Next Article in Journal
Biofilm Formation, and Related Impacts on Healthcare, Food Processing and Packaging, Industrial Manufacturing, Marine Industries, and Sanitation–A Review
Next Article in Special Issue
Pterostilbene, a Natural Methoxylated Analog of Resveratrol, Exhibits Antifungal Activity Induced by Reactive Oxygen Species Production and Plasma Membrane Injury
Previous Article in Journal / Special Issue
Asteltoxins: Synthesis and Biological Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

β-Lactams and Ureas as Cross Inhibitors of Prokaryotic Systems

by
Monika I. Konaklieva
1,* and
Balbina J. Plotkin
2
1
Department of Chemistry, American University, 4400 Massachusetts Ave. NW, Washington, DC 20016, USA
2
Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA
*
Author to whom correspondence should be addressed.
Appl. Microbiol. 2023, 3(3), 605-628; https://doi.org/10.3390/applmicrobiol3030043
Submission received: 31 May 2023 / Revised: 21 June 2023 / Accepted: 24 June 2023 / Published: 25 June 2023

Abstract

:
β-Lactams in the last thirty years have been viewed as universal acylating agents of serine and cysteine enzymes of both prokaryotic and eukaryotic systems. More recently, their use has been propelled by the COVID-19 pandemic, thus broadening their application as inhibitors of viral enzymes. The urea-based drugs have been extensively studied as inhibitors of the aforementioned enzymes. The focus of this review is the last decade’s drug discovery strategies, as well as new strategies that show utility in the expansion of β-lactams and ureas in the development of new antimicrobial and antiviral drugs.

1. Introduction

The majority of currently approved FDA drugs bind with reversible noncovalent interactions to their biological (molecular) targets. However, in the last decade, there has been increased interest in covalently binding enzyme inhibitors as potential antimicrobials. Covalent drugs incorporate an electrophilic functional group that forms a covalent bond with a nucleophile on their molecular targets. In fact, most compounds that can form covalent bonds with their drug targets were discovered serendipitously through the use of naturally occurring enzyme inhibitors, e.g., amides.
The structural features of amides have been utilized by nature for millennia. One of the most successful examples of naturally occurring amide-based enzyme products targeting proteins is the group of β-lactams. For close to a century, the β-lactams have served as a rich source of marketed drugs and have opened new areas of target-based drug discovery. They are representatives of the mechanism-based covalent modification of a target enzyme as a key step in achieving potent inhibition of that target. They are also an example of how nature utilizes small molecules to modify key catalytic residues at an enzyme active site. Newly developed covalent modulators of non-catalytic amino acid residues have also been successful as anticancer drugs.
More recently, there has been a turn in the direction of drug development towards the utilization of ureas as targeted covalent enzyme inhibitors. Originally, ureas were marginalized by organic/medicinal chemistry since they lacked the natural ubiquity of amides, which were the historical focus of enzyme inhibition research. However, after a slow start as antimicrobials, initiated by Paul Ehrlich, urea-based inhibitors have gained traction in the past twenty years as HIV and other viral inhibitors, as well as anticancer drugs.
Our focus herein will be on the general electrophile–nucleophile interaction utilized by β-lactams and ureas and their bioisosters in the context of developing anti-infectives in the current age of rapid microbial resistance growth.
  • General Chemistry and Structure
Most current β -lactams that target microbes modify their targets, mainly proteases, and are viewed as universal acylating agents of prokaryotic and eukaryotic serine and cysteine enzymes through covalent modification [1,2,3]. These targets and mechanisms of action are shared by a broad range of β-lactam structures, with penicillin being the first-identified antimicrobial of this drug class. The value of the β-lactam ring, a small heterocycle which features a reactive electrophilic center, has been demonstrated for a multitude of molecular targets (Figure 1).
There are also other examples, such as clinically relevant Ezetimibe (Figure 2), intended as an inhibitor (acylating agent) of one of the enzymes associated with the mammalian lipid metabolism (Acyl-CoA:cholesterol acyltransferase, ACAT); however, its molecular target has not yet been completely identified. Currently, Ezetimibe’s mechanism of action is accepted to be an inhibition of a transport protein target called Niemann-Pick C1- Like 1 (NPC1L1) [4,5]. Although non-acylating β-lactams, e.g., alkylthio-β-lactams (Figure 2), also contain the β-lactam functionality, their anti-microbial activity it is not based on enzyme-acylating, but on S-alkylation of the bacterial coenzyme A (CoA) [6,7]. The focus of this review will be on the acylating agents, which are the largest β-lactam class developed to target infectious agents (Figure 1).
  • Lactams—Cyclic Amides
The stability and acylating function of a simple β-lactam ring is comparable to that of simple acyclic amide. Briefly, the carbonyl group of either the acyclic amide, or its cyclic counterpart, e.g., the β-lactam, engages in hydrogen bonding in the so-called “oxyanion” hole of a given enzyme. This binding allows for stabilization of the negative charge on the oxygen atom of the carbonyl, which secures the general acid–base catalysis, namely an attack of a nucleophile, most often a primary alcohol, such as the one from serine on the amide carbonyl carbon, as the electrophile. The nucleophilic attack on the latter leads to the formation of a tetrahedral intermediate, a species whose collapse results in acylation of the enzyme. This covalent binding of an amide/β-lactam with an enzyme has essential requirements, which have been extensively described in the literature [1,8,9].
Overall, β-lactams exhibit good pharmacokinetics/pharmacodynamics. Thus, they continue to be on the forefront of recently approved antimicrobial drugs, with the newest being the “pan”-antimicrobial cephalosporin Cefiderocol (Figure 3). This cephalosporin is the only one out of the twelve antimicrobials approved in the past five years, with activity against critical class pathogens as defined by the World Health Organization (WHO) [10]. Cefiderocol has a catechol moiety in its side chain which acts as an iron chelator. This allows Cefiderol to be actively transported across the cell membrane via iron transporter channels [11]. This transport process avoids current resistance mechanisms associated with passive diffusion. Furthermore, Cefiderocol is minimally affected by multidrug efflux pumps and is stable against hydrolysis by β-lactamases, including metallo-β-lactamases [11,12]. This ability to bypass several major mechanisms of bacterial resistance makes Cefiderocol suitable for the treatment of Gram-negative bacteria [11,12].
There are a number of other amide-based clinically approved acyclic amides and β-lactam drugs, including lidocaine, paracetamol, β-lactam antibiotics, and β-lactamase inhibitors, e.g., penicillin, clavulanic acid, and monocyclic β-lactams, as well as compounds with various other activities, e.g., ezetimibe, atorvastatin, chloramphenicol, moclobemide, captopril, acetazolamide, ponatinib, methotrexate, and trimethobenzamide. Although the best-known mode of action for β-lactam antibiotics is through inhibition of cell wall synthesis, their activity has also been demonstrated to interfere with bacterial quorum sensing [13]. In addition to their activity against prokaryotes, they have been shown to regulate innate immune responses through their effect on myeloid cells [14].
  • Bioisosters of Amides—Ureas
A short-fall of amide-based structures is that they have limited stability in vivo. Therefore, use of these compounds requires replacing the initial amide-based lead compound with a peptide mimic, i.e., a bioisoster. There are different heterocyclic molecules that are broadly used in drug design as amide bioisosters. Their properties are elegantly described in recent perspectives and reviews [15,16,17]. The focus herein is on the urea-based scaffold commonly used in drug development for the treatment of bacterial and viral infections.
The earliest example of a urea compound developed as an anti-infective having anti-parasitic (anti-trypanosome) activity was developed by the Nobel laureate Paul Ehrlich (Figure 4). Recently, some compounds that were developed for mammalian (human) diseases (e.g., cancer) have been re-purposed as anti-microbials, including anti-virals, via exploitation of the mammalian metabolic pathways that are hijacked by the viruses.
The ability of urea to form stable hydrogen bonds with a drug target is the core quality that allows for specificity in drug action. Modification of the physicochemical properties of the molecule by increasing its stability also leads to the development of new drugs [15,16]. It is the use of various types of substituents on the urea nitrogen atoms that significantly alters the conformational preferences of the urea derivatives as they appear to have a wide variety of drug targets [16]. Ureas typically are more tempered electrophiles (as compared to β-lactams) covalently inhibiting individual serine hydrolases with excellent potency and selectivity [18,19,20,21]. Ureas’ utility as antimicrobials are being further expanded through the testing of currently FDA-approved urea-based drugs which have potential for drug repurposing. These drugs are anticancer agents (e.g., the kinase inhibitors Sorafenib and Regorafenib, Figure 5), stearoyl-CoA desaturase 1, as well as inhibitors of the insulin-like growth factor I receptor.

2. Microbe-Specific Targeting by β-Lactams and Ureas

2.1. Gram-Positive Staphylococcus aureus and Enterococcus faecalis

Despite the increasing resistance to antibiotics, β-lactams, particularly the anti-staphylococcal penicillins (ASPs), e.g., nafcillin, oxacillin, and flucloxacillin and the first-generation cephalosporins, e.g., cefazolin, remain the primary treatments for methicillin-sensitive Staphylococcus aureus (MSSA) infections, including bacteremia and infective endocarditis, although with restrictions [22,23]. An overview of the structure–function relationship for β-lactams and ureas with activity against Staphylococcus aureus and Enterococcus faecalis is shown in Table 1. Similarly, Enterococcus faecalis clinical isolates are resistant against most of the clinically relevant β-lactams with only the third- and fourth-generation cephalosporins and the carbapenems remaining that are efficacious. Monocyclic β-lactams substituted on the lactam nitrogen with the sulphonyl benzene pharmacophore were prepared recently in search of novel antimicrobial and antiviral compounds [24]. Compound 1 (Table 1) demonstrated one of the best antimicrobial activities of all compounds tested against both Gram-positive and Gram-negative organisms, with an MIC of 1 μg/mL for S. aureus. The N-phenylsulfonyl ureas (e.g., 1, Table 1) have moderate antiviral activity, but are not promising enough for further optimization of the scaffold as antivirals [24].
Beyond involvement with penicillin binding proteins (PBP) as the mode of action, kinase inhibitors, such as Sorafenib and Regorafenib (Figure 5, Table 6) FDA-approved anticancer agents), have demonstrated antibacterial and antiviral activity. The Sorafenib analog PK150 (Table 1) has demonstrated antimicrobial activity against MSSA strains and a 10-fold enhanced anti-MRSA activity. In addition, PK150 is as effective in the elimination of pre-existing biofilms as urea 2 (Table 1) with better activity than either Sorafenib or Regorafenib (3 µM for both Sorafenib and Regorafenib) [25]. Additionally, PK150 is an inhibitor of demethylmenaquinone methyltranferase (MenG) biosynthesis, which is involved in S. aureus menaquinone metabolism. Furthermore, it is not affected by environmentally acquired resistance as it causes over-activation of SpsB, an S. aureus signal peptidase I enzyme [25]. PK150 also shows oral bioavailability and antibacterial activity against several pathogenic strains at submicromolar concentrations [25].
Triclocarban belongs to the same chemotype as Sorafenib and Regorafenib, but was FDA approved specifically as an antimicrobial. Triclocarban’s mechanism of action, which is similar to Sorafenib, has MenG as its molecular target [26]. However, due to its numerous side effects, and its lack of timely degradation in the environment, it was banned by the FDA in 2016 for use in hygiene consumer products [27,28,29]. The development of triclocarban analogs with reduced toxicity led to the development of a series of diarylureas, some of which can dissipate pre-existing S. aureus biofilms. The most promising diarylurea in this library is the thiofluoro-substituted urea 2 (Table 1) that has increased selectivity against MRSA (MIC, 0.05 μg/mL) [30].
Other substitutions include PQ 401 (Table 1), an inhibitor of insulin-like growth factor I receptor (IGF-1R) signaling, which is important in both breast cancer and osteosarcoma [31]. PQ 401, which has demonstrated bactericidal activity against S. aureus strains, including MRSA, functions by disruption of the lipid bilayer [32]. Meanwhile, incorporating an aminoguanidine group into the diarylurea scaffold, such as in compounds 3 and 4 (Table 1), results in compounds that interfere with cell wall biosynthesis and produce re-sensitization of vancomycin-resistant S. aureus (VRSA) to vancomycin [33]. In high concentrations, these guanidinolated ureas also have S. aureus anti-biofilm activity [34]. Alternatively, substituting one of the aryl groups in the diarylureas with a pyrazoyl group led to the preparation of pyrazoyl-ureas 5 (Table 1) [35]. These compounds, when evaluated for antimicrobial activity against S. aureus, E. coli, and B. subtilis, demonstrated a moderate bacteriostatic/bacteriocidal activity of 250 μg/mL for all organisms tested. Furthermore, pyrazoyl-substituted ureas of type 6 (Table 1) demonstrated good antimicrobial activity against both Gram-positive and Gram-negative bacteria with S. aureus MICs in the 0.8 mg/mL range for both the trifluoromethyl- and chloro-substituted aromatic ring [36]. The pyrazoyl-substituted diarylureas have additional activities, such as antifungal and anti-malarial activity [37]. Diarylureas 7 and 8 also demonstrate activity against Enterococcus faecalis (MIC = 31.3 µg/mL, Table 1) and Klebsiella pneumoniae (MIC = 31.3 µg/mL, Table 2). Interestingly, both 7 and 8 are the only ones from the ureas described that contain a hydroxy group and a Michael acceptor. Whether this chemical difference affects their activity against these organisms remains to be answered [38]. In all cases, a disubstituted urea is necessary for activity and active compounds require, in addition to the pyrazole scaffold, an N-aryl substituent preferably 3,4 or 3,5-disubstituted with halogen atoms and/or CF3 groups. Unfortunately, these ureas demonstrate low bioavailability in vivo, a result that could be attributed to low solubility [36].
Table 1. Representative examples of Gram-positive bacteria with recognized/reported β-lactams and ureas as antimicrobials: Staphylococcus aureus and Enterococcus faecalis. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
Table 1. Representative examples of Gram-positive bacteria with recognized/reported β-lactams and ureas as antimicrobials: Staphylococcus aureus and Enterococcus faecalis. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
Microorg.Structure—β-Lactam/ReferenceStructure—Urea/Reference
S. aureusApplmicrobiol 03 00043 i001
Cephazolin (first-generation cephalosporin) [22]
Applmicrobiol 03 00043 i003
Ceftaroline (first-generation cephalosporin) [23]
Applmicrobiol 03 00043 i005
Cefiderocol (approved for medical use in the United States in November 2019, and in the European Union in April 2020) [10,11,12]
Applmicrobiol 03 00043 i007
1 [24]
MIC = 1 mg/mL


Applmicrobiol 03 00043 i002
Triclocarban (TCC) [26,27,28,29]







Applmicrobiol 03 00043 i004
2 [25,30]
S. aureus (ATCC, MIC50 = 0.5 µg/mL) as well as MRSA (MIC50 = 0.05 µg/mL)
Applmicrobiol 03 00043 i006
PK150 [25]
MIC 0.3 µM for MSSA, MIC 0.03 µM for MRSA.

Applmicrobiol 03 00043 i008
PQ 401 [31,32]
MRSA/
Biofilm
Applmicrobiol 03 00043 i009
3 [33,34]
Applmicrobiol 03 00043 i010
4 [33,34] MIC values for 3 and 4 between 2.0 and 4.0 µg/mL as compared with vancomycin (from 0.5 to 1 µg/mL). Molecular target: cell wall synthesis
S. aureus Applmicrobiol 03 00043 i011
5 [35] MIC = 250 mg/mL
Compound 5 was evaluated as an antibacterial and antifungal agent against Staphylococcus aureus (ATCC 25923), Escherichia coli (ATCC 25922), Bacillus subtilis (ATCC 8236F800), and Candida albicans (ATCC 885-653), showing a moderate antimicrobial activity.
Applmicrobiol 03 00043 i012
6 [36]
IC50 ranging from 0.054 to 4.24 μM
E. faecalisApplmicrobiol 03 00043 i013
Cefepime—fourth-generation cephalosporin [39]
Applmicrobiol 03 00043 i016
Meropenem [40]
Applmicrobiol 03 00043 i014
7 [38]
Applmicrobiol 03 00043 i015
8 [38]

2.2. Gram-Negative Multi-Drug Resistant Bacteria: Proteus mirabilis, Klebsiella pneumoniae, and Acinetobacter baumannii

Resistance to β-lactam drugs is even more pronounced with respect to Gram-negative pathogenic bacteria, such as Proteus mirabilis, Klebsiella pneumoniae, and Acinetobacter baumannii. While some strains of P. mirabilis are sensitive to ampicillin, in general, broad-spectrum penicillins (e.g., ticarcillin and piperacillin), first-, second-, and third-generation cephalosporins (e.g., imipenem and aztreonam), carbapenems, and piperacillin/tazobactam are required for use in the treatment of extended-spectrum β-lactamase-producing P. mirabilis bacteremia [39]. With respect to the treatment of K. pneumoniae infections, the use of β-lactams is limited to third- and fourth-generation cephalosporins and carbapenems. The only clinically used monobactam, aztreonam, has good activity against many Gram-negative bacteria; however, its activity against some of the most problematic multidrug-resistant (MDR) strains of P. aeruginosa and A. baumannii is limited [40]. Interestingly, coupling of the aztreonam side chain free acid with a bis-catechol siderophore mimetic (lactam 9, Table 2) significantly improves the activity against the MDR strains of Gram-negative bacteria that are of most significant concern [41].
With regard to the activity of diarylureas, there are several representatives of this class of compounds with activity against Gram-negative bacteria. The chloro, fluoro-, and trifluorpmethyl-substituted diarylureas 10 and 11 (Table 2) demonstrated activity against P. mirabilis that is comparable to that of ciprofloxacin [42]. In compound 12 (Table 2), one of the aryl groups of the prototypic diarylurea was substituted by an adamantane group, a feature that increases the lipophilicity of the compound [43]. Compound 12 (Table 2) demonstrated very good selectivity against A. baumannii (94% inhibition at 32 μg/mL) as compared with the rest of the bacteria tested, such as S. aureus, K. pneumonia, E. coli, P. aeruginosa, and C. albicans. Using several other analogs containing the adamantane group, it was determined that these compounds bind to the active site of enoyl-(acyl-carrier-protein) reductase (ENR). Of the library of adamantine-based analogs screened, compound 12 (Table 2) remains the best compound against A. baumannii, with analogs of diarylureas 7 and 8 (Table 1 and Table 2) exhibiting good activity against E. faecalis (MIC = 31.3 µg/mL, Table 1) and K. pneumoniae (MIC = 31.3 µg/mL, Table 2).
Table 2. Representative examples of Gram-negative bacteria with recognized/reported β-lactams and ureas as antimicrobials: Proteus mirabilis, Klebsiella pneumoniae, Acinetobacter baumannii. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
Table 2. Representative examples of Gram-negative bacteria with recognized/reported β-lactams and ureas as antimicrobials: Proteus mirabilis, Klebsiella pneumoniae, Acinetobacter baumannii. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
Microorg.Structure—β-Lactam/ReferenceStructure—Urea/Reference
P. mirabilisApplmicrobiol 03 00043 i017
Cefepime [39]
fourth-generation cephalosporin
Applmicrobiol 03 00043 i018
Meropenem [39]


Applmicrobiol 03 00043 i019
10 [42]




Applmicrobiol 03 00043 i020
11 [42]
K. pneumoniaeApplmicrobiol 03 00043 i021
Cefepime [40]
fourth-generation cephalosporin
Applmicrobiol 03 00043 i022
Meropenem [40]
Applmicrobiol 03 00043 i023
Aztreonam [41]
Applmicrobiol 03 00043 i024
7 [38]
Applmicrobiol 03 00043 i025
8 [38]
A. baumanniiApplmicrobiol 03 00043 i026
Aztreonam [41]
Applmicrobiol 03 00043 i027
9 [41]
Aztreonam with siderophore mimetic
Applmicrobiol 03 00043 i028
12 [43]

2.3. Mycobacterium tuberculosis (Mtb)

Until over a decade ago, Mycobacterium tuberculosis (Mtb) was considered refractory to β-lactam antibiotics. However, more recent studies show that carbapenems have anti-Mtb activity and are now used in the treatment of disease caused by MDR and extremely drug-resistant (XDR) Mtb strains [44,45]. Following this recognition of efficacy, further drug screening (8900 β-lactams) by a consortium of pharmaceutical companies, the NIH, and academic institutions identified about 1600 compounds that exhibited activity against Mtb, including many minus the necessity for the presence of a β-lactamase inhibitor [46]. Representative structures of cephalosporins from this work (lactam 13, which is active against replicating (R), and 14, which is active against both R and non-replicating (NR) Mtb) are shown in Table 3 [46]. Identification of β-lactams that are cidal for both R and NR Mtb was an additional reason to propel this massive screen undertaking [46,47]. Among the cephalosporins developed, six have a pyrithione-leaving group and are dual-acting compounds with activity against both NR and R Mtb. The release of the pyrithione upon cleavage of the β-lactam ring by β-lactamases, and/or engagement of the β-lactam ring with its target, allows for these two parts of the molecule to contribute to antitubercular activity.
A new family of anti-Mtb agents consists of C4-phenylthio β-lactams with the 15 scaffold (Table 3) [48]. This family shares similarities to the monocyclic structures 20 (Table 4) with the majority of structures having a benzylthio substituent at C4 and a urea moiety at N1 [49]. Lactams such as 15 having a phenylthio group at C4, and a urea moiety at N1, have shown activity against R and NR Mtb, as well as in macrophages [48]. Lactams with the 20 scaffold (Table 4) have antiviral activity as inhibitors of human cytomegalovirus (HCMV) protease with acceptable anti-HCMV activity [49]. Improvements upon the physicochemical properties of the monocyclic β-lactams 15 (Table 3) in our laboratories are ongoing.
Table 3. Representative examples with recognized/reported β-lactams and ureas as antimicrobials: Mycobacterium tuberculosis (Mtb). The structures of the lactams and ureas associated with Mtb and the corresponding references (bolded, in parentheses) are shown.
Table 3. Representative examples with recognized/reported β-lactams and ureas as antimicrobials: Mycobacterium tuberculosis (Mtb). The structures of the lactams and ureas associated with Mtb and the corresponding references (bolded, in parentheses) are shown.
Microorg.Structure—β-Lactam/ReferenceStructure—Urea/Reference
Mtb

Applmicrobiol 03 00043 i029
Meropenem [44,45]







Applmicrobiol 03 00043 i030
13 [46,47]
Applmicrobiol 03 00043 i031
14 [46,47]
(TBBL-0000316)
Applmicrobiol 03 00043 i032
15 [48]
R = o-F, MIC 1.5 µg/mL (M. tuberculosis pathogenic strain H37Rv)
Applmicrobiol 03 00043 i033
16 [50]
MIC 6.25 µg/mL (M. tuberculosis pathogenic strain H37Rv)
Applmicrobiol 03 00043 i034
17 [50]
MIC 3.125 µg/mL (M. tuberculosis pathogenic strain H37Rv)
Applmicrobiol 03 00043 i035
18 [31]
MIC 6.0 µg/mL (M. tuberculosis pathogenic strain H37Rv)
Applmicrobiol 03 00043 i036
19 [31]
MIC 5.2 µg/mL (M. tuberculosis pathogenic strain H37Rv)
Nitro-substituted diarylurea 16 and methoxy-substituted diarylurea 17 (Table 3) inhibited Mtb H37Rv with an MIC of 6.25 μg/mL and 3.12 μg/mL, respectively [50]. These MIC values are higher than those of isoniazid (0.05–0.2 μg/mL), the most often used drug as a control for evaluation of anti-Mtb activity, but are encouraging for further compound development. Nitro-substituted diarylurea 18 and methyl-substituted diaryl urea 19 (Table 3) have similar MIC values of 6.0 and 5.2 µg/mL, respectively, to compounds 16 and 17 (Table 3) against Mtb H37Rv. Interestingly, these diaryl-substituted ureas proved to be selective inhibitors of mycolic acid biosynthesis [51].
In summary, the predominant development of antimicrobials continues to be focused on the bacterial molecular targets exploring dual action, e.g., anti-Mtb inhibitors of both the PBPs and β-lactamases. Cefiderocol is the only antibiotic, out of the 12 approved since 2017, able to target all the pathogens considered critical by the WHO. The rest of the antibiotics are under development in Phase 1 to 3 clinical trials. As of this year (2023), from a total of 27 compounds, only four have new mechanisms of action, only six are considered innovative enough to meet the WHO criteria for the treatment of bacteria-resistant organisms, and only two of these six target the most difficult organisms to treat, such as A. baumannii and P. aeruginosa. Most of the drugs under development are the next generation of existing classes.

3. β-Lactams and Ureas as Antiviral Agents

3.1. Herpesviridae (DNA viruses): Human Cytomegalovirus (HCMV)

Early examples of monocyclic β-lactams as antivirals are monocyclic structures of type 20 (Table 4) having a carbamoyl moiety at N1. This scaffold contains at C-4 a side chain, thiomethyl (21 and 22, Table 4), or a one-carbon chain (23, Table 4), with either a phenyl or a heterocyclic ring [49,52,53]. These lactams were all developed as inhibitors of human cytomegalovirus (HCMV) serine protease. In the 21/22 series (heterocyclic thiomethyl side chain at C-4), the N-methyl thiotetrazole-containing compounds 22 (Table 4) demonstrated the best activity in the enzymatic assay. Members of the 23 series (C4 carbon-chain-substituted lactams) use both tri- and tetrasubstituted carbamoyl functionalities, which result in effective inhibitors of HCMV protease. However, for all compounds tested only modest antiviral activity was found in plaque reduction assays [53]. Further development led to a library of monocyclic β-lactams of type 24 (Table 4) that function as covalent and noncovalent HCMV protease inhibitors, e.g., 1-acylazetidines of scaffold type 25 (Table 4) [54,55]. Two of the 1-acylazetidines (of the 25 series) have activity as low micromolar inhibitors of HCMV replication (EC50, 0.6–7.0 mM) [55].
Shortly after the synthesis of the first mono-β-lactams (20, Table 4) to be used as inhibitors of the HCMV viral serine protease, an investigation of disubstituted ureas as inhibitors of that HMCV enzyme followed [49,56]. This novel primaquine-containing series consists of ureas having different alkyl, or a combination of alkyl and aryl, substituents, e.g., compounds 26 and 27 (Table 4). Those having an aryl group as one of the substituents on the urea moiety demonstrate the best inhibitory activity against HMCV with compounds 26 and 27 being the most active of the series (EC50 = 1.2–13.4 μM). However, their activity is lower than that of ganciclovir (a purine-based FDA-approved antiviral against HCMV, which preferentially inhibits viral DNA polymerases more than cellular DNA polymerases) [56]. Another representative of the disubstituted ureas with one aryl and a piperidine as the second urea substituent is compound A939572 (Table 4). It was developed originally as an inhibitor of stearoyl-CoA desaturase 1 (SCD1, IC50 < 4 nM) [57]. SCD1 is an enzyme involved in lipid metabolism. Its high activity in humans has been correlated with elevated plasma triglyceride levels and it has been observed to be expressed in abnormally high levels in cell renal cell carcinoma (ccRCC) [58,59].
Table 4. Representative examples of Herpesviridae (DNA viruses) with recognized/reported β-lactams and ureas as antimicrobials: human cytomegalovirus (HCMV). The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
Table 4. Representative examples of Herpesviridae (DNA viruses) with recognized/reported β-lactams and ureas as antimicrobials: human cytomegalovirus (HCMV). The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
VirusStructure—β-Lactam/ReferenceStructure—Urea/Reference
HCMVApplmicrobiol 03 00043 i037
20 [49]
Applmicrobiol 03 00043 i038
21 [52]
Applmicrobiol 03 00043 i039
22 [52]
The most active compound of the series.
Applmicrobiol 03 00043 i040
23 [53]
Applmicrobiol 03 00043 i041    Applmicrobiol 03 00043 i042
24 [54,55]         25 [55]
EC50 0.6−7.4 μM
Applmicrobiol 03 00043 i043
26 [56]
(R1 = Ar, R2 = H); 27 (R1 = Ar, R2 = H)
(EC50 = 1.2–13.4 μM)
















Applmicrobiol 03 00043 i044
A939572 [57,58,59,60,61,62]
IC50 < 4 nM (mSCD1)

3.2. Flaviviridae, Picornaviridae, Retroviridae, and Coronaviridae (Single-Stranded Positive Sense RNA Viruses): DENV, ZKV, WNV, HCV, HRV, HIV, and SARS-CoV-2

Peptide-β-lactams 28 (Table 5) are reported as inhibitors of NS2BNS3 protease, a serine protease important in Dengue (DENV) and West Nile (WNV) virus replication [63]. Tripeptide-β-lactams are considerably more active as compared with the di-peptide-β-lactams, wherein the (3S)-β-lactam moiety displayed the highest activity. The IC50 and EC50 values of the S-tripeptide-β-lactams 28 (Table 5) are in the lower micromolar range in both biochemical and cellular assays (IC50, 1.9–2.8 μM). These compounds demonstrated two distinct binding modes to the NS2BNS3 protease: a covalent but reversible acylation of the active site serine by the β-lactams and another that leads to cleavage of the peptide at the C3 amide. Determining these two distinct modes of action on the two different amide bonds is the first experimental evidence that the benzyloxyphenylglycine in flaviviral protease inhibitors is positioned at the prime site of the enzyme [63]. Spirolactams of type BSS-730A (Table 5) have been recently evaluated for anti-HIV activity [64]. Compound BSS-730A inhibited both HIV-1 and HIV-2 replication with an IC50 of 13 ± 9.59 nM, the best antiviral activity of a β-lactam. This potency may be attributed to its ability to affect all stages of the HIV replicative cycle. BSS-730A also affects multidrug-resistant HIV isolates, with a median 2.4-fold higher IC50 relative to control isolates [64].
Other β-lactams containing purines, which are well established as potent antiviral agents, have been synthesized [65,66,67]. The idea behind molecular hybridization is to identify novel combinations of biologically active compounds able to circumvent drug resistance. This is envisioned to occur by linking bioactive units to moieties that are recognized and actively transported into mammalian cells. Hybrid systems also allow for the design of new organic structures through selective modification of one or both of the individual parts. The hybrid monocyclic β-lactam/purine hybrid structures 29 and 30 (Table 6) included the lactam ring that was subsequently opened (compounds 31 and 32, Table 6) [67]. This is similar to the approach reported for HCMV inhibition (Table 4) [54,55]. These two types of purine-containing compounds (20 in total) were evaluated against nine viruses. Eight of the β-lactam/purine hybrids and two of the compounds with an opened lactam ring showed very good antiviral activity in the micromolar range against five viruses (RSV, ChikV, CMV, HBV, and CoxV), with the best activity against RSV (the EC50 for the β-lactams was 2.24–8.05 μM and the EC50 for the opened lactam structures was 1.92–23.55 μM) [67].
C-4-diethoxyphosphoryl β-lactams of type 33 (Table 6) have been synthesized [68] and evaluated for their antiviral (against both DNA and RNA viruses) as well as their antimicrobial and cytostatic activity against several cancer cell lines. The isomers of the most promising structures, 33c and 33t (Table 6), differ in activity, i.e., the cis-isomer 33c has activity against influenza virus (H1N1) at an EC50 of 8.3–12 μM, depending on the type of assay used, while the trans-isomer 33t showed activity against a representative of the coronaviruses (229E) at a higher concentration (EC50, 45 μM). The trans-33t lactam also demonstrated activity against the drug-resistant S. aureus (HEMSA 5) strain as an adjuvant of oxacillin with a significant ability to enhance the efficacy of this antibiotic toward that strain, presumably by binding to the S. aureus β-lactamase, thus sparing the oxacillin from hydrolysis [68].
Table 5. Representative examples of Flaviviridae, Retroviridae, Picornavididae, and Coronaviridae (single-stranded positive-sense RNA viruses) with recognized/reported β-lactams and ureas as antimicrobials. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
Table 5. Representative examples of Flaviviridae, Retroviridae, Picornavididae, and Coronaviridae (single-stranded positive-sense RNA viruses) with recognized/reported β-lactams and ureas as antimicrobials. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
VirusStructure—β-Lactam/ReferenceStructure—Urea/Reference
Hepatitis C virus
Flaviviridae
Applmicrobiol 03 00043 i045
28 [63]
Applmicrobiol 03 00043 i046
38 [57,58,59,60,61,62]
Applmicrobiol 03 00043 i047
A939572 [59,60,61]
Applmicrobiol 03 00043 i048
Boceprevir (SCH 503034) [69,70,71,72]
(Ki = 14 nM, EC90 = 0.35 mM)
Dengue virus
Flaviviridae
Applmicrobiol 03 00043 i049
A939572 [59,60,61]
Applmicrobiol 03 00043 i050
39 [73]
IC50 = 91 mM
Applmicrobiol 03 00043 i051
40 [73]
IC50 = 110 mM
Applmicrobiol 03 00043 i052
41 [74]
IC50 = 26 mM
Applmicrobiol 03 00043 i053
42 [74]
IC50 = 24 mM
West Nile virus
Flaviviridae
Applmicrobiol 03 00043 i054
28 [63]
Applmicrobiol 03 00043 i055
39 [73]
IC50 = 51 mM
Applmicrobiol 03 00043 i056
40 [73]
IC50 = 71 mM
Zika virus
Flaviviridae
Applmicrobiol 03 00043 i057
41 [74]
IC50 = 28 mM
Applmicrobiol 03 00043 i058
42 IC50 = 19 mM [74]
Applmicrobiol 03 00043 i059
43 IC50 = 35 mM [75]
Applmicrobiol 03 00043 i060
44 [75]
ZIKV MTase IC50 = 23–48 mM
ZIKV EC50 = 1.67–25 mM
Applmicrobiol 03 00043 i061
ASN 25 [76,77]
Applmicrobiol 03 00043 i062
Suramin [78]
Rhinovirus
Picornaviridae
Applmicrobiol 03 00043 i063
45 [79,80,81,82]
Applmicrobiol 03 00043 i064
46 [79,80,81,82]
Applmicrobiol 03 00043 i065
47 [83]
HIV
Retroviridae
Applmicrobiol 03 00043 i066
BSS-730A [64]
Applmicrobiol 03 00043 i067
Ritonavir [84]
Applmicrobiol 03 00043 i068
38 [85,86,87,88]
Coronaviruses
SARS-CoV-2
Coronaviridae
Applmicrobiol 03 00043 i069
34 and 35 [89,90]
Applmicrobiol 03 00043 i070
36 and 37 [89,90]
Applmicrobiol 03 00043 i071
38 [85,86,87,88]
As is well documented in the literature, the SARS-CoV-2 main protease (Mpro), a cysteine protease, is a molecular target for developing SARS-CoV-2 treatments. Recently, the sulfoxide- and sulfone-penicillins 34 and 35 (Table 5) were synthesized and their molecular target -(Mpro) enzyme confirmed by mass-spectrometry (MS) [89]. It is interesting to note the very subtle structural difference between the compound 36 (Table 5), which demonstrates Mpro inhibition, and the corresponding penicillin V sulfone, 37 (Table 5), which is inactive against this protease. The ability of the C6-phenoxyacetyl ether oxygen to function as a hydrogen bond acceptor/Lewis acid/conformation restrictor may be important for efficient Mpro inhibition, as supported by preliminary molecular docking studies [90]. Compound 36 inhibited Mpro (IC50, ~6.5 μM), with further optimization leading to derivatives of compound 36 having an IC50 of ~0.6mM, superior to its corresponding sulfoxide (IC50, ~22.9 mM). Neither Penicillin V nor its benzyl ester inhibited Mpro efficiently [90].
There are also urea-based drugs that are specifically FDA-approved as antiviral agents, e.g., ritonavir (Table 5) and boceprevir (Table 5). Ritonavir, a disubstituted urea (Table 5) initially approved as an HIV protease inhibitor, has also been shown to inhibit cytochrome P450-3A4 [84]. Clinically, it is usually used in combination with other HIV or Hepatitis C (Table 5) drugs, e.g., Lopinavir, to treat the corresponding infections. The urea-based compounds 38 (Table 5), similarly substituted at the urea moiety, are inhibitors of cyclophilins, which are peptidyl–prolyl cis/trans isomerases (PPIase) that catalyze the interconversion of the peptide bond [85]. It has been shown that human cyclophilins, mainly CypA, play an important role in the life cycle of viruses, including HIV, HCV, DENV, Japanese encephalitis virus, yellow fever virus, coronaviruses, HBV, cytomegalovirus, influenza A virus, and enteroviruses [85,86]. Thus, human cyclophilin inhibitors are an attractive target in antiviral therapy though blockage of an enzyme essential for viral replication. The currently known active site inhibitors of cyclophilins are cyclosporine A (CsA) and non-immunosuppressive macrocyclic analogs of CsA and sanglifehrin A (SfA). These two inhibitors have shown effectiveness against HIV, HCV, and HBV replication [86]. Alispovir, a CsA analog (synthesized from cyclosporin), has demonstrated potent anti-HCV in vivo activity but its clinical development did not progress beyond Phase III clinical trials due to severe adverse events unrelated to cyclophilin inhibition [87,88]. A fragment-based drug discovery (FBDD) approach was used to identify compounds 38 (Table 5) as cyclophilin inhibitors [85] which show antiviral activity for HIV (EC50s ranging from 3.6 to 15 μM, Table 5), HCV (EC50s ranging from 0.4 to 8.4 μM, Table 5), and coronaviruses (EC50s ranging from 7.2 to 71.5 μM, Table 5). These inhibitors appear to be non-cytotoxic at their effective concentrations [85].
An HCV protease inhibitor, Boceprevir (Table 5), played a valuable role in the treatment of patients with an HCV genotype 1 infection. However, production was discontinued by the manufacturer in 2015 due to the superiority of newer agents (e.g., ledipasvir/sofosbuvir) [69]. The relevance of Boceprevir was considered historical until, during the search for effective treatments against SARS-CoV-2, it was demonstrated that Boceprevir inhibits SARS-CoV-2 (Table 5) by inhibiting its main protease [70,71,72].
Viruses are known to hijack critical metabolic pathways in host cells to convert their activity for their replication. Flaviviruses, such as DENV and HCV, hijack lipid metabolic pathways for their energy and substrate requirements. It was shown that SCD1 is critical to virus replication and infectious particle formation [60]. Compound A939572 (Table 5) has been demonstrated to have antiviral activity against HCV and DENV infection in a cell culture [60]. Inhibition of SCD1 continues to be targeted for new drug development [61,62]. What is notable, with respect to the use of the urea-based inhibitor A939572, is that it targets the activity of the host enzyme, rather than the viral enzymes, which is a potential way to slow down the development of viral drug resistance, a too common occurrence with the conventional virus-targeted antivirals.
Incorporating a sulfonamide as well as a carboxylic acid group in one of the aryl substituents in the scaffold of the disubstituted ureas such as PK150 (Table 1) and compounds 10 and 11 (Table 1) using the FBDD resulted in compounds 39 and 40 (Table 5) having activity against DENV and WNV NS5 methyltransferase [73]. The optimization of the fragment linking hit led to another series of diarylureas with compounds 41 and 42 (Table 5) with improved IC50s (DENV IC50 = 26 μM and 24 μM, respectively) as compared with compounds 39 (DENV IC50 = 91 µM; WNV IC50 = 51 µM) and 40 (DENV IC50 = 110 µM; IC50 = 71 µM). Interestingly, compounds 41 and 42 also demonstrate activity against ZIKV (IC50 = 28 μM and 19 μM, respectively (Table 5)) [74].
Compound 43 (Table 5) is an example of the discovery of a hit compound using a structure-based virtual screening approach using the ZIKV NS5-MTase (43 IC50 35 μM) [75]. Optimization of compound 43 led to compounds 44 (Table 5) with improved activity against ZIKV (44 ZIKV Mtase IC50 23–48 μM; ZIKV EC50 1.67–25 μM) [75]. Diarylureas of type ASN-25 (Table 5) exhibit nanomolar to picomolar activity against ZIKV in vitro (IC50 = 160.3 nM, 189.2 pM, 317.7 pM, and 26.9 nM, respectively) [76]. In a continuation of their work, the authors prepared compound ASN-25, which exhibited an excellent IC50 = 85.1 pM [77]. It is interesting to note that it was shown earlier by a high-throughput screening method that a host kinase is a key regulatory factor in Zika infection [91].

3.3. Pneumoviridae and Orthomixoviridae (Single-Stranded Negative-Sense RNA Viruses): RSV and IV

Other representatives of diarylureas containing pyrazoyl groups are compounds of type 45 and 46 (Table 5) as well as compound 47 (Table 5 and Table 6). Compound 46 is structurally related to BIRB 796 (Doramapimod), a pan p38 MAP kinase inhibitor with a Kd of 0.1 nM in THP-1 cells [79]. Initially developed as p38 MAP kinase inhibitors, three derivatives of the 46 scaffold are the most active of the series with an IC50 < 5 μM against the p38α enzyme [80]. They were also described as antivirals against several viruses, including influenza [81,82]. Compound 47 (Table 5 and Table 6) has been described as representative of pyrazoyl ureas for the treatment of inflammatory diseases of the respiratory system (asthma, COPD), and also for the treatment, or prevention, of inflammation mediated by viral infectious diseases (such as influenza virus, rhinovirus, or RSV) [83].
RNAi-based small-molecule repositioning revealed clinically approved urea-based kinase inhibitors (UBKIs) to be broadly active antivirals. Regorafenib and Sorafenib (Figure 1, Table 6) exhibited a superior therapeutic window of high IV antiviral activity and low cytotoxicity. Both UBKIs appeared to block a cell signaling pathway involved in IV replication after internalization, yet prior to vRNP uncoating. Interestingly, both compounds were also active against unrelated viruses including cowpox virus (CPXV), hantavirus (HTV), herpes simplex virus 1 (HSV-1), and vesicular stomatitis virus (VSV) [78]. Pyrazoyl-ureas, such as compound 47 (Table 5 and Table 6), have demonstrated anti-IV activity as well [83].
Table 6. Representative examples of Pneumoviridae (RSV) and Orthomixoviridae (IVA and IVB) (single-stranded negative-sense RNA viruses) recognized/reported β-lactams and ureas as antimicrobials. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
Table 6. Representative examples of Pneumoviridae (RSV) and Orthomixoviridae (IVA and IVB) (single-stranded negative-sense RNA viruses) recognized/reported β-lactams and ureas as antimicrobials. The structures of the lactams and ureas associated with a given microorganism with their corresponding references (bolded, in parentheses) are shown.
VirusStructure—β-Lactam/ReferenceStructure—Urea/Reference
RSV
Pneumoviridae
Applmicrobiol 03 00043 i072
29 [67]
Applmicrobiol 03 00043 i073
30 [67]
Applmicrobiol 03 00043 i074
31 [67]
Applmicrobiol 03 00043 i075
32 [67]
Applmicrobiol 03 00043 i076
47 [83]
Influenza virus
Orthomixoviridae
Applmicrobiol 03 00043 i077
33c [68]
Applmicrobiol 03 00043 i078
33t [68]
Applmicrobiol 03 00043 i079
Regorafenib [78]
Applmicrobiol 03 00043 i080
Sorafenib [78]
Applmicrobiol 03 00043 i081
47 [83]

4. Conclusions

Recent advances in drug design with potential as pan-antimicrobials are based on the two “classical” electrophilic chemotypes, i.e., β-lactams and urea-based derivatives and their bioisosteres. The illustrative β-lactam and urea covalent protein-reactive drugs discussed represent important advances since they target two commonly present enzyme active-site nucleophiles (serine and cysteine); thus, they are not restricted to a specific taxonomic kingdom affecting fungi, bacteria, and viruses, i.e., pan-activity. The included utility of this enzyme-targeted approach is illustrated by the ability of β-lactams to inhibit representatives of the ESKAPE pathogens. Currently, the focus is on the development of new moieties added to the β-lactam scaffold which exhibit siderophore activity. These new lactams engage with the microbes’ active transport systems, rather than cell entry via diffusion. In addition, their siderophore’s metal chelating properties target an alternative system essential for microbe survival, enhancing the ability of these compounds, especially when used in combination with chemotherapy, to affect drug-resistant organisms since the siderophore moieties allow for evasion of the bacterial efflux pumps as well as β-lactamases. To date, this type of β-lactam structure has found utility in Mtb treatment, including activity against the latent Mtb. This exploitation of the lactam scaffold and ureas takes advantage of the carbonyl group in the lactams and ureas as an electrophile that inhibits serine and cysteine hydrolases via catalytic modification of these active-site residues, resulting in a covalent inhibition of a variety of target proteins. Unfortunately, the selectivity of the β-lactam towards the serine enzymes appears to be rather poor. In contrast, ureas, as more tempered electrophiles, covalently inhibit individual serine hydrolases with excellent potency and selectivity. The advantage of the ureas is that they represent a simple chemical core permitting modifications directed toward the electrophilicity of the carbonyl group, as well as the number of hydrogens on the urea moiety. Thus, they allow for rapid preparation of suitable chemical probes for activity-based protein profiling (ABPP). The ability to inhibit and/or detect changes in the activity and localization of specific enzymes in tissues or whole organisms that contain many related enzymes with similar substrate specificities remains a primary goal of contemporary ABPP research. For example, triazole ureas appear to be some of the scaffolds capable of generating probes with moderate selectivity for serine enzymes. It might be that the urea scaffold, explored a century ago by Paul Erlich, will allow us to achieve the features necessary for better enzyme selectivity. Currently, more than 40 covalent drugs are on the market with many of them in clinical trials. Several ureas have been utilized as antimicrobials at the turn of this century, but their mechanism of action has not been unveiled until recently. Several very successful repurposing studies of ureas from anti-cancer to antimicrobial (antiviral) have occurred, with follow-up FDA approval for use as antivirals. The lessons that might be learned from these protein-reactive drug classes could assist in the further design of the next generation of antimicrobials, especially in the current age of elevated microbial resistance towards the clinically relevant antimicrobials.

Author Contributions

Conceptualization, writing—original draft preparation, M.I.K.; writing—review and editing, B.J.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Acknowledgments

The authors wish to thank the American University, the Midwestern University Offices of Research and Sponsored Programs, and the Midwestern University College of Graduate Studies for their support.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

HCMVHuman Cytomegalovirus
DENVDengue virus
ZKVZika virus
WNVWest Nile virus
HCVHepatitis C virus
HIVHuman Immunodeficiency virus
HRVHuman Rhinovirus
RSVRespiratory Syncytial virus
IVInfluenza virus
SARS-CoV-2Severe Acute Respiratory Syndrome Coronavirus-2

References

  1. Fisher, J.F.; Mobashery, S. Chapter 3: The β-Lactam (Azetidin-2-one) as a Privileged Ring in Medicinal Chemistry. In Privileged Scaffolds in Medicinal Chemistry: Design, Synthesis, Evaluation; Bräse, S., Ed.; Royal Chemical Society of Chemictry, RSC Drug Discovery, 2015; Available online: https://books.rsc.org/books/edited-volume/529/Privileged-Scaffolds-in-Medicinal-Chemistry-Design (accessed on 21 March 2023).
  2. Konaklieva, M.I.; Plotkin, B.J. The relationship between inhibitors of eukaryotic and prokaryotic serine proteases. Mini Rev. Med. Chem. 2004, 4, 721–739. [Google Scholar] [CrossRef]
  3. Kluge, A.F.; Petter, R.S. Acylating drugs: Redesigning natural covalent inhibitors. Curr. Opin. Chem. Biol. 2010, 14, 421–427. [Google Scholar] [CrossRef] [PubMed]
  4. Burnett, D.A. β-Lactam cholesterol absorption inhibitors. Curr. Med. Chem. 2004, 11, 1873–1887. [Google Scholar] [CrossRef] [PubMed]
  5. Garcia-Calvo, M.; Lisnock, J.-M.; Bull, H.G.; Hawes, B.E.; Burnett, D.A.; Braun, M.P.; Crona, J.H.; Davis, H.R., Jr.; Dean, D.C.; Detmers, P.A. The target of ezetimibe is Niemann-Pick C1-Like 1 (NPC1L1). Proc. Natl. Acad. Sci. USA 2005, 102, 8132–8137. [Google Scholar] [CrossRef] [PubMed]
  6. Revell, K.D.; Heldreth, B.; Long, T.E.; Jang, S.; Turos, E. N-thiolated β-lactams: Studies on the mode of action and identification of a primary cellular target in Staphylococcus aureus. Bioorg. Med. Chem. 2007, 15, 2453–2467. [Google Scholar] [CrossRef]
  7. Konaklieva, M.I. Molecular Targets of β-Lactam-Based Antimicrobials: Beyond the Usual Suspects. Antibiotics 2014, 3, 128–142. [Google Scholar] [CrossRef]
  8. Fisher, J.F.; Mobashery, S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem. Rev. 2021, 121, 3412–3463. [Google Scholar] [CrossRef]
  9. Fisher, J.F.; Mobashery, S. β-Lactams from the Ocean. Mar. Drugs 2023, 21, 86. [Google Scholar] [CrossRef]
  10. WHO: More New Antibiotics Needed. Available online: https://www.idse.net/Bacterial-Infections/Article/03-23/WHO-More-New-Antibiotics-Needed/69730 (accessed on 21 March 2023).
  11. Zhanel, G.G.; Golden, A.R.; Zelenitsky, S.; Wiebe, K.; Lawrence, C.K.; Adam, H.J.; Idowu, T.; Domalaon, R.; Schweizer, F.; Zhanel, M.A.; et al. Cefiderocol: A siderophore cephalosporin with activity against carbapenem-resistant and multidrug-resistant Gram-negative bacilli. Drugs 2019, 79, 271–289. [Google Scholar] [CrossRef]
  12. Sato, T.; Yamawaki, K. Cefiderocol: Discovery, chemistry, and in vivo profiles of a novel siderophore cephalosporin. Clin. Infect. Dis. 2019, 69, S538–S543. [Google Scholar] [CrossRef]
  13. Kumar, L.; Brenner, N.; Brice, J.; Klein-Seetharaman, J.; Sarkar, S.K. Cephalosporins Interfere with Quorum Sensing and Improve the Ability of Caenorhabditis elegans to Survive Pseudomonas aeruginosa Infection. Front. Microbiol. 2021, 12, 598498. [Google Scholar] [CrossRef] [PubMed]
  14. Mondeme, M.; Carnoy, C.; Sirard, J.-C.; Faveeuw, C. Treatment of Bacterial Infections with β-Lactams: Cooperation with Innate Immunity. Infect. Immun. 2023, 2, 1–11. [Google Scholar] [CrossRef] [PubMed]
  15. Kumari, S.; Carmona, A.V.; Towari, A.K.; Trippier, P.C. Amide Bond Bioisosteres: Strategies, Synthesis, and Successes. J. Med. Chem. 2020, 63, 12290–12358. [Google Scholar] [CrossRef] [PubMed]
  16. Ghosh, A.K.; Brindisi, M. Urea Derivatives in Modern Drug Discovery and Medicinal Chemistry. J. Med. Chem. 2020, 63, 2751–2788. [Google Scholar] [CrossRef]
  17. Ronchetti, R.; Moroni, G.; Carotti, A.; Gioiello, A.; Camaioni, E. Recent advances in urea- and thiourea-containing compounds: Focus on innovative approaches in medicinal chemistry and organic synthesis. RSC Med. Chem. 2021, 12, 1046. [Google Scholar] [CrossRef]
  18. Adibekian, A.; Martin, B.R.; Wang, C.; Hsu, K.-L.; A Bachovchin, D.; Niessen, S.; Hoover, H.; Cravatt, B.F. Click-generated triazole ureas as ultrapotent in vivo-active serine hydrolase inhibitors. Nat. Chem. Biol. 2011, 7, 469–478. [Google Scholar] [CrossRef]
  19. Chang, J.W.; Nomura, D.K.; Cravatt, B.F. A potent and selective inhibitor of KIAA1363/AADACL1 that impairs prostate cancer pathogenesis. Chem. Biol. 2011, 18, 476–484. [Google Scholar] [CrossRef]
  20. Hsu, K.L.; Tsuboi, K.; Adibekian, A.; Pugh, H.; Masuda, K.; Cravatt, B.F. DAGLbeta inhibition perturbs a lipid network involved in macrophage infammatory responses. Nat. Chem. Biol. 2012, 8, 999–1007. [Google Scholar] [CrossRef]
  21. Remsberg, J.R.; Suciu, R.M.; Zambetti, N.A.; Hanigan, T.W.; Firestone, A.J.; Inguva, A.; Long, A.; Ngo, N.; Lum, K.M.; Henry, C.L.; et al. ABHD17 regulation of plasma membrane palmitoylation and N-Ras-dependent cancer growth. Nat. Chem. Biol. 2021, 17, 856–864. [Google Scholar] [CrossRef]
  22. Li, J.; Echevarria, K.L.; Traugott, K.A. β-Lactam therapy for methicillin-susceptible Staphylococcus aureus bacteremia: A comparative review of cefazolin versus antistaphylococcal penicillins. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2017, 37, 346–360. [Google Scholar] [CrossRef]
  23. Dingle, T.C.; Gamage, D.; Gomez-Villegas, S.; Hanson, B.M.; Reyes, J.; Abbott, A.; Burnham, C.-A.D.; Bard, J.D.; Fritz, S.; Miller, W.R.; et al. Prevalence and Characterization of the Cefazolin Inoculum Effect in North American Methicillin-Susceptible Staphylococcus aureus Isolates. J. Clin. Microbiol. 2022, 60. [Google Scholar] [CrossRef] [PubMed]
  24. Mandal, M.K.; Ghosh, S.; Bhat, H.R.; Naesens, L.; Singh, U.P. Synthesis and biological evaluation of substituted phenyl azetidine-2-one sulphonyl derivatives as potential antimicrobial and antiviral agents. Bioorg. Chem. 2020, 104, 104320. [Google Scholar] [CrossRef]
  25. Le, P.; Kunold, E.; Macsics, R.; Rox, K.; Jennings, M.C.; Ugur, I.; Reinecke, M.; Chaves-Moreno, D.; Hackl, M.W.; Fetzer, C.; et al. Repurposing human kinase inhibitors to create an antibiotic active against drug-resistant Staphylococcus aureus, persisters and biofilms. Nat. Chem. 2020, 12, 145–158. [Google Scholar] [CrossRef] [PubMed]
  26. Macsics, R.; Hackl, M.W.; Fetzer, C.; Mostert, D.; Bender, J.; Layer, F.; Sieber, S.A. Comparative Target Analysis of Chlorinated Biphenyl Antimicrobials Highlights MenG as Molecular Target of Triclocarban. Appl. Environ. Microbiol. 2020, 86. [Google Scholar] [CrossRef]
  27. FDA Issues Final Rule on Safety and Effectiveness of Antibacterial Soaps. Available online: https://www.fda.gov/news-events/press-announcements/fda-issues-final-rule-safety-and-effectiveness-antibacterial-soaps (accessed on 12 February 2023).
  28. Wang, Y.; Teng, Y.; Wang, D.; Han, K.; Wang, H.; Kang, L. The fate of triclocarban in activated sludge and its influence on biological wastewater treatment system. J. Environ. Manag. 2020, 276, 111237. [Google Scholar] [CrossRef]
  29. Catalano, A.; Iacopetta, D.; Pellegrino, M.; Aquaro, S.; Franchini, C.; Sinicropi, M.S. Diarylureas: Repositioning from Antitumor to Antimicrobials or Multi-Target Agents against New Pandemics. Antibiotics 2021, 10, 92. [Google Scholar] [CrossRef]
  30. Pujol, E.; Blanco-Cabra, N.; Julián, E.; Leiva, R.; Torrents, E.; Vázquez, S. Pentafluorosulfanyl-containing triclocarban analogs with potent antimicrobial activity. Molecules 2018, 23, 2853. [Google Scholar] [CrossRef] [PubMed]
  31. Catalano, A.; Iacopetta, D.; Sinicropi, M.S.; Franchini, C. Diarylureas as antitumor agents. Appl. Sci. 2021, 11, 374. [Google Scholar] [CrossRef]
  32. Kim, W.; Zou, G.; Pan, W.; Fricke, N.; Faizi, H.A.; Kim, S.M.; Khader, R.; Li, S.; Lee, K.; Escorba, L.; et al. The neutrally charged diarylurea compound PQ401 kills antibiotic-resistant and antibiotic-tolerant Staphylococcus aureus. Mbio 2020, 11, 603. [Google Scholar] [CrossRef]
  33. Eissa, I.H.; Mohammad, H.; Qassem, O.A.; Younis, W.; Abdelghany, T.M.; Elshafeey, A.; Moustafa, M.M.A.R.; Seleem, M.N.; Mayhoub, A.S. Diphenylurea derivatives for combating methicillin- and vancomycin-resistant Staphylococcus aureus. Eur. J. Med. Chem. 2017, 130, 73–85. [Google Scholar] [CrossRef]
  34. Mohammad, H.; Younis, W.; Ezzat, H.G.; Peters, C.E.; AbdelKhalek, A.; Cooper, B.; Pogliano, K.; Pogliano, J.; Mayhoub, A.S.; Seleem, M.N. Bacteriological profiling of diphenylureas as a novel class of antibiotics against methicillin-resistant Staphylococcus aureus. PLoS ONE 2017, 12, e0182821. [Google Scholar] [CrossRef] [PubMed]
  35. Bratenko, M.K.; Barus, M.M.; Rotar, D.V.; Vovk, M.V. Polyfunctional Pyrazoles. 9*. Synthesis of 1-Alkyl(Aryl)-3-[4-(Hydroxymethyl)-1H-Pyrazol-3-Yl]ureas. Chem. Heterocyc. Compd. 2014, 50, 1252–1258. [Google Scholar] [CrossRef]
  36. Kane, J.L.; Hirth, B.H.; Liang, B.; Gourlie, B.B.; Nahill, S.; Barsomian, G. Ureas of 5-aminopyrazole and 2-aminothiazole inhibit growth of gram-positive bacteria. Bioorg. Med. Chem. Lett. 2003, 13, 4463–4466. [Google Scholar] [CrossRef] [PubMed]
  37. Brullo, C.; Rapetti, F.; Bruno, O. Pyrazolyl-Ureas as Interesting Scaffold in Medicinal Chemistry. Molecules 2020, 25, 3457. [Google Scholar] [CrossRef]
  38. Gezegen, H.; Hepokur, C.; Tutar, U.; Ceylan, M. Synthesis and Biological Evaluation of Novel 1-(4-(Hydroxy(1-oxo-1,3-dihydro-2H-inden-2-ylidene)methyl)phenyl)-3-phenylurea Derivatives. Chem. Biodivers. 2017, 14, e1700223. [Google Scholar] [CrossRef]
  39. Jamil, R.T.; Foris, L.A.; Snowden, J. Proteus Mirabilis Infections. Available online: https://www.ncbi.nlm.nih.gov/books/NBK442017/ (accessed on 30 March 2023).
  40. Ashurst, J.V.; Dawson, A. Klebsiella Pneumoniae. Available online: https://www.ncbi.nlm.nih.gov/books/NBK519004/ (accessed on 30 March 2023).
  41. Liu, R.; Miller, P.A.; Miller, M.J. Conjugation of Aztreonam, a Synthetic Monocyclic β-Lactam Antibiotic, to a Siderophore Mimetic Significantly Expands Activity Against Gram-Negative Bacteria. ACS Infect. Dis. 2021, 7, 2979–2986. [Google Scholar] [CrossRef] [PubMed]
  42. Sarveswari, S.; Vijayakumar, V. A Facile Synthesis of diaylureas and their antimicrobial evaluation. Chiang Mai J. Sci. 2018, 45, 997–1007. [Google Scholar]
  43. Patil, M.; Noonikara-Poyil, A.; Joshi, S.D.; Patil, S.A.; Patil, S.A.; Bugarin, A. New Urea Derivatives as Potential Antimicrobial Agents: Synthesis, Biological Evaluation, and Molecular Docking Studies. Antibiotics 2019, 8, 178. [Google Scholar] [CrossRef]
  44. Diacon, A.H.; van der Merwe, L.; Barnard, M.; von Groote-Bidlingmaier, F.; Lange, C.; García-Basteiro, A.L.; Sevene, E.; Ballell, L.; Barros-Aguirre, D. β-Lactams against Tuberculosis--New Trick for an Old Dog? N. Engl. J. Med. 2016, 375, 393–394. [Google Scholar] [CrossRef] [PubMed]
  45. van Rijn, S.P.; Zuur, M.A.; Anthony, R.; Wilffert, B.; van Altena, R.; Akkerman, O.W.; de Lange, W.C.M.; van der Werf, T.S.; Kosterink, J.G.W.; Alffenaar, J.C. Evaluation of Carbapenems for Treatment of Multi- and Extensively Drug-Resistant Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2019, 63, e01489-18. [Google Scholar] [CrossRef] [PubMed]
  46. Gold, B.; Zhang, J.; Quezada, L.L.; Roberts, J.; Ling, Y.; Wood, M.; Shinwari, W.; Goullieux, L.; Roubert, C.; Fraisse, L.; et al. Identification of β-Lactams Active against Mycobacterium tuberculosis by a Consortium of Pharmaceutical Companies and Academic Institutions. ACS Infect. Dis. 2022, 8, 557–573. [Google Scholar] [CrossRef]
  47. Lopez Quezada, L.; Li, K.; McDonald, S.L.; Nguyen, Q.; Perkowski, A.J.; Pharr, C.W.; Gold, B.; Roberts, J.; McAulay, K.; Saito, K.; et al. Dual-Pharmacophore Pyrithione-Containing Cephalosporins Kill Both Replicating and Nonreplicating Mycobacterium tuberculosis. ACS Infect. Dis. 2019, 5, 1433–1445. [Google Scholar] [CrossRef] [PubMed]
  48. Kuskovsky, R.; Lloyd, D.; Arora, K.; Plotkin, B.; Green, J.; Boshoff, H.; Barry, C.; Deschamps, J.; Konaklieva, M.I. C4-Phenylthio β-lactams: Effect of the Chirality of the β-Lactam Ring on Antimicrobial Activity. Bioorg. Med. Chem. 2019, 27, 115050. [Google Scholar] [CrossRef]
  49. Déziel, R.; Malenfant, E. Inhibition of human cytomegalovirus protease No with monocyclic β-lactams. Bioorg. Med. Chem. Lett. 1998, 8, 1437–1442. [Google Scholar] [CrossRef]
  50. Upadhayaya, R.S.; Kulkarni, G.M.; Vasireddy, N.R.; Vandavasi, J.K.; Dixit, S.S.; Sharma, V. Chattapadhayaya, Design, synthesis and biological evaluation of novel triazole, urea and thiourea derivatives of quinoline against Mycobacterium tuberculosis. Bioorg. Med. Chem. 2009, 17, 4681–4692. [Google Scholar] [CrossRef] [PubMed]
  51. Velappan, A.B.; Raja, M.R.C.; Datta, D.; Tsai, Y.T.; Halloum, I.; Wan, B.; Kremer, L.; Gramajo, H.; Franzblau, S.G.; Mahapatra, S.K.; et al. Attenuation of Mycobacterium species through direct and macrophage mediated pathway by unsymmetrical diaryl urea. Eur. J. Med. Chem. 2017, 125, 825–841. [Google Scholar] [CrossRef]
  52. Yoakim, C.; Ogilvie, W.W.; Cameron, D.R.; Chabot, C.; Grand-Maître, C.; Guse, I.; Haché, B.; Kawai, S.; Naud, J.; O’Meara, J.A.; et al. Potent β-Lactam Inhibitors of Human Cytomegalovirus Protease. Antivir. Chem. Chemother. 1998, 9, 379–387. [Google Scholar] [CrossRef]
  53. Yoakim, C.; Ogilvie, W.W.; Cameron, D.R.; Chabot, C.; Guse, I.; Haché, B.; Naud, J.; O’Meara, J.A.; Plante, R.; Déziel, R. β-Lactam Derivatives as Inhibitors of Human Cytomegalovirus Protease. J. Med. Chem. 1998, 41, 2882–2891. [Google Scholar] [CrossRef]
  54. Gerona-Navarro, G.; Pérez de Vega, M.J.; García-López, M.T.; Andrei, G.; Snoeck, R.; Balzarini, J.; De Clercq, E.; González-Muñiz, R. Synthesis and anti-HCMV activity of 1-acyl-β-lactams and 1-acylazetidines derived from phenylalanine. Bioorg. Med. Chem. Lett. 2004, 14, 2253–2256. [Google Scholar] [CrossRef]
  55. Gerona-Navarro, G.; Pérez de Vega, M.J.; García-López, M.T.; Andrei, G.; Snoeck, R.; De Clercq, E.; Balzarini, J.; González-Muñiz, R. From 1-Acyl-β-lactam Human Cytomegalovirus Protease Inhibitors to 1-Benzyloxycarbonylazetidines with Improved Antiviral Activity. A Straightforward Approach to Convert Covalent to Noncovalent Inhibitors. J. Med. Chem. 2005, 48, 2612–2621. [Google Scholar] [CrossRef]
  56. Džimbeg, G.; Zorc, B.; Kralj, M.; Ester, K.; Pavelić, K.; Andrei, G.; Snoeck, R.; Balzarini, J.; De Clercq, E.; Mintas, M. The novel primaquine derivatives of N-alkyl, cycloalkyl or aryl urea: Synthesis, cytostatic and antiviral activity evaluations. Eur. J. Med. Chem. 2008, 43, 1180–1187. [Google Scholar] [CrossRef]
  57. Xin, Z.; Zhao, H.; Serby, M.D.; Liu, B.; Liu, M.; Szczepankiewicz, B.G.; Nelson, L.T.; Smith, H.T.; Suhar, T.S.; Janis, R.S.; et al. Discovery of piperidine-aryl urea-based stearoyl-CoA desaturase 1 inhibitors. Bioorg. Med. Chem. Lett. 2008, 18, 4298–4302. [Google Scholar] [CrossRef] [PubMed]
  58. Attie, A.D.; Krauss, M.R.; Gray-Keller, M.P.; Brownlie, A.; Miyazaki, M.; Kastelein, J.J.; Lusis, A.J.; Stalenhoef, A.F.; Stoehr, J.P.; Hayden, M.R.; et al. Relationship between stearoyl-CoA desaturase activity and plasma triglycerides in human and mouse hypertriglyceridemia. J. Lipid Res. 2002, 43, 1899–1907. [Google Scholar] [CrossRef] [PubMed]
  59. von Roemeling, C.A.; Marlow, L.A.; Wei, J.J.; Cooper, S.J.; Caulfield, T.R.; Wu, K.; Tan, W.W.; Tun, H.W.; Copland, J.A. Stearoyl-CoA desaturase 1 is a novel molecular therapeutic target for clear cell renal cell carcinoma. Clin. Cancer Res. 2013, 19, 2368–2380. [Google Scholar] [CrossRef] [PubMed]
  60. Gullberg, R.C.; Steel, J.J.; Pujari, V.; Rovnak, J.; Crick, D.C.; Perera, R. Stearoly-CoA desaturase 1 differentiates early and advanced dengue virus infections and determines virus particle infectivity. PLoS Pathog. 2018, 14, e1007261. [Google Scholar] [CrossRef]
  61. Uto, Y. Recent progress in the discovery and development of stearoyl CoA desaturase inhibitors. Chem. Phys. Lipids 2016, 197, 3–12. [Google Scholar] [CrossRef]
  62. Hamilton, L.K.; Moquin-Beaudry, G.; Mangahas, C.L.; Pratesi, F.; Aubin, M.; Aumont, A.; Joppé, S.E.; Légiot, A.; Vachon, A.; Plourde, M.; et al. Stearoyl-CoA Desaturase inhibition reverses immune, synaptic and cognitive impairments in an Alzheimer’s disease mouse model. Nat. Commun. 2002, 13, 2061. [Google Scholar] [CrossRef]
  63. Dražić, T.; Kopf, S.; Corridan, J.; Leuthold, M.M.; Bertoša, B.D.; Klein, C.D. Peptide-β-lactam Inhibitors of Dengue and West Nile Virus NS2B-NS3 Protease Display Two Distinct Binding Modes. J. Med. Chem. 2020, 63, 140–156. [Google Scholar] [CrossRef]
  64. Bártolo, I.; Santos, S.S.; Fontinha, D.; Machado, M.; Francisco, D.; Sepodes, B.; Rocha, J.; Mota-Filipe, H.; Pinto, R.; Figueira, M.E.; et al. Spiro-β-lactam BSS-730A Displays Potent Activity against HIV and Plasmodium. ACS Infect. Dis. 2021, 7, 421–434. [Google Scholar] [CrossRef]
  65. Dömling, A.; Starnecker, M.; Ugi, I. The β-lactam-nucleoside chimera. Angew. Chem. Int. Ed. 1995, 34, 2238–2239. [Google Scholar] [CrossRef]
  66. Dang, Q.; Zhang, Z.; Bai, Y.; Sun, R.; Yin, J.; Chen, T.; Bogen, S.; Girijavallabhan, V.; Olsen, D.B.; Meinke, P.T. Syntheses of nucleosides with a 1′,2′-β-lactam moiety as potential inhibitors of hepatitis C virus NS5B polymerase. Tetrahedron Lett. 2014, 55, 5576–5579. [Google Scholar] [CrossRef]
  67. D’hooghe, M.; Mollet, K.; De Vreese, R.; Jonckers, T.H.M.; Dams, G.; De Kimpe, N. Design, Synthesis, and Antiviral Evaluation of Purine-β-lactam and Purine-aminopropanol Hybrids. J. Med. Chem. 2012, 55, 5637–5641. [Google Scholar] [CrossRef]
  68. Głowacka, I.E.; Grabkowska-Drużyc, M.; Andrei, G.; Schols, D.; Snoeck, R.; Witek, K.; Podlewska, S.; Handzlik, J.; Piotrowska, D.G. Novel N-Substituted 3-Aryl-4-(diethoxyphosphoryl)azetidin-2-ones as Antibiotic Enhancers and Antiviral Agents in Search for a Successful Treatment of Complex Infections. Int. J. Mol. Sci. 2021, 22, 8032. [Google Scholar] [CrossRef]
  69. Oerlemans, R.; Ruiz-Moreno, A.J.; Cong, Y.; Dinesh Kumar, N.; Velasco-Velazquez, M.A.; Neochoritis, C.G.; Smith, J.; Reggiori, F.; Groves, M.R.; Dömling, A. Repurposing the HCV NS3-4A protease drug boceprevir as COVID-19 therapeutics. RSC Med. Chem. 2020, 12, 370–379. [Google Scholar] [CrossRef] [PubMed]
  70. Kneller, D.W.; Li, H.; Phillips, G.; Weiss, K.L.; Zhang, Q.; Arnould, M.A.; Jonsson, C.B.; Surendranathan, S.; Parvathareddy, J.; Blakeley, M.P.; et al. Covalent narlaprevir- and boceprevir-derived hybrid inhibitors of SARS-CoV-2 main protease. Nat. Commun. 2022, 13, 2268. [Google Scholar] [CrossRef] [PubMed]
  71. Benmansour, F.; Trist, I.; Coutard, B.; Decroly, E.; Querat, G.; Brancale, A.; Barral, K. Discovery of novel dengue virus NS5 methyltransferase non-nucleoside inhibitors by fragment-based drug design. Eur. J. Med. Chem. 2017, 125, 865–880. [Google Scholar] [CrossRef]
  72. Hernandez, J.; Hoffer, L.; Coutard, B.; Querat, G.; Roche, P.; Morelli, X.; Decroly, E.; Barral, K. Optimization of a fragment linking hit toward Dengue and Zika virus NS5 methyltransferases inhibitors. Eur. J. Med. Chem. 2019, 161, 323–333. [Google Scholar] [CrossRef] [PubMed]
  73. Spizzichino, S.; Mattedi, G.; Lauder, K.; Valle, C.; Aouadi, W.; Canard, B.; Decroly, E.; Kaptein, S.J.F.; Neyts, J.; Graham, C.; et al. Design, Synthesis and Discovery of N,N′-Carbazoyl-aryl-urea Inhibitors of Zika NS5 Methyltransferase and Virus Replication. ChemMedChem 2020, 15, 385–390. [Google Scholar] [CrossRef]
  74. Micewicz, E.D.; Khachatoorian, R.; French, S.W.; Ruchala, P. Identification of novel small-molecule inhibitors of Zika virus infection. Bioorg. Med. Chem. Lett. 2018, 28, 452–458. [Google Scholar] [CrossRef]
  75. Khachatoorian, R.; Micewicz, E.D.; Micewicz, A.; French, S.W.; Ruchala, P. Optimization of 1, 3-disubstituted urea-based inhibitors of Zika virus infection. Bioorg. Med. Chem. Lett. 2019, 29, 126626. [Google Scholar] [CrossRef]
  76. Xu, H.; Cheng, M.; Chi, X.; Liu, X.; Zhou, J.; Lin, T.; Yang, W. High-Throughput Screening Identifies Mixed-Lineage Kinase 3 as a Key Host Regulatory Factor in Zika Virus Infection. J. Virol. 2019, 93, e00758-19. [Google Scholar] [CrossRef]
  77. Voss, S.; Nitsche, C. Inhibitors of the Zika virus protease NS2B-NS3. Bioorg. Med. Chem. Lett. 2020, 30, 126965. [Google Scholar] [CrossRef]
  78. Lesch, M.; Luckner, M.; Meyer, M.; Weege, F.; Gravenstein, I.; Raftery, M.; Sieben, C.; Martin-Sancho, L.; Imai-Matsushima, A.; Welke, R.-W.; et al. RNAi-based small molecule repositioning reveals clinically approved urea-based kinase inhibitors as broadly active antivirals. PLoS Pathog. 2019, 15, e1007601. [Google Scholar] [CrossRef] [PubMed]
  79. Longshaw, A.I.; Fordyce, E.A.F.; Onions, S.T.; King-Underwood, J.; Venable, J.D. Preparation of Aromatic Heterocyclic Compounds as p38 MAP Kinase Inhibitors with Antiinflammatory Activity. WO Patent 2,015,121,444, 20 August 2015. [Google Scholar]
  80. King-Underwood, J.; Murray, P.J.; Williams, J.G.; Buck, I.; Onions, S.T. Preparation of Pyrazolyl Naphthyl Ureas as p38 MAP Kinase Inhibitors. WO Patent 2,011,124,930, 13 October 2011. [Google Scholar]
  81. King-Underwood, J.; Ito, K.; Strong, P.; Rapeport, W.G.; Charron, C.E.; Murray, P.J.; Williams, J.G.; Onions, S.T. 1-(5-Pyrazolyl)-3-(1-naphthyl)ureas as Enzyme Inhibitors and Their Preparation, Pharmaceutical Compositions and Use in the Treatment of Inflammatory Disorders. WO Patent 2,011,124,923, 13 October 2011. [Google Scholar]
  82. Ito, K.; Strong, P.; Rapeport, W.G.; Murray, P.J.; King-Underwood, J.; Williams, J.G.; Onions, S.T.; Joly, K.; Charron, C.E. Preparation of Pyrazolyl Ureas as p38 MAP Kinase Inhibitors. WO Patent 2,010,067,130, 21 April 2011. [Google Scholar]
  83. Murray, P.J.; Onions, S.T.; Williams, J.G.; Joly, K. Preparation of Pyrido[2,3-b]pyrazine Compounds for Use in Drug Formulations for Treating Inflammation, Respiratory Disorders, and Viral Infections. WO2012119690A1, 22 December 2011. Available online: https://www.ncbi.nlm.nih.gov/books/NBK544312 (accessed on 3 April 2023).
  84. Ahmed-Belkacem, A.; Colliandre, L.; Ahnou, N.; Nevers, Q.; Gelin, M.; Bessin, Y.; Brillet, R.; Cala, O.; Douguet, D.; Bourguet, W.; et al. Fragment-based discovery of a new family of non-peptidic small-molecule cyclophilin inhibitors with potent antiviral activities. Nat. Commun. 2016, 7, 12777. [Google Scholar] [CrossRef] [PubMed]
  85. Hopkins, S.; Gallay, P.A. The role of immunophilins in viral infection. Biochim. Biophys. Acta 2014, 1850, 2103–2110. [Google Scholar] [CrossRef] [PubMed]
  86. Flisiak, R.; Feinman, S.V.; Jablkowski, M.; Horban, A.; Kryczka, W.; Pawlowska, M.; Heathcote, J.E.; Mazzella, G.; Vandelli, C.; Nicolas-Métral, V.; et al. The cyclophilin inhibitor Debio-025 combined with PEG-IFN alpha-2a significantly reduces viral load in treatment-naïve hepatitis C patients. Hepatology 2009, 49, 1460–1468. [Google Scholar] [CrossRef] [PubMed]
  87. Pawlotsky, J.M.; Flisiak, R.; Sarin, S.K.; Rasenack, J.; Piratvisuth, T.; Chuang, W.L.; Peng, C.Y.; Foster, G.R.; Shah, S.; Wedemeyer, H.; et al. Alisporivir plus ribavirin, interferon-free or in combination with peg-interferon, for HCV genotype 2 or 3 infection. Hepatology 2015, 62, 1013–1023. [Google Scholar] [CrossRef]
  88. Fu, L.; Ye, F.; Feng, Y.; Yu, F.; Wang, Q.; Wu, Y.; Zhao, C.; Sun, H.; Huang, P.; Niu, P.; et al. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat. Commun. 2020, 11, 4417. [Google Scholar] [CrossRef]
  89. Malla, T.R.; Tumber, A.; John, T.; Brewitz, L.; Strain-Damerell, C.; Owen, C.D.; Schofield, C.J. Mass spectrometry reveals potential of β-lactams as SARS-CoV-2 M pro inhibitors. Chem. Commun. 2021, 57, 1430–1433. [Google Scholar] [CrossRef]
  90. Malla, T.R.; Brewitz, L.; Muntean, D.-G.; Aslam, H.; Owen, C.D.; Salah, E.; Tumber, A.; Lukacik, P.; Strain-Damerell, C.; Mikolajek, H.; et al. Penicillin Derivatives Inhibit the SARS-CoV-2 Main Protease by Reaction with Its Nucleophilic Cysteine. J. Med. Chem. 2022, 65, 7682–7696. [Google Scholar] [CrossRef]
  91. Niphakis, M.J.; Cravatt, B.F. Enzyme inhibitor discovery by activity-based protein profling. Annu. Rev. Biochem. 2014, 83, 341–377. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Illustrative examples of the “classical” β-lactams: acylating agents of proteases.
Figure 1. Illustrative examples of the “classical” β-lactams: acylating agents of proteases.
Applmicrobiol 03 00043 g001
Figure 2. Illustrative examples of β-lactams: interfering with bacterial fatty acid synthesis, N-alkylthio β-lactams and adsorption of mammalian cholesterol, Ezetimide, respectively.
Figure 2. Illustrative examples of β-lactams: interfering with bacterial fatty acid synthesis, N-alkylthio β-lactams and adsorption of mammalian cholesterol, Ezetimide, respectively.
Applmicrobiol 03 00043 g002
Figure 3. The latest FDA-approved pan-antimicrobial, Cefiderocol.
Figure 3. The latest FDA-approved pan-antimicrobial, Cefiderocol.
Applmicrobiol 03 00043 g003
Figure 4. Trypan Red analog—the molecule at the foundation of modern chemotherapy (Paul Ehrlich sought for the first time to correlate the chemical structure of a synthetic drug with its biological effects).
Figure 4. Trypan Red analog—the molecule at the foundation of modern chemotherapy (Paul Ehrlich sought for the first time to correlate the chemical structure of a synthetic drug with its biological effects).
Applmicrobiol 03 00043 g004
Figure 5. Urea-based FDA-approved kinase inhibitors Sorafenib and Regorafenib (anti-cancer agents), respectively.
Figure 5. Urea-based FDA-approved kinase inhibitors Sorafenib and Regorafenib (anti-cancer agents), respectively.
Applmicrobiol 03 00043 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Konaklieva, M.I.; Plotkin, B.J. β-Lactams and Ureas as Cross Inhibitors of Prokaryotic Systems. Appl. Microbiol. 2023, 3, 605-628. https://doi.org/10.3390/applmicrobiol3030043

AMA Style

Konaklieva MI, Plotkin BJ. β-Lactams and Ureas as Cross Inhibitors of Prokaryotic Systems. Applied Microbiology. 2023; 3(3):605-628. https://doi.org/10.3390/applmicrobiol3030043

Chicago/Turabian Style

Konaklieva, Monika I., and Balbina J. Plotkin. 2023. "β-Lactams and Ureas as Cross Inhibitors of Prokaryotic Systems" Applied Microbiology 3, no. 3: 605-628. https://doi.org/10.3390/applmicrobiol3030043

Article Metrics

Back to TopTop