Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Comput Chem ; 45(20): 1762-1778, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38647338

RESUMEN

Protein-ligand binding prediction typically relies on docking methodologies and associated scoring functions to propose the binding mode of a ligand in a biological target. Significant challenges are associated with this approach, including the flexibility of the protein-ligand system, solvent-mediated interactions, and associated entropy changes. In addition, scoring functions are only weakly accurate due to the short time required for calculating enthalpic and entropic binding interactions. The workflow described here attempts to address these limitations by combining supervised molecular dynamics with dynamical averaging quantum mechanics fragment molecular orbital. This combination significantly increased the ability to predict the experimental binding structure of protein-ligand complexes independent from the starting position of the ligands or the binding site conformation. We found that the predictive power could be enhanced by combining the residence time and interaction energies as descriptors in a novel scoring function named the P-score. This is illustrated using six different protein-ligand targets as case studies.


Asunto(s)
Simulación de Dinámica Molecular , Unión Proteica , Proteínas , Ligandos , Proteínas/química , Proteínas/metabolismo , Sitios de Unión , Teoría Cuántica , Termodinámica
3.
Nature ; 560(7717): 192-197, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30046105

RESUMEN

Visceral leishmaniasis causes considerable mortality and morbidity in many parts of the world. There is an urgent need for the development of new, effective treatments for this disease. Here we describe the development of an anti-leishmanial drug-like chemical series based on a pyrazolopyrimidine scaffold. The leading compound from this series (7, DDD853651/GSK3186899) is efficacious in a mouse model of visceral leishmaniasis, has suitable physicochemical, pharmacokinetic and toxicological properties for further development, and has been declared a preclinical candidate. Detailed mode-of-action studies indicate that compounds from this series act principally by inhibiting the parasite cdc-2-related kinase 12 (CRK12), thus defining a druggable target for visceral leishmaniasis.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Leishmania donovani/efectos de los fármacos , Leishmania donovani/enzimología , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/parasitología , Terapia Molecular Dirigida , Pirazoles/farmacología , Pirimidinas/farmacología , Animales , Quinasa 9 Dependiente de la Ciclina/química , Quinasas Ciclina-Dependientes/química , Quinasas Ciclina-Dependientes/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Simulación del Acoplamiento Molecular , Proteoma/efectos de los fármacos , Proteómica , Pirazoles/química , Pirazoles/uso terapéutico , Pirimidinas/química , Pirimidinas/uso terapéutico , Reproducibilidad de los Resultados , Especificidad por Sustrato
4.
Antimicrob Agents Chemother ; 66(1): e0153521, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34606338

RESUMEN

Phenotypic screening identified an arylsulfonamide compound with activity against Trypanosoma cruzi, the causative agent of Chagas' disease. Comprehensive mode of action studies revealed that this compound primarily targets the T. cruzi proteasome, binding at the interface between ß4 and ß5 subunits that catalyze chymotrypsin-like activity. A mutation in the ß5 subunit of the proteasome was associated with resistance to compound 1, while overexpression of this mutated subunit also reduced susceptibility to compound 1. Further genetically engineered and in vitro-selected clones resistant to proteasome inhibitors known to bind at the ß4/ß5 interface were cross-resistant to compound 1. Ubiquitinated proteins were additionally found to accumulate in compound 1-treated epimastigotes. Finally, thermal proteome profiling identified malic enzyme as a secondary target of compound 1, although malic enzyme inhibition was not found to drive potency. These studies identify a novel pharmacophore capable of inhibiting the T. cruzi proteasome that may be exploitable for anti-chagasic drug discovery.


Asunto(s)
Enfermedad de Chagas , Trypanosoma cruzi , Enfermedad de Chagas/tratamiento farmacológico , Descubrimiento de Drogas , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Trypanosoma cruzi/química
5.
Proc Natl Acad Sci U S A ; 116(19): 9318-9323, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30962368

RESUMEN

Visceral leishmaniasis (VL), caused by the protozoan parasites Leishmania donovani and Leishmania infantum, is one of the major parasitic diseases worldwide. There is an urgent need for new drugs to treat VL, because current therapies are unfit for purpose in a resource-poor setting. Here, we describe the development of a preclinical drug candidate, GSK3494245/DDD01305143/compound 8, with potential to treat this neglected tropical disease. The compound series was discovered by repurposing hits from a screen against the related parasite Trypanosoma cruzi Subsequent optimization of the chemical series resulted in the development of a potent cidal compound with activity against a range of clinically relevant L. donovani and L. infantum isolates. Compound 8 demonstrates promising pharmacokinetic properties and impressive in vivo efficacy in our mouse model of infection comparable with those of the current oral antileishmanial miltefosine. Detailed mode of action studies confirm that this compound acts principally by inhibition of the chymotrypsin-like activity catalyzed by the ß5 subunit of the L. donovani proteasome. High-resolution cryo-EM structures of apo and compound 8-bound Leishmania tarentolae 20S proteasome reveal a previously undiscovered inhibitor site that lies between the ß4 and ß5 proteasome subunits. This induced pocket exploits ß4 residues that are divergent between humans and kinetoplastid parasites and is consistent with all of our experimental and mutagenesis data. As a result of these comprehensive studies and due to a favorable developability and safety profile, compound 8 is being advanced toward human clinical trials.


Asunto(s)
Antiprotozoarios/administración & dosificación , Leishmania donovani/efectos de los fármacos , Leishmania infantum/efectos de los fármacos , Leishmaniasis Visceral/diagnóstico por imagen , Inhibidores de Proteasoma/administración & dosificación , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Antiprotozoarios/química , Sitios de Unión , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Leishmania donovani/química , Leishmania donovani/enzimología , Leishmania infantum/química , Leishmania infantum/enzimología , Leishmaniasis Visceral/parasitología , Masculino , Ratones , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/química , Conformación Proteica , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo
6.
J Comput Aided Mol Des ; 35(10): 1025-1036, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34458939

RESUMEN

Water molecules play a crucial role in protein-ligand binding, and many tools exist that aim to predict the position and relative energies of these important, but challenging participants of biomolecular recognition. The available tools are, in general, capable of predicting the location of water molecules. However, predicting the effects of their displacement is still very challenging. In this work, a linear-scaling quantum mechanics-based approach was used to assess water network energetics and the changes in network stability upon ligand structural modifications. This approach offers a valuable way to improve understanding of SAR data and help guide compound design.


Asunto(s)
Proteínas/metabolismo , Termodinámica , Agua/química , Sitios de Unión , Ligandos , Modelos Moleculares , Unión Proteica , Proteínas/química , Agua/metabolismo
7.
Nature ; 522(7556): 315-20, 2015 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-26085270

RESUMEN

There is an urgent need for new drugs to treat malaria, with broad therapeutic potential and novel modes of action, to widen the scope of treatment and to overcome emerging drug resistance. Here we describe the discovery of DDD107498, a compound with a potent and novel spectrum of antimalarial activity against multiple life-cycle stages of the Plasmodium parasite, with good pharmacokinetic properties and an acceptable safety profile. DDD107498 demonstrates potential to address a variety of clinical needs, including single-dose treatment, transmission blocking and chemoprotection. DDD107498 was developed from a screening programme against blood-stage malaria parasites; its molecular target has been identified as translation elongation factor 2 (eEF2), which is responsible for the GTP-dependent translocation of the ribosome along messenger RNA, and is essential for protein synthesis. This discovery of eEF2 as a viable antimalarial drug target opens up new possibilities for drug discovery.


Asunto(s)
Antimaláricos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Malaria/parasitología , Plasmodium/efectos de los fármacos , Plasmodium/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Quinolinas/farmacología , Animales , Antimaláricos/administración & dosificación , Antimaláricos/efectos adversos , Antimaláricos/farmacocinética , Descubrimiento de Drogas , Femenino , Estadios del Ciclo de Vida/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/parasitología , Malaria/tratamiento farmacológico , Masculino , Modelos Moleculares , Factor 2 de Elongación Peptídica/antagonistas & inhibidores , Factor 2 de Elongación Peptídica/metabolismo , Plasmodium/genética , Plasmodium/crecimiento & desarrollo , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/fisiología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Plasmodium vivax/efectos de los fármacos , Plasmodium vivax/metabolismo , Quinolinas/administración & dosificación , Quinolinas/química , Quinolinas/farmacocinética
8.
Proc Natl Acad Sci U S A ; 115(38): 9616-9621, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30185555

RESUMEN

African trypanosomes cause lethal and neglected tropical diseases, known as sleeping sickness in humans and nagana in animals. Current therapies are limited, but fortunately, promising therapies are in advanced clinical and veterinary development, including acoziborole (AN5568 or SCYX-7158) and AN11736, respectively. These benzoxaboroles will likely be key to the World Health Organization's target of disease control by 2030. Their mode of action was previously unknown. We have developed a high-coverage overexpression library and use it here to explore drug mode of action in Trypanosoma brucei Initially, an inhibitor with a known target was used to select for drug resistance and to test massive parallel library screening and genome-wide mapping; this effectively identified the known target and validated the approach. Subsequently, the overexpression screening approach was used to identify the target of the benzoxaboroles, Cleavage and Polyadenylation Specificity Factor 3 (CPSF3, Tb927.4.1340). We validated the CPSF3 endonuclease as the target, using independent overexpression strains. Knockdown provided genetic validation of CPSF3 as essential, and GFP tagging confirmed the expected nuclear localization. Molecular docking and CRISPR-Cas9-based editing demonstrated how acoziborole can specifically block the active site and mRNA processing by parasite, but not host CPSF3. Thus, our findings provide both genetic and chemical validation for CPSF3 as an important drug target in trypanosomes and reveal inhibition of mRNA maturation as the mode of action of the trypanocidal benzoxaboroles. Understanding the mechanism of action of benzoxaborole-based therapies can assist development of improved therapies, as well as the prediction and monitoring of resistance, if or when it arises.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Tripanocidas/farmacología , Trypanosoma brucei brucei/fisiología , Tripanosomiasis Africana/prevención & control , Animales , Benzamidas/farmacología , Benzamidas/uso terapéutico , Compuestos de Boro/farmacología , Compuestos de Boro/uso terapéutico , Sistemas CRISPR-Cas , Núcleo Celular/genética , Núcleo Celular/metabolismo , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Técnicas de Silenciamiento del Gen , Biblioteca de Genes , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Simulación del Acoplamiento Molecular , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , ARN Protozoario/metabolismo , Tripanocidas/uso terapéutico , Trypanosoma brucei brucei/efectos de los fármacos , Tripanosomiasis Africana/transmisión , Tripanosomiasis Africana/veterinaria , Valina/análogos & derivados , Valina/farmacología , Valina/uso terapéutico
9.
Chemistry ; 24(54): 14448-14460, 2018 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-29975429

RESUMEN

A library of mannose- and fucose-based glycomimetics was synthesized and screened in a microarray format against a set of C-type lectin receptors (CLRs) that included DC-SIGN, DC-SIGNR, langerin, and dectin-2. Glycomimetic ligands able to interact with dectin-2 were identified for the first time. Comparative analysis of binding profiles allowed their selectivity against other CLRs to be probed.

11.
Bioorg Med Chem ; 22(15): 4135-50, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24980703

RESUMEN

In the last decade the heat shock protein 90 (Hsp90) has emerged as a major therapeutic target and many efforts have been dedicated to the discovery of Hsp90 inhibitors as new potent anticancer agents. Here we report the identification of a novel class of Hsp90 inhibitors by means of a biophysical FAXS-NMR based screening of a library of fragments. The use of X-ray structure information combined with modeling studies enabled the fragment evolution of the initial triazoloquinazoline hit to a class of compounds with nanomolar potency and drug-like properties suited for further lead optimization.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Quinazolinas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Enlace de Hidrógeno , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Estructura Terciaria de Proteína , Quinazolinas/síntesis química , Quinazolinas/farmacología , Relación Estructura-Actividad
12.
ChemMedChem ; 19(13): e202400025, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38581280

RESUMEN

Identification and assessment of novel targets is essential to combat drug resistance in the treatment of HIV/AIDS. HIV Capsid (HIV-CA), the protein playing a major role in both the early and late stages of the viral life cycle, has emerged as an important target. We have applied an NMR fragment screening platform and identified molecules that bind to the N-terminal domain (NTD) of HIV-CA at a site close to the interface with the C-terminal domain (CTD). Using X-ray crystallography, we have been able to obtain crystal structures to identify the binding mode of these compounds. This allowed for rapid progression of the initial, weak binding, fragment starting points to compounds 37 and 38, which have 19F-pKi values of 5.3 and 5.4 respectively.


Asunto(s)
Fármacos Anti-VIH , Cristalografía por Rayos X , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/síntesis química , Sitios de Unión , Descubrimiento de Drogas , VIH-1/efectos de los fármacos , Unión Proteica , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/antagonistas & inhibidores , Humanos , Estructura Molecular , Modelos Moleculares , Espectroscopía de Resonancia Magnética , Relación Estructura-Actividad
13.
Bioorg Med Chem ; 21(22): 7047-63, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24100158

RESUMEN

Novel small molecule inhibitors of heat shock protein 90 (Hsp90) were discovered with the help of a fragment based drug discovery approach (FBDD) and subsequent optimization with a combination of structure guided design, parallel synthesis and application of medicinal chemistry principles. These efforts led to the identification of compound 18 (NMS-E973), which displayed significant efficacy in a human ovarian A2780 xenograft tumor model, with a mechanism of action confirmed in vivo by typical modulation of known Hsp90 client proteins, and with a favorable pharmacokinetic and safety profile.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Isoxazoles/química , Isoxazoles/farmacología , Animales , Antineoplásicos/uso terapéutico , Sitios de Unión , Biomarcadores de Tumor/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Femenino , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Isoxazoles/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad , Trasplante Heterólogo
14.
ACS Omega ; 8(17): 15083-15098, 2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37151542

RESUMEN

The optimization of compounds' binding affinity for a biological target is a crucial aspect of the drug development process. Being able to accurately predict binding energies in advance of synthesizing compounds would have a massive impact on the speed of the drug discovery process. The ideal binding affinity prediction method should combine accuracy, reliability, and speed. In this paper, we present SophosQM, a quantum mechanics (QM)-based approach, which can accurately predict the binding affinities of compounds to proteins. The binding affinity predictive models generated by SophosQM are based on the fragment molecular orbital (FMO) method to estimate the enthalpic component of the binding free energy, and a macroscopic descriptor, clog P, is used as an approximation of the entropic component. The affinity prediction is performed using multilinear regression, fitting the experimental values against the FMO-computed enthalpic term and clog P. The quality of the prediction can be assessed in terms of the correlation coefficient between experimental and predicted values. In this work, the method's reliability and accuracy are exemplified by applying SophosQM to 70 compounds binding to six different targets of pharmaceutical relevance. Overall, the results show a very satisfactory performance with a global correlation coefficient in the order of 0.9. Our predictions also show a satisfactory performance compared to data based on free energy perturbation. Finally, SophosQM can also be applied in high-throughput mode by using semiempirical QM methods to evaluate large portions of chemical space, while retaining a good level of accuracy, but decreasing the computing time to just a few seconds per compound.

15.
ACS Omega ; 8(14): 12787-12804, 2023 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-37065080

RESUMEN

There is an urgent need for the development of new therapeutics with novel modes of action to target Gram-negative bacterial infections, due to resistance to current drugs. Previously, FabA, an enzyme in the bacterial type II fatty acid biosynthesis pathway, was identified as a potential drug target in Pseudomonas aeruginosa, a Gram-negative bacteria of significant clinical concern. A chemical starting point was also identified. There is a cysteine, Cys15, in the active site of FabA, adjacent to where this compound binds. This paper describes the preparation of analogues containing an electrophilic warhead with the aim of covalent inhibition of the target. A wide variety of analogues were successfully prepared. Unfortunately, these analogues did not increase inhibition, which may be due to a loop within the enzyme partially occluding access to the cysteine.

16.
J Med Chem ; 66(22): 15380-15408, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37948640

RESUMEN

There is an urgent need for new tuberculosis (TB) treatments, with novel modes of action, to reduce the incidence/mortality of TB and to combat resistance to current treatments. Through both chemical and genetic methodologies, polyketide synthase 13 (Pks13) has been validated as essential for mycobacterial survival and as an attractive target for Mycobacterium tuberculosis growth inhibitors. A benzofuran series of inhibitors that targeted the Pks13 thioesterase domain, failed to progress to preclinical development due to concerns over cardiotoxicity. Herein, we report the identification of a novel oxadiazole series of Pks13 inhibitors, derived from a high-throughput screening hit and structure-guided optimization. This new series binds in the Pks13 thioesterase domain, with a distinct binding mode compared to the benzofuran series. Through iterative rounds of design, assisted by structural information, lead compounds were identified with improved antitubercular potencies (MIC < 1 µM) and in vitro ADMET profiles.


Asunto(s)
Benzofuranos , Mycobacterium tuberculosis , Sintasas Poliquetidas , Antituberculosos/química , Mycobacterium tuberculosis/metabolismo , Benzofuranos/química , Pruebas de Sensibilidad Microbiana
17.
J Med Chem ; 66(15): 10413-10431, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37506194

RESUMEN

There is an urgent need for new treatments for Chagas disease, a parasitic infection which mostly impacts South and Central America. We previously reported on the discovery of GSK3494245/DDD01305143, a preclinical candidate for visceral leishmaniasis which acted through inhibition of the Leishmania proteasome. A related analogue, active against Trypanosoma cruzi, showed suboptimal efficacy in an animal model of Chagas disease, so alternative proteasome inhibitors were investigated. Screening a library of phenotypically active analogues against the T. cruzi proteasome identified an active, selective pyridazinone, the development of which is described herein. We obtained a cryo-EM co-structure of proteasome and a key inhibitor and used this to drive optimization of the compounds. Alongside this, optimization of the absorption, distribution, metabolism, and excretion (ADME) properties afforded a suitable compound for mouse efficacy studies. The outcome of these studies is discussed, alongside future plans to further understand the series and its potential to deliver a new treatment for Chagas disease.


Asunto(s)
Enfermedad de Chagas , Leishmaniasis Visceral , Tripanocidas , Trypanosoma cruzi , Ratones , Animales , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Complejo de la Endopetidasa Proteasomal , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Leishmaniasis Visceral/tratamiento farmacológico , Tripanocidas/farmacología , Tripanocidas/uso terapéutico , Tripanocidas/química
18.
Sci Transl Med ; 15(726): eadg8105, 2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-38091410

RESUMEN

Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects millions of people in the Americas and across the world, leading to considerable morbidity and mortality. Current treatment options, benznidazole (BNZ) and nifurtimox, offer limited efficacy and often lead to adverse side effects because of long treatment durations. Better treatment options are therefore urgently required. Here, we describe a pyrrolopyrimidine series, identified through phenotypic screening, that offers an opportunity to improve on current treatments. In vitro cell-based washout assays demonstrate that compounds in the series are incapable of killing all parasites; however, combining these pyrrolopyrimidines with a subefficacious dose of BNZ can clear all parasites in vitro after 5 days. These findings were replicated in a clinically predictive in vivo model of chronic Chagas disease, where 5 days of treatment with the combination was sufficient to prevent parasite relapse. Comprehensive mechanism of action studies, supported by ligand-structure modeling, show that compounds from this pyrrolopyrimidine series inhibit the Qi active site of T. cruzi cytochrome b, part of the cytochrome bc1 complex of the electron transport chain. Knowledge of the molecular target enabled a cascade of assays to be assembled to evaluate selectivity over the human cytochrome b homolog. As a result, a highly selective and efficacious lead compound was identified. The combination of our lead compound with BNZ rapidly clears T. cruzi parasites, both in vitro and in vivo, and shows great potential to overcome key issues associated with currently available treatments.


Asunto(s)
Enfermedad de Chagas , Parásitos , Tripanocidas , Trypanosoma cruzi , Animales , Humanos , Citocromos b , Tripanocidas/efectos adversos , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/inducido químicamente , Enfermedad de Chagas/parasitología
19.
Sci Transl Med ; 15(726): eadh9902, 2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-38091406

RESUMEN

New drugs for visceral leishmaniasis that are safe, low cost, and adapted to the field are urgently required. Despite concerted efforts over the last several years, the number of new chemical entities that are suitable for clinical development for the treatment of Leishmania remains low. Here, we describe the discovery and preclinical development of DNDI-6174, an inhibitor of Leishmania cytochrome bc1 complex activity that originated from a phenotypically identified pyrrolopyrimidine series. This compound fulfills all target candidate profile criteria required for progression into preclinical development. In addition to good metabolic stability and pharmacokinetic properties, DNDI-6174 demonstrates potent in vitro activity against a variety of Leishmania species and can reduce parasite burden in animal models of infection, with the potential to approach sterile cure. No major flags were identified in preliminary safety studies, including an exploratory 14-day toxicology study in the rat. DNDI-6174 is a cytochrome bc1 complex inhibitor with acceptable development properties to enter preclinical development for visceral leishmaniasis.


Asunto(s)
Leishmaniasis Visceral , Leishmaniasis , Ratas , Animales , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/parasitología , Modelos Animales de Enfermedad
20.
J Med Chem ; 65(7): 5606-5624, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35303411

RESUMEN

African animal trypanosomiasis or nagana, caused principally by infection of the protozoan parasites Trypanosoma congolense and Trypanosoma vivax, is a major problem in cattle and other livestocks in sub-Saharan Africa. Current treatments are threatened by the emergence of drug resistance and there is an urgent need for new, effective drugs. Here, we report the repositioning of a compound series initially developed for the treatment of human African trypanosomiasis. A medicinal chemistry program, focused on deriving more soluble analogues, led to development of a lead compound capable of curing cattle infected with both T. congolense and T. vivax via intravenous dosing. Further optimization has the potential to yield a single-dose intramuscular treatment for this disease. Comprehensive mode of action studies revealed that the molecular target of this promising compound and related analogues is the cyclin-dependent kinase CRK12.


Asunto(s)
Trypanosoma congolense , Tripanosomiasis Africana , Animales , Bovinos , Quinasas Ciclina-Dependientes , Reposicionamiento de Medicamentos , Trypanosoma vivax , Tripanosomiasis Africana/tratamiento farmacológico , Tripanosomiasis Africana/parasitología , Tripanosomiasis Africana/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA