Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Antibiotics (Basel) ; 11(12)2022 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-36551427

RESUMO

It is urgent to find new antibiotic classes against multidrug-resistant bacteria as the rate of discovery of new classes of antibiotics has been very slow in the last 50 years. Recently, pyrrolobenzodiazepines (PBDs) with a C8-linked aliphatic-heterocycle have been identified as a new broad-spectrum antibiotic class with activity against Gram-negative bacteria. The active imine moiety of the reported lead pyrrolobenzodiazepine compounds was replaced with amide to obtain the non-DNA binding and noncytotoxic dilactam analogues to understand the structure-activity relationship further and improve the safety potential of this class. The synthesised compounds were tested against panels of multidrug-resistant Gram-positive and Gram-negative bacteria, including WHO priority pathogens. Minimum inhibitory concentrations for the dilactam analogues ranged from 4 to 32 mg/L for MDR Gram-positive bacteria, compared to 0.03 to 2 mg/L for the corresponding imine analogues. At the same time, they were found to be inactive against MDR Gram-negative bacteria, with a MIC > 32 mg/L, compared to a MIC of 0.5 to 32 mg/L for imine analogues. A molecular modelling study suggests that the lack of imine functionality also affects the interaction of PBDs with DNA gyrase. This study suggests that the presence of N10-C11 imine moiety is crucial for the broad-spectrum activity of pyrrolobenzodiazepines.

2.
J Med Chem ; 63(13): 6941-6958, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32515951

RESUMO

It is urgent to find new antibiotic classes with activity against multidrug-resistant (MDR) Gram-negative pathogens as the pipeline of antibiotics is essentially empty. Modified pyrrolobenzodiazepines with a C8-linked aliphatic heterocycle provide a new class of broad-spectrum antibacterial agents with activity against MDR Gram-negative bacteria, including WHO priority pathogens. The structure-activity relationship established that the third ring was particularly important for Gram-negative activity. Minimum inhibitory concentrations for the lead compounds ranged from 0.125 to 2 mg/L for MDR Gram-negative, excluding Pseudomonas aeruginosa, and between 0.03 and 1 mg/L for MDR Gram-positive species. The lead compounds were rapidly bactericidal with >5 log reduction in viable count within 4 h for Acinetobacter baumannii and Klebsiella pneumoniae. The lead compound inhibited DNA gyrase in gel-based assays, with an IC50 of 3.16 ± 1.36 mg/L. This study provides a new chemical scaffold for developing novel broad-spectrum antibiotics which can help replenish the pipeline of antibiotics.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Benzodiazepinas/química , Benzodiazepinas/farmacologia , Desenho de Fármacos , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Antibacterianos/metabolismo , Benzodiazepinas/metabolismo , Linhagem Celular , DNA Girase/química , DNA Girase/metabolismo , Bactérias Gram-Negativas/enzimologia , Humanos , Simulação de Acoplamento Molecular , Conformação Proteica
3.
Mol Pharm ; 17(7): 2354-2369, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32352791

RESUMO

Gram-negative bacteria possess numerous defenses against antibiotics, due to the intrinsic permeability barrier of their outer membrane (OM), explaining the recalcitrance of some common and life-threatening infections. We report the formulation of a new drug, PPA148, which shows promising activity against all Gram-negative bacteria included in the ESKAPEE pathogens. PPA148 was solubilized by inclusion complexation with cyclodextrin followed by encapsulation in liposomes. The complex and liposomal formulation presented increased activity against E. coli compared to the pure drug when assessed with the Kirby Bauer assay. The novel formulation containing 1 µg PPA148 reached similar efficacy levels equivalent to those of 30 µg of pure rifampicin. A range of biophysical techniques was used to explore the mechanism of drug uptake. Langmuir trough (LT) and neutron reflectivity (NR) techniques were employed to monitor the interactions between the drug and the formulation with model membranes. We found evidence for liposome fusion with the model Gram-negative outer membrane and for cyclodextrins acting as inner membrane (IM) permeation enhancers without presenting intrinsic antimicrobial activity. An antibiotic-in-cyclodextrin-in-liposomes (ACL) formulation was developed, which targets both the bacterial OM and IM, and offers promise as a means to breach the Gram-negative cell envelope.


Assuntos
Antibacterianos/administração & dosagem , Antibacterianos/farmacocinética , Membrana Externa Bacteriana/metabolismo , Benzodiazepinas/administração & dosagem , Benzodiazepinas/farmacocinética , Ciclodextrinas/química , Composição de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/métodos , Escherichia coli/metabolismo , Pirróis/administração & dosagem , Pirróis/farmacocinética , Antibacterianos/química , Membrana Externa Bacteriana/efeitos dos fármacos , Benzodiazepinas/química , Permeabilidade da Membrana Celular/efeitos dos fármacos , Farmacorresistência Bacteriana , Escherichia coli/efeitos dos fármacos , Bicamadas Lipídicas/metabolismo , Lipossomos , Fusão de Membrana , Modelos Biológicos , Pirróis/química , Rifampina/farmacologia , Solubilidade
4.
RSC Adv ; 10(24): 13944-13948, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35498455

RESUMO

The functionalization of halloysite nanotube (HNT) surfaces with aminosilanes is an important strategy for their further decoration with organic molecules to obtain hybrid inorganic-organic nanoarchitectures to be used in catalysis and drug delivery. The exact quantification of amino groups on the surface is an important aspect in view of the obtainment of systems with a known number of loaded molecules. In the present study, we describe a simple and reliable method for the correct quantification of groups present on HNT surfaces after their reaction with aminopropyltriethoxysilane (APTES). This method, applied for the first time to HNT chemistry, was based on the use of Fmoc groups as probes covalently bound to APTES and quantified by UV-Vis after release from the HNT-APTES-Fmoc system. Interestingly, this method showed great accordance with the already employed quantitative thermogravimetric analysis (TGA), with some benefits such as simple and non-destructive procedure, besides the possibility to monitor the deprotection reaction.

5.
ACS Omega ; 4(25): 20873-20881, 2019 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-31867477

RESUMO

The development of new antitubercular agents for the treatment of infections caused by multidrug-resistant (MDR) Mycobacterium tuberculosis is an urgent priority. Pyrrolobenzodiazepines (PBDs) are a promising class of antibacterial agents that were initially discovered and isolated from a range of Streptomyces species. Recently, C8-linked PBD monomers have been shown to work by inhibiting DNA gyrase and have demonstrated activity against M. tuberculosis. However, both PBD monomers and dimers are toxic to eukaryotic cells, limiting their development as antibacterial agents. To eliminate the toxicity associated with PBDs and explore the effect of C8-modification with a known antibacterial agent with the same mechanism of action (i.e., ciprofloxacin, a gyrase inhibitor), we synthesized a C8-linked PBD-ciprofloxacin (PBD-CIP, 3) hybrid. The hybrid compound displayed minimum inhibitory concentration values of 0.4 or 2.1 µg/mL against drug-sensitive and drug-resistant M. tuberculosis strains, respectively. A molecular modeling study showed good interaction of compound 3 with wild-type M. tuberculosis DNA gyrase, suggesting gyrase inhibition as a possible mechanism of action. Compound 3 is a nontoxic combination hybrid that can be utilized as a new scaffold and further optimized to develop new antitubercular agents.

6.
ACS Infect Dis ; 4(2): 158-174, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29260545

RESUMO

Antimicrobial resistance has become a major global concern. Development of novel antimicrobial agents for the treatment of infections caused by multidrug resistant (MDR) pathogens is an urgent priority. Pyrrolobenzodiazepines (PBDs) are a promising class of antibacterial agents initially discovered and isolated from natural sources. Recently, C8-linked PBD biaryl conjugates have been shown to be active against some MDR Gram-positive strains. To explore the role of building block orientations on antibacterial activity and obtain structure activity relationship (SAR) information, four novel structures were synthesized in which the building blocks of previously reported compounds were inverted, and their antibacterial activity was studied. The compounds showed minimum inhibitory concentrations (MICs) in the range of 0.125-32 µg/mL against MDR Gram-positive strains with a bactericidal mode of action. The results showed that a single inversion of amide bonds reduces the activity while the double inversion restores the activity against MDR pathogens. All inverted compounds did not stabilize DNA and lacked eukaryotic toxicity. The compounds inhibit DNA gyrase in vitro, and the most potent compound was equally active against both wild-type and mutant DNA gyrase in a biochemical assay. The observed activity of the compounds against methicillin resistant S. aureus (MRSA) strains with equivalent gyrase mutations is consistent with gyrase inhibition being the mechanism of action in vivo, although this has not been definitively confirmed in whole cells. This conclusion is supported by a molecular modeling study showing interaction of the compounds with wild-type and mutant gyrases. This study provides important SAR information about this new class of antibacterial agents.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Benzodiazepinas/química , Benzodiazepinas/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Pirróis/química , Pirróis/farmacologia , Antibacterianos/efeitos adversos , Benzodiazepinas/efeitos adversos , Linhagem Celular Tumoral , DNA/química , Relação Dose-Resposta a Droga , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Pirróis/efeitos adversos , Relação Estrutura-Atividade
7.
Bioorg Med Chem ; 25(15): 3971-3979, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28600080

RESUMO

A novel series of pyridyl nitrofuranyl isoxazolines were synthesized and evaluated for their antibacterial activity against multiple drug resistant (MDR) Staphylococcus strains. Compounds with piperazine linker between the pyridyl group and isoxazoline ring showed better activity when compared to compounds without the piperazine linker. 3-Pyridyl nitrofuranyl isoxazoline with a piperazine linker was found to be more active than corresponding 2-and 4-pyridyl analogues with MICs in the range of 4-32µg/mL against MDR Staphylococcus strains. The eukaryotic toxicity of the compounds was tested by MTT assay and were found to be non-toxic against both non-tumour lung fibroblast WI-38 and cervical cancer cell line HeLa. The most active pyridyl nitrofuranyl isoxazoline compound showed improved activity against a panel of Staphylococcus strains compared to nitrofuran group containing antibiotic nitrofurantoin.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Nitrofurantoína/química , Oxazóis/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/química , Linhagem Celular Tumoral , Humanos , Testes de Sensibilidade Microbiana , Oxazóis/química , Análise Espectral , Relação Estrutura-Atividade
8.
J Med Chem ; 60(14): 6045-6059, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28650661

RESUMO

A new class of nontoxic triaryl benzimidazole compounds, derived from existing classes of DNA minor groove binders, were designed, synthesized, and evaluated for their antibacterial activity against multidrug resistant (MDR) Gram-positive and Gram-negative species. Molecular modeling experiments suggest that the newly synthesized class cannot be accommodated within the minor groove of DNA due to a change in the shape of the molecules. Compounds 8, 13, and 14 were found to be the most active of the series, with MICs in the range of 0.5-4 µg/mL against the MDR Staphylococci and Enterococci species. Compound 13 showed moderate activity against the MDR Gram-negative strains, with MICs in the range of 16-32 µg/mL. Active compounds showed a bactericidal mode of action, and a mechanistic study suggested the inhibition of bacterial gyrase as the mechanism of action (MOA) of this chemical class. The MOA was further supported by the molecular modeling study.


Assuntos
Antibacterianos/química , Benzimidazóis/química , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Antibacterianos/síntese química , Antibacterianos/farmacologia , Benzimidazóis/síntese química , Benzimidazóis/farmacologia , DNA Girase/metabolismo , Farmacorresistência Bacteriana Múltipla , Células HeLa , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Relação Estrutura-Atividade , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/química , Inibidores da Topoisomerase II/farmacologia
9.
Bioorg Med Chem ; 21(9): 2577-86, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23498917

RESUMO

Strong pharmacological evidences indicate that σ1 receptors are implicated in the pathophysiology of all major CNS disorders. In the last years our research group has conducted extensive studies aimed at discovering novel σ1 ligands and we recently selected (R/S)-RC-33 as a novel potent and selective σ1 receptor agonist. As continuation of our work in this field, here we report our efforts in the development of this new σ1 receptor agonist. Initially, we investigated the binding of (R) and (S) enantiomers of RC-33 to the σ1 receptor by in silico experiments. The close values of the predicted affinity of (R)-RC-33 and (S)-RC-33 for the protein evidenced the non-stereoselective binding of RC-33 to the σ1 receptor; this, in turn, supported further development and characterization of RC-33 in its racemic form. Subsequently, we set-up a scaled-up, optimized synthesis of (R/S)-RC-33 along with some compound characterization data (e.g., solubility in different media and solid state characterization by thermal analysis techniques). Finally, metabolic studies of RC-33 in different biological matrices (e.g., plasma, blood, and hepatic S9 fraction) of different species (e.g., rat, mouse, dog, and human) were performed. (R/S)-RC-33 is generally stable in all examined biological matrices, with the only exception of rat and human liver S9 fractions in the presence of NADPH. In such conditions, the compound is subjected to a relevant oxidative metabolism, with a degradation of approximately 65% in rat and 69% in human. Taken together, our results demonstrated that (R/S)-RC-33 is a highly potent, selective, metabolically stable σ1 agonist, a promising novel neuroprotective drug candidate.


Assuntos
Compostos de Bifenilo/farmacologia , Fator de Crescimento Neural/metabolismo , Neuritos/efeitos dos fármacos , Piperidinas/farmacologia , Receptores sigma/agonistas , Animais , Compostos de Bifenilo/química , Compostos de Bifenilo/metabolismo , Físico-Química , Cães , Humanos , Camundongos , Modelos Moleculares , Estrutura Molecular , Células PC12 , Piperidinas/química , Piperidinas/metabolismo , Ratos , Receptores sigma/metabolismo , Receptor Sigma-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...