Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Chem Biol Drug Des ; 103(6): e14569, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38877369

ABSTRACT

Staphylococcus aureus has the ability to invade cortical bone osteocyte lacuno-canalicular networks (OLCNs) and cause osteomyelitis. It was recently established that the cell wall transpeptidase, penicillin-binding protein 4 (PBP4), is crucial for this function, with pbp4 deletion strains unable to invade OLCNs and cause bone pathogenesis in a murine model of S. aureus osteomyelitis. Moreover, PBP4 has recently been found to modulate S. aureus resistance to ß-lactam antibiotics. As such, small molecule inhibitors of S. aureus PBP4 may represent dual functional antimicrobial agents that limit osteomyelitis and/or reverse antibiotic resistance. A high throughput screen recently revealed that the phenyl-urea 1 targets PBP4. Herein, we describe a structure-activity relationship (SAR) study on 1. Leveraging in silico docking and modeling, a set of analogs was synthesized and assessed for PBP4 inhibitory activities. Results revealed a preliminary SAR and identified lead compounds with enhanced binding to PBP4, more potent antibiotic resistance reversal, and diminished PBP4 cell wall transpeptidase activity in comparison to 1.


Subject(s)
Anti-Bacterial Agents , Molecular Docking Simulation , Penicillin-Binding Proteins , Staphylococcus aureus , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Structure-Activity Relationship , Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Urea/chemistry , Urea/pharmacology , Urea/analogs & derivatives , Animals , Mice , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors
2.
ACS Infect Dis ; 10(4): 1339-1350, 2024 04 12.
Article in English | MEDLINE | ID: mdl-38491938

ABSTRACT

Increasing antimicrobial resistance, coupled with the absence of new antibiotics, has led physicians to rely on colistin, a polymyxin with known nephrotoxicity, as the antibiotic of last resort for the treatment of infections caused by Gram-negative bacteria. One approach to increasing antibiotic efficacy and thereby reducing dosage is the use of small-molecule potentiators that augment antibiotic activity. We recently identified the aporphine alkaloid (±)-variabiline, which lowers the minimum inhibitory concentration of colistin in Acinetobacter baumannii and Klebsiella pneumoniae. Herein, we report the first total synthesis of (±)-variabiline to confirm structure and activity, the resolution, and evaluation of both enantiomers as colistin potentiators, and a structure-activity relationship study that identifies more potent variabiline derivatives. Preliminary mechanistic studies indicate that (±)-variabiline and its derivatives potentiate colistin by targeting the Gram-negative outer membrane.


Subject(s)
Acinetobacter baumannii , Alkaloids , Aporphines , Colistin/pharmacology , Klebsiella pneumoniae , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Alkaloids/pharmacology
3.
Chembiochem ; 25(8): e202400127, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38451872

ABSTRACT

The development of novel therapeutic approaches is crucial in the fight against multi-drug resistant (MDR) bacteria, particularly gram-negative species. Small molecule adjuvants that enhance the activity of otherwise gram-positive selective antibiotics against gram-negative bacteria have the potential to expand current treatment options. We have previously reported adjuvants based upon a 2-aminoimidazole (2-AI) scaffold that potentiate macrolide antibiotics against several gram-negative pathogens. Herein, we report the discovery and structure-activity relationship (SAR) investigation of an additional class of macrolide adjuvants based upon a 2-aminobenzimidazole (2-ABI) scaffold. The lead compound lowers the minimum inhibitory concentration (MIC) of clarithromycin (CLR) from 512 to 2 µg/mL at 30 µM against Klebsiella pneumoniae 2146, and from 32 to 2 µg/mL at 5 µM, against Acinetobacter baumannii 5075. Preliminary investigation into the mechanism of action suggests that the compounds are binding to lipopolysaccharide (LPS) in K. pneumoniae, and modulating lipooligosaccharide (LOS) biosynthesis, assembly, or transport in A. baumannii.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria , Benzimidazoles/pharmacology , Macrolides , Microbial Sensitivity Tests
4.
Antibiotics (Basel) ; 12(11)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37998765

ABSTRACT

In 2019, five million deaths associated with antimicrobial resistance were reported by The Centers for Disease Control and Prevention (CDC). Acinetobacter baumannii, a Gram-negative bacterial pathogen, is among the list of urgent threats. Previously, we reported 2-aminoimidazole (2-AI) adjuvants that potentiate macrolide activity against A. baumannii. In this study, we identify several of these adjuvants that sensitize A. baumannii to aminoglycoside antibiotics. Lead compounds 1 and 7 lower the tobramycin (TOB) minimum inhibitory concentration (MIC) against the TOB-resistant strain AB5075 from 128 µg/mL to 2 µg/mL at 30 µM. In addition, the lead compounds lower the TOB MIC against the TOB-susceptible strain AB19606 from 4 µg/mL to 1 µg/mL and 0.5 µg/mL, respectively, at 30 µM and 15 µM. The evolution of resistance to TOB and 1 in AB5075 revealed mutations in genes related to protein synthesis, the survival of bacteria under environmental stressors, bacteriophages, and proteins containing Ig-like domains.

5.
Int J Mol Sci ; 24(12)2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37372952

ABSTRACT

Ovarian cancer is the sixth leading cause of cancer-related death in women, and both occurrence and mortality are increased in women over the age of 60. There are documented age-related changes in the ovarian cancer microenvironment that have been shown to create a permissive metastatic niche, including the formation of advanced glycation end products, or AGEs, that form crosslinks between collagen molecules. Small molecules that disrupt AGEs, known as AGE breakers, have been examined in other diseases, but their efficacy in ovarian cancer has not been evaluated. The goal of this pilot study is to target age-related changes in the tumor microenvironment with the long-term aim of improving response to therapy in older patients. Here, we show that AGE breakers have the potential to change the omental collagen structure and modulate the peritoneal immune landscape, suggesting a potential use for AGE breakers in the treatment of ovarian cancer.


Subject(s)
Glycation End Products, Advanced , Ovarian Neoplasms , Humans , Female , Aged , Pilot Projects , Collagen , Ovarian Neoplasms/drug therapy , Tumor Microenvironment
6.
Org Biomol Chem ; 21(16): 3373-3380, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37013457

ABSTRACT

Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are difficult to treat due to their resistance to many ß-lactam antibiotics, and their highly coordinated excretion of virulence factors. One way in which MRSA accomplishes this is by responding to environmental stimuli using two-component systems (TCS). The ArlRS TCS has been identified as having a key role in regulating virulence in both systemic and local infections caused by S. aureus. We recently disclosed 3,4'-dimethoxyflavone as a selective ArlRS inhibitor. In this study we explore the structure-activity relationship (SAR) of the flavone scaffold for ArlRS inhibition and identify several compounds with increased activity compared to the parent. Additionally, we identify a compound that suppresses oxacillin resistance in MRSA, and begin to probe the mechanism of action behind this activity.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Humans , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Structure-Activity Relationship , Microbial Sensitivity Tests
7.
mBio ; 14(3): e0013723, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37074183

ABSTRACT

Antibiotic tolerance within a biofilm community presents a serious public health challenge. Here, we report the identification of a 2-aminoimidazole derivative that inhibits biofilm formation by two pathogenic Gram-positive bacteria, Streptococcus mutans and Staphylococcus aureus. In S. mutans, the compound binds to VicR, a key response regulator, at the N-terminal receiver domain, and concurrently inhibits expression of vicR and VicR-regulated genes, including the genes that encode the key biofilm matrix producing enzymes, Gtfs. The compound inhibits S. aureus biofilm formation via binding to a Staphylococcal VicR homolog. In addition, the inhibitor effectively attenuates S. mutans virulence in a rat model of dental caries. As the compound targets bacterial biofilms and virulence through a conserved transcriptional factor, it represents a promising new class of anti-infective agents that can be explored to prevent or treat a host of bacterial infections. IMPORTANCE Antibiotic resistance is a major public health issue due to the growing lack of effective anti-infective therapeutics. New alternatives to treat and prevent biofilm-driven microbial infections, which exhibit high tolerance to clinically available antibiotics, are urgently needed. We report the identification of a small molecule that inhibits biofilm formation by two important pathogenic Gram-positive bacteria, Streptococcus mutans and Staphylococcus aureus. The small molecule selectively targets a transcriptional regulator leading to attenuation of a biofilm regulatory cascade and concurrent reduction of bacterial virulence in vivo. As the regulator is highly conserved, the finding has broad implication for the development of antivirulence therapeutics that selectively target biofilms.


Subject(s)
Anti-Infective Agents , Dental Caries , Staphylococcal Infections , Rats , Animals , Virulence , Staphylococcus aureus/genetics , Dental Caries/drug therapy , Dental Caries/prevention & control , Biofilms , Anti-Bacterial Agents/metabolism , Anti-Infective Agents/pharmacology , Staphylococcal Infections/drug therapy , Streptococcus mutans/metabolism
8.
Eur J Med Chem ; 253: 115329, 2023 May 05.
Article in English | MEDLINE | ID: mdl-37023677

ABSTRACT

The Centers for Disease Control and Prevention (CDC) reports that hospital acquired infections have increased by 65% since 2019. One of the main contributors is the gram-negative bacterium Acinetobacter baumannii. Previously, we reported aryl 2-aminoimidazole (2-AI) adjuvants that potentiate macrolide antibiotics against A. baumannii. Macrolide antibiotics are typically used to treat infections caused by gram-positive bacteria, but are ineffective against most gram-negative bacteria. We describe a new class of dimeric 2-AIs that are highly active macrolide adjuvants, with lead compounds lowering minimum inhibitory concentrations (MICs) to or below the gram-positive breakpoint level against A. baumannii. The parent dimer lowers the clarithromycin (CLR) MIC against A. baumannii 5075 from 32 µg/mL to 1 µg/mL at 7.5 µM (3.4 µg/mL), and a subsequent structure activity relationship (SAR) study identified several compounds with increased activity. The lead compound lowers the CLR MIC to 2 µg/mL at 1.5 µM (0.72 µg/mL), far exceeding the activity of both the parent dimer and the previous lead aryl 2-AI. Furthermore, these dimeric 2-AIs exhibit considerably reduced mammalian cell toxicity compared to aryl-2AI adjuvants, with IC50s of the two lead compounds against HepG2 cells of >200 µg/mL, giving therapeutic indices of >250.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Imidazoles/pharmacology , Microbial Sensitivity Tests , Adjuvants, Immunologic/pharmacology , Gram-Negative Bacteria , Polymers/pharmacology , Macrolides/pharmacology , Mammals
9.
RSC Med Chem ; 14(2): 247-252, 2023 Feb 22.
Article in English | MEDLINE | ID: mdl-36846374

ABSTRACT

Multidrug resistant (MDR) bacterial infections have become increasingly common, leading clinicians to rely on last-resort antibiotics such as colistin. However, the utility of colistin is becoming increasingly compromised as a result of increasing polymyxin resistance. Recently we discovered that derivatives of the eukaryotic kinase inhibitor meridianin D abrogate colistin resistance in several Gram-negative species. A subsequent screen of three commercial kinase inhibitor libraries led to the identification of several scaffolds that potentiate colistin activity, including 6-bromoindirubin-3'-oxime, which potently suppresses colistin resistance in Klebsiella pneumoniae. Herein we report the activity of a library of 6-bromoindirubin-3'-oxime analogs and identify four derivatives that show equal or increased colistin potentiation activity compared to the parent compound.

10.
ACS Infect Dis ; 9(2): 283-295, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36651182

ABSTRACT

Colistin, typically viewed as the antibiotic of last resort to treat infections caused by multidrug-resistant (MDR) Gram-negative bacteria, had fallen out of favor due to toxicity issues. The recent increase in clinical usage of colistin has resulted in colistin-resistant isolates becoming more common. To counter this threat, we have investigated previously reported compounds, HSD07 and HSD17, and developed 13 compounds with more desirable drug-like properties for colistin sensitization against 16 colistin-resistant bacterial strains, three of which harbor the plasmid-borne mobile colistin resistance (mcr-1). Lead compound HSD1624, which has a lower LogDpH7.4 (2.46) compared to HSD07 (>5.58), reduces the minimum inhibitory concentration (MIC) of colistin against Pseudomonas aeruginosa strain TRPA161 to 0.03 µg/mL from 1024 µg/mL (34,000-fold reduction). Checkerboard assays revealed that HSD1624 and analogues are also synergistic with colistin against colistin-resistant strains of Escherichia coli, Acinetobacter baumannii, and Klebsiella pneumoniae. Preliminary mechanism of action studies indicate that HSD1624 exerts its action differently depending on the bacterial species. Time-kill studies suggested that HSD1624 in combination with 0.5 µg/mL colistin was bactericidal to extended-spectrum beta-lactamase (ESBL)-producing E. coli, as well as to E. coli harboring mcr-1, while against P. aeruginosa TRPA161, the combination was bacteriostatic. Mechanistically, HSD1624 increased membrane permeability in K. pneumoniae harboring a plasmid containing the mcr-1 gene but did not increase radical oxygen species (ROS), while a combination of 15 µM HSD1624 and 0.5 µg/mL colistin significantly increased ROS in P. aeruginosa TRPA161. HSD1624 was not toxic to mammalian red blood cells (up to 226 µM).


Subject(s)
Anti-Bacterial Agents , Colistin , Gram-Negative Bacteria , Anti-Bacterial Agents/pharmacology , Bacteria , Colistin/pharmacology , Escherichia coli , Gram-Negative Bacteria/drug effects , Pseudomonas aeruginosa , Reactive Oxygen Species , Drug Resistance, Multiple, Bacterial
11.
Bioorg Med Chem Lett ; 80: 129113, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36566797

ABSTRACT

Gram-negative bacteria are intrinsically resistant to many classes of antibiotics, predominantly due to the impermeability of the outer membrane and the presence of efflux pumps. Small molecule adjuvants that circumvent these resistance mechanisms have the potential to expand therapeutic options for treating Gram-negative infections to encompass antibiotic classes that are otherwise limited to treating Gram-positive infections. Adjuvants that effect increased antibiotic permeation, either by physical disruption of the outer membrane or through interference with synthesis, transport, or assembly of membrane components, and adjuvants that limit efflux, are discussed as potential avenues to overcoming intrinsic resistance in Gram-negative bacteria.


Subject(s)
Drug Resistance, Multiple, Bacterial , Gram-Negative Bacteria , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biological Transport , Adjuvants, Immunologic/pharmacology
12.
13.
ChemMedChem ; 17(16): e202200286, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35704751

ABSTRACT

Multidrug resistant (MDR) bacteria are an increasing public health problem. One promising alternative to the development of new antibiotics is the use of antibiotic adjuvants, which would allow the continued use of FDA-approved antibiotics that have been rendered ineffective due to resistance. Herein, we report a series of dipyrrins and pyrrole derivatives designed as analogues of prodigiosin and obatoclax, several of which potentiate the activity of colistin against Klebsiella pneumoniae, with lead compounds also potentiating colistin against Acinetobacter baumannii and Pseudomonas aeruginosa.


Subject(s)
Acinetobacter baumannii , Colistin , Adjuvants, Pharmaceutic/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Colistin/pharmacology , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests , Prodigiosin/pharmacology , Pseudomonas aeruginosa
14.
Methods Enzymol ; 665: 153-176, 2022.
Article in English | MEDLINE | ID: mdl-35379433

ABSTRACT

The identification of antibiotic adjuvants, small molecules that potentiate the activity of conventional antibiotics, provides an orthogonal approach to the development of new antibiotics in the fight against drug resistant bacterial infections. Methods to identify novel adjuvants could potentially aid efforts to overcome the increasing prevalence of resistance and stave off the onset of a "post-antibiotic era." Phenotypic whole cell screens allow for the identification of hits with the necessary properties to access their biomolecular target, and may also facilitate the discovery of novel adjuvant targets. A phenotypic screening platform is outlined, in which a natural product library was explored for activity with antibiotics from several mechanistically distinct classes against clinically important bacterial species. General approaches to delineating the mechanism of action of hit compounds identified from phenotypic screens are described, followed by specific approaches to uncovering the mechanism of action of the colistin adjuvants identified from the natural product screen.


Subject(s)
Biological Products , Colistin , Adjuvants, Pharmaceutic/pharmacology , Anti-Bacterial Agents/pharmacology , Biological Products/pharmacology , Colistin/pharmacology , Small Molecule Libraries
15.
Angew Chem Int Ed Engl ; 61(17): e202117458, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35167718

ABSTRACT

Over the past decades, antibiotic resistance has grown to a point where orthogonal approaches to combating infections caused by resistant bacteria are needed. One such approach is the development of non-microbicidal small molecules that potentiate the activity of conventional antibiotics, termed adjuvants. The diterpene natural product 12(S),16ϵ-dihydroxycleroda-3,13-dien-15,16-olide, which we refer to as (-)-LZ-2112, is known to synergize with oxacillin against methicillin-resistant Staphylococcus aureus (MRSA). To explore this activity, (-)-LZ-2112 was synthesized and the structure confirmed through X-ray analysis. Preliminary structure-activity relationship studies following the synthesis of several analogs identified key structural elements responsible for activity and indicate that scaffold simplification is possible. A preliminary mode of action study suggests mecA plays a role in the adjuvant activity of (-)-LZ-2112.


Subject(s)
Diterpenes, Clerodane , Methicillin-Resistant Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Diterpenes, Clerodane/pharmacology , Microbial Sensitivity Tests , Oxacillin/pharmacology , beta-Lactams/pharmacology
16.
Eur J Med Chem ; 232: 114203, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35219950

ABSTRACT

Salmonella enterica serovars cause millions of infections each year that result either in typhoid fever or salmonellosis. Among those serovars that cause typhoid fever, Salmonella enterica subspecies Typhi can form biofilms on gallstones in the gallbladders of acutely-infected patients, leading to chronic carriage of the bacterium. These biofilms are recalcitrant to antibiotic-mediated eradication, leading to chronic fecal shedding of the bacteria, which results in further disease transmission. Herein, we report the synthesis and anti-biofilm activity of a 55-member library of small molecules based upon a previously identified hit that both inhibits and disrupts S. Typhi and S. Typhimurium (a nontyphoidal model serovar for S. Typhi) biofilms. Lead compounds inhibit S. Typhimurium biofilm formation in vitro at sub-micromolar concentrations, and disperse biofilms with five-fold greater potentency than the parent compound. Three of the most promising compounds demonstrated synergy with ciprofloxacin in a murine model of chronic Salmonella carriage. This work furthers the development of effective anti-biofilm agents as a promising therapeutic avenue for the eradication of typhoidal Salmonella.


Subject(s)
Ciprofloxacin , Gallbladder , Animals , Anti-Bacterial Agents/pharmacology , Biofilms , Ciprofloxacin/pharmacology , Humans , Mice , Salmonella typhi
17.
ACS Infect Dis ; 7(12): 3303-3313, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34752055

ABSTRACT

Multidrug-resistant bacterial infections have become a global threat. We recently disclosed that the known IKK-ß inhibitor IMD-0354 and subsequent analogues abrogate colistin resistance in several Gram-negative strains. Herein, we report the activity of a second-generation library of IMD-0354 analogues incorporating a benzimidazole moiety as an amide isostere. We identified several analogues that show increased colistin potentiation activity against Gram-negative bacteria.


Subject(s)
Colistin , Salicylanilides , Anti-Bacterial Agents/pharmacology , Benzimidazoles/pharmacology , Colistin/pharmacology , Microbial Sensitivity Tests
18.
RSC Med Chem ; 12(2): 293-296, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-34046617

ABSTRACT

Infections that stem from bacterial biofilms are difficult to eradicate. Within a biofilm state, bacteria are upwards of 1000-fold more resistant to conventional antibiotics, necessitating the development of alternative approaches to treat biofilm-based infections. One such approach is the development of small molecule adjuvants that can inhibit/disrupt bacterial biofilms. When such molecules are paired with conventional antibiotics, these dual treatments present a combination approach to eradicate biofilm-based infections. Previously, we have demonstrated that small molecules containing either a 2-amino pyrimidine (2-AP) or a 2-aminoimidazole (2-AI) heterocycle are potent anti-biofilm agents. Herein, we now report a scaffold hopping strategy to generate new aryl 2-AP analogs that inhibit biofilm formation by methicillin-resistant Staphylococcus aureus (MRSA). These molecules also suppress colistin resistance in colistin resistant Klebsiella pneumoniae, lowering the minimum inhibitory concentration (MIC) by 32-fold.

19.
ACS Med Chem Lett ; 11(9): 1723-1731, 2020 Sep 10.
Article in English | MEDLINE | ID: mdl-32944140

ABSTRACT

Approximately 1.7 million Americans develop hospital associated infections each year, resulting in more than 98,000 deaths. One of the main contributors to such infections is the Gram-negative pathogen Acinetobacter baumannii. Recently, it was reported that aryl 2-aminoimidazole (2-AI) compounds potentiate macrolide antibiotics against a highly virulent strain of A. baumannii, AB5075. The two lead compounds in that report increased clarithromycin (CLR) potency against AB5075 by 16-fold, lowering the minimum inhibitory concentration (MIC) from 32 to 2 µg/mL at a concentration of 10 µM. Herein, we report a structure-activity relationship study of a panel of derivatives structurally inspired by the previously reported aryl 2-AI leads. Substitutions around the core phenyl ring yielded a lead that potentiates clarithromycin by 64- and 32-fold against AB5075 at 10 and 7.5 µM, exceeding the dose response of the original lead. Additional probing of the amide linker led to the discovery of two urea containing adjuvants that suppressed clarithromycin resistance in AB5075 by 64- and 128-fold at 7.5 µM. Finally, the originally reported adjuvant was tested for its ability to suppress the evolution of resistance to clarithromycin over the course of nine consecutive days. At 30 µM, the parent compound reduced the CLR MIC from 512 to 2 µg/mL, demonstrating that the original lead remained active against a more CLR resistant strain of AB5075.

20.
Bioorg Med Chem Lett ; 30(23): 127550, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32927027

ABSTRACT

Synthesis of novel 4(3H)-quinazolinonyl aminopyrimidine derivatives has been achieved via quinazolinonyl enones which in turn were obtained from 2-acyl-4(3H)-quinazolinone. They have been assayed for biofilm inhibition against Gram-positive (methicillin-resistant Staphylococcus aureus (MRSA)) and Gram-negative bacteria (Acinetobacter baumannii). The analogues with 2,4,6-trimethoxy phenyl, 4-methylthio phenyl, and 3-bromo phenyl substituents (5h, 5j & 5k) have been shown to inhibit biofilm formation efficiently in MRSA with IC50 values of 20.7-22.4 µM). The analogues 5h and 5j have demonstrated low toxicity in human cells in vitro and can be investigated further as leads.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Pyrimidines/pharmacology , Quinazolinones/pharmacology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/physiology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Cell Line , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Microbial Sensitivity Tests , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/toxicity , Quinazolinones/chemical synthesis , Quinazolinones/toxicity , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...