Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Antiviral Res ; 222: 105789, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38158129

RESUMEN

The recent pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posed a major threat to global health. Although the World Health Organization ended the public health emergency status, antiviral drugs are needed to address new variants of SARS-CoV-2 and future pandemics. To identify novel broad-spectrum coronavirus drugs, we developed a high-content imaging platform compatible with high-throughput screening. The platform is broadly applicable as it can be adapted to include various cell types, viruses, antibodies, and dyes. We demonstrated that the antiviral activity of compounds against SARS-CoV-2 variants (Omicron BA.5 and Omicron XBB.1.5), SARS-CoV, and human coronavirus 229E could easily be assessed. The inclusion of cellular dyes and immunostaining in combination with in-depth image analysis enabled us to identify compounds that induced undesirable phenotypes in host cells, such as changes in cell morphology or in lysosomal activity. With the platform, we screened ∼900K compounds and triaged hits, thereby identifying potential candidate compounds carrying broad-spectrum activity with limited off-target effects. The flexibility and early-stage identification of compounds with limited host cell effects provided by this high-content imaging platform can facilitate coronavirus drug discovery. We anticipate that its rapid deployability and fast turnaround can also be applied to combat future pandemics.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento/métodos , Colorantes/farmacología , Colorantes/uso terapéutico , Pandemias
2.
Antiviral Res ; 210: 105495, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36567021

RESUMEN

While progress has been made in fighting diseases disproportionally affecting underserved populations, unmet medical needs persist for many neglected tropical diseases. The World Health Organization has encouraged strong public-private partnerships to address this issue and several public and private organizations have set an example in the past showing a strong commitment to combat these diseases. Pharmaceutical companies are contributing in different ways to address the imbalance in research efforts. With this review, we exemplify the role of a public-private partnership in research and development by the journey of our dengue antiviral molecule that is now in early clinical development. We detail the different steps of drug development and outline the contribution of each partner to this process. Years of intensive collaboration resulted in the identification of two antiviral compounds, JNJ-A07 and JNJ-1802, the latter of which has advanced to clinical development.


Asunto(s)
Dengue , Asociación entre el Sector Público-Privado , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Serogrupo , Industria Farmacéutica , Enfermedades Desatendidas/tratamiento farmacológico , Enfermedades Desatendidas/prevención & control , Dengue/tratamiento farmacológico , Dengue/prevención & control
3.
J Med Chem ; 63(15): 8046-8058, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32407115

RESUMEN

Respiratory syncytial virus (RSV) is a seasonal virus that infects the lungs and airways of 64 million children and adults every year. It is a major cause of acute lower respiratory tract infection and is associated with significant morbidity and mortality. Despite the large medical and economic burden, treatment options for RSV-associated bronchiolitis and pneumonia are limited and mainly consist of supportive care. This publication covers the medicinal chemistry efforts resulting in the identification of JNJ-53718678, an orally bioavailable RSV inhibitor that was shown to be efficacious in a phase 2a challenge study in healthy adult subjects and that is currently being evaluated in hospitalized infants and adults. Cocrystal structures of several new derivatives helped in rationalizing some of the structure-activity relationship (SAR) trends observed.


Asunto(s)
Antivirales/química , Descubrimiento de Drogas/métodos , Imidazolidinas/química , Indoles/química , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Inhibidores de Proteínas Virales de Fusión/química , Administración Oral , Antivirales/administración & dosificación , Cristalografía por Rayos X/métodos , Células HeLa , Humanos , Imidazolidinas/administración & dosificación , Indoles/administración & dosificación , Estructura Secundaria de Proteína , Virus Sincitial Respiratorio Humano/fisiología , Inhibidores de Proteínas Virales de Fusión/administración & dosificación
4.
J Med Chem ; 62(21): 9680-9690, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31647875

RESUMEN

In the search for novel influenza inhibitors we evaluated 7-fluoro-substituted indoles as bioisosteric replacements for the 7-azaindole scaffold of Pimodivir, a PB2 (polymerase basic protein 2) inhibitor currently in clinical development. Specifically, a 5,7-difluoroindole derivative 11a was identified as a potent and metabolically stable influenza inhibitor. 11a demonstrated a favorable oral pharmacokinetic profile and in vivo efficacy in mice. In addition, it was found that 11a was not at risk of metabolism via aldehyde oxidase, an advantage over previously described inhibitors of this class. The crystal structure of 11a bound to influenza A PB2 cap region is disclosed here and deposited to the PDB.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Diseño de Fármacos , Indoles/síntesis química , Indoles/farmacología , Proteínas Virales/efectos de los fármacos , Células A549 , Animales , Antivirales/química , Antivirales/farmacocinética , Cristalografía por Rayos X , Perros , Humanos , Indoles/química , Indoles/farmacocinética , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Células de Riñón Canino Madin Darby , Pruebas de Sensibilidad Microbiana , Estructura Molecular
5.
J Infect Dis ; 218(5): 748-756, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29684148

RESUMEN

Background: Respiratory syncytial virus (RSV) disease has no effective treatment. JNJ-53718678 is a fusion inhibitor with selective activity against RSV. Methods: After confirmation of RSV infection or 5 days after inoculation with RSV, participants (n = 69) were randomized to JNJ-53718678 75 mg (n = 15), 200 mg (n = 17), 500 mg (n = 18), or placebo (n = 17) orally once daily for 7 days. Antiviral effects were evaluated by assessing RSV RNA viral load (VL) area under the curve (AUC) from baseline (before the first dose) until discharge, time-to-peak VL, duration of viral shedding, clinical symptoms, and quantity of nasal secretions. Results: Mean VL AUC was lower for individuals treated with different doses of JNJ-53718678 versus placebo (203.8-253.8 vs 432.8 log10 PFUe.hour/mL). Also, mean peak VL, time to peak VL, duration of viral shedding, mean overall symptom score, and nasal secretion weight were lower in each JNJ-53718678-treated group versus placebo. No clear exposure-response relationship was observed. Three participants discontinued due to treatment-emergent adverse events of grade 2 and 1 electrocardiogram change (JNJ-53718678 75 mg and 200 mg, respectively) and grade 2 urticaria (placebo). Conclusions: JNJ-53718678 at all 3 doses substantially reduced VL and clinical disease severity, thus establishing clinical proof of concept and the compound's potential as a novel RSV treatment. Clinical trials registration: ClinicalTrials.gov: NCT02387606; EudraCT number: 2014-005041-41.


Asunto(s)
Antivirales/administración & dosificación , Imidazolidinas/administración & dosificación , Indoles/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Administración Oral , Adolescente , Adulto , Antivirales/farmacología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Voluntarios Sanos , Humanos , Imidazolidinas/farmacología , Indoles/farmacología , Masculino , Persona de Mediana Edad , Placebos/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Resultado del Tratamiento , Carga Viral , Esparcimiento de Virus , Adulto Joven
6.
Nat Commun ; 8(1): 167, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28761099

RESUMEN

Respiratory syncytial virus is a major cause of acute lower respiratory tract infection in young children, immunocompromised adults, and the elderly. Intervention with small-molecule antivirals specific for respiratory syncytial virus presents an important therapeutic opportunity, but no such compounds are approved today. Here we report the structure of JNJ-53718678 bound to respiratory syncytial virus fusion (F) protein in its prefusion conformation, and we show that the potent nanomolar activity of JNJ-53718678, as well as the preliminary structure-activity relationship and the pharmaceutical optimization strategy of the series, are consistent with the binding mode of JNJ-53718678 and other respiratory syncytial virus fusion inhibitors. Oral treatment of neonatal lambs with JNJ-53718678, or with an equally active close analog, efficiently inhibits established acute lower respiratory tract infection in the animals, even when treatment is delayed until external signs of respiratory syncytial virus illness have become visible. Together, these data suggest that JNJ-53718678 is a promising candidate for further development as a potential therapeutic in patients at risk to develop respiratory syncytial virus acute lower respiratory tract infection.Respiratory syncytial virus causes lung infections in children, immunocompromised adults, and in the elderly. Here the authors show that a chemical inhibitor to a viral fusion protein is effective in reducing viral titre and ameliorating infection in rodents and neonatal lambs.


Asunto(s)
Imidazolidinas/metabolismo , Indoles/metabolismo , Virus Sincitial Respiratorio Humano/metabolismo , Inhibidores de Proteínas Virales de Fusión/metabolismo , Proteínas Virales de Fusión/metabolismo , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Chlorocebus aethiops , Células Epiteliales , Humanos , Imidazolidinas/farmacología , Imidazolidinas/uso terapéutico , Indoles/farmacología , Indoles/uso terapéutico , Estructura Molecular , Neumonía Viral/tratamiento farmacológico , Ratas , Mucosa Respiratoria/citología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/metabolismo , Ovinos , Relación Estructura-Actividad , Células Vero , Inhibidores de Proteínas Virales de Fusión/farmacología , Inhibidores de Proteínas Virales de Fusión/uso terapéutico
7.
mBio ; 8(2)2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28400527

RESUMEN

Drug-resistant mycobacterial infections are a serious global health challenge, leading to high mortality and socioeconomic burdens in developing countries worldwide. New innovative approaches, from identification of new targets to discovery of novel chemical scaffolds, are urgently needed. Recently, energy metabolism in mycobacteria, in particular the oxidative phosphorylation pathway, has emerged as an object of intense microbiological investigation and as a novel target pathway in drug discovery. New classes of antibacterials interfering with elements of the oxidative phosphorylation pathway are highly active in combating dormant or latent mycobacterial infections, with a promise of shortening tuberculosis chemotherapy. The regulatory approval of the ATP synthase inhibitor bedaquiline and the discovery of Q203, a candidate drug targeting the cytochrome bc1 complex, have highlighted the central importance of this new target pathway. In this review, we discuss key features and potential applications of inhibiting energy metabolism in our quest for discovering potent novel and sterilizing drug combinations for combating tuberculosis. We believe that the combination of drugs targeting elements of the oxidative phosphorylation pathway can lead to a completely new regimen for drug-susceptible and multidrug-resistant tuberculosis.


Asunto(s)
Antituberculosos/farmacología , Descubrimiento de Drogas/tendencias , Metabolismo Energético/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/metabolismo , Fosforilación Oxidativa/efectos de los fármacos
8.
Nat Chem Biol ; 12(2): 87-93, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26641933

RESUMEN

Respiratory syncytial virus (RSV) is a leading cause of pneumonia and bronchiolitis in young children and the elderly. Therapeutic small molecules have been developed that bind the RSV F glycoprotein and inhibit membrane fusion, yet their binding sites and molecular mechanisms of action remain largely unknown. Here we show that these inhibitors bind to a three-fold-symmetric pocket within the central cavity of the metastable prefusion conformation of RSV F. Inhibitor binding stabilizes this conformation by tethering two regions that must undergo a structural rearrangement to facilitate membrane fusion. Inhibitor-escape mutations occur in residues that directly contact the inhibitors or are involved in the conformational rearrangements required to accommodate inhibitor binding. Resistant viruses do not propagate as well as wild-type RSV in vitro, indicating a fitness cost for inhibitor escape. Collectively, these findings provide new insight into class I viral fusion proteins and should facilitate development of optimal RSV fusion inhibitors.


Asunto(s)
Antivirales/farmacología , Modelos Moleculares , Virus Sincitiales Respiratorios/efectos de los fármacos , Proteínas Virales de Fusión/antagonistas & inhibidores , Antivirales/química , Bioensayo , Colorimetría , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Sci Adv ; 1(4): e1500106, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26601184

RESUMEN

Multidrug-resistant tuberculosis (MDR-TB) is more prevalent today than at any other time in human history. Bedaquiline (BDQ), a novel Mycobacterium-specific adenosine triphosphate (ATP) synthase inhibitor, is the first drug in the last 40 years to be approved for the treatment of MDR-TB. This bactericidal compound targets the membrane-embedded rotor (c-ring) of the mycobacterial ATP synthase, a key metabolic enzyme required for ATP generation. We report the x-ray crystal structures of a mycobacterial c9 ring without and with BDQ bound at 1.55- and 1.7-Å resolution, respectively. The structures and supporting functional assays reveal how BDQ specifically interacts with the rotor ring via numerous interactions and thereby completely covers the c-ring's ion-binding sites. This prevents the rotor ring from acting as an ion shuttle and stalls ATP synthase operation. The structures explain how diarylquinoline chemicals specifically inhibit the mycobacterial ATP synthase and thus enable structure-based drug design of next-generation ATP synthase inhibitors against Mycobacterium tuberculosis and other bacterial pathogens.

10.
PLoS One ; 10(5): e0126959, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26010881

RESUMEN

BACKGROUND: The study assessed the antiviral activity of TMC353121, a respiratory syncytial virus (RSV) fusion inhibitor, in a preclinical non-human primate challenge model with a viral shedding pattern similar to that seen in humans, following continuous infusion (CI). METHODS: African green monkeys were administered TMC353121 through CI, in 2 studies. Study 1 evaluated the prophylactic and therapeutic efficacy of TMC353121 at a target plasma level of 50 ng/mL (n=15; Group 1: prophylactic arm [Px50], 0.033 mg/mL TMC353121, flow rate 2.5 mL/kg/h from 24 hours pre-infection to 10 days; Group 2: therapeutic arm [Tx50], 0.033 mg/mL TMC353121 from 24 hours postinfection to 8 days; Group 3: control [Vh1] vehicle, 24 hours post-infection to 8 days). Study 2 evaluated the prophylactic efficacy of TMC353121 at target plasma levels of 5 and 500 ng/mL (n=12; Group 1: prophylactic 5 arm [Px5], 0.0033 mg/mL TMC353121, flow rate 2.5 mL/kg/h from 72 hours pre-infection to 14 days; Group 2: prophylactic 500 arm [Px500], 0.33 mg/mL TMC353121; Group 3: control [Vh2] vehicle, 14 days). Bronchoalveolar lavage fluid and plasma were collected every 2 days from day 1 postinfection for pharmacokinetics and safety analysis. FINDINGS: TMC353121 showed a dose-dependent antiviral activity, varying from 1 log10 reduction of peak viral load to complete inhibition of the RSV replication. Complete inhibition of RSV shedding was observed for a relatively low plasma exposure (0.39 µg/mL) and was associated with a dose-dependent reduction in INFγ, IL6 and MIP1α. TMC353121 administered as CI for 16 days was generally well-tolerated. CONCLUSION: TMC353121 exerted dose-dependent antiviral effect ranging from full inhibition to absence of antiviral activity, in a preclinical model highly permissive for RSV replication. No new safety findings emerged from the study.


Asunto(s)
Antivirales/farmacología , Bencimidazoles/farmacología , Piridinas/farmacología , Virus Sincitiales Respiratorios/fisiología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/farmacocinética , Área Bajo la Curva , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacocinética , Líquido del Lavado Bronquioalveolar/virología , Chlorocebus aethiops , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Piridinas/administración & dosificación , Piridinas/farmacocinética , Virus Sincitiales Respiratorios/efectos de los fármacos , Factores de Tiempo , Esparcimiento de Virus/efectos de los fármacos
11.
Sci Rep ; 5: 10333, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26015371

RESUMEN

Targeting respiration and ATP synthesis has received strong interest as a new strategy for combatting drug-resistant Mycobacterium tuberculosis. Mycobacteria employ a respiratory chain terminating with two branches. One of the branches includes a cytochrome bc1 complex and an aa3-type cytochrome c oxidase while the other branch terminates with a cytochrome bd-type quinol oxidase. In this communication we show that genetic inactivation of cytochrome bd, but not of cytochrome bc1, enhances the susceptibility of Mycobacterium smegmatis to hydrogen peroxide and antibiotic-induced stress. The type-II NADH dehydrogenase effector clofazimine and the ATP synthase inhibitor bedaquiline were bacteriostatic against wild-type M. smegmatis, but strongly bactericidal against a cytochrome bd mutant. We also demonstrated that the quinone-analog aurachin D inhibited mycobacterial cytochrome bd at sub-micromolar concentrations. Our results identify cytochrome bd as a key survival factor in M. smegmatis during antibiotic stress. Targeting the cytochrome bd respiratory branch therefore appears to be a promising strategy that may enhance the bactericidal activity of existing tuberculosis drugs.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Peróxido de Hidrógeno/toxicidad , Mycobacterium smegmatis/efectos de los fármacos , Complejos de ATP Sintetasa/antagonistas & inhibidores , Complejos de ATP Sintetasa/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Clofazimina/farmacología , Diarilquinolinas/farmacología , Complejo IV de Transporte de Electrones/genética , Técnicas de Inactivación de Genes , Mutación , Mycobacterium smegmatis/enzimología , NADH Deshidrogenasa/química , NADH Deshidrogenasa/metabolismo , Quinolonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico
12.
J Antimicrob Chemother ; 70(7): 2028-37, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25754998

RESUMEN

OBJECTIVES: It is not fully understood why inhibiting ATP synthesis in Mycobacterium species leads to death in non-replicating cells. We investigated the bactericidal mode of action of the anti-tubercular F1Fo-ATP synthase inhibitor bedaquiline (Sirturo™) in order to further understand the lethality of ATP synthase inhibition. METHODS: Mycobacterium smegmatis strains were used for all the experiments. Growth and survival during a bedaquiline challenge were performed in multiple media types. A time-course microarray was performed during initial bedaquiline challenge in minimal medium. Oxygen consumption and proton-motive force measurements were performed on whole cells and inverted membrane vesicles, respectively. RESULTS: A killing of 3 log10 cfu/mL was achieved 4-fold more quickly in minimal medium (a glycerol carbon source) versus rich medium (LB with Tween 80) during bedaquiline challenge. Assessing the accelerated killing condition, we identified a transcriptional remodelling of metabolism that was consistent with respiratory dysfunction but inconsistent with ATP depletion. In glycerol-energized cell suspensions, bedaquiline caused an immediate 2.3-fold increase in oxygen consumption. Bedaquiline collapsed the transmembrane pH gradient, but not the membrane potential, in a dose-dependent manner. Both these effects were dependent on binding to the F1Fo-ATP synthase. CONCLUSIONS: Challenge with bedaquiline results in an electroneutral uncoupling of respiration-driven ATP synthesis. This may be a determinant of the bactericidal effects of bedaquiline, while ATP depletion may be a determinant of its delayed onset of killing. We propose that bedaquiline binds to and perturbs the a-c subunit interface of the Fo, leading to futile proton cycling, which is known to be lethal to mycobacteria.


Asunto(s)
Antituberculosos/farmacología , Diarilquinolinas/farmacología , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/fisiología , Desacopladores/farmacología , Medios de Cultivo/química , Perfilación de la Expresión Génica , Humanos , Análisis por Micromatrices , Técnicas Microbiológicas
13.
PLoS One ; 9(7): e102135, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25010492

RESUMEN

Bedaquiline (BDQ), an ATP synthase inhibitor, is the first drug to be approved for treatment of multi-drug resistant tuberculosis in decades. In vitro resistance to BDQ was previously shown to be due to target-based mutations. Here we report that non-target based resistance to BDQ, and cross-resistance to clofazimine (CFZ), is due to mutations in Rv0678, a transcriptional repressor of the genes encoding the MmpS5-MmpL5 efflux pump. Efflux-based resistance was identified in paired isolates from patients treated with BDQ, as well as in mice, in which it was confirmed to decrease bactericidal efficacy. The efflux inhibitors verapamil and reserpine decreased the minimum inhibitory concentrations of BDQ and CFZ in vitro, but verapamil failed to increase the bactericidal effect of BDQ in mice and was unable to reverse efflux-based resistance in vivo. Cross-resistance between BDQ and CFZ may have important clinical implications.


Asunto(s)
Diarilquinolinas/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Animales , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Clofazimina/farmacología , Diarilquinolinas/uso terapéutico , Genes Bacterianos , Aptitud Genética , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Biológicos , Datos de Secuencia Molecular , Mutación/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/aislamiento & purificación , Reserpina/farmacología , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Regulación hacia Arriba/efectos de los fármacos , Verapamilo/farmacología
15.
Nat Commun ; 5: 3369, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24569628

RESUMEN

Bedaquiline (BDQ), an ATP synthase inhibitor, is the first drug to be approved for treatment of multidrug-resistant tuberculosis in decades. Though BDQ has shown excellent efficacy in clinical trials, its early bactericidal activity during the first week of chemotherapy is minimal. Here, using microfluidic devices and time-lapse microscopy of Mycobacterium tuberculosis, we confirm the absence of significant bacteriolytic activity during the first 3-4 days of exposure to BDQ. BDQ-induced inhibition of ATP synthesis leads to bacteriostasis within hours after drug addition. Transcriptional and proteomic analyses reveal that M. tuberculosis responds to BDQ by induction of the dormancy regulon and activation of ATP-generating pathways, thereby maintaining bacterial viability during initial drug exposure. BDQ-induced bacterial killing is significantly enhanced when the mycobacteria are grown on non-fermentable energy sources such as lipids (impeding ATP synthesis via glycolysis). Our results show that BDQ exposure triggers a metabolic remodelling in mycobacteria, thereby enabling transient bacterial survival.


Asunto(s)
Diarilquinolinas/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Antituberculosos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica/métodos , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/genética , Técnicas Analíticas Microfluídicas , Microscopía Fluorescente , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de la Célula Individual/métodos , Factores de Tiempo , Imagen de Lapso de Tiempo
16.
Drug Discov Today ; 18(5-6): 250-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23032727

RESUMEN

Discovery of new antibacterial agents is crucial to counter the challenge of drug-resistant bacterial infections. In this review we discuss the issue of bacterial metabolic resting states, observed for a variety of pathogenic bacteria, which display low susceptibility for most antibacterials. We present examples of how bacterial metabolic states may be controlled, target pathways may be validated and screening on metabolically resting bacteria can be designed. A deeper understanding of bacterial metabolic states may provide valuable input for the design of efficient screening approaches in the discovery of new antibacterial agents.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Animales , Bacterias/metabolismo , Descubrimiento de Drogas
17.
Antimicrob Agents Chemother ; 56(8): 4131-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22615276

RESUMEN

Emergence of drug-resistant bacteria represents a high, unmet medical need, and discovery of new antibacterials acting on new bacterial targets is strongly needed. ATP synthase has been validated as an antibacterial target in Mycobacterium tuberculosis, where its activity can be specifically blocked by the diarylquinoline TMC207. However, potency of TMC207 is restricted to mycobacteria with little or no effect on the growth of other Gram-positive or Gram-negative bacteria. Here, we identify diarylquinolines with activity against key Gram-positive pathogens, significantly extending the antibacterial spectrum of the diarylquinoline class of drugs. These compounds inhibited growth of Staphylococcus aureus in planktonic state as well as in metabolically resting bacteria grown in a biofilm culture. Furthermore, time-kill experiments showed that the selected hits are rapidly bactericidal. Drug-resistant mutations were mapped to the ATP synthase enzyme, and biochemical analysis as well as drug-target interaction studies reveal ATP synthase as a target for these compounds. Moreover, knockdown of the ATP synthase expression strongly suppressed growth of S. aureus, revealing a crucial role of this target in bacterial growth and metabolism. Our data represent a proof of principle for using the diarylquinoline class of antibacterials in key Gram-positive pathogens. Our results suggest that broadening the antibacterial spectrum for this chemical class is possible without drifting off from the target. Development of the diarylquinolines class may represent a promising strategy for combating Gram-positive pathogens.


Asunto(s)
Complejos de ATP Sintetasa/antagonistas & inhibidores , Antibacterianos/farmacología , Bacterias Grampositivas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Quinolinas/farmacología , Staphylococcus aureus/efectos de los fármacos , Complejos de ATP Sintetasa/genética , Adenosina Trifosfato/biosíntesis , Secuencia de Aminoácidos , Biopelículas/efectos de los fármacos , Línea Celular Tumoral , Farmacorresistencia Bacteriana/genética , Bacterias Grampositivas/crecimiento & desarrollo , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Quinolinas/química , Quinolinas/toxicidad , Alineación de Secuencia , Staphylococcus aureus/crecimiento & desarrollo
18.
PLoS One ; 6(8): e23575, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21858172

RESUMEN

Infections with Mycobacterium tuberculosis are substantially increasing on a worldwide scale and new antibiotics are urgently needed to combat concomitantly emerging drug-resistant mycobacterial strains. The diarylquinoline TMC207 is a highly promising drug candidate for treatment of tuberculosis. This compound kills M. tuberculosis by binding to a new target, mycobacterial ATP synthase. In this study we used biochemical assays and binding studies to characterize the interaction between TMC207 and ATP synthase. We show that TMC207 acts independent of the proton motive force and does not compete with protons for a common binding site. The drug is active on mycobacterial ATP synthesis at neutral and acidic pH with no significant change in affinity between pH 5.25 and pH 7.5, indicating that the protonated form of TMC207 is the active drug entity. The interaction of TMC207 with ATP synthase can be explained by a one-site binding mechanism, the drug molecule thus binds to a defined binding site on ATP synthase. TMC207 affinity for its target decreases with increasing ionic strength, suggesting that electrostatic forces play a significant role in drug binding. Our results are consistent with previous docking studies and provide experimental support for a predicted function of TMC207 in mimicking key residues in the proton transfer chain and blocking rotary movement of subunit c during catalysis. Furthermore, the high affinity of TMC207 at low proton motive force and low pH values may in part explain the exceptional ability of this compound to efficiently kill mycobacteria in different microenvironments.


Asunto(s)
Adenosina Trifosfato/metabolismo , ATPasas de Translocación de Protón Bacterianas/metabolismo , Mycobacterium smegmatis/enzimología , Quinolinas/metabolismo , Adenosina Trifosfato/química , Antituberculosos/química , Antituberculosos/metabolismo , ATPasas de Translocación de Protón Bacterianas/química , ATPasas de Translocación de Protón Bacterianas/genética , Sitios de Unión/genética , Unión Competitiva/efectos de los fármacos , Diarilquinolinas , Relación Dosis-Respuesta a Droga , Concentración de Iones de Hidrógeno , Ionóforos/farmacología , Cinética , Modelos Moleculares , Estructura Molecular , Mutación , Nitrilos/farmacología , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Fuerza Protón-Motriz , Protones , Quinolinas/química , Electricidad Estática , Resonancia por Plasmón de Superficie
19.
Nature ; 469(7331): 483-90, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21270886

RESUMEN

Tuberculosis (TB) is more prevalent in the world today than at any other time in human history. Mycobacterium tuberculosis, the pathogen responsible for TB, uses diverse strategies to survive in a variety of host lesions and to evade immune surveillance. A key question is how robust are our approaches to discovering new TB drugs, and what measures could be taken to reduce the long and protracted clinical development of new drugs. The emergence of multi-drug-resistant strains of M. tuberculosis makes the discovery of new molecular scaffolds a priority, and the current situation even necessitates the re-engineering and repositioning of some old drug families to achieve effective control. Whatever the strategy used, success will depend largely on our proper understanding of the complex interactions between the pathogen and its human host. In this review, we discuss innovations in TB drug discovery and evolving strategies to bring newer agents more quickly to patients.


Asunto(s)
Antituberculosos/uso terapéutico , Descubrimiento de Drogas , Tuberculosis/tratamiento farmacológico , Antituberculosos/química , Antituberculosos/farmacología , Respiración de la Célula/efectos de los fármacos , Ensayos Clínicos como Asunto , Farmacorresistencia Bacteriana , Resistencia a Múltiples Medicamentos , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Mycobacterium tuberculosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
20.
Antimicrob Agents Chemother ; 54(11): 4534-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20823290

RESUMEN

Human respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in infants, young children, elderly persons, and severely immunocompromised patients. Effective postinfection treatments are not widely available, and currently there is no approved vaccine. TMC353121 is a potent RSV fusion inhibitor in vitro, and its ability to reduce viral loads in vivo was demonstrated in cotton rats following prophylactic intravenous administration. Here, the pharmacokinetics of TMC353121 in the cotton rat, which is semipermissive for RSV replication, were further explored to build a pharmacokinetic-pharmacodynamic (PK-PD) model and to estimate the plasma drug levels needed for significant antiviral efficacy. TMC353121 reduced the viral titers in bronchoalveolar lavage fluid in a dose-dependent manner after a single subcutaneous administration and intranasal RSV inoculation 24 h after compound administration. The viral titer reduction and plasma TMC353121 concentration at the time of RSV inoculation were well described using a simple E(max) model with a maximal viral titer reduction (E(max)) of 1.5 log(10). The plasma drug level required to achieve 50% of the E(max) (200 ng/ml) was much higher than the 50% inhibitory concentration observed in vitro in HeLaM cells (0.07 ng/ml). In conclusion, this simple PK-PD approach may be useful in predicting efficacious exposure levels for future RSV inhibitors.


Asunto(s)
Antivirales/farmacocinética , Antivirales/uso terapéutico , Bencimidazoles/farmacocinética , Bencimidazoles/uso terapéutico , Piridinas/farmacocinética , Piridinas/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/efectos de los fármacos , Animales , Antivirales/sangre , Bencimidazoles/sangre , Líquido del Lavado Bronquioalveolar/química , Femenino , Masculino , Piridinas/sangre , Ratas , Ratas Sprague-Dawley , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sigmodontinae
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...