evaluated for their inhibitory activity against LasB. Structure-based optimization led to the identification of compound 12 as the most potent inhibitor (Ki = 0.92 μM), exhibiting high selectivity for LasB over human matrix metalloproteinases. Cell-based assays demonstrated its ability to inhibit LasB proteolytic activity and reduce biofilm formation without affecting bacterial viability. These findings highlight the potential of LasB inhibitors as pathoblockers, providing a targeted approach to disarm bacterial virulence rather than exerting bactericidal pressure.
1. Introduction
Over the past decade, the healthcare system worldwide has been faced with enormous challenges due to the burden of ravaging bacterial infections. Such issues include nosocomial infections (NIs) usually caused by both Gram-negative and Gram-positive bacterial pathogens1 which primarily affect patients with compromised immune systems. Among the major contributors to NIs are the “ESKAPE” group of pathogens, which includes Gram-positive bacteria such as Enterococcus faecium and Staphylococcus aureus, and Gram-negative bacteria, which include Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.2,3 Due to their antibiotic resistance, ESKAPE pathogens are associated with the highest risk of mortality thereby resulting in increased health care costs.4 The bacterial priority pathogens list released by the World Health Organization in 2024 included P. aeruginosa in the high priority category. P. aeruginosa is generally found in patients with chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), different types of cancer, traumatic wounds, septicemia, and ventilator-associated pneumonia (VAP).5–12 In addition to the rapid appearance of genetic mutations and adaptive resistance mechanisms to antibiotics, the difficulty of eradicating P. aeruginosa infections is due to its ability to form biofilm.13 Biofilm-residing bacteria exhibit significantly increased antibiotic resistance, up to 1000 times more than planktonic bacteria.14 The treatment of P. aeruginosa infections urgently requires the development of new antibacterials with novel mechanisms of action to overcome these resistance strategies.15 Considering these issues, medicinal chemistry research has focused on the development of new putative therapeutics known as “pathoblockers” which are described as antibacterial agents that specifically target key pathogenic processes and seek to reduce bacterial virulence rather than kill the bacteria. The aim is to reduce selection pressure and the likelihood of resistance development.16–18 P. aeruginosa secretes several virulence factors, including four major proteases: elastase B (LasB), elastase A (LasA), alkaline protease (AprA), and protease IV.19 Among these, LasB plays a key role in the virulence of P. aeruginosa and therefore has become a central focus of research. LasB, also known as pseudolysin, is an extracellular zinc-dependent metalloprotease with multiple activities. It has been clearly shown to degrade several important host proteins, including elastin, collagen, fibrinogen, immunoglobulins, and complement components.20 This degradation leads to extensive tissue damage and enables P. aeruginosa to evade the host immune response. Furthermore, emerging evidence indicates that LasB is implicated in respiratory infections, particularly in patients with cystic fibrosis (CF).10 In these patients, LasB compromises lung structural integrity by degrading elastin and components of the extracellular matrix. Other studies have shown that LasB can modulate immune responses through the degradation of cytokines and other immune mediators, resulting in the weakening of the host defense mechanisms.21 Moreover, LasB promotes biofilm formation, which increases the persistence of bacteria and their resistance to antimicrobial therapy.22 Given its central role in pathogenesis, LasB is considered a promising therapeutic target for the treatment of P. aeruginosa infections. Consequently, a combined approach using a LasB inhibitor and common antibiotics could improve therapeutic outcomes by hindering tissue degradation and immune evasion.23
The MEROPS database classifies LasB as a member of the M4 family of metalloproteases that are characterized by the presence of a Zn2+ located at the catalytic site.24 The ion is tetrahedrally coordinated by the two histidines of the conserved HEXXH motif and a glutamic acid located 20 amino acid C-terminal of this motif and a water molecule. The glutamic acid of the HEXXH motif is an important catalytical residue. The zinc-binding group (ZBG) is a critical feature of metalloenzyme inhibitors that represents a valuable tool for enzyme inhibition.25 Commonly studied ZBGs include hydroxamate, carboxylic acids, tropolones, and 3-hydroxypyridine-4(1H)-thiones.26–31 The development of selective inhibitors for LasB represents a significant challenge due to its strong structural similarity to human matrix metalloproteinases (MMPs) and A disintegrin and metalloproteinases (ADAMs).32,33 This similarity hampered the development of highly specific therapeutic agents. Notwithstanding this high degree of homology, the depth of the S1′ sub-pocket represents a putative selectivity factor between LasB and MMPs.34 Specifically, the Tyr114 and Trp115 residues in LasB are replaced by leucine residues in MMPs, with the side chains of these leucine residues pointing towards the binding site of MMPs, thereby limiting the accessibility of the S1′ sub-pocket. Furthermore, an arginine residue (Arg198 in LasB) is positioned between S1′ and S2′ sub-pockets in the M4 enzymes, influencing the access and size of the S1′ sub-pocket. In summary, the S1′ sub-pocket of LasB is not as deep as the S1′ sub-pocket of the MMPs and this pocket also differs in size and shape among MMPs.35,36 Thereby, a good fitting with the S1′ sub-pocket may allow to develop selective LasB inhibitors. Previous studies have reported several LasB inhibitors with sub-micromolar activity, bearing diverse ZBGs. Among these, carboxylate derivative A and thiol B demonstrated significant activity (Fig. 1).29,37 In 2023, Hirsch's research group developed a promising compound, the phosphonic acid C (Fig. 1), which emerged as a potent inhibitor of the elastase LasB from P. aeruginosa.23 In vivo, compound C combined with levofloxacin significantly reduced bacterial burden and LasB protein levels in a neutropenic murine lung infection model, confirming its efficacy and target engagement. In a previous effort to discover new LasB inhibitors, we performed an enzymatic screening of an in-house chemical library of MMP inhibitors. This screening identified a N-benzyloxy-aminoacid-hydroxamate, LM-2 (Fig. 2), as the most effective LasB inhibitor, with high activity and selectivity for M4 enzymes over MMPs.38 In the present study, the structure of LM-2 and its binding mode, as predicted through computational studies (Fig. 2), were exploited to synthesize a series of LM-2 analogues presenting three kinds of structural modifications, compounds 1–24 (Fig. 3). The synthesis of these analogues was guided by a structure-based design, with the aim of enhancing efficacy and selectivity against LasB. To demonstrate the potential of the new derivatives, an initial enzymatic screening was conducted on isolated enzymes to identify the most promising candidates. Subsequently, their antibacterial activity, cytotoxicity, and ability to inhibit the secreted LasB of P. aeruginosa were evaluated in vitro. Based on these findings, four compounds were selected, and their capacity to prevent P. aeruginosa biofilm formation was further assessed in the PAO1 strain.
 |
| Fig. 1 Structures of reported LasB inhibitors and their corresponding inhibitory activities.23,29,37 | |
 |
| Fig. 2 (A) Chemical structure of LM-2.38,39 (B) LM-2 (S-enantiomer, violet-red) binding-mode into LasB (PDB ID: 1U4G, H-bonds in orange).38 Image generated with Chimera, version 1.16. | |
 |
| Fig. 3 General structures of the newly synthesized derivatives of LM-2, compounds 1–24. | |
2. Results and discussion
2.1. Design strategy
According to previous docking predictions in the LasB catalytic site, LM-2 placed its hydroxamate group to form a bidentate interaction with the catalytic Zn2+, exploiting the oxygen atoms of the C
O and OH groups. The aromatic ring of LM-2 occupied the S1′ sub-site, while the n-butyl chain extended into the S2′ sub-site. Additionally, the hydroxamate OH group donated a proton to Glu141, and its NH group established a hydrogen bond with the backbone of Ala113 (Fig. 2B).38 Before designing and synthesizing novel inhibitors, three previously reported N-benzyl-oxy derivatives of LM-2 from our in-house library, 1, 8, and 9 (Fig. 3, Table 1),39 were tested against LasB using a fluorometric assay. In compounds 1 and 8 the n-butyl chain of LM-2 (R group in Fig. 3) was replaced by a sec-butyl and a methyl group, respectively, and 8 also presented a chlorine atom in the para position of the benzyl group (R1). These modifications did not enhance inhibitory activity (Table 1). Instead, compound 9, which features an n-butyl chain and a chlorine atom in R1, exhibited activity comparable to the reference compound (Table 1). This suggested that the nature of the chain in α to the hydroxamate group plays a crucial role in inhibiting LasB.
Table 1 Structure and inhibitory activitya (Ki μM) of racemic N-benzyloxy-amino acid derivatives 1–16 against LasB

|
Compound |
R1 |
R |
X |
Ki μM |
Data performed in triplicate. Values reported as average ± SD. |
LM-2 |
H |
(CH2)3CH3 |
NHOH |
4.5 ± 0.1 |
1 |
H |
CH(CH3)CH2CH3 |
NHOH |
20 ± 1 |
2 |
H |
(CH2)7CH3 |
NHOH |
1.71 ± 0.09 |
3 |
H |
CH2CH(CH2CH3)CH2(CH2)2CH3 |
NHOH |
>100 |
4 |
H |
(CH2)3CN |
NHOH |
4.5 ± 0.2 |
5 |
H |
CH2COOH |
NHOH |
>100 |
6 |
H |
CH2COOt-But |
NHOH |
>100 |
7 |
H |
CH2(CO)NH–Phe–COOH |
NHOH |
35 ± 5 |
8 |
Cl |
CH3 |
NHOH |
13 ± 1 |
9 |
Cl |
(CH2)3CH3 |
NHOH |
5.6 ± 0.2 |
10 |
Cl |
(CH2)7CH3 |
NHOH |
4.3 ± 0.4 |
11 |
Cl |
CH2COOH |
NHOH |
>100 |
12 |
Cl |
CH2COOt-But |
NHOH |
0.92 ± 0.06 |
13 |
CF3 |
CH2CH3 |
NHOH |
21 ± 1 |
14 |
CF3 |
(CH2)3CH3 |
NHOH |
10.9 ± 0.4 |
15 |
CF3 |
CH2COOt-But |
NHOH |
1.09 ± 0.08 |
16 |
H |
(CH2)3CH3 |
NHSO2CF3 |
>100 |
Based on the predicted binding mode of LM-2 (Fig. 2) and these preliminary data, the n-butyl chain of LM-2 was further replaced by longer chains to explore potential hydrophobic interactions within the S2′ sub-pocket (compounds 2–4). Additionally, the n-butyl fragment was substituted with functionalized moieties to facilitate interaction with the key Arg198 (compounds 5–7). Moreover, analogues with a chlorine atom or a trifluoromethyl group in R1 were synthesized (compounds 10–15), and a novel derivative endowed with a ZBG different from hydroxamate was also prepared (compound 16). Given the quite hydrophobic nature of the S2′ sub-pocket,40 the n-butyl chain was initially replaced with an n-octyl fragment (compound 2), a longer branched chain (compound 3), or a butyl chain terminating with a cyano group (compound 4). The S2′ sub-pocket, characterized by a solvent-exposed environment and hydrophobic residues lining its walls provided a favorable site for such modifications. For these reasons, compounds 3 and 4 were prepared to investigate how the extended aliphatic chains along the S2′ sub-site might influence inhibitory activity, while the cyano group was selected for its potential to form hydrogen bonds through its electron-rich nitrogen.
Subsequently, our study focused on the importance of the configuration of the chiral center in alpha position to the ZBG. Computational studies41 carried out on LM-2 enantiomers, indicated that the S-enantiomer ((S)-17, Table 2) showed better interactions than the R-enantiomer ((R)-18). To verify this prediction, (S)-17 and (R)-18 were synthesized together with compound (S)-19 (Table 2), a bioisoster of (S)-17 devoid of the oxygen atom close to the benzyl fragment. This compound and its derivatives (S)-20 and (S)-21 were prepared to elucidate the specific role of this oxygen atom in the pharmacophore of this class of inhibitors. Finally, a small series of N-benzyloxy iminoacetic derivatives (compounds 22–24, Fig. 3 and Table 3) with an oxime-ether group and a carboxylate in R was synthesized to seek for further interactions with Arg198.
Table 2 Structure and inhibitory activitya (Ki μM) of optically active LM-2 derivatives 17–21 against LasB

|
Compound |
R |
Y |
n |
Ki μM |
Data performed in triplicate. Values reported as average ± SD. |
LM-2 |
(CH2)3CH3 |
O |
1 |
4.5 ± 0.1 |
(S)-17 |
(CH2)3CH3 |
O |
1 |
3.31 ± 0.08 |
(R)-18 |
(CH2)3CH3 |
O |
1 |
34 ± 2 |
(S)-19 |
(CH2)3CH3 |
CH2 |
1 |
87 ± 5 |
(S)-20 |
(CH2)3CH3 |
CH2 |
0 |
>100 |
(S)-21 |
(CH2)7CH3 |
CH2 |
0 |
>100 |
Table 3 Structure and inhibitory activitya (Ki μM) of N-benzyloxy iminoacetic derivatives 22–24 against LasB

|
Compound |
R1 |
R |
X |
Ki μM |
Data performed in triplicate. Values reported as average ± SD. |
22 |
H |
(CH2)2COOH |
OH |
>100 |
23 |
H |
(CH2)2COOCH3 |
NHOH |
>100 |
24 |
Cl |
H |
NHOH |
>100 |
2.2. Chemistry
N-Benzyloxy amino acid derivatives 2, 3, 7, 10 and 16 were prepared as reported in Scheme 1. The previously described (E)-2-((benzyloxy)imino) acetic acid 25 and (E)-2-(((4-chlorobenzyl)oxy)imino)acetic acid 26 (ref. 39) were protected as methyl esters by treatment with thionyl chloride (SOCl2) in methanol (MeOH). This step was essential for facilitating the subsequent nucleophilic addition reaction with Grignard reagents. The appropriate α-oxime esters 27 and 28 were then converted into the proper alkyl-esters 29–31 by an alkyl addition in a mixture of zinc chloride (ZnCl2), boron trifluoride diethyl etherate (BF3·OEt2), and the corresponding commercially available magnesium bromide (RMgBr, octyl magnesium bromide or (2-ethylhexyl)magnesium bromide) in DCM.42 Subsequently, a mild hydrolysis in basic conditions of methyl esters 29–31 afforded the carboxylates 32–34, respectively. Finally, a two-steps reaction was performed to convert carboxylates 32–34 into the corresponding hydroxamates 2, 3 and 10 by condensation with O-(tert-butyl-dimethyl-silyl)-hydroxylamine (TBDMSiONH2) to give the silyl intermediates 35–37, followed by acid cleavage by trifluoroacetic acid (TFA) in DCM. The synthesis of 7 involved the treatment of methyl ester 27 with tert-butyl 2-iodoacetate (38), which previously had been synthesized via a reported halogen substitution reaction.43 An electrophilic radical addition catalyzed by triethyl borane (Et3B) between the oxime ether 27 and the iodide compound 38 in a MeOH/H2O mixture resulted in the formation of the tert-butyl ester 39.44 The orthogonally protected ester 39 was treated with TFA in controlled condition, to selectively remove the tert-butyl protection on the α-chain leading to the formation of carboxylic acid 40. Subsequently, the amino acid ester 41 was obtained by a condensation reaction between the carboxylic acid 40 and L-phenylalanine tert-butyl ester, using tetramethyl uronium hexafluorophosphate (HBTU) as carboxylic acid activator and N,N-diisopropylethylamine (DIPEA) as a base.45 The hydroxamate 42 was obtained directly from methyl ester 41 under strongly basic reaction conditions using a methanolic solution both of potassium hydroxide (KOH) and hydroxylamine hydrochloride (NH2OH·HCl).46 The instability of the substrate, particularly the N-benzyloxy fragment under strongly basic conditions, might have affected the reaction yield, as confirmed by NMR analysis of the reaction byproducts, which revealed aliphatic moieties lacking the N-benzyloxy aromatic ring. Finally, the tert-butyl ester 42 was deprotected by acid hydrolysis with a solution of TFA in DCM, to give final compound 7. The sulfonamide derivative 16 was synthesized by condensing the previously described N-alkyloxy amino acid 43 (ref. 39) with trifluoromethanesulfonamide in the presence of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and 4-dimethylaminopyridine (DMAP) in DCM solution.47
 |
| Scheme 1 Synthesis of 2, 3, 7, 10 and 16. Reagents and conditions: (a) SOCl2, MeOH, 2–4 h, rt, (27: 96%; 28: 100%); (b) RMgBr, ZnCl2, BF3·OEt2, DCM, 0 °C – rt, 16 h (29: 14%; 30: 14%; 31: 15%); (c) LiOH, THF/H2O 2 : 1, rt, 16 h (32: 52%, 33: 64%; 34: 57%); (d) TBDMSiONH2, EDC, DCM, rt, 16 h (35: 79%; 36: 25%; 37: 57%); (e) TFA, DCM, 0 °C, 2–5 h (2: 17%; 3: 45%; 10: 79%); (f) n-BuLi, THF, −78 °C, 1 h, (43: 71%); (g) CF3SO2NH2, EDC, DMAP, DCM, rt, 24 h (16: 43%); (h) Et3B in hexane 1.0 M, MeOH/H2O 1 : 1, rt, 4 h (39: 60%); (i) TFA, DCM, 0 °C – rt, 0.5 h, (40: 100%); (l) L-phenylalanine tert-butyl ester, HBTU, DIPEA, CH3CN, 0 °C, 1.5 h, (41: 52%); (m) NH2OH·HCl, KOH, MeOH, 0 °C – rt, 2 h, (42: 7%); (n) TFA, DCM, 0 °C – rt, 0.5 h, (7: 38%). | |
The synthesis of N-benzyloxy amino acids 4–6 and 11–15 is described in Scheme 2. The carboxylic acids 47–52 were obtained through a Et3B-catalyzed intermolecular radical addition reaction in water between the oxime ethers 25, 26 and 44 and the corresponding iodo-derivatives 38, 45 and 46, which were previously synthesized.44,48 Carboxylic acid 52, was obtained as by-product of the reaction in a mixture with 51. Subsequently, carboxylic acids 47 and 49–52 were reacted with TBDMSiONH2 and EDC as a condensing agent in DCM. The selective hydrolysis of the silyl hydroxylamine functionality was achieved by washing the crude with HCl during the reaction work-up, which resulted in the formation of the hydroxamic derivatives 6 and 12–15. The same reaction conducted with carboxylic acids 47–49 in the absence of HCl treatment resulted in the isolation of the O-silylated hydroxamates 53–55, which gave the hydroxamates 4, 5 and 11 by treatment with TFA in DCM.
 |
| Scheme 2 Synthesis of 4–6 and 11–15. Reagents and conditions: (a) Et3B in hexane 1.0 M, H2O, rt, 4 h, 17–76%; (b) 1) O-TBDMSiNH2·HCl, EDCI, DCM, rt, 24 h, 2) HCl, SiO2, 18–83%; (c) O-TBDMSiNH2·HCl, EDCI, DCM, rt, 24 h, 21–88%; (d) TFA, DCM, rt, 5 h, 21–75%. | |
The synthesis of optically active N-benzyloxy amino acid derivatives 17–21 is described in Scheme 3. The corresponding α-hydroxy amino acids 59 and 60 were obtained by a substitution reaction via diazonium salt formation from commercially available enantiomerically pure D-norleucine 56 and L-norleucine 57, respectively.49 Methyl esters 61 and 62 were synthesized by reaction of carboxylic acids 59 and 60 with SOCl2 in MeOH. Then, the N-benzyloxy intermediates 63, 64 were prepared by an SN2 reaction from the corresponding hydroxyl derivatives, 61 and 62, using triflic anhydride (Tf2O), 2,6-lutidine as base and O-benzylhydroxylamine as nucleophile in DCM. Subsequently, the final enantiomers (S)-17 and (R)-18 were obtained as described above. For the synthesis of (S)-19, L-norleucine was treated with SOCl2 in MeOH to give methyl ester 69. The latter was used as a nucleophile to react with the (2-bromoethyl)benzene to give the alkylated compound 70. Due to the low nucleophilicity of the methyl ester amino acid 69, this alkylation reaction proceeded with low yield. Finally, the hydroxamate 19 was prepared by a MW-assisted reaction in strongly basic conditions in the presence of KOH, using directly the methyl ester 70 with NH2OH·HCl in a MeOH solution. The synthesis of (S)-N-benzyl amino acid derivatives 20 and 21 was conducted from the corresponding (S)-amino acids 57 and 58, as illustrated in Scheme 3. The esters 69 and 71 were reacted with benzyl bromide to afford the corresponding N-benzyl derivatives 72 and 73 via a nucleophilic substitution reaction (SN2). Finally, (S)-N-benzyl amino acids 20 and 21 were achieved by treating the corresponding methyl esters 72 and 73 with a methanolic solution of hydroxylamine hydrochloride and KOH in methanol.
 |
| Scheme 3 Synthesis of 17–21. Reagents and conditions: (a) H2SO4 0.5 M, NaNO2, H2O, 0 °C – rt, 27 h (59: 83%; 60: 78%); (b) SOCl2, MeOH, rt, 1.5 h (61: 70%; 62: 80%); (c) Tf2O, 2,6-lutidine, O-benzylhydroxylamine, DCM, 0 °C – rt, 16 h (63: 69%; 64: 68%); (d) LiOH, THF/H2O 2:1, 0 °C, 6 h (65: 61%; 66: 68%); (e) TBDMSiONH2, EDC, DCM, rt, 16 h (67: 44%; 68: 13%); (f) TFA, DCM, 0 °C, 5 h (17: 100%; 18: 90%); (g) SOCl2, MeOH, rt, 16 h, 100%; (h) (2-bromoethyl)benzene, K2CO3, THF, reflux, 6 days, 18%; (i) NH2OH·HCl, KOH, MeOH, MW-assisted, T = 80 °C, initial power: 150 W, t = 15 min, 5 cycles, 27%; (l) benzyl bromide, K2CO3, CH3CN, rt, 16 h (72: 32%; 73: 41%); (m) NH2OH·HCl, KOH, MeOH, 0 °C – rt, 4 h (20: 78%, 21: 20%). | |
Scheme 4 shows the synthesis of N-benzyloxy iminoacetic acids 22–24. The carboxylic acid 22 was prepared by a condensation reaction between commercial O-benzylhydroxylamine hydrochloride (74) and commercial α-ketoglutaric acid in a solution of THF and H2O. To achieve the hydroxamate 23, the carboxylic acid groups of 22 were protected as O-(tetrahydropyranyl) derivatives to obtain 75 by condensation with O-(tetrahydropyranyl)hydroxylamine (THPONH2), using EDC as a condensing agent, N-methyl morpholine (NMM) and 1-hydroxybenzotriazole (HOBt) in dry DMF. The protecting groups were then removed by acid hydrolysis (HCl 4 N) in a mixture MeOH/dioxane. The use of MeOH as solvent promoted the replacement of THPONH– on the side chain by a methyl ester, giving the final compound 23. Finally, carboxylic acid 26 was converted into the corresponding hydroxamate 24 according to the synthetic procedure described above.
 |
| Scheme 4 Synthesis of 22–24. Reagents and conditions: (a) α-ketoglutaric acid, THF/H2O 1 : 1, rt, 0.5 h, 73%; (b) THPONH2, EDC, HOBT, NMM, DMF dry, rt, 16 h (75: 98%, 76: 66%); (c) HCl 4 N, MeOH/dioxane 1 : 1, rt, 3 h, 12%. | |
2.3. LasB inhibitory activity and SAR study
All newly synthesized compounds were initially tested by a fluorometric assay measuring hydrolysis of a fluorogenic substrate McaRPPGFSAFK(Dnp)-OH by purified LasB and Ki values were determined (Tables 1–3). The previously reported LM-2 was used as reference compound, while its derivatives 1, 8 and 9, already published as MMPs inhibitors,39 were also evaluated against LasB.
Among non-R1 substituted hydroxamates (1–7, Table 1), the cyanoalkyl derivative 4 showed an inhibitory activity comparable to that of LM-2 (Ki = 4.5 μM) while compound 2 with a n-octyl group in R resulted 2-fold more active than LM-2 (Ki = 1.7 μM). Otherwise, derivative 3 with a long and branched chain in R was unable to reduce the enzymatic activity by 50% even at a concentration of 100 μM (Table 1). Remarkably, these results found support in molecular docking studies. As shown in Fig. 4, the long and linear side chain of 2 was correctly positioned in the S2′ sub-pocket by forming hydrophobic interactions, the aromatic ring was retained in the S1′ sub-pocket, while the hydroxamic acid coordinated in a bidentate manner the Zn2+. On the other hand, the analysis of compound 3 within the LasB binding site revealed two potential scenarios. In the first one, the branched side chain induced important steric clashes whereas the hydroxamate chelated the Zn2+ and the aromatic ring occupied the S1′ sub-site. Alternatively, when the longer portion of the side chain was located within the groove of the S2′ sub-pocket and its shorter branch was oriented similarly to the n-butyl fragment of LM-2, the carbonyl group of the hydroxamate moiety was too distant to effectively coordinate the zinc ion in a bidentate manner (Fig. 4). Since chelation is an essential feature of this class of compounds, the lack of proper zinc chelation is most likely the reason for the inactivity of compound 3.
 |
| Fig. 4 (A) Binding-mode of compound 2 (lime green) in the catalytic site of LasB (PDB ID: 1U4G). (B) Derivative 3 (turquoise) and LM-2 (violet-red) in the binding site of LasB. The carbonyl group of 3 is not oriented as the one of LM-2. Dashed lines indicate zinc chelation by both the coordinating residues and the ligand(s). Image generated with Chimera, version 1.16. | |
Subsequently, the substitution of the alkyl chain in R with a functionalized fragment was explored in derivatives 5–7 (Table 1). The rationale was to establish an interaction with Arg198 by introducing a carboxylic group on the α chain, keeping the N-benzyloxy moiety and the hydroxamate as the ZBG. As mentioned above, Arg198 is strategically located between S1′ and S2′ and influences the dimensions and depth of S1′ sub-pocket. Unfortunately, the hydroxamate 5 did not show even 50% enzyme inhibition at a concentration of 100 μM. Computational models suggested that the observed lack of inhibition may result from compound 5 coordinating the catalytic Zn2+ via its carboxyl group, while the hydroxamate formed hydrogen bonds with the side chain of the crucial Arg198 (Fig. S1†). It is well known that hydroxamates are stronger chelating groups than carboxylates, which are more prone to forming monodentate interactions with the zinc ion.50 To prevent this undesirable shift in chelation, the carboxyl group in compound 5 was then substituted with a tert-butyl ester in compound 6, or the carboxylic acid was condensed with L-phenylalanine as for compound 7. Esterification failed to preserve inhibitory activity (Ki > 100 μM), likely because the carbonyl group of the tert-butyl ester in compound 6 is positioned too far to effectively accept a hydrogen bond from the Arg198 side chain. This could be due to a deeper positioning of the benzyloxy fragment within the S1′ sub-pocket. In contrast, the condensation with L-phenylalanine preserved some affinity, although it was 8-fold lower than that of LM-2 (Ki = 35 μM). It is possible that the amino acid fragment in compound 7 is too hydrophobic and bulky to remain solvent-exposed and properly position itself within the S2′ sub-pocket, with the amino acid size potentially impeding optimal alignment and reducing the inhibitory effect. Based on these findings, the next step was targeting the interactions within the S1′ sub-pocket to improve activity. To leverage the hydrophobic nature of this sub-pocket (e.g. Leu132, Val137), a chlorine atom was introduced in R1 position of the benzyl group in compound 2, to give hydroxamate 10 (Table 1). Similarly, a chlorine atom was added in compound 6 to try to preserve the correct orientation of the benzyl group within the S1′ sub-pocket. This modification was expected to be essential for maintaining the optimal positioning of the hydroxamate group and ensuring effective bidentate chelation of the zinc ion, leading to the development of tert-butyl ester 12. As a matter of fact, while the p-Cl derivative 10 exhibited an activity comparable to LM-2 and 2 (Ki = 4.3 μM), 12 was found to be 5-fold more potent than LM-2, with a Ki = 0.92 μM. As displayed in Fig. 5, the hydroxamate group formed a bidentate coordination with the catalytic Zn2+, as observed for LM-2. Additionally, the incorporation of the p-chlorine atom on the benzyl group slightly forced out the latter from the S1′ sub-pocket, enabling the carbonyl group of the tert-butyl ester to form a hydrogen bond with Arg198. Consequently, derivative 12 was firmly anchored within the LasB binding site through a synergistic network of interactions: the bidentate coordination of the hydroxamate with Zn2+, the hydrophobic interaction of the p-chlorine benzyl moiety, and two critical hydrogen bonds, one involving the NH group of the –NH–O-benzyl functionality with Asn112 and the other, as previously mentioned, between the carbonyl group of the tert-butyl ester and Arg198. Notably, the N–O-benzyl moiety—typically considered chemically labile—participates in a key hydrogen bond within the catalytic pocket, suggesting a stabilizing effect from its engagement in the binding site. To further support this observation, we evaluated the integrity of this functionality under physiologically and chemically relevant conditions. Compound 12 was incubated at 37 °C in PBS at pH 1, 7.4, and 10, and no significant degradation of the N–O bond was observed, indicating good chemical stability across a broad pH range (ESI†).
 |
| Fig. 5 Compound 12 (gold) located in the catalytic site of LasB (PDB ID: 1U4G, H-bonds in orange, and dashed lines indicate zinc chelation by the coordinating residues and the ligand). The amino acids of the binding site are characterized by their hydrophobicity values, assigned according to the hydrophobicity scale of Kyte and Doolittle.51 The image was generated using Chimera, version 1.16. | |
To verify if the introduction of the p-Cl substituent could induce a similar increase in potency also in compound 5 with a carboxylate in R, its analogue 11 was synthesized and tested. On the contrary, this derivative showed no activity (Ki > 100 μM), suggesting that the chlorine atom alone did not facilitate hydrogen bond formation between Arg198 and the carboxyl group of 11, thereby preventing the switch of the ZBG. To enlarge the SAR study, the p-CF3 substituted analogues of LM-2 and 6 were synthesized, to give compounds 14 and 15 respectively (compound 13 was obtained as a by-product). While the introduction of a CF3 in R1 led to a slight decrease of activity in 14 with respect to LM-2, the same substituent in 15 caused a strong increase in potency with respect to 6, with a Ki = 1.1 μM. As aforementioned for the p-Cl substituted 12, this enhanced activity could be due to the positioning of a substituent in the para position of the benzyl group, which slightly moved the aromatic group out of the S1′ sub-site thus facilitating the interaction of the carbonyl group of the tert-butyl ester with the key Arg198.
The last N-benzyloxy amino acid LM-2 analogue to be synthesized was compound 16, bearing an N-acyl(trifluoromethane)sulfonamide as ZBG. However, the introduction of this “soft binder”47 for the catalytic Zn2+ led to complete loss of affinity (Ki > 100 μM).
As regards LM-2 enantiomers, the enzymatic assays confirmed a stronger binding of (S)-17 than of (R)-18 (Table 2). Particularly, (S)-17 had 10-fold stronger affinity than (R)-18 on LasB, but the improvement of affinity with respect to racemic LM-2 was negligible (Table 2). Considering the complex synthesis to obtain optically pure N-benzyloxy derivatives (as described in Scheme 3), and the small difference of activity between the (S) enantiomer and the respective racemate, the idea to obtain (S) enantiomers of inhibitors was abandoned.
At this point, with the aim to prove the role of the oxygen atom in the N–O benzyl moiety, a bioisoster of (S)-17 devoid of the oxygen atom, compound (S)-19, was synthesized followed by two N-benzyl derivatives, (S)-20 and (S)-21. These last two compounds were designed to optimize the selectivity for M4 enzymes over human MMPs. The preference for the N-benzyl fragment was based on its shorter length compared to the N-benzyloxy counterpart thus reducing the probability of penetrating deeply into the S1′ sub-pocket of MMPs, which is a feature to obtain M4 inhibitors selective over MMPs. N-Benzyl derivatives (S)-20 and (S)-21 bore a hydroxamate moiety as ZBG and an n-butyl and n-octyl chain in R, respectively. As a result of this isosteric substitution, compound (S)-19 showed approximately a 26-fold weaker binding against LasB compared to (S)-17, and both N-benzyl derivatives, (S)-20 and (S)-21, were inactive, with a Ki > 100 μM (Table 2). These findings clearly pointed out that the N–O– fragment is crucial for a correct orientation of the aromatic system within the S1′ sub-pocket of the enzyme.
As a final step, the structure of the inactive compound 5 was further modified to synthesize unsaturated oxime-ether derivatives (–C
N–O–R, 22–24, Fig. 3) to obtain compounds potentially able to interact with Arg198 in LasB. Compared to 5, its analogue 22 had a carboxylate group as ZBG, and its side chain was extended by one carbon atom. On the other hand, both 23 and 24 contained a hydroxamate as ZBG but differed from 5 for the aliphatic chain in R. Specifically, 23 had an additional carbon atom like 22 and a methyl ester group in R, while 24 lacked the aliphatic side chain in R and contained a chlorine atom in R1. Unfortunately, all these compounds were ineffective (Table 3) probably due to the geometric constraints introduced by the oxime double bond (–C
N–O–), which did not allow a proper orientation of the ZBG. Thus, the design and synthesis of other unsaturated compounds was interrupted due to these unsuccessful results.
2.4. Molecular dynamics simulations
To investigate the stability and dynamic behavior of compound 12, the most promising inhibitor in this series, a 100 ns molecular dynamics (MD) simulation was performed in explicit solvent (TIP3P) at 300 K. The root mean square deviation (RMSD) of the protein heavy atoms (yellow) and the ligand (aligned to the protein, blue), as shown in Fig. S2,† was analyzed to assess the conformational stability of the complex. The protein RMSD remained stable, indicating that LasB retains its structural integrity, while the ligand RMSD exhibited initial fluctuations (10–15 ns), followed by stabilization, suggesting conformational adaptation within the binding pocket (Fig. S3†). A transient RMSD increase in the final phase suggests localized intra-pocket reorganization, likely due to dynamic interactions with active-site residues. This aligns with the Ki value of 0.92 μM, suggesting that while compound 12 remains stably engaged within the catalytic pocket, its residual flexibility, particularly due to the solvent-exposed tert-butyl fragment, may limit its potency. Overall, these MD results confirm the stability of compound 12 within LasB, supporting its potential as an effective inhibitor while highlighting opportunities for structural refinement to enhance affinity.
2.5. Selectivity against MMPs
The most promising LasB inhibitors were selected for further characterization, including selectivity studies against human recombinant MMP-2, -9 and -14. These enzymes were chosen among the 23 human MMPs because represent the most well-studied MMPs, due to their involvement in cancer-induced angiogenesis and invasion.52 MMP-2 and -9 (gelatinase A and B) are secreted MMPs, while MMP-14 is a collagenase membrane-type MMP (MT1-MMP), so they could represent two different sub-types of human MMPs.53 Compounds 2, 10, 12, 15 and (S)-17 were tested on MMPs by a fluorometric assay which uses a fluorogenic peptide54 as the substrate, in comparison with the parent compound LM-2.
As can be seen from data in Table 4, all compounds were highly selective over MMP-14, which is anchored to the cell membrane and has important roles in cell migration implicated in many physiological and pathological events.55 A good selectivity for LasB over MMP-2 and MMP-9 was also found, even if to a lower extent. In particular, hydroxamate 12 displayed a 90-fold selectivity for LasB over MMP-14 and a 24-fold selectivity over MMP-2. The same selectivity profile was found for the CF3-analogue of 12, compound 15. Overall, compounds 12 and 15 presented an improved activity and selectivity for LasB with respect to the parent compound LM-2.
Table 4 In vitroa inhibitory activity (IC50 μM) of the best LasB inhibitors against selected MMPs
Compound |
MMP-2 |
MMP-9 |
MMP-14 |
Data performed in duplicate. Values reported as average ± SD. |
2 |
47.5 ± 10.1 |
24 ± 2 |
>100 |
10 |
>100 |
35 ± 3 |
>100 |
12 |
22.5 ± 3.0 |
15.8 ± 0.6 |
83.4 ± 2.6 |
15 |
39.7 ± 2.6 |
22.2 ± 0.8 |
89 ± 15 |
(S)-17 |
42.1 ± 2.6 |
55 ± 4 |
>100 |
LM-2 |
34 ± 4 |
81 ± 5 |
>100 |
Based on the results of enzymatic assays, the LasB inhibitors LM-2, 2, 10, 12, 15, and (S)-17 were selected for further investigation in vitro against Pseudomonas aeruginosa PAO1 strain.
2.6. Antibacterial activity
None of the N-benzyloxy amino acid derivatives tested against the PAO1 strain showed any bacteriostatic effect when tested up to a concentration of 200 μM (minimum inhibitory concentration, MIC > 200 μM), confirming their potential use as pathoblockers.
2.7. Cytotoxicity
To further explore the translational potential of LM-2, 2, 10, 12, 15, and (S)-17, the cytotoxic effect of these inhibitors was tested on the human lung epithelial cell line NCI H441. After incubation for 24 h, cell viability was evaluated using the WST-1 assay. All compounds did not exhibit any evident effect on cell viability at the tested concentrations, except 2 and 10, which markedly reduced viability at the concentration of 200 μM (Fig. 6).
 |
| Fig. 6 Cytotoxic effect of LM-2, 2, 10, 12, 15, and (S)-17. Inhibitors were incubated for 24 h with the human lung epithelial cell line NCI-H441 and cell viability was evaluated using the WST-1 assay. Results are shown as mean ± standard error of the mean values (n = 3). Statistical significance was evaluated by one-way ANOVA followed by the Tukey–Kramer post hoc test. *p < 0.05, **p < 0.01. Statistical significance refers to comparisons between the optical density values (wavelength at 450 nm) of each sample incubated with inhibitors with respect to its control represented by DMSO at the corresponding concentration. | |
2.8. Inhibition of secreted LasB activity of PAO1 strain
The ability of LM-2, 2, 10, 12, 15, and (S)-17 to inhibit the enzymatic activity of native LasB produced by P. aeruginosa was evaluated (Fig. 7). For this purpose, PAO1-CFS (cell free supernatant) was pre-incubated for 30 min with each derivative, and then the mixture was incubated for 18 h with LasB substrate (elastin Congo-red). The elastase activity was determined by measuring the absorbance at OD490 and expressed as percent of LasB activity compared to the untreated controls. Among the tested compounds, 12 exhibited the highest efficacy, reducing LasB activity by 50% at 10.7 μM (Fig. 7) while LM-2, 2, 10, 15, and (S)-17 showed similar efficacy in inhibiting LasB activity (IC50 from 21.2 to 29.7 μM) (Fig. 7). Notably, these data were in accordance with the inhibitory activity determined on the isolated enzyme LasB (Table 1), since compound 12 resulted the most active in both assays.
 |
| Fig. 7 Effects of LM-2, 2, 10, 12, 15, and (S)-17 on the elastinolytic activity of P. aeruginosa. After 30 minutes of incubation of inhibitors with PAO1 CFS, the mixture was incubated for 18 h with elastin-Congo red. The elastase activity was determined by measuring the absorbance at OD490 and expressed as % of LasB activity as compared to the untreated controls. The IC50 values were calculated from at least three independent experiments performed in duplicate. Values represent the mean ± standard deviation (SD). | |
2.9. Antibiofilm activity
Although the biofilm formation is a multifactorial process, it has been found that the increased expression of LasB elastase leads to higher rhamnolipid production, which in turn has a role in the biofilm formation in P. aeruginosa.56 Thus, to evaluate the potential antibiofilm activity of N-benzyloxy amino acids, we tested whether selected compounds, LM-2, 2, 12, and (S)-17, at sub-MIC concentrations (25, 50, 100, 200 μM) inhibited P. aeruginosa PAO1 biofilm after 24 h of treatment. The biofilm biomass was evaluated by crystal-violet staining. Compounds 2 and (S)-17 significantly reduced the biofilm biomass in a dose-dependent manner (Fig. 8). Both reduced the biofilm biomass by approximately 50% at concentrations as low as 50 μM. In sharp contrast, a similar effect was observed with LM-2 and compound 12 but only when tested at 200 μM (Fig. 8). This apparent discrepancy between LasB inhibition and antibiofilm activity of compound 12 could be explained considering that there may be other factors which contribute to biofilm formation, in addition to LasB activity. The importance of quorum sensing (QS) system in P. aeruginosa biofilm has been largely documented.57
 |
| Fig. 8 Antibiofilm activity of LM-2, 2, 12, and (S)-17. Bacterial suspensions were seeded in 96-well polystyrene microtiter plates and exposed to sub-MIC concentrations of each inhibitor for 24 h. Biofilm biomass was quantified by crystal-violet staining. Values represent the mean ± standard error of the mean. *p < 0.05; **p < 0.01. Statistical significance refers to comparisons between the optical density values (wavelength at 570 nm) of each sample incubated with inhibitors with respect to its control represented by DMSO at the corresponding concentration. | |
3. Conclusions
This study presents the design and synthesis of novel N-benzyloxy-amino acid derivatives as potential pathoblockers targeting the LasB enzyme secreted by Pseudomonas aeruginosa. Through structure-based design, we identified key modifications that could enhance selectivity and inhibitory activity, particularly focusing on interactions within the S1′ and S2′ sub-pockets of the catalytic site. Hydroxamate 12, bearing a p-Cl benzyl group and a tert-butyl ester moiety in R, demonstrated the highest efficacy with significant inhibition of LasB enzymatic activity and promising antibiofilm properties. Importantly, the selectivity of these compounds against LasB over human matrix metalloproteinases underscores their therapeutic potential while minimizing off-target effects. The inhibitors were effective in attenuating LasB-mediated virulence without exhibiting antibacterial effects, confirming their potential use as pathoblockers. Cytotoxicity evaluations on human lung epithelial cells revealed a favorable safety profile for most compounds at relevant concentrations, further supporting their translational viability. This work highlights the importance of targeting bacterial virulence mechanisms to combat multidrug-resistant pathogens such as P. aeruginosa.
4. Experimental section
4.1. Chemistry
Air sensitive reactions were carried out under an inert atmosphere (N2 or Ar) and monitored by thin layer chromatography (TLC) on Merck aluminum silica gel sheets (60 F254). TLCs were visualized with a UV lamp at 254 or 365 nm and, for non-chromophore substances, stained with phosphomolybdic acid solution and then heated. In addition, an aqueous solution of FeCl3 was used to detect hydroxamic acids. Evaporation was performed in vacuo (rotating evaporator). Na2SO4 was used as the drying agent for organic solutions. Reaction mixtures were purified by chromatographic separations on silica gel using an automated system or prepacked ISOLUTE Flash Si II cartridges (Biotage). The automated system was the Isolera Prime from Biotage (Uppsala, Sweden) equipped with a UV detector with a range of 200–400 wavelengths (λ) and using SFÄR HC Duo silica cartridges (Biotage, Uppsala, Sweden). The microwave-assisted reactions were carried out in a Biotage Initiator+ Microwave Synthesizer. The purified compounds were characterized by 1H, 13C, and 19F NMR spectra using a Bruker Avance III HD 400 MHz spectrometer. Chemical shifts (δ) are given in ppm, J in Hz, and various abbreviations have been used for the multiplicities (s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet, dd = double doublet, dt = double triplet, br s = broad singlet). Melting points were determined on a Leica Galen III microscope (Leica/Cambridge Instruments) and were not corrected. The ESI-MS spectra were recorded by direct injection at 5 (positive) and 7 (negative) μL min−1 flow rate in an Orbitrap high-resolution mass spectrometer (Thermo, San Jose, CA, USA), equipped with HESI source. Optical rotations were measured at 20 °C using an ATAGO AP-300 polarimeter equipped with a continuous sodium lamp (λ = 589 nm). The final compounds 2–7 and 10–24 were synthesized with a purity of at least 95%, as confirmed by combustion analysis and by NMR analysis reported in the ESI.† Analytical results are within ±0.4% of the theoretical values. Commercially available chemicals were purchased from Sigma-Aldrich (Merck) or ABCR. Compounds 1, 8 and 9 were synthesized as previously described.39
General procedure to synthesize methyl esters 27–28, 61–62, 69, and 71. The appropriate carboxylic acid (1 equiv.) was dissolved in MeOH (1.95 mL mmol−1) and thionyl chloride (SOCl2, 1.6 equiv.) was added dropwise at 0 °C. The reaction mixture was stirred at rt for 1.5 h – overnight. After, the reaction solution was concentrated under vacuum conditions and methyl esters were obtained without any further purification.
Methyl (E)-2-((benzyloxy)imino)acetate (27). The title compound was prepared from carboxylic acid 25 following the general procedure. Methyl ester 27 was obtained as a yellow oil (5.8 g). Yield: 96%. 1H NMR (400 MHz, CDCl3) δ: 7.56 (s, 1H); 7.38–7.34 (m, 5H); 5.3 (s, 2H); 3.86 (s, 3H).
Methyl (E)-2-(((4-chlorobenzyl)oxy)imino)acetate (28). The title compound was synthesized from carboxylic acid 26 following the general procedure. Methyl ester 28 was obtained as a yellow oil (936.30 mg). Yield: 100%. 1H NMR (400 MHz, CDCl3) δ: 7.55 (s, 5H), 7.36–7.29 (m, 4H); 5.25 (s, 2H); 3.85 (s, 3H).
Methyl (R)-2-hydroxyhexanoate (61). The title compound was obtained from (R)-carboxylic acid 59 following the general procedure. (R)-Methyl ester 61 was obtained as a yellow oil (331.2 mg). Yield: 70%. 1H NMR (400 MHz, DMSO-d6) δ: 5.32 (d, J = 6 Hz, 1H); 4.03–3.99 (m, 1H); 3.62 (s, 3H); 1.63–1.58 (m, 1H); 1.57–1.49 (m, 1H); 1.32–1.23 (m, 4H); 0.85 (t, J = 6.8 Hz, 3H). Characterization data are in agreement with the previous literature.58
Methyl (S)-2-hydroxyhexanoate (62). The title compound was synthesized from (S)-carboxylic acid 60 following the general procedure. (S)-Methyl ester 62 was obtained as a yellow oil (349 mg). Yield: 80%. 1H NMR (400 MHz, DMSO-d6) δ: 5.31 (br s, 1H); 4.02–3.99 (m, 1H); 3.62 (s, 3H); 1.63–1.58 (m, 1H); 1.57–1.49 (m, 1H); 1.32–1.23 (m, 4H); 0.58 (t, J = 7.2 Hz, 3H). Characterization data are in agreement with the previous literature.59
Methyl (R)-2-aminohexanoate (69). The title compound was prepared from L-norleucine (57) following the general procedure. Methyl ester 69 was obtained as a yellow oil (332.07 mg). Yield: 100%. 1H NMR (400 MHz, DMSO-d6) δ: 8.34 (br s, 2H); 3.96 (t, J = 6.4 Hz, 1H); 3.74 (s, 3H); 1.79–1.73 (m, 2H); 1.37–1.20 (m, 4H); 3.17 (t, J = 6.8 Hz, 3H). Characterization data are in agreement with the previous literature.60
Methyl (S)-2-aminodecanoate (71). The title compound was obtained from (S)-2-aminodecanoic acid (58) following the general procedure. The enantiopure ester 71 was a white solid (540 mg). Yield: 100%. 1H NMR (400 MHz, DMSO-d6) δ: 8.53 (br s, 2H); 3.98 (t, J = 6.4 Hz, 1H); 3.73 (s, 3H); 1.77–1.75 (m, 2H); 1.37–1.23 (m, 12H); 0.85 (t, J = 6.8 Hz, 3H). Characterization data are in agreement with the previous literature.61
General procedure to synthesize derivatives 29–31, and 24. Solution A: the appropriate Grignard reagent (RMgBr 1 M, 6 mL mmol−1) was added to a heterogeneous mixture of dried ZnCl2 (2 equiv.) in dry DCM (4 mL mmol−1) under argon atmosphere. Solution A was stirred at rt for 1 h. The resulting mixture was matt white.Solution B: the appropriate protected carboxylic acid (1 equiv.) was dissolved in dry DCM (10 mL mmol−1) and BF3·OEt2 (1 equiv.) was added dropwise under argon atmosphere. The mixture was stirred for 10 min at 0 °C. Then, solution A was added portionwise to solution B and stirred overnight leaving the ice bath which melted during the reaction time (0 °C – rt). After quenching the reaction with a saturated solution of NH4Cl, the solution was extracted with DCM (3×). The combined organic phases were dried over Na2SO4, filtered, and concentrated under vacuum conditions.
Methyl 2-((benzyloxy)amino)decanoate (29). The title compound was synthesized from methyl ester 27 and octylmagnesium bromide following the general procedure. After extraction, the crude product was purified by flash chromatography using an ISOLUTE Si II 20 g cartridge (n-hexane/EtOAc gradient from n-hexane 100% to 90
:
1 v/v), and alkyl compound 29 was obtained as a yellow oil (44 mg). Yield: 14%. 1H NMR (400 MHz, CDCl3) δ: 7.34–7.28 (m, 5H); 5.91 (br s, 1H); 4.69 (s, 2H); 3.75 (s, 3H); 3.58–3.56 (m, 1H); 1.50–1.41 (m, 3H); 1.23–1.12 (m, 11H); 0.88 (t, J = 6 Hz, 3H).
Methyl 2-((benzyloxy)amino)-4-ethyloctanoate (30). The title compound was synthesized from methyl ester 27 and (2-ethylhexyl)magnesium bromide following the general procedure. After extraction, the product was purified by flash chromatography using an ISOLUTE Si II 20 g cartridge (n-hexane/EtOAc gradient from n-hexane 100% to 90
:
1 v/v), and alkyl compound 30 was obtained as a yellow oil (135 mg). Yield: 14%. 1H NMR (400 MHz, CDCl3) δ: 7.36–7.29 (m, 5H); 5,87 (d, J = 10.4 Hz, 1H); 4.68 (s, 2H); 3.64 (s, 3H); 3.62–3.60 (m, 1H); 1.40–1.18 (m, 11H); 0.88 (t, J = 6.8 Hz, 3H); 0.76 (t, J = 6 Hz, 3H).
Methyl 2-(((4-chlorobenzyl)oxy)amino)decanoate (31). The title compound was prepared starting from methyl ester 28 and octylmagnesium bromide following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 20 g cartridge (n-hexane/petroleum ether (PE) gradient from 100
:
1 to 90
:
1 v/v), and 31 was obtained as a yellow oil (83.3 mg). Yield: 15%. 1H NMR (400 MHz, CDCl3) δ: 7.29–7.28 (m, 4H); 5.91 (br s, 1H); 4.63 (s, 2H); 3.74 (s, 3H); 3.57–3.54 (m, 1H); 1.49–1.38 (m, 3H); 1.36–1.18 (m, 11H); 0.88 (t, J = 6.4 Hz, 3H).
(E)-2-(((4-Chlorobenzyl)oxy)imino)-N-hydroxyacetamide (24). The title compound was obtained from the piranyl-derivative 76 and (2-ethylhexyl)magnesium bromide following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (DCM/MeOH gradient from DCM 100% to 30
:
1 v/v), and 24 was obtained as a yellow solid (45 mg). Yield: 35%. Mp: 113–116 °C. 1H NMR (400 MHz, DMSO-d6) δ: 11.1 (br s, 1H); 9.40–9.28 (m, 1H); 8.32 (s, 1H); 7.57–7.39 (m, 4H); 5.15 (s, 2H). 13C NMR (100 MHz, DMSO-d6) δ: 157.8; 143.2; 136.0; 132.7; 130.2; 128.4; 79.2; 75.2; 22.1; 13.9. Elemental analysis for C9H9ClN2O3, calculated: % C, 47.28; % H, 3.97; % N, 12.25 found: % C, 47.10; % H, 4.05; % N, 12.33.
General procedure to synthesize carboxylic acids 32–34, and 65–66. To a solution of the appropriate methyl ester (1 equiv.) in THF (3.60 mL mmol−1), an aqueous solution of LiOH 0.86 M (1.6 equiv. of LiOH powder) was added portionwise at 0 °C. The resulting mixture was stirred at rt or 0 °C for 2 h – overnight. Then, the reaction mixture was concentrated under vacuum conditions to be purified by flash chromatography.
2-((Benzyloxy)amino)decanoic acid (32). The title compound was prepared from methyl ester 29 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (CHCl3/MeOH gradient from 10
:
1 to 8
:
1 v/v, and the cartridge was washed with EtOAc/MeOH 8
:
1 v/v). The carboxylic acid 32 was achieved as a white solid (45 mg). Yield: 52%. 1H NMR (400 MHz, CD3OD-d4) δ: 7.35–7.26 (m, 5H); 4.68 (s, 2H); 3.52 (t, J = 6.8 Hz, 1H); 1.53–1.45 (m, 3H); 1.40–1.28 (m, 11H); 0.90 (t, J = 6.4 Hz, 3H).
2-((Benzyloxy)amino)-4-ethyloctanoic acid (33). The title compound was prepared from methyl ester 30 according to the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (DCM/MeOH gradient from DCM 100% to 25
:
1 v/v, and the cartridge was washed with EtOAc/MeOH 8
:
1 v/v). Carboxylic acid 33 was obtained as a white solid (79.8 mg). Yield: 64%. 1H NMR (400 MHz, DMSO-d6) δ: 7.33–7.27 (m, 6H); 4.57 (s, 2H); 3.50 (m, 1H); 1.33–1.20 (m, 11H); 0.85 (t, J = 6.4 Hz, 3H); 0.76 (t, J = 6.8 Hz, 3H).
2-(((4-Chlorobenzyl)oxy)amino)decanoic acid (34). The title compound was obtained from methyl ester 31 following the general procedure. The crude product was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 10
:
1 v/v). Carboxylic acid 34 was obtained as a white solid (46.1 mg). Yield: 57%. 1H NMR (400 MHz, CD3OD-d4) δ: 7.35–7.31 (m, 4H); 4.65 (s, 2H); 3.48–3.47 (m, 1H); 1.70–1.59 (m, 3H); 1.57–1.36 (m, 11H); 0.87 (t, J = 6.4 Hz, 3H).
(S)-2-((Benzyloxy)amino)hexanoic acid (65). The title compound was prepared from (S)-methyl ester 63 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 10
:
1 v/v). Purified carboxylic acid 65 was obtained as a white solid. Yield: 61%. 1H NMR (400 MHz, DMSO-d6) δ: 7.35–7.25 (m, 5H); 4.58 (s, 2H); 3.20 (t, J = 6.4 Hz, 1H); 1.50–1.34 (m, 2H); 1.33–1.15 (m, 4H); 0.84 (t, J = 6.8 Hz, 3H).
(R)-2-((Benzyloxy)amino)hexanoic acid (66). The title compound was synthesized from the (R)-methyl ester 64 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 10
:
1 v/v) to give the desired compound 66 as a white solid. Yield: 68%. 1H NMR (400 MHz, DMSO-d6) δ: 7.34–7.25 (m, 5H); 4.58 (s, 2H); 3.30 (t, J = 6.4 Hz, 1H); 1.48–1.39 (m, 1H); 1.37–1.32 (m, 1H); 1.31–1.18 (m, 4H); 0.83 (t, J = 6.8 Hz, 3H).
General procedure to synthesize silyl-derivatives 35–37, 53–55, 67–68. To a solution of the corresponding carboxylic acid (1 equiv.) in dry DCM (6.54 mL mmol−1), O-TBDMSNH2 (1 equiv. × each carboxylic acid group) was added. Finally, the carboxylic activating agent, EDC (1 equiv. × each carboxylic acid group), was added at 0 °C to the reaction mixture. The reaction was stirred at rt overnight. The reaction solution was diluted with DCM and washed with H2O (2×). The organic phase was dried over Na2SO4, filtered, and concentrated under reduced pressure.
2-((Benzyloxy)amino)-N-((tert-butyldimethylsilyl)oxy)decanamide (35). The title compound was prepared from carboxylic acid 32 following the general procedure. The desired compound 35 (51 mg) was obtained as a yellow oil, without any further purification. Yield: 79%. 1H NMR (400 MHz, CDCl3) δ: 8.21 (s, 1H); 7.52–7.34 (m, 5H); 5.66–5.65 (br s, 1H); 4.69 (s, 2H); 3.55–3.49 (m, 1H); 1.87–1.49 (m, 5H); 1.40–1.11 (m, 9H); 1.01–0.87 (m, 12H); 0.17 (s, 6H).
2-((Benzyloxy)amino)-N-((tert-butyldimethylsilyl)oxy)-4-ethyloctanamide (36). The title compound was obtained from carboxylic acid 33 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (isocratic eluent DCM 100%) to give the silyl derivative 36 as a yellow oil (29.1 mg). Yield: 25%. 1H NMR (400 MHz, CDCl3) δ: 8.14 (s, 1H); 7.38–7.30 (m, 5H); 5.63 (d, J = 7.20 Hz, 1H); 4.68 (s, 2H); 3.36–3.33 (m, 1H); 1.42–1.11 (m, 11H); 0.95 (s, 9H); 0.89–0.87 (m, 3H); 0.86–0.78 (m, 3H); 0.16 (s, 6H).
N-((tert-Butyldimethylsilyl)oxy)-2-(((4-chlorobenzyl)oxy)amino)decanamide (37). The title compound was obtained from the appropriate carboxylic acid 34 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 2 g cartridge (DCM/MeOH gradient from DCM 100% to 10
:
1 v/v) to afford the silyl derivative 37 as a yellow oil (13 mg). Yield: 57%. 1H NMR (400 MHz, CDCl3) δ: 8.09 (s, 1H); 7.36–7.27 (m, 4H); 5.67 (d, J = 6.8 Hz, 1H); 4.64 (s, 2H); 3.32–3.30 (m, 1H); 1.40–1.23 (m, 14H); 0.98 (s, 9H); 0.87 (t, J = 6.4 Hz, 3H); 0.17 (s, 6H).
tert-Butyl 3-((benzyloxy)amino)-4-(((tert-butyldimethylsilyl)oxy)amino)-4-oxobutanoate (53). The title compound was prepared from carboxylic acid 47 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (n-hexane/CHCl3 from 2
:
1 to 1
:
1 v/v, subsequently, the cartridge was washed with a mixture of DCM/MeOH 30
:
1 v/v) to give the desired compound 53 (95 mg). Yield: 83%. 1H NMR (400 MHz, CDCl3) δ: 8.57 (s, 1H); 7.41–7.30 (m, 5H); 6.19 (d, J = 8 Hz, 1H); 4.27–4.66 (m, 2H); 3.76–3.75 (m, 1H); 2.74–2.65 (m, 2H); 1.42 (s, 9H); 0.95 (s, 9H); 0.15 (s, 6H).
tert-Butyl 4-(((tert-butyldimethylsilyl)oxy)amino)-3-(((4-chlorobenzyl)oxy)amino)-4-oxobutanoate (54). The title compound was synthesized from carboxylic acid 49 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (isocratic eluent CHCl3 100%) to give the desired compound 54 as a yellow oil (23.5 mg). Yield: 39%. 1H NMR (400 MHz, CDCl3) δ: 8.52 (s, 1H); 7.36–7.30 (m, 4H); 6.20 (d, J = 8 Hz, 1H); 4.64 (s, 2H); 3.75–3.73 (m, 1H); 2.67–2.63 (m, 2H); 1.40 (s, 9H); 0.95–0.92 (m, 15H).
2-((Benzyloxy)amino)-N-((tert-butyldimethylsilyl)oxy)-5-cyanopentanamide (55). The title compound was obtained from the appropriate carboxylic acid 48 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 2 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 10
:
1 v/v) to afford the silyl derivative 55 as a yellow oil (29 mg). Yield: 48%. 1H NMR (400 MHz, CDCl3) δ: 8.22 (s, 1H); 7.35–7.30 (m, 5H); 5.81 (br s, 1H); 4.69 (s, 2H); 3.29 (t, J = 6 Hz,1H); 1.70–1.64 (m, 1H); 0.96 (s, 9H); 0.96–0.94 (m, 2H); 0.95–0.93 (m, 2H); 0.17 (s, 6H).
(S)-2-((Benzyloxy)amino)-N-((tert-butyldimethylsilyl)oxy)hexanamide (67). The title compound was synthesized from carboxylic acid 65 according to the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si 2 g cartridge (isocratic eluent PE/EtOAc 15
:
1 v/v) to give the desired compound 67 as a colorless oil (36.6 mg). Yield: 44%. 1H NMR (400 MHz, DMSO-d6) δ: 10.78 (s, 1H); 7.33–7.25 (m, 5H); 6.42 (d, J = 10.8 Hz, 1H); 4.57 (s, 2H); 3.33–3.31 (m, 1H); 1.30–1.15 (m, 6H); 0.87 (s, 9H); 0.85–0.79 (m, 3H); 0.10 (s, 6H).
(R)-2-((Benzyloxy)amino)-N-((tert-butyldimethylsilyl)oxy)hexanamide (68). The title compound was prepared from carboxylic acid 66 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (PE/EtOAc 15
:
1 v/v). Silyl-derivative 68 was obtained as a colorless oil (48 mg). Yield: 13%. 1H NMR (400 MHz, DMSO-d6) δ: 10.78 (s, 1H); 7.31–7.25 (m, 5H); 6.45 (d, J = 10.8 Hz, 1H); 4.62 (s, 2H); 3.31–3.28 (m, 1H); 1.32–1.15 (m, 6H); 0.87 (s, 9H); 0.85–0.79 (m, 3H); 0.19 (s, 6H).
General procedure to synthesize hydroxamic acids 2–5, 7, 10–11, 17–18, and 40. To a solution of the appropriate silyl-derivative (1 equiv.) in dry DCM (4.57 mL mmol−1), TFA (57 equiv. × each group to be hydrolyzed) was added portionwise at 0 °C. The resulting mixture was stirred at 0 °C – rt for 0.5 h – overnight. The reaction mixture was concentrated under reduced pressure by co-evaporating with Et2O or toluene.
2-((Benzyloxy)amino)-N-hydroxydecanamide (2). The title compound was obtained from silyl derivative 35 according to the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 2 g cartridge (CHCl3/MeOH 60
:
1 v/v). Trituration of the crude product recovered from flash chromatography with n-hexane afforded compound 2 (6.4 mg), as a white solid. Yield: 17%. Mp: 123–126 °C. 1H NMR (400 MHz, DMSO-d6) δ: 10.54 (s, 1H); 8.87 (s, 1H); 7.35–7.27 (m, 5H); 6.40 (d, J = 10.4 Hz, 1H); 4.60–4.53 (m, 2H); 3.32–3.26 (m, 1H); 1.27–1.21 (m, 14H); 0.87 (t, J = 6.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 169.6; 138.1; 128.2; 128.1; 127.5; 74.8; 61.1; 31.3; 29.2; 29.0; 28.9; 28.6; 25.5; 22.1; 14.0. HRMS (ESI, m/z) calculated for C17H29N2O3 [M + H]+: 309.21727; found: 309.21698. Elemental analysis for C17H28N2O3, calculated: % C, 66.20; % H, 9.15; % N, 9.08 found: % C, 66.30; % H, 8.90; % N, 9.12.
2-((Benzyloxy)amino)-4-ethyl-N-hydroxyoctanamide (3). The title compound was prepared from silyl derivative 36 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 2 g cartridge (DCM/MeOH gradient from DCM 100% to 50
:
1 v/v) to give the desired compound 3 as a white solid (9.8 mg). Yield: 45%. Mp: 108–111 °C. 1H NMR (400 MHz, DMSO-d6) δ: 10.60 (s, 1H); 8.87 (s, 1H); 7.35–7.26 (m, 5H); 6.37 (d, J = 11.2 Hz, 1H); 4.59–4.56 (m, 2H); 3.32–3.29 (m, 1H); 1.25–1.16 (m, 11H); 0.86 (t, J = 6.8 Hz, 3H); 0.77–0.75 (m, 3H). HRMS (ESI, m/z) calculated for C17H27N2O3 [M − H]−: 307.20272; found: 307.20346; calculated for C17H28N2O3Cl [M + Cl]−: 343.17939; found: 343.18011. Elemental analysis for C17H28N2O3, calculated: % C, 67.82; % H, 9.59; % N, 8.33 found: % C, 67.91; % H, 9.63; % N, 8.40.
2-((Benzyloxy)amino)-5-cyano-N-hydroxypentanamide (4). The title compound was synthesized from silyl derivative 55 following the general procedure. Trituration of the crude with n-hexane/Et2O afforded compound 4 as a white solid (16 mg). Yield: 88%. Mp: 76–79 °C. 1H NMR (400 MHz, DMSO-d6) δ: 10.55 (s, 1H); 8.89 (br s, 1H); 7.35–7.30 (m, 5H); 4.62–4.57 (m, 2H); 3.23 (t, J = 7.2 Hz,1H); 1.57–1.42 (m, 2H); 1.35 (q, J = 7.2 Hz, 2H); 0.82 (t, J = 7.2 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 169.33, 137.97, 128.09, 128.07, 127.44, 74.75, 62.62, 22.41, 10.46. HRMS (ESI, m/z) calculated for C13H18N3O3 [M + H]+: 264.13427; found: 264.13416. Elemental analysis for C13H17N3O3, calculated: % C, 59.30; % H, 6.51; % N, 15.96 found: % C, 59.10; % H, 6.61; % N, 15.98.
3-((Benzyloxy)amino)-4-(hydroxyamino)-4-oxobutanoic acid (5). The title compound was prepared from tert-butyl 3-((benzyloxy)amino)-4-(((tert-butyldimethylsilyl)oxy)amino)-4-oxobutanoate (53) following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (DCM/MeOH gradient from DCM 100% to 10
:
1 v/v) to give the desired compound 5 a sticky white solid (8 mg). Yield: 15%. 1H NMR (400 MHz, CDCl3) δ: 12.32 (br s, 1H); 10.68 (s, 1H); 8.94 (s, 1H); 7.39–7.27 (m, 5H); 6.59 (s, 1H); 4.62–4.55 (m, 2H); 3.76 (s, 1H); 3.38–3.35 (m, 1H). 13C NMR (100 MHz, CDCl3) δ: 175.5; 164.3; 136.4; 129.6; 129.1; 128.9; 128.8; 128.3; 82.8; 78.4; 61.6; 31.6. Elemental analysis for C11H14N2O5, calculated: % C, 51.97; % H, 5.55; % N, 11.02 found: % C, 51.72; % H, 5.63; % N, 11.20.
(3-((Benzyloxy)amino)-4-(hydroxyamino)-4-oxobutanoyl)-L-phenylalanine (7). The title compound was prepared from hydroxamic acid 42 according to the general procedure. After trituration with n-hexane, the desired compound 7 was achieved as a white solid (2 mg). Yield: 38%. 1H NMR (400 MHz, DMSO-d6) δ: 10.59–10.57 (m, 1H); 8.87 (br s, 1H); 8.25–8.15 (m, 1H); 7.30–7.22 (m, 10H); 6.65–6.40 (2× br s, 1H); 4.56–4.46 (m, 3H); 3.73–3.72 (m, 1H); 3.20–3.15 (m, 2H); 3.00–2.86 (m, 1H); 2.29–2.28 (m, 1H). 13C NMR (100 MHz, DMSO-d6) δ: 169.4; 137.8; 137.5; 129.2; 129.1; 129.0; 128.2; 128.1; 128.0; 127.5; 126.4; 74.9; 58.0; 53.7; 53.5; 36.9; 35.0. HRMS (ESI, m/z) calculated for C20H22N3O6 [M − H]−: 400.15141; found: 400.15002. Elemental analysis for C20H23N3O6, calculated: % C, 59.84; % H, 5.78; % N, 10.47 found: % C, 59.70; % H, 5.80; % N, 10.35.
2-(((4-Chlorobenzyl)oxy)amino)-N-hydroxydecanamide (10). The title compound was synthesized from silyl derivative 37 following the general procedure. Trituration of the crude with n-hexane/Et2O afforded compound 10, as a white solid (7.3 mg). Yield: 79%. Mp: 132–135 °C. 1H NMR (400 MHz, DMSO-d6) δ: 10.54 (s, 1H); 8.87 (s, 1H); 7.40–7.38 (m, 2H); 7.34–7.31 (m, 2H); 6.44 (d, J = 10.8 Hz, 1H); 4.55 (s, 2H); 3.25–3.22 (m, 1H); 1.27–1.20 (m, 14H); 0.85 (t, J = 6.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 169.6; 137.3; 132.1; 132.0; 128.1; 73.8; 61.1; 31.3; 30.7; 29.2; 28.9; 28.8; 28.6; 25.5; 22.1; 14.0. HRMS (ESI, m/z) calculated for C17H27ClN2O3 [M − H]−: 341.16374; found: 341.16345. Elemental analysis for C17H27ClN2O3, calculated: % C, 59.55; % H, 7.94; % N, 8.17 found: % C, 59.15; % H, 7.80; % N, 8.23.
3-(((4-Chlorobenzyl)oxy)amino)-4-(hydroxyamino)-4-oxobutanoic acid (11). The title compound was prepared from silyl derivative 54 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 20
:
1 v/v) to give the desired compound 11 as an orange solid (12 mg). Yield: 21%. Mp: 98–101 °C. 1H NMR (400 MHz, CD3CN) δ: 9.42 (br s, 1H); 7.41–7.28 (m, 4H); 4.63–4.55 (m, 2H); 3.79–3.77 (m, 1H); 2.64–2.55 (m, 1H); 2.51–2.45 (m, 1H). 13C NMR (100 MHz, CD3CN) δ: 172.4137.6, 134.0, 131.1, 131.0, 129.5, 129.3, 75.6, 59.3, 34.3. Elemental analysis for C11H13ClN2O5, calculated: % C, 45.77; % H, 4.54; % N, 9.70 Found: % C, 45.37; % H, 4.50; % N, 9.85.
(S)-2-((Benzyloxy)amino)-N-hydroxyhexanamide ((S)-17). The title compound was synthesized from silyl derivative 67 following the general procedure. The crude was triturated with n-hexane/Et2O to yield pure compound (S)-17 as a white solid (20 mg). Yield: 100%. Mp: 93–96 °C. [α]D20 = −25.6 (c = 0.454, MeOH). 1H NMR (400 MHz, DMSO-d6) δ: 10.6 (s, 1H); 7.45–7.27 (m, 5H); 4.65–4.58 (m, 2H); 3.33 (t, J = 6.8 Hz, 1H); 1.34–1.29 (m, 2H); 1.27–1.24 (m, 4H); 0.83 (t, J = 6.8 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 169.1; 137.7; 128.5; 128.2; 128.1; 127.6; 74.8; 61.0; 28.7; 27.6; 22.0; 13.8. HRMS (ESI, m/z) calculated for C13H21N2O3 [M + H]+: 253.15467; found: 253.15462; calculated for C13H20N2O3Na [M + Na]+: 275.13661; found: 275.13647; calculated for C13H19N2O3 [M − H]−: 251.14012; found: 251.14032. Elemental analysis for C13H20N2O3, calculated: % C, 61.88; % H, 7.99; % N, 11.10 found: % C, 61.57; % H, 8.13; % N, 11.37.
(R)-2-((Benzyloxy)amino)-N-hydroxyhexanamide ((R)-18). The title compound was achieved starting from the corresponding silyl derivative 68 according to the general procedure. The desired compound (R)-18 was obtained by crude trituration with n-hexane/Et2O as a white solid (29.8 mg). Yield: 90%. Mp: 102–105 °C. [α]D20 = +25.5 (c = 0.517, MeOH). 1H NMR (400 MHz, DMSO-d6) δ: 10.61 (s, 1H); 7.36–7.27 (m, 5H); 4.65–4.58 (m, 2H); 3.33 (t, J = 6.8 Hz, 1H); 1.36–1.26 (m, 2H); 1.24–1.15 (m, 4H); 0.83 (t, J = 6.8 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 169.0; 137.7; 128.2; 128.1; 127.6; 74.8; 61.0; 28.7; 27.6; 21.9; 13.8. Elemental analysis for C13H20N2O3, calculated: % C, 61.88; % H, 7.99; % N, 11.10 found: % C, 61.77; % H, 8.22; % N, 11.25.
3-((Benzyloxy)amino)-4-methoxy-4-oxobutanoic acid (40). The title compound was synthesized from tert-butyl ester 39 following the general procedure. Trituration of the crude product with n-hexane/Et2O afforded the desired product 40 as a white solid (520 mg). Yield: 100%. 1H NMR (400 MHz, CDCl3) δ: 7.35–7.32 (m, 5H); 6.38 (br s, 1H); 4.75–4.69 (m, 2H); 4.00 (dd, J1 = 6 Hz, J2 = 8 Hz, 1H); 3.77 (s, 3H); 2.88 (dd, J1 = 6 Hz, J2 = 16.8 Hz, 1H); 2.72 (dd, J1 = 7.6 Hz, J2 = 16.8 Hz, 1H).
Synthesis of 2-((benzyloxy)amino)-N-((trifluoromethyl)sulfonyl)hexanamide (16). EDC (105 mg, 0.547 mmol) was added portionwise to a stirred solution of carboxylic acid 43 (100 mg, 0.421 mmol), DMAP (67 mg, 0.547 mmol) and trifluoromethanesulfonamide (82 mg, 0.547 mmol) in dry CH2Cl2 (4.2 mL). After stirring for 4 h at rt, the mixture was washed with HCl 1 N and brine. The organic phase was dried and evaporated in vacuo. The product was purified by a trituration in Et2O/Hex/CHCl3 to afford 67 mg of compound 16 as a white solid (43% yield). Mp: 128–131 °C. 1H NMR (400 MHz, DMSO-d6) δ: 7.46–7.41 (m, 5H); 5.05 (s, 2H); 3.95 (t, J = 5.2 Hz, 1H); 1.77–1.75 (m, 2H); 1.44–1.30 (m, 4H); 0.88 (t, J = 6.8 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 172.2, 134.0, 129.2, 129.0, 128.6, 121.9, 118.6, 75.2, 64.4, 27.5, 26.5, 21.9, 13.6. 19F NMR (376 MHz, DMSO-d6) δ: 77.4. Elemental analysis for C14H19F3N2O4S, calculated: % C, 45.65; % H, 5.20; % N, 7.60 found: % C, 45.55; % H, 5.36; % N, 7.71.
General procedure to synthesize iodo derivatives 38 and 45. The appropriate bromo derivative (tert-butyl-2-bromoacetate or 4-bromobutyronitrile) (1 equiv.) was dissolved in acetone (0.59 mL mmol−1) and NaI (1.5 equiv.) was added. The obtained suspension was refluxed overnight. The resulting dark orange suspension was concentrated under vacuum conditions. The crude was diluted with H2O and extracted with CHCl3 (5×). The combined organic phases were dried over Na2SO4, filtered, and evaporated to yield the desired compounds 30 or 31 as dark oils.
tert-Butyl-2-iodoacetate (38). The title compound was synthesized from commercially available tert-butyl-2-bromoacetate and NaI following the general procedure. Iodo derivative 38 was obtained without any further purification as a dark oil (4.840 g). Yield: 100%. 1H NMR (400 MHz, CDCl3) δ: 3.6 (s, 2H); 1.46 (s, 9 H). Characterization data are in agreement with the previous literature.43
4-Iodobutyronitrile (45). The title compound was synthesized from commercially available 4-bromobutyronitrile and NaI following the general procedure. Iodo derivative 45 was obtained without any further purification as a dark oil (1.500 g). Yield: 100%. 1H NMR (400 MHz, CDCl3) δ: 3.30 (t, J = 6.4 Hz, 2H); 2.53 (t, J = 7.2 Hz, 2H); 2.14 (dt, J1–2 = 7.2 Hz, J1–3 = 13.2 Hz, 2H). Characterization data are in agreement with the previous literature.62
General procedure to synthesize methyl esters 39, 47–52. A commercial solution of BF3·Et2O in hexane (0.67 or 0.83 equiv.) was added every 40 min, for a total of 4 or 5 equiv., to a solution of methyl ester 27 (1 equiv.) or the appropriate carboxylic acid (25–26, and 44) and the respective iodo derivatives (38, 45, and 46, 2 equiv.) in a mixture of H2O/MeOH 1
:
1 (3.86 mL mmol−1 for ester 27) or only H2O (4.27 mL mmol−1 for the carboxylic acids). The reaction was stirred at rt for 4 h. The reaction mixture was then concentrated under vacuum conditions.
4-(tert-Butyl)-1-methyl(benzyloxy)aspartate (39). The title compound was synthesized from methyl ester 27 and iodo derivative 38 according to the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 20 g cartridge (n-hexane/EtOAc gradient from 30
:
1 to 20
:
1 v/v) to afford 39 as a colorless oil (564 mg). Yield: 60%. 1H NMR (400 MHz, CDCl3) δ: 7.36–7.28 (m, 5H); 6.18 (d, J = 6.8 Hz, 1H); 4.70 (s, 2H); 3.99 (dd, J1 = 6.8 Hz, J2 = 13.2 Hz, 1H); 3.75 (s, 3H); 2.71 (dd, J1 = 6 Hz, J2 = 16 Hz, 1H); 2.58 (dd, J1 = 7.2 Hz, J2 = 16 Hz, 1H); 1.44 (s, 9H).
2-((Benzyloxy)amino)-4-(tert-butoxy)-4-oxobutanoic acid (47). The title compound was prepared from carboxylic acid 25 and the iodo derivative 38 following the general procedure. The product (47) was obtained as a pure yellow oil (39 mg) after flash chromatography using an ISOLUTE Si II 2 g cartridge (DCM/MeOH gradient from DCM 100% to 8
:
1 v/v). Yield: 48%. 1H NMR (400 MHz, CDCl3) δ: 7.38–7.30 (m, 5H); 4.72–4.71 (m, 2H); 3.96–3.93 (m, 1H); 2.78 (dd, J1 = 5.6 Hz, J2 = 16.8 Hz, 1H); 2.69 (dd, J1 = 5.6 Hz, J2 = 16.8 Hz, 1H); 1.44 (s, 9H).
2-((Benzyloxy)amino)-5-cyanopentanoic acid (48). The title compound was achieved from iminoacetic acid 25 and iodo derivative 38 according to the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 50
:
1 v/v) to afford 48 as a white solid (228 mg). Yield: 55%. 1H NMR (400 MHz, CDCl3) δ: 7.35–7.31 (m, 5H); 4.71 (s, 2H); 3.56 (t, J = 6.8 Hz, 1H); 1.67–1.63 (m, 2H); 1.61–1.56 (m, 2H), 0.97 (t, J = 7.6 Hz, 2H).
4-(tert-Butoxy)-2-(((4-chlorobenzyl)oxy)amino)-4-oxobutanoic acid (49). The title compound was synthesized from carboxylic acid 26 and iodo derivative 38 according to the general procedure. The crude reaction was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 30
:
1 v/v). Carboxylic acid 49 was obtained as a yellow oil (111 mg). Yield: 27%. 1H NMR (400 MHz, CDCl3) δ: 7.32–7.25 (m, 4H); 4.67 (s, 2H); 3.93–3.92 (m, 1H); 2.78–2.73 (m, 1H); 2.68–2.64 (m, 1H); 1.43 (s, 9H).
4-(tert-Butoxy)-4-oxo-2-(((4-(trifluoromethyl)benzyl)oxy)amino)butanoic acid (50). The title compound was prepared from iminoacetic acid 44 and iodo derivative 38 according to the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (n-hexane/EtOAc gradient from 9
:
1 to 1
:
1 v/v) to give 50 as a colorless oil (225 mg). Yield: 76%. 1H NMR (400 MHz, CDCl3) δ: 7.61–7.59 (m, 2H); 7.45–7.43 (m, 2H); 4.78 (s, 2H); 4.02–4.00 (m, 1H); 2.77 (dd, J1–2 = 5.6 Hz, J1–3 = 16.4 Hz, 1H); 2.65 (dd, J1–2 = 7.2 Hz, J1–3 = 16.4 Hz, 1H); 1.43 (s, 9H).
2-(((4-(Trifluoromethyl)benzyl)oxy)amino)hexanoic acid (51) and 2-(((4-(trifluoromethyl)benzyl)oxy)amino)butanoic acid (52). The title compounds were prepared from iminoacetic acid 44 and commercially available butyl iodide 46 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (n-hexane/EtOAc gradient from 5
:
1 to 1
:
10 v/v) to afford 17 mg of pure compound 51 as a white solid and 121 mg of compound 51 mixed with derivative 52 as a white solid. (51) 1H NMR (400 MHz, CD3OD) δ: 7.64–7.62 (m, 2H); 7.54–7.52 (m, 2H); 4.76 (s, 2H); 3.53 (t, J1–2 = 8 Hz, 1H); 1.55–1.47 (m, 2H); 1.36–1.25 (m, 4H); 0.88 (t, J1–2 = 4 Hz, 3H).
Synthesis of methyl N2-(benzyloxy)-N4-((S)-1-(tert-butoxy)-1-oxo-3-phenylpropan-2-yl)asparaginate (41). DIPEA (0.48 mL, 2.764 mmol, 2 equiv.) was added to a solution of commercially available L-phenylalanine tert-butyl ester hydrochloride (712 mg, 2.764 mmol, 2 equiv.) in CH3CN (4 mL). This solution was added dropwise at 0 °C to a mixture of carboxylic acid 40 (350 mg, 1.382 mmol, 1 equiv.) in CH3CN (4 mL) to which 0.24 mL of DIPEA (1.382 mmol, 1 equiv.) and HBTU (629 mg, 1.685 mmol, 1.2 equiv.) had previously been added dropwise at 0 °C. The resulting reaction mixture was stirred at 0 °C for 1.5 h, concentrated under vacuum conditions and then diluted with EtOAc. The organic phase was washed with HCl 10% (3 × 50 mL), and then with a saturated solution of NaHCO3 (1 × 50 mL). The organic phase was dried over Na2SO4, filtered, and concentrated in vacuo. To afford the tert-butyl ester 41 as a yellow oil (99 mg), the crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (n-hexane/EtOAc 6
:
1 gradient from 6
:
1 to 4
:
1 v/v). Yield: 52%. 1H NMR (400 MHz, DMSO-d6) δ: 7.35–7.12 (m, 10H); 6.77 (t, J = 6.4 Hz, 1H); 4.76–4.71 (m, 1H); 4.67,4.62 (2× s, 2H); 4.01,3.97 (2× dd, J1 = 4.8 Hz, J2 = 8.4 Hz, 1H); 3.75,3.73 (2× s, 3H); 3.15–3.00 (m, 2H); 2.68–2.63 (m, 1H); 2.57–2.47 (m, 1H); 1.41,1.40 (2× s, 9H).
General procedure to synthesize hydroxamic acids (S)-20–(S)-21, and 42. 1.25 mL mmol−1 of a methanol solution of KOH (12 equiv.) was added at 0 °C to a mixture of NH2OH·HCl (8 equiv.) in MeOH (1.25 mL mmol−1), previously heated at 40 °C. The resulting suspension was added directly, at 0 °C, to a solution of the appropriate methyl ester (1 equiv.) in MeOH (4.35 mL mmol−1). The resulting mixture was gradually heated from 0 °C to rt and stirred for 2–4 h.
(S)-2-(Benzylamino)-N-hydroxyhexanamide ((S)-20). The title compound was prepared from the methyl ester 72 following the general procedure. The crude was first concentrated under vacuum conditions, then diluted with H2O and extracted with EtOAc (1× 125 mL). The organic phase was dried over Na2SO4, filtered, and concentrated under reduced pressure leading to the pure compound (S)-20 without any further purification, as a white solid (96 mg). Yield: 78%. Mp: 174–177 °C. [α]D20 = −35.9 (c = 0.501, MeOH). 1H NMR (400 MHz, DMSO-d6) δ: 10.52 (br s, 1H); 8.85 (s, 1H); 7.30–7.21 (m, 5H); 3.70–3.66 (m, 1H); 3.46–3.43 (m, 1H); 2.85–2.81 (t, J = 6.4 Hz, 1H); 1.47–1.43 (m, 2H); 1.29–1.22 (m, 4H); 0.83 (t, J = 6.8 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 170.7; 140.6; 128.1; 127.8; 126.6; 58.6; 50.6; 33.1; 27.7; 22.1; 13.9. Elemental analysis for C13H20N2O2, calculated: % C, 66.07; % H, 8.53; % N, 11.85 found: % C, 66.26; % H, 8.33; % N, 11.92.
(S)-2-(Benzylamino)-N-hydroxydecanamide (((S)-21)). The title compound was synthesized from methyl ester 73 according to the general procedure. The reaction solution was concentrated under vacuum conditions, diluted with H2O, and extracted with EtOAc (1× 125 mL). The organic phase was dried over Na2SO4, filtered, and evaporated in vacuo. The pure final compound ((S)-21) was obtained without any further purification, as a white solid (96 mg). Yield: 20%. Mp: 156–159 °C. [α]D20 = −38.8 (c = 0.487, MeOH). 1H NMR (400 MHz, DMSO-d6) δ: 10.48 (br s, 1H); 8.82 (s, 1H); 7.34–7.20 (m, 5H); 3.70–3.67 (m, 1H); 3.46–3.43 (m, 1H); 2.83 (t, J = 6.8 Hz, 1H); 1.44–1.42 (m, 2H); 1.31–1.15 (m, 12H); 0.86 (t, J = 6.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 170.7; 140.6; 128.1; 127.7; 126.5; 58.6; 50.7; 33.4; 31.3; 28.9; 28.6; 25.5; 22.1; 14.0. Elemental analysis for C17H28N2O2, calculated: % C, 69.83; % H, 9.65; % N, 9.58 found: % C, 69.75; % H, 9.55; % N, 9.78.
tert-Butyl-(3-((benzyloxy)amino)-4-(hydroxyamino)-4-oxobutanoyl)-L-phenylalaninate (42). The title compound was synthesized from methyl ester 41 following the general procedure. The crude was concentrated in vacuo, diluted with H2O (1× 50 mL), and extracted with CHCl3. The chloroform phase was dried over Na2SO4, filtered, and concentrated under vacuum conditions. To afford the pure amide 42 as a yellow oil (7.5 mg), the crude was purified by flash chromatography using an ISOLUTE Si II 2 g cartridge (CHCl3/MeOH 70
:
1 v/v). Yield: 7%. 1H NMR (400 MHz, DMSO-d6) δ: 10.62, 10.60 (2× s, 1H); 8.88 (s, 1H); 8.32–8.23 (m, 1H); 7.31–7.20 (m, 10H); 6.49, 6.43 (2× d, J = 8.4 Hz, 1H); 4.56–4.50 (m, 2H); 4.42–4.31 (m, 1H); 3.78–3.75 (m, 1H); 2.99–2.95 (m, 2H); 2.92–2.87 (m, 1H); 2.31–2.29 (m, 1H); 1.35, 1.30 (2× s, 9H).
General procedure to synthesize hydroxamic acids 6, 12–15. To a solution of the corresponding carboxylic acid (1 equiv.) in dry DCM (6.54 mL mmol−1), O-TBDMSNH2 (1 equiv.) was added. Finally, the condensing agent, EDC (1 equiv.), was added at 0 °C to the reaction mixture. The reaction was stirred at rt overnight. The reaction solution was then diluted with DCM and acid cleaved by washing with 1 N HCl (3×). The organic phase was dried over Na2SO4, filtered, and concentrated under vacuum conditions.
tert-Butyl-3-((benzyloxy)amino)-4-(hydroxyamino)-4-oxobutanoate (6). The title compound was obtained from carboxylic acid 47 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (CHCl3/MeOH gradient from CHCl3 100% to 50
:
1 v/v) to give acid 6 as a yellow oil (83 mg). Yield: 59%. 1H NMR (400 MHz, DMSO-d6) δ: 9.87 (s, 1H), 8.31 (s,1H); 7.41–7.31 (m, 5H); 4.75 (dd, J1 = 2.8 Hz, J2 = 7.6 Hz, 2H); 3.82–3.80 (m, 1H); 2.58–2.50 (m, 2H); 1.41 (s, 9H); 13C NMR (100 MHz, DMSO-d6) δ: 169.4; 137.7; 128.9; 128.3; 128.1; 81.1; 80.2; 79.2; 77.3; 61.4; 27.8. Elemental analysis for C15H22N2O5, calculated: % C, 58.05; % H, 7.15; % N, 9.03 found: % C, 57.91; % H, 7.10; % N, 9.15.
tert-Butyl-3-(((4-chlorobenzyl)oxy)amino)-4-(hydroxylamino)-4-oxobutanoate (12). The title compound was obtained from carboxylic acid 49 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (n-hexane/EtOAc gradient from 5
:
1 to AcOEt 100%, v/v) to give hydroxamic acid 12 as a colourless oil (89 mg). Yield: 42%. 1H NMR (400 MHz, DMSO-d6) δ: 10.67 (s, 1H), 8.94 (s, 1H); 7.45–7.32 (m, 4H); 6.57 (d, J = 9.6 Hz, 1H); 4.59–4.52 (m, 2H); 3.73–3.67 (m, 1H); 2.58–2.47–2.41 (m, 1H); 2.25–2.19 (m, 1H); 1.36 (s, 9H); 13C NMR (100 MHz, DMSO-d6) δ: 169.4; 167.7; 137.0; 132.1; 129.9; 128.1; 128.0; 80.1; 73.9; 57.9; 35.4; 27.7. HRMS (ESI, m/z) calculated for C15H20ClN2O5 [M − H]−: 343.10662; found: 343.10678; calculated for C15H21Cl2N2O5 [M + Cl]−: 379.08330; found: 379.08368. Elemental analysis for C15H21ClN2O5, calculated: % C, 52.25; % H, 6.14; % N, 8.12 found: % C, 52.01; % H, 6.27; % N, 8.08.
N-Hydroxy-2-(((4-(trifluoromethyl)benzyl)oxy)amino)butanamide (13) and N-hydroxy-2-(((4-(trifluoromethyl)benzyl)oxy)amino)hexanamide (14). The title compounds were obtained from the corresponding carboxylic acid (51–52) following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (n-hexane/EtOAc gradient from 4
:
1 to AcOEt 100%, v/v) to afford 43 mg of product 13 and 26 mg of product 14 as white solids (13 yield: 33%; 14 yield: 18%).(13) Mp: 125–128 °C. 1H NMR (400 MHz, DMSO-d6) δ: 10.56 (s, 1H); 8.88 (s, 1H); 7.71–7.69 (m, 2H); 7.54–7.52 (m, 2H); 6.55 (d, J = 10.4 Hz, 1H); 4.68 (s, 2H); 3.22 (dt, J1–2 = 7.2 Hz, J1–3 = 10.4 Hz, 1H); 1.35–1.32 (m, 2H); 0.80 (t, J = 8 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 170.7, 141.1, 130.5 (q, JC–F = 32 Hz); 128.2, 125.6 (q, JC–F = 3.7 Hz); 124.2 (q, JC–F = 270 Hz); 75.7, 64.6, 22.9, 10.4. 19F NMR (376 MHz, DMSO-d6) δ: 60.8. Elemental analysis for C12H15F3N2O3, calculated: % C, 49.32; % H, 5.17; % N, 9.59 found: % C, 49.27; % H, 4.99; % N, 9.63.
(14) Mp: 123–126 °C. 1H NMR (400 MHz, DMSO-d6) δ: 10.56 (s, 1H); 8.88 (s, 1H); 7.70–7.68 (m, 2H); 7.54–7.52 (m, 2H); 6.54 (d, J = 10.4 Hz, 1H); 4.66 (s, 2H); 3.28–3.25 (m, 1H); 1.29–1.19 (m, 6H); 0.81 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 169.5, 143.2, 128.5, 128.2 (q, JC–F = 32 Hz); 124.9 (q, JC–F = 3.7 Hz); 73.8, 61.1, 28.8, 27.7, 21.9, 13.8. 19F NMR (376 MHz, DMSO-d6) δ: 60.8. HRMS (ESI, m/z) calculated for C14H19N2O3F3 [M − H]−: 319.12750; found: 319.12744. Elemental analysis for C14H19F3N2O3, calculated: % C, 52.50; % H, 5.98; % N, 8.75 found: % C, 52.55; % H, 5.75; % N, 8.80.
tert-Butyl 4-(hydroxylamino)-4-oxo-3-(((4-(trifluoromethyl)benzyl)oxy)amino)butanoate (15). The title compound was obtained from carboxylic acid 50 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (n-hexane/EtOAc gradient from 5
:
1 to AcOEt 100%, v/v) to give acid 15 as a white solid (95 mg). Yield: 41%. Mp: 105–108 °C. 1H NMR (400 MHz, DMSO-d6) δ: 10.66 (s, 1H); 8.92 (s, 1H); 7.69–7.67 (m, 2H); 7.53–7.51 (m, 2H); 6.66 (d, J = 8 Hz, 1H); 4.67 (s, 2H); 3.75–3.70 (m, 1H); 2.48–2.42 (m, 1H); 2.21 (dd, J1–2 = 8 Hz, J1–3 = 16 Hz, 1H); 1.34 (s, 9H). 13C NMR (100 MHz, CD3OD) δ: 171.3, 170.9, 143.9, 130.8 (q, JC–F = 32 Hz); 129.6, 126.2 (q, JC–F = 3.7 Hz); 125.8 (q, JC–F = 269 Hz); 82.3, 75.9, 59.8, 36.3, 28.3. 19F NMR (376 MHz, CDCl3) δ: 62.6. HRMS (ESI, m/z) calculated for C16H20F3N2O5 [M − H]−: 377.13298; found: 377.13330. Elemental analysis for C16H21F3N2O5, calculated: % C,50.79; % H, 5.59; % N, 7.40 found: % C, 50.67; % H, 5.45; % N, 7.25.
General procedure to synthesize α-hydroxyl carboxylic acids 59–60. The appropriate amino acid was dissolved in an aqueous solution of H2SO4 0.5 M (4.0 mL mmol−1) at 0 °C. Then, an aqueous solution of NaNO2 4 M (1.3 mL mmol−1) was added over 30 min to the reaction mixture at 0 °C. The reaction mixture was stirred at 0 °C for 3 h and for 24 h at rt. Then, the reaction was extracted with Et2O (3×) and the organic phase was dried over Na2SO4, filtered, and concentrated under vacuum conditions.
(R)-2-Hydroxyhexanoic acid (59). The title compound was prepared from commercially available optically pure D-norleucine (56) following the general procedure. The crude reaction was triturated with Et2O/n-hexane to give the carboxylic acid 59 as a white solid (418.2 mg). Yield: 83%. 1H NMR (400 MHz, DMSO-d6) δ: 12.33 (br s, 1H); 5.08 (s, 1H); 3.90 (dd, J1 = 4.4 Hz, J2 = 7.6 Hz, 1H); 1.65–1.55 (m, 1H); 1.53–1.46 (m, 1H); 1.35–1.21 (m, 4H); 0.87 (t, J = 6.8 Hz, 3H). Characterization data are in agreement with the previous literature.49
(S)-2-Hydroxyhexanoic acid (60). The title compound was prepared from commercially available optically pure L-norleucine (57) following the general procedure. Trituration of the crude product with Et2O/n-hexane afforded pure carboxylic acid 60, as a white solid (391.7 mg). Yield: 78%. 1H NMR (400 MHz, DMSO-d6) δ: 12.29 (br s, 1H); 3.91 (dd, J1 = 4.4 Hz, J2 = 7.6 Hz, 1H); 1.65–1.57 (m, 1H); 1.54–1.46 (m, 1H); 1.35–1.19 (m, 4H); 0.87 (t, J = 10.8 Hz, 3H). Characterization data are in agreement with the previous literature.63
General procedure to synthesize N–O-benzyl derivatives 63–64. The appropriate methyl ester (1 equiv.) was dissolved in dry DCM (0.89 mL mmol−1) and the trifluoromethanesulfonic anhydride (Tf2O, 1.1 equiv.) was added portionwise at 0 °C. The solution was stirred for 15 min at 0 °C and 2,6-lutidine (1.19 equiv.) was added dropwise. Finally, a solution of O-benzylhydroxyl amine (1.99 equiv.) in DCM (0.22 mL mmol−1) was also added slowly. The reaction mixture was stirred at 0 °C for 10 min and then was stirred at rt overnight. The reaction solution was washed with H2O (1×), HCl (1×), and NaCl (2×). The organic phase was dried over Na2SO4, filtered, and evaporated under vacuum conditions.
Methyl (S)-2-((benzyloxy)amino)hexanoate (63). The title compound was prepared from alkyl methyl ester 61 following the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (PE/CHCl3 gradient from 15
:
1 to 8
:
1 v/v) to give the desired compound 63 as a yellow oil (121.1 mg). Yield: 69%. 1H NMR (400 MHz, DMSO-d6) δ: 7.36–7.27 (m, 5H); 6.72 (d, J = 9.6 Hz, 1H); 4.59 (s, 2H); 3.64 (s, 3H); 3.53 (dt, J1 = 7.2 Hz, J2 = 9.6 Hz, 1H); 1.44–1.40 (m, 2H); 1.26–1.18 (m, 4H); 0.82 (t, J = 7.2 Hz, 3H).
Methyl (R)-2-((benzyloxy)amino)hexanoate (64). The title compound was synthesized from (S)-methyl ester 62 according to the general procedure. The crude was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (PE/CHCl3 gradient from 30
:
1 to 8
:
1 v/v) to afford compound 64 as yellow oil (405.8 mg). Yield: 68%. 1H NMR (400 MHz, DMSO-d6) δ: 7.42–7.2 (m, 5H); 6.73 (d, J = 9.6 Hz, 1H); 4.59 (s, 2H); 3.62 (s, 3H); 3.54 (dt, J1 = 7.2 Hz, J2 = 9.6 Hz, 1H); 1.79–1.39 (m, 2H); 1.37–1.01 (m, 4H); 0.81 (t, J = 7.2 Hz, 3H).
Synthesis of methyl (S)-2-(phenethylamino)hexanoate (70). Methyl ester 69 (53 mg, 0.365 mmol, 1.2 equiv.) was dissolved in THF (3.04 mL), then (2-bromomethyl)benzene (0.04 mL, 0.304 mmol, 1 equiv.) and K2CO3 (42.01 mg, 0.304 mmol, 1 equiv.) were added. The reaction was refluxed for six days. THF was removed under vacuum conditions, and the crude was diluted with DCM and washed with H2O (1× 50 mL), and HCl (1× 50 mL). The organic phase was dried over Na2SO4, filtered, and concentrated under vacuum conditions. Trituration with n-hexane afforded methyl ester 70, as a white solid (14 mg). Yield: 18%. 1H NMR (400 MHz, DMSO-d6) δ: 9.62 (br s, 1H); 7.36–7.32 (m, 2H); 7.27–7.24 (m, 3H); 4.10 (m, 1H); 3.78 (s, 3H); 3.35–3.15 (m, 2H); 3.12–2.94 (m, 2H); 1.96–1.79 (m, 2H); 1.40–1.29 (m, 3H); 1.27–1.23 (m, 1H); 0.88 (t, J = 6.8 Hz, 3H).
Synthesis of (S)-N-hydroxy-2-(phenethylamino)hexanamide ((S)-19). To a solution of methyl ester 70 (22 mg, 0.088 mmol, 1 equiv.) in dry MeOH (0.5 mL), NH2OH·HCl (55.04 mg, 0.792 mmol, 9 equiv.), KOH (88.88 mg, 1.584 mmol, 18 equiv.) were added. The reaction was MW-conducted with the following parameters: T = 80 °C, initial power: 150 W, t = 15 min, 5 cycles. The crude reaction was first concentrated under vacuum conditions and then purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (isocratic eluent CHCl3/MeOH 50
:
1 v/v) to give the final compound (S)-19 as a white solid (6 mg). Yield: 27%. Mp: 149–152 °C. [α]D20 = −16.7 (c = 0.420, MeOH). 1H NMR (400 MHz, DMSO-d6) δ: 10.46 (s, 1H); 8.77 (s, 1H); 7.33–7.26 (m, 2H); 7.25–7.17 (m, 3H); 2.83 (t, J = 6.8 Hz, 1H); 2.69–2.56 (m, 4H); 1.39–1.35 (m, 2H); 1.24–1.17 (m, 5H); 0.84 (t, J = 6.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 170.7; 140.3; 128.5; 128.2; 125.8; 59.2; 49.0; 36.0; 32.9; 27.7; 22.0; 13.9. Elemental analysis for C14H22N2O2, calculated: % C, 67.17; % H, 8.86; % N, 11.19 found: % C, 67.00; % H, 8.93; % N, 11.15.
General procedure to synthesize N-benzyl derivatives 72–73. To a solution of the appropriate amino acid (69 or 71, 1 equiv.) and K2CO3 (1 equiv.) in CH3CN (2.90 mL mmol−1), 0.12 mL mmol−1 of benzyl bromide was added dropwise. The reaction mixture was stirred at rt overnight. The organic solvent was evaporated under reduced pressure and the crude was diluted with EtOAc and H2O with the addition of NaOH 1 N to a pH = 8 and washed with H2O (3×). The organic phase was dried over Na2SO4, filtered, and concentrated in vacuo.
Methyl (S)-2-(benzylamino)hexanoate (72). The title compound was prepared from methyl ester 69 following the general procedure. The crude reaction was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (n-hexane/EtOAc 20
:
1 v/v) to give the desired compound 72 as a colorless oil (263 mg). Yield: 32%. 1H NMR (400 MHz, DMSO-d6) δ: 7.30–7.20 (m, 5H); 3.75–3.71 (m, 1H); 3.62 (s, 3H); 3.54–3.50 (m, 1H); 3.13–3.11 (m, 1H); 2.39 (br s, 1H); 1.57–1.52 (m, 2H); 1.30–1.20 (m, 4H); 0.83 (t, J = 6.8 Hz, 3H).
Methyl (S)-2-(benzylamino)decanoate (73). The title compound was synthesized from methyl ester 71 according to the general procedure. The crude reaction was purified by flash chromatography using an ISOLUTE Si II 10 g cartridge (n-hexane/EtOAc 35
:
1 v/v) to give the desired compound 73 as a colorless oil (239 mg). Yield: 41%. 1H NMR (400 MHz, DMSO-d6) δ: 7.32–7.19 (m, 5H); 3.75–3.71 (m, 1H); 3.62 (s, 3H); 3.56–3.49 (m, 1H); 3.13–3.10 (m, 1H); 2.38 (br s, 1H); 1.55–1.50 (m, 2H); 1.31–1.16 (m, 12H); 0.85 (t, J = 6.4 Hz, 3H).
Synthesis of (E)-2-((benzyloxy)imino)pentanedioic acid (22). The commercial α-ketoglutaric acid (457 mg, 3.13 mmol, 1 equiv.) was added to a solution of the commercial O-benzyl-hydroxylamine hydrochloride 74 (500 mg, 3.13 mmol, 1 equiv.) in a mixture of THF/H2O (1
:
1, v/v, 1.96 mL mmol−1). The reaction was stirred at rt for 0.5 h. THF was removed under vacuum conditions and the aqueous solution was extracted with EtOAc (3×). The combined organic phases were dried over Na2SO4, filtered, and evaporated. Carboxylic acid 22 was obtained without any further purification methods as a white solid (570.4 mg). Yield: 73%. Mp: 109–111 °C. 1H NMR (400 MHz, DMSO-d6) δ: 12.59 (br s, 2H); 7.41–7.31 (m, 5H); 5.23 (s, 2H); 2.68 (t, J = 7.2 Hz, 2H); 2.41 (t, J = 7.2 Hz, 2H). 13C NMR (100 MHz, CDCl3) δ: 178.4; 165.2; 150.4; 136.0; 128.8; 128.7; 128.5; 78.5; 30.0; 20.3. Elemental analysis for C12H13NO5, calculated: % C, 57.37; % H, 5.22; % N, 5.58 found: % C, 57.23; % H, 5.45; % N, 5.78.
Synthesis of methyl (E)-4-((benzyloxy)imino)-5-(hydroxyamino)-5-oxopentanoate (23). Pyranyl derivative 75 (185 mg, 0.412 mmol, 1 mmol) was dissolved in a mixture of MeOH/1,4-dioxane 1
:
1 (11 mL) and an aqueous solution of HCl 4 N (11 mL) was added dropwise. The solution was stirred at rt for 3 h, the solvent was co-evaporated in vacuo with toluene to obtain a crude solid, which was purified by flash chromatography. The latter was performed using an ISOLUTE 5 g cartridge (CHCl3/MeOH from CHCl3 100% to 10
:
1 v/v) to give the desired product 23 as a yellow oil (14 mg). Yield: 12%. 1H NMR (400 MHz, DMSO-d6) δ: 10.90 (s, 1H); 9.06 (s, 1H); 7.39–7.33 (m, 5H); 5.25 (s, 2H); 3.56 (s, 3H); 2.69 (t, J = 7.6 Hz, 2H); 2.46 (t, J = 7.6 Hz, 2H). 13C NMR (100 MHz, CD3CN) δ: 173.3; 161.7; 153.1; 138.1; 129.4; 129.3; 129.2; 129.1; 78.40; 77.84; 52.12; 30.66; 30.24; 20.98. Elemental analysis for C13H16N2O5, calculated: % C, 55.71; % H, 5.75; % N, 10.00 found: % C, 55.59; % H, 5.67; % N, 9.99.
General procedure to synthesize O-tetrahydropyranyl derivatives 75–76. To a solution of the appropriate carboxylic acid (1 equiv.) in dry DMF (2.21 mL mmol−1), HOBt (1.2 equiv. for each carboxylic acid group), NMM (3 equiv. for each carboxylic acid function), THPONH2 (3.1 equiv. for each carboxylic acid group), and lastly EDC (1.4 equiv. for each carboxylic acid moiety) were added under nitrogen atmosphere. The reaction was stirred at rt overnight under nitrogen atmosphere. The reaction mixture was treated with NaHCO3 and extracted with DCM (3×). The combined organic phases were dried over Na2SO4, filtered, and evaporated in vacuo.
(E)-2-((Benzyloxy)imino)-N1,N5-bis((tetrahydro-2H-pyran-2-yl)oxy)pentanediamide (75). The title compound was prepared from di-carboxylic acid 22 according to the general procedure. After solvent evaporation, the desired product 75 was obtained without any further purification, as a yellow oil and as a mixture of four diastereoisomers (175 mg). Yield: 98%. 1H NMR (400 MHz, DMSO-d6) δ: 11.32 (s, 1H); 11.02 (s, 1H); 7.39–7.32 (m, 5H); 5.02 (s, 2H); 4.92–4.89 (m, 1H); 4.78–4.75 (m, 1H); 4.56–4.55 (m, 1H); 4.05–4.00 (m, 1H); 3.91–3.87 (m, 1H); 3.78–3.74 (m, 1H); 2.64–2.62 (m, 2H); 2.19–2.10 (m, 6H); 1.66–1.60 (m, 10H); 1.57–1.44 (m, 14H).
(E)-2-(((4-Chlorobenzyl)oxy)imino)-N-((tetrahydro-2H-pyran-2-yl)oxy)acetamide (76). The title compound was obtained from carboxylic acid 26 following the general procedure. After solvent evaporation, the crude was purified by flash chromatography using an ISOLUTE Si II 5 g cartridge (isocratic eluent DCM 100%) to afford pyranyl derivative 76, as a yellow oil (173.7 mg). Yield: 66%. 1H NMR (400 MHz, DMSO-d6) δ: 11.58 (s, 1H); 7.61 (s, 1H); 7.46–7.34 (m, 4H); 5.18 (s, 2H); 4.91 (s, 1H); 4.03–3.96 (m, 1H); 3.58–3.50 (m, 1H); 1.83–1.81 (m, 3H); 1.67–1.55 (m, 3H).
4.2. Biology assays
4.2.1. LasB inhibition assay. LasB was from Calbiochem (Cat# 324676; Lot# D00100981; San Diego, CA, USA), while the fluorescence quenched substrate McaRPPGFSAFK(Dnp)-OH was from R&D Systems (Minneapolis, MN, USA). LasB activity with McaRPPGFSAFK(Dnp)-OH and reaction velocities/initial rates (v) were determined at 37 °C at an excitation wavelength of 320 nm and an emission wavelength of 405 nm with a slit width of 10 nm, using either a Perkin Elmer LS 50 Luminescence spectrometer with the FL WinLab software package (Perkin Elmer) or a Clario Star micro plate reader (CLARIOstar® BMG LABTECH).The test compounds were dissolved in 100% DMSO giving a concentration of 10 mM. A substrate concentration of 4.0 μM in a total volume of 100 μL of 0.1 M Hepes (pH 7.5), 10 mM CaCl2, 0.005% Brij-35, 1.0% DMSO and LasB concentration of 0.21 nM were used in all assays. To determine if the compounds are slow binders or not, time dependent inhibitory experiments were performed with all compounds. Briefly, time dependent inhibitory experiments with 100 μM of compound were performed as follows: at time zero the reaction was started by adding LasB to a mixture of substrate and compound. Further, compounds were pre-incubated with LasB for 15 and 30 min at room temperature and then the enzymatic reaction was started by adding the substrate. Control tests without compound were performed in the same way. The reaction was followed for 30 min at 37 °C using the Clario Star micro plate reader.
As none of the compounds were found to be slow binders, all IC50 and Ki values were determined without pre-incubation. IC50 and Ki values were determined for all compounds that gave a vi/v0 value smaller than 0.5. For compounds giving vi/v0 value larger than 0.5, Ki values are reported as >100 μM. In the determination of IC50 and Ki values, the concentration of the inhibitory compounds varied from 0 up to maximum 100 μM depending on the strength of the inhibition, using a constant concentration of substrate, enzyme and buffer as described above. For each compound, the number of different concentrations tested was at least 8, all in triplicates, and at least two independent assays were performed. The IC50 values were calculated with GraphPad Prism 5 using eqn (1)
|
vi/vo = 1/(1 + [I]/IC50)
| (1) |
where
vi represents the enzyme activity in the presence of the inhibitor, while
v0 denotes activity in its absence.
Eqn (2) describe the relationship between IC
50 and
Ki values for substrate competitive inhibitors, considering the fixed substrate concentration and the enzyme's
Km value for the substrate:
|
IC50 = Ki(1 + [S]/Km)
| (2) |
The
Km value for LasB under the experimental conditions used in the present study was 24 ± 8 μM.
64
4.2.2. Selectivity assays against human MMPs. Recombinant human MMP-14 catalytic domain was a kind gift of Prof. Gillian Murphy (Department of Oncology, University of Cambridge, UK). Pro-MMP-2 (PF037) and pro-MMP-9 (PF038) were purchased from Merck Millipore (Burlington, MA, USA). p-Aminophenylmercuric acetate (APMA, A9563) was from Sigma-Aldrich (Milan, Italy). Proenzymes were activated immediately prior to use with APMA 2 mM for 1 h at 37 °C for MMP-2 and APMA 1 mM for 1 h at 37 °C for MMP-9. For assay measurements, the compound stock solution (10 mM in DMSO) was further diluted for each MMP in the fluorometric assay buffer (FAB: Tris 50 mM, pH = 7.5, NaCl 150 mM, CaCl2 10 mM, Brij 35 0.05% and DMSO 1%). Activated enzyme (final concentration 0.56 nM for MMP-2, 1.3 nM for MMP-9, 1.0 nM for MMP-14 cd) and inhibitor solutions were incubated in the assay buffer for 3 h at 25 °C. After the addition of 200 μM solution of the fluorogenic substrate Mca–Lys–Pro–Leu–Gly–Leu–Dap(Dnp)–Ala–Arg–NH2 (444282, Merck Millipore) in DMSO (final concentration 2 μM), the hydrolysis was monitored for 15 min recording the increase in fluorescence (λex = 325 nm, λem = 400 nm) using a SpectraMax Gemini XPS plate reader (Molecular Devices, Sunnyvale, CA). The assays were performed in duplicate in a total volume of 200 μL per well in 96-well microtitre plates (Corning, black, NBS). The MMP inhibition activity was expressed in relative fluorescence units (RFU) and percent of inhibition was calculated from control reactions without inhibitor. IC50 was determined using the formula: vi/vo = 1/(1 + [I]/IC50), where vi is the initial velocity of substrate cleavage in the presence of the inhibitor at concentration [I] and vo is the initial velocity in the absence of the inhibitor. Results were analyzed using SoftMax Pro software version 5.4.3 and Origin 6.0 software.
4.2.3. Bacterial strain cultivation and supernatant preparation. The bacterial strain used in this study was PAO1 (ATCC), a reference strain of P. aeruginosa. Bacterial culture supernatant of PAO1 was prepared from overnight cultures in Luria–Bertani broth medium (LB; Sigma-Aldrich) for 24 h. The cultures were centrifuged at 3250 × g and filtered using a 0.2 μm non-pyrogenic sterile filter.
4.2.4. Determination minimum inhibitory concentrations (MICs). MICs of inhibitors against PAO1 were determined following the standard broth microdilution method recommended by The European Committee on Antimicrobial Susceptibility Testing (EUCAST https://www.eucast.org/clinical_breakpoints). Briefly, exponential phase cultures of PAO1 were diluted in Mueller Hinton broth (MHB; Sigma-Aldrich) to obtain 1 × 106 CFU mL−1, and 50 μL was added to 96-well plates (Euroclone). Inhibitors were serially diluted in MHB, and a volume of 50 μL was added to each well. DMSO that was used to dissolve the compounds was added to control samples. Inhibitors were tested up to 200 μM, corresponding to a DMSO concentration of 2%. MIC values were defined as the lowest concentration of inhibitors that inhibited visible growth after 24 h of incubation at 37 °C.
4.2.5. Cytotoxicity. The cytotoxic potential of the compounds was tested against the human lung adenocarcinoma cell line NCI-H441 (ATCC, HTB-174). NCI-H441 cells were seeded on a 96-well plate at a density of 35
000 cells per well in Royal Park Memorial Institute 1640 medium (RPMI) added with 10% fetal bovine serum (FBS) and 2 mM L-glutamine [complete RPMI]. Cells were incubated in a humidified atmosphere containing 5% CO2 for 24 h at 37 °C to reach approximately 90–100% confluence. Non-adherent cells were then removed by washing, and adherent cells were added with 100 μL of inhibitors diluted at 25, 50, 100, and 200 μM in complete RPMI or with 100 μL of complete RPMI added with DMSO at 0.25, 0.5, 1 and 2%, and further incubated for 24 h at 37 °C. At the end of the incubation, the wells were washed once with 200 μL warm PBS. The WST-1 assay was used to measure the metabolic activity of the cells. To this aim, 10 μL of WST-1 reagent was added to the wells. After 45 min of incubation at 37 °C, the absorbance was measured using a microplate reader (Multiskan Ascent, Thermo Fisher Scientific, Waltham, MA, USA) at 450 nm.
4.2.6. Elastolytic assay to evaluate LasB activity in P. aeruginosa PAO1. This assay measures the elastolytic activity of secreted LasB in the supernatant of P. aeruginosa cultures using elastin Congo red (ECR, Elastin Company, Missouri, USA) as substrate. LasB digests elastin and releases the Congo red dye into the supernatant which can then be measured spectrophotometrically. The supernatant of PAO1 was incubated with inhibitors used at concentrations ranging from 3.125 to 200 μM and incubated for 30 min at 37 °C. The pre-treated supernatant was then mixed 1
:
1 with 2× ECR solution (final concentration of 10 mg mL−1 ECR in 50 mM Tris-HCl pH 7.4 and 0.5 mM CaCl2) and incubated for 18 h in a 37 °C shaking incubator. The supernatants were centrifuged, and the released Congo-red dye was quantitated by its absorbance at 495 nm. As a negative control, supernatants were replaced by LB medium and DMSO 2%.
4.2.7. Biofilm inhibition assay. PAO1 grown overnight in LB at 37 °C was diluted 1
:
100 in LB. Bacterial suspensions were dispensed into flat-bottom polystyrene 96-well microplates (Corning Costar, Lowell, MA, USA) in the presence of sub-MIC concentrations of inhibitors (from 25 to 200 μM). Plates were incubated for 24 h in static conditions at 37 °C to let the biofilms grow. DMSO was added to the samples used as a control. Following incubation, wells were washed thrice with PBS to remove non-biofilm-embedded bacteria, dried for 1 h at 60 °C and incubated for 15 min with 1% (w/v) crystal-violet (CV) (bioMérieux, Florence, Italy). Following incubation, unbound CV was removed by extensively washing the plate with PBS. After drying the plates at 37 °C for 30 min, biofilm-associated CV was extracted with 33% acid acetic (Sigma Aldrich) and quantified by measuring the optical density at 570 nm (OD570) in a microplate reader (Multiskan FC, Thermo-Fisher Scientific, Monza, Italy).
4.2.8. Statistical analysis. Data were analyzed using GraphPad Prism (Dotmatics, Boston, MA, USA). All the experiments were conducted at least in triplicate. Differences between mean values were evaluated by one-way or two-way analysis of variance (ANOVA), followed by the Tukey–Kramer post hoc test. A p-value of <0.05 was considered significant.
4.3. Computational methods
The crystal structure used to perform non-covalent docking was downloaded from the Protein Data Bank (PDB). PDB ID of LasB is 1U4G. Modelling studies were performed with Maestro, release 2023-01, version 13.1, with the tool named Glide.
4.3.1. Ligand preparation. Prior to docking, the ligands were drawn in Maestro's 2D Sketcher, then converted to their 3D structure and energy minimized using the LigPrep65 module. Default parameters were selected in the LigPrep panel, except for the following options: (a) retain specified chiralities (vary other chiral centers); (b) use Ionizer or Epik66 (including and excluding metal binding states) for ionization and tautomerization; (c) use 7.0 as effective pH; and (d) use 1.0 as pH tolerance for generated structures.
4.3.2. Protein preparation. Before the protein preparation, the electron density maps of the structure 1U4G was analyzed using Maestro's PrimeX tool.67 Then it was prepared using Maestro's Protein Preparation Wizard68 according to the following procedure: (a) use 7.0 as simulation pH; (b) cap termini; (c) fill in missing side chains; (d) assign bond order; (e) use CCD (chemical component dictionary) database. The chemical component dictionary is an external reference file for PDB entries that provides detailed information on standard and modified amino acids/nucleotides, small molecule components and solvent molecules. The dictionary provides comprehensive information such as stereochemical assignments, chemical descriptions, systematic names and ideal coordinates. It is essential for understanding the chemical composition of PDB biomolecular structures;69 (f) replace hydrogens; (g) convert selenomethionines into methionines; (h) fill in missing loops; (i) generate het states (with Epik) at pH 7.0 ± 1.0; (j) PROPKA as optimization. In fact, based on the 3D structure, PROPKA is able to predict the pKa of the ionizable group of the protein and the ligand–protein complex;70,71 (k) converge heavy atoms to RMSD 0.30. After Wizard preparation, water molecules were removed.
4.3.3. Grid preparation. The grid for protein–ligand docking was generated with Maestro72 by using default parameter values and the co-crystallized ligand as a centroid.
4.3.4. Docking. Protein–ligand docking was performed with Glide73 using default parameter values except for the following settings: (a) standard precision (SP) and extra precision (XP), with the different multiple ionic and tautomeric states produced with Ionizer or Epik, respectively; (b) write a maximum of 50 poses per ligand; (c) include 50 poses per ligand in the post-docking minimization; (d) shape constraints were set by selecting all atoms of the ligand within the complex.
4.3.5. Molecular dynamics. Molecular dynamics simulations were conducted using the Desmond package (Schrödinger, Maestro, release 2023-01, version 13.1) to analyze the dynamic behavior of compound 12. The simulation was performed under the NPT ensemble for 100 ns to assess the structural stability and interaction patterns of the ligand–protein complex. The system was solvated in a periodic octahedral box with TIP3P water molecules, ensuring at least 10 Å of solvation around the solutes. Charge neutrality was maintained with Na+ and Cl− counter ions, and the ionic strength was adjusted to 0.15 M NaCl. System relaxation followed Desmond's standard protocol, including energy minimization and equilibration, before the MD simulation.74,75 Temperature was controlled at 300 K using the Nose-Hoover thermostat, while pressure was maintained at 1.01325 bar with the Martyna–Tobias–Klein method. Short-range nonbonded interactions used a 9 Å cutoff.75,76 Due to the +2 charge of the zinc ion and surrounding anionic residues, special treatment was required to maintain active site integrity. The zinc ion was coordinated by two histidines, a glutamic acid, and a water molecule, with inhibitors typically replacing the water via a ZBG. To preserve coordination stability, a force constant of 50 kcal Å−1 was applied between the zinc ion and its amino acid ligands: His 140 (NE2), His 144 (NE2), and Glu 164 (OE1).38
Data availability
The data supporting this article have been included as part of the ESI.†
Author contributions
Conceptualization, E. N., A. R., I. S., and J.-O. W.; investigation, R. D. L., E. C., A. B., M. B., I. W., and F. A. R.; formal analysis, J.-O. W., G. M., and R. D. L.; data curation, E. N., D. C., and R. D. L.; writing – original draft, R. D. L., E. C., and E. N.; writing – review and editing, E. N., D. C., I. S., G. M., and J.-O. W.; supervision, E. N., A. R., I. S., J.-O. W., and G. B.; funding acquisition, A. R., I. S., E. N., G. B., and G. M. All authors have read and approved the final version of the manuscript.
Conflicts of interest
There are no conflicts to declare.
Acknowledgements
This work was supported by the Northern Norway Health Authorities (Helse Nord) (grant number HNF 1514-20) to I. S. and A. R., by University of Pisa (PRA_2022_19) to E. N., by the European Union – Next Generation EU, PNRR MUR M4 C2 Inv. 1.5 CUP I53C22000780001 to G. B. and by Telethon Foundation “Seed Grant Spring 2024 PCD” ref. number 5737 to G. M. We thank CISUP – Centre for Instrumentation Sharing – University of Pisa for the acquisition and elaboration of the HRMS spectra. We are grateful to the Cheminformatics & Nutrition research group from the Universitat Rovira i Virgili (Tarragona, Spain) for allowing us to use the Schrödinger Drug Discovery suite in their computers.
References
- M. Haque, M. Sartelli, J. McKimm and M. B. Abu Bakar, Infect. Drug Resist., 2018, 11, 2321–2333 CrossRef PubMed
. - M. Rodríguez-Alvarado, R. Russo, N. D. Connell and S. E. Brenner-Moyer, Org. Biomol. Chem., 2020, 18, 5867–5878 RSC
. - J.-L. Vincent, JAMA, J. Am. Med. Assoc., 2009, 302, 2323 CrossRef CAS PubMed
. - U. Theuretzbacher, R. P. Jumde, A. Hennessy, J. Cohn and L. J. V. Piddock, Nat. Rev. Microbiol., 2025, 23, 474–490 CrossRef CAS PubMed
. - N. Safdar, C. J. Crnich and D. G. Maki, Respir. Care, 2005, 50, 725–739 Search PubMed
; discussion 739–741. - N. A. M. Cobben, M. Drent, M. Jonkers, E. F. M. Wouters, M. Vaneechoutte and E. E. Stobberingh, J. Hosp. Infect., 1996, 33, 63–70 CrossRef CAS PubMed
. - S. Schelenz and G. French, J. Hosp. Infect., 2000, 46, 23–30 CrossRef CAS PubMed
. - H. A. Khan, F. K. Baig and R. Mehboob, Asian Pac. J. Trop. Biomed., 2017, 7, 478–482 CrossRef
. - S. Qin, W. Xiao, C. Zhou, Q. Pu, X. Deng, L. Lan, H. Liang, X. Song and M. Wu, Signal Transduction Targeted Ther., 2022, 7, 199 CrossRef CAS PubMed
. - S. Malhotra, D. Hayes and D. J. Wozniak, Clin. Microbiol. Rev., 2019, 32, e00138-18 CrossRef PubMed
. - T. E. Woo, J. Duong, N. M. Jervis, H. R. Rabin, M. D. Parkins and D. G. Storey, Microbiology, 2016, 162, 2126–2135 CrossRef CAS PubMed
. - J. L. L. Ferreiro, J. Á. Otero, L. G. González, L. N. Lamazares, A. A. Blanco, J. R. B. Sanjurjo, I. R. Conde, M. F. Soneira and J. De La Fuente Aguado, PLoS One, 2017, 12, e0178178 CrossRef PubMed
. - A. David, A. Tahrioui, A.-S. Tareau, A. Forge, M. Gonzalez, E. Bouffartigues, O. Lesouhaitier and S. Chevalier, Antibiotics, 2024, 13, 688 CrossRef CAS PubMed
. - N. Høiby, C. Moser and O. Ciofu, Microorganisms, 2024, 12, 2115 CrossRef PubMed
. - Z. Pang, R. Raudonis, B. R. Glick, T.-J. Lin and Z. Cheng, Biotechnol. Adv., 2019, 37, 177–192 CrossRef CAS PubMed
. - S. Wagner, R. Sommer, S. Hinsberger, C. Lu, R. W. Hartmann, M. Empting and A. Titz, J. Med. Chem., 2016, 59, 5929–5969 CrossRef CAS PubMed
. - M. B. Calvert, V. R. Jumde and A. Titz, Beilstein J. Org. Chem., 2018, 14, 2607–2617 CrossRef CAS PubMed
. - A. B. H. Khalifa, D. Moissenet, H. Vu Thien and M. Khedher, Ann. Biol. Clin., 2011, 69, 393–403 Search PubMed
. - Y. Kida, Y. Higashimoto, H. Inoue, T. Shimizu and K. Kuwano, Cell. Microbiol., 2008, 10, 1491–1504 CrossRef CAS PubMed
. - E. Kessler and D. E. Ohman, in Handbook of Proteolytic Enzymes, Elsevier, 2013, pp. 582–592 Search PubMed
. - B. Wretlind and O. R. Pavlovskis, Clin. Infect. Dis., 1983, 5, S998–S1004 CrossRef CAS PubMed
. - S. Sharma, J. Mohler, S. D. Mahajan, S. A. Schwartz, L. Bruggemann and R. Aalinkeel, Microorganisms, 2023, 11, 1614 CrossRef CAS PubMed
. - J. Konstantinović, A. M. Kany, A. Alhayek, A. S. Abdelsamie, A. Sikandar, K. Voos, Y. Yao, A. Andreas, R. Shafiei, B. Loretz, E. Schönauer, R. Bals, H. Brandstetter, R. W. Hartmann, C. Ducho, C.-M. Lehr, C. Beisswenger, R. Müller, K. Rox, J. Haupenthal and A. K. H. Hirsch, ACS Cent. Sci., 2023, 9, 2205–2215 CrossRef PubMed
. - N. D. Rawlings, A. J. Barrett, P. D. Thomas, X. Huang, A. Bateman and R. D. Finn, Nucleic Acids Res., 2018, 46, D624–D632 CrossRef CAS PubMed
. - O. A. Adekoya and I. Sylte, Chem. Biol. Drug Des., 2009, 73, 7–16 CrossRef CAS PubMed
. - G. R. A. Cathcart, D. Quinn, B. Greer, P. Harriott, J. F. Lynas, B. F. Gilmore and B. Walker, Antimicrob. Agents Chemother., 2011, 55, 2670–2678 CrossRef CAS PubMed
. - J. Zhu, X. Cai, T. L. Harris, M. Gooyit, M. Wood, M. Lardy and K. D. Janda, Chem. Biol., 2015, 22, 483–491 CrossRef CAS PubMed
. - D. Grobelny, L. Poncz and R. E. Galardy, Biochemistry, 1992, 31, 7152–7154 CrossRef CAS PubMed
. - M. J. Everett, D. T. Davies, S. Leiris, N. Sprynski, A. Llanos, J. M. Castandet, C. Lozano, C. N. LaRock, D. L. LaRock, G. Corsica, J.-D. Docquier, T. D. Pallin, A. Cridland, T. Blench, M. Zalacain and M. Lemonnier, ACS Infect. Dis., 2023, 9, 270–282 CrossRef CAS PubMed
. - J. L. Fullagar, A. L. Garner, A. K. Struss, J. A. Day, D. P. Martin, J. Yu, X. Cai, K. D. Janda and S. M. Cohen, Chem. Commun., 2013, 49, 3197 RSC
. - A. L. Garner, A. K. Struss, J. L. Fullagar, A. Agrawal, A. Y. Moreno, S. M. Cohen and K. D. Janda, ACS Med. Chem. Lett., 2012, 3, 668–672 CrossRef CAS PubMed
. - C. Camodeca, D. Cuffaro, E. Nuti and A. Rossello, Curr. Med. Chem., 2019, 26, 2661–2689 CrossRef CAS PubMed
. - L. G. N. De Almeida, H. Thode, Y. Eslambolchi, S. Chopra, D. Young, S. Gill, L. Devel and A. Dufour, Pharmacol. Rev., 2022, 74, 714–770 CrossRef CAS PubMed
. - D. Cuffaro, E. Nuti, F. D'Andrea and A. Rossello, Pharmaceuticals, 2020, 13, 376 CrossRef CAS PubMed
. - R. Di Leo, D. Cuffaro, A. Rossello and E. Nuti, Molecules, 2023, 28, 4378 CrossRef CAS PubMed
. - B. Fabre, A. Ramos and B. De Pascual-Teresa, J. Med. Chem., 2014, 57, 10205–10219 CrossRef CAS PubMed
. - C. Kaya, I. Walter, A. Alhayek, R. Shafiei, G. Jézéquel, A. Andreas, J. Konstantinović, E. Schönauer, A. Sikandar, J. Haupenthal, R. Müller, H. Brandstetter, R. W. Hartmann and A. K. H. Hirsch, ACS Infect. Dis., 2022, 8, 1010–1021 CrossRef CAS PubMed
. - S. Sjøli, E. Nuti, C. Camodeca, I. Bilto, A. Rossello, J.-O. Winberg, I. Sylte and O. A. Adekoya, Eur. J. Med. Chem., 2016, 108, 141–153 CrossRef PubMed
. - E. Nuti, E. Orlandini, S. Nencetti, A. Rossello, A. Innocenti, A. Scozzafava and C. T. Supuran, Bioorg. Med. Chem., 2007, 15, 2298–2311 CrossRef CAS PubMed
. - O. A. Adekoya, S. Sjøli, Y. Wuxiuer, I. Bilto, S. M. Marques, M. A. Santos, E. Nuti, G. Cercignani, A. Rossello, J.-O. Winberg and I. Sylte, Eur. J. Med. Chem., 2015, 89, 340–348 CrossRef CAS PubMed
. - H. Sabishiro, Y. Nagura, N. Chimura, M. Yuguchi, C. Nakatani, S. Takenaka, H. Sugizaki and N. Kurita, J. Mol. Graphics Modell., 2025, 137, 108977 CrossRef CAS PubMed
. - M. Mitani, Y. Tanaka, A. Sawada, A. Misu and Y. Matsumoto, Eur. J. Org. Chem., 2008, 2008, 1383–1391 CrossRef
. - L. Gu, P. G. Luo, H. Wang, M. J. Meziani, Y. Lin, L. M. Veca, L. Cao, F. Lu, X. Wang, R. A. Quinn, W. Wang, P. Zhang, S. Lacher and Y.-P. Sun, Biomacromolecules, 2008, 9, 2408–2418 CrossRef CAS PubMed
. - S. B. McNabb, M. Ueda and T. Naito, Org. Lett., 2004, 6, 1911–1914 CrossRef CAS PubMed
. - N. Venugopal, J. Moser, M. Vojtíčková, I. Císařová, B. König and U. Jahn, Adv. Synth. Catal., 2022, 364, 405–412 CrossRef CAS
. - L. Baldini, E. Lenci, F. Bianchini and A. Trabocchi, Molecules, 2022, 27, 1249 CrossRef CAS PubMed
. - E. Nuti, D. Cuffaro, E. Bernardini, C. Camodeca, L. Panelli, S. Chaves, L. Ciccone, L. Tepshi, L. Vera, E. Orlandini, S. Nencetti, E. A. Stura, M. A. Santos, V. Dive and A. Rossello, J. Med. Chem., 2018, 61, 4421–4435 CrossRef CAS PubMed
. - T. Naito, Chem. Pharm. Bull., 2008, 56, 1367–1383 CrossRef CAS PubMed
. - J. Müller, S. C. Feifel, T. Schmiederer, R. Zocher and R. D. Süssmuth, ChemBioChem, 2009, 10, 323–328 CrossRef PubMed
. - K. Kawai and N. Nagata, Eur. J. Med. Chem., 2012, 51, 271–276 CrossRef CAS PubMed
. - J. Kyte and R. F. Doolittle, J. Mol. Biol., 1982, 157, 105–132 CrossRef CAS PubMed
. - E. Nuti, A. R. Cantelmo, C. Gallo, A. Bruno, B. Bassani, C. Camodeca, T. Tuccinardi, L. Vera, E. Orlandini, S. Nencetti, E. A. Stura, A. Martinelli, V. Dive, A. Albini and A. Rossello, J. Med. Chem., 2015, 58, 7224–7240 CrossRef CAS PubMed
. - M. Raeeszadeh-Sarmazdeh, L. Do and B. Hritz, Cells, 2020, 9, 1313 CrossRef CAS PubMed
. - U. Neumann, H. Kubota, K. Frei, V. Ganu and D. Leppert, Anal. Biochem., 2004, 328, 166–173 CrossRef CAS PubMed
. - V. Gifford and Y. Itoh, Biochem. Soc. Trans., 2019, 47, 811–826 CrossRef CAS PubMed
. - H. Yu, X. He, W. Xie, J. Xiong, H. Sheng, S. Guo, C. Huang, D. Zhang and K. Zhang, Can. J. Microbiol., 2014, 60, 227–235 CrossRef CAS PubMed
. - F. Soukarieh, P. Williams, M. J. Stocks and M. Cámara, J. Med. Chem., 2018, 61, 10385–10402 CrossRef CAS PubMed
. - Z. Lin, J. Li, Q. Huang, Q. Huang, Q. Wang, L. Tang, D. Gong, J. Yang, J. Zhu and J. Deng, J. Org. Chem., 2015, 80, 4419–4429 CrossRef CAS PubMed
. - Y. Ji, P. Xue, D.-D. Ma, X.-Q. Li, P. Gu and R. Li, Tetrahedron Lett., 2015, 56, 192–194 CrossRef CAS
. - C. R. Ganellin, P. B. Bishop, R. B. Bambal, S. M. T. Chan, J. K. Law, B. Marabout, P. M. Luthra, A. N. J. Moore, O. Peschard, P. Bourgeat, C. Rose, F. Vargas and J.-C. Schwartz, J. Med. Chem., 2000, 43, 664–674 CrossRef CAS PubMed
. - W. A. Gibbons, R. A. Hughes, M. Charalambous, M. Christodoulou, A. Szeto, A. E. Aulabaugh, P. Mascagni and I. Toth, Liebigs Ann. Chem., 1990, 1990, 1175–1183 CrossRef
. - R. K. Bowman and J. S. Johnson, Org. Lett., 2006, 8, 573–576 CrossRef CAS PubMed
. - E. Busto, N. Richter, B. Grischek and W. Kroutil, Chem. – Eur. J., 2014, 20, 11225–11228 CrossRef CAS PubMed
. - F. Rahman, T.-M. Nguyen, O. A. Adekoya, C. Campestre, P. Tortorella, I. Sylte and J.-O. Winberg, J. Enzyme Inhib. Med. Chem., 2021, 36, 819–830 CrossRef CAS PubMed
. - Schrödinger Release 2016-3: LigPrep, Schrödinger, LLC, New York, NY, USA, 2016 Search PubMed
. - Schrödinger Release 2016-3: Epik, Schrödinger, LLC, New York, NY, USA, 2016 Search PubMed
. - J. A. Bell, Y. Cao, J. R. Gunn, T. Day, E. Gallicchio, Z. Zhou, R. Levy and R. Farid, International Tables for Crystallography, Wiley, 2012, ch. 18.10, vol. F, pp. 534–538 Search PubMed
. - Schrödinger Release 2016-3: Schrödinger Suite 2016-3 Protein Preparation Wizard; Epik, Schrödinger, LLC: New York, NY, USA; and Impact, Schrödinger, LLC: New York, NY, USA; Prime, Schrödinger, LLC: New York, NY, USA, 2016 Search PubMed.
- J. D. Westbrook, C. Shao, Z. Feng, M. Zhuravleva, S. Velankar and J. Young, Bioinformatics, 2015, 31, 1274–1278 CrossRef PubMed
. - M. H. M. Olsson, C. R. Søndergaard, M. Rostkowski and J. H. Jensen, J. Chem. Theory Comput., 2011, 7, 525–537 CrossRef CAS PubMed
. - C. R. Søndergaard, M. H. M. Olsson, M. Rostkowski and J. H. Jensen, J. Chem. Theory Comput., 2011, 7, 2284–2295 CrossRef PubMed
. - Schrödinger Release 2016-3: Maestro, Schrödinger, LLC, New York, NY, USA, 2016 Search PubMed
. - Schrödinger Release 2016-3: Glide, Schrödinger, LLC, New York, NY, USA, 2016 Search PubMed
. - D. A. Anil, M. F. Polat, R. Saglamtas, A. H. Tarikogullari, M. A. Alagoz, I. Gulcin, O. Algul and S. Burmaoglu, Comput. Biol. Chem., 2022, 100, 107748 CrossRef CAS PubMed
. - O. Ozten, B. Zengin Kurt, F. Sonmez, B. Dogan and S. Durdagi, Bioorg. Chem., 2021, 115, 105225 CrossRef CAS PubMed
. - G. J. Martyna, D. J. Tobias and M. L. Klein, J. Chem. Phys., 1994, 101, 4177–4189 CrossRef CAS
.
Footnote |
† Electronic supplementary information (ESI) available: 1H-NMR spectra of final compounds 2–7 and 10–24; 13C-NMR spectra of final compounds 2, 4–6, 10–24; 19F-NMR spectra of final compounds 13–16; HRMS spectra of final compounds 2–4, 7, 10, 12, 14, 15 and 17; Fig. S1: binding of compound 5 to LasB; Fig. S2: RMSD plot of compound 12 and protein heavy atoms in the LasB complex during MD simulations; Fig. S3: LasB residues interacting with compound 12 and their interaction frequency during the MD simulation; chemical stability of compound 12. See DOI: https://doi.org/10.1039/d5md00393h |
|
This journal is © The Royal Society of Chemistry 2025 |
Click here to see how this site uses Cookies. View our privacy policy here.