Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
bioRxiv ; 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38826291

RESUMEN

PARP1 and PARP2 recognize DNA breaks immediately upon their formation, generate a burst of local PARylation to signal their location, and are co-targeted by all current FDA-approved forms of PARP inhibitors (PARPi) used in the cancer clinic. Recent evidence indicates that the same PARPi molecules impact PARP2 differently from PARP1, raising the possibility that allosteric activation may also differ. We find that unlike for PARP1, destabilization of the autoinhibitory domain of PARP2 is insufficient for DNA damage-induced catalytic activation. Rather, PARP2 activation requires further unfolding of an active site α-helix absent in PARP1. Only one clinical PARPi, Olaparib, stabilizes the PARP2 active site α-helix, representing a structural feature with the potential to discriminate small molecule inhibitors. Collectively, our findings reveal unanticipated differences in local structure and changes in activation-coupled backbone dynamics between PARP1 and PARP2.

2.
Biochem J ; 481(6): 437-460, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38372302

RESUMEN

Catalytic poly(ADP-ribose) production by PARP1 is allosterically activated through interaction with DNA breaks, and PARP inhibitor compounds have the potential to influence PARP1 allostery in addition to preventing catalytic activity. Using the benzimidazole-4-carboxamide pharmacophore present in the first generation PARP1 inhibitor veliparib, a series of 11 derivatives was designed, synthesized, and evaluated as allosteric PARP1 inhibitors, with the premise that bulky substituents would engage the regulatory helical domain (HD) and thereby promote PARP1 retention on DNA breaks. We found that core scaffold modifications could indeed increase PARP1 affinity for DNA; however, the bulk of the modification alone was insufficient to trigger PARP1 allosteric retention on DNA breaks. Rather, compounds eliciting PARP1 retention on DNA breaks were found to be rigidly held in a position that interferes with a specific region of the HD domain, a region that is not targeted by current clinical PARP inhibitors. Collectively, these compounds highlight a unique way to trigger PARP1 retention on DNA breaks and open a path to unveil the pharmacological benefits of such inhibitors with novel properties.


Asunto(s)
Antineoplásicos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Bencimidazoles/farmacología , Reparación del ADN , Roturas del ADN , Daño del ADN
3.
Bioorg Med Chem Lett ; 88: 129288, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37094724

RESUMEN

NIMA Related Kinase 2 (Nek2) kinase is an attractive target for the development of therapeutic agents for several types of highly invasive cancers. Despite this, no small molecule inhibitor has advanced to the late clinical stages thus far. In this work, we have identified a novel spirocyclic inhibitor (V8) of Nek2 kinase, utilizing a high-throughput virtual screening (HTVS) approach. Using recombinant Nek2 enzyme assays, we show that V8 can inhibit Nek2 kinase activity (IC50 = 2.4 ± 0.2 µM) by binding to the enzyme's ATP pocket. The inhibition is selective, reversible and is not time dependent. To understand the key chemotype features responsible for Nek2 inhibition, a detailed structure-activity relationships (SAR) was performed. Using molecular models of the energy-minimized structures of Nek2-inhibitory complexes, we identify key hydrogen-bonding interactions, including two from the hinge-binding region, likely responsible for the observed affinity. Finally, using cell-based studies, we show that V8 attenuates (a) pAkt/PI3 Kinase signaling in a dose-dependent manner, and (b) proliferative and migratory phenotypes of highly aggressive human MDA-MB-231 breast and A549 lung cancer cell lines. Thus, V8 is an important novel lead compound for the development of highly potent and selective Nek2 inhibitory agents.


Asunto(s)
Quinasas Relacionadas con NIMA , Humanos , Línea Celular Tumoral , Neoplasias Pulmonares , Modelos Moleculares , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Fosforilación , Relación Estructura-Actividad
4.
Molecules ; 27(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35056661

RESUMEN

Cell cycle kinases represent an important component of the cell machinery that controls signal transduction involved in cell proliferation, growth, and differentiation. Nek2 is a mitotic Ser/Thr kinase that localizes predominantly to centrosomes and kinetochores and orchestrates centrosome disjunction and faithful chromosomal segregation. Its activity is tightly regulated during the cell cycle with the help of other kinases and phosphatases and via proteasomal degradation. Increased levels of Nek2 kinase can promote centrosome amplification (CA), mitotic defects, chromosome instability (CIN), tumor growth, and cancer metastasis. While it remains a highly attractive target for the development of anti-cancer therapeutics, several new roles of the Nek2 enzyme have recently emerged: these include drug resistance, bone, ciliopathies, immune and kidney diseases, and parasitic diseases such as malaria. Therefore, Nek2 is at the interface of multiple cellular processes and can influence numerous cellular signaling networks. Herein, we provide a critical overview of Nek2 kinase biology and discuss the signaling roles it plays in both normal and diseased human physiology. While the majority of research efforts over the last two decades have focused on the roles of Nek2 kinase in tumor development and cancer metastasis, the signaling mechanisms involving the key players associated with several other notable human diseases are highlighted here. We summarize the efforts made so far to develop Nek2 inhibitory small molecules, illustrate their action modalities, and provide our opinion on the future of Nek2-targeted therapeutics. It is anticipated that the functional inhibition of Nek2 kinase will be a key strategy going forward in drug development, with applications across multiple human diseases.


Asunto(s)
Enfermedades Óseas/patología , Inhibidores Enzimáticos/farmacología , Enfermedades del Sistema Inmune/patología , Enfermedades Renales/patología , Malaria/patología , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Neoplasias/patología , Enfermedades Óseas/tratamiento farmacológico , Enfermedades Óseas/enzimología , Resistencia a Medicamentos , Humanos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Enfermedades del Sistema Inmune/enzimología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/enzimología , Malaria/tratamiento farmacológico , Malaria/enzimología , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología
5.
Nat Cell Biol ; 24(1): 62-73, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35013556

RESUMEN

Poly (ADP-ribose) polymerase (PARP) inhibitors elicit antitumour activity in homologous recombination-defective cancers by trapping PARP1 in a chromatin-bound state. How cells process trapped PARP1 remains unclear. Using wild-type and a trapping-deficient PARP1 mutant combined with rapid immunoprecipitation mass spectrometry of endogenous proteins and Apex2 proximity labelling, we delineated mass spectrometry-based interactomes of trapped and non-trapped PARP1. These analyses identified an interaction between trapped PARP1 and the ubiquitin-regulated p97 ATPase/segregase. We found that following trapping, PARP1 is SUMOylated by PIAS4 and subsequently ubiquitylated by the SUMO-targeted E3 ubiquitin ligase RNF4, events that promote recruitment of p97 and removal of trapped PARP1 from chromatin. Small-molecule p97-complex inhibitors, including a metabolite of the clinically used drug disulfiram (CuET), prolonged PARP1 trapping and enhanced PARP inhibitor-induced cytotoxicity in homologous recombination-defective tumour cells and patient-derived tumour organoids. Together, these results suggest that p97 ATPase plays a key role in the processing of trapped PARP1 and the response of tumour cells to PARP inhibitors.


Asunto(s)
Cromatina/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteína que Contiene Valosina/metabolismo , Línea Celular Tumoral , Disulfiram/análogos & derivados , Disulfiram/farmacología , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Ubiquitinación
7.
Mol Med ; 27(1): 79, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34271850

RESUMEN

BACKGROUND: High mobility group box 1 protein (HMGB1) is an alarmin following its release by immune cells upon cellular activation or stress. High levels of extracellular HMGB1 play a critical role in impairing the clearance of invading pulmonary pathogens and dying neutrophils in the injured lungs of cystic fibrosis (CF) and acute respiratory distress syndrome (ARDS). A heparin derivative, 2-O, 3-O desulfated heparin (ODSH), has been shown to inhibit HMGB1 release from a macrophage cell line and is efficacious in increasing bacterial clearance in a mouse model of pneumonia. Thus, we hypothesized that ODSH can attenuate the bacterial burden and inflammatory lung injury in CF and we conducted experiments to determine the underlying mechanisms. METHODS: We determined the effects of ODSH on lung injury produced by Pseudomonas aeruginosa (PA) infection in CF mice with the transmembrane conductance regulator gene knockout (CFTR-/-). Mice were given ODSH or normal saline intraperitoneally, followed by the determination of the bacterial load and lung injury in the airways and lung tissues. ODSH binding to HMGB1 was determined using surface plasmon resonance and in silico docking analysis of the interaction of the pentasaccharide form of ODSH with HMGB1. RESULTS: CF mice given 25 mg/kg i.p. of ODSH had significantly lower PA-induced lung injury compared to mice given vehicle alone. The CF mice infected with PA had decreased levels of nitric oxide (NO), increased levels of airway HMGB1 and HMGB1-impaired macrophage phagocytic function. ODSH partially attenuated the PA-induced alteration in the levels of NO and airway HMGB1 in CF mice. In addition, ODSH reversed HMGB1-impaired macrophage phagocytic function. These effects of ODSH subsequently decreased the bacterial burden in the CF lungs. In a surface plasmon resonance assay, ODSH interacted with HMGB1 with high affinity (KD = 3.89 × 10-8 M) and induced conformational changes that may decrease HMGB1's binding to its membrane receptors, thus attenuating HMGB1-induced macrophage dysfunction. CONCLUSIONS: The results suggest that ODSH can significantly decrease bacterial infection-induced lung injury in CF mice by decreasing both HMGB1-mediated impairment of macrophage function and the interaction of HMGB1 with membrane receptors. Thus, ODSH could represent a novel approach for treating CF and ARDS patients that have HMGB1-mediated lung injury.


Asunto(s)
Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Proteína HMGB1/genética , Heparina/análogos & derivados , Macrófagos/inmunología , Macrófagos/metabolismo , Neumonía Bacteriana/etiología , Neumonía Bacteriana/metabolismo , Animales , Carga Bacteriana , Biomarcadores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Proteína HMGB1/química , Proteína HMGB1/metabolismo , Heparina/química , Heparina/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Óxido Nítrico/metabolismo , Fagocitosis/inmunología , Neumonía Bacteriana/patología , Unión Proteica , Células RAW 264.7 , Relación Estructura-Actividad
8.
Expert Opin Ther Pat ; 31(7): 609-623, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33554679

RESUMEN

INTRODUCTION: Discovery of small molecules that impede the activity of single-strand DNA repair enzyme, PARP1, has led to four marketed drugs for the treatment of advanced-stage cancers. Hence, there is a renewed enthusiasm in the PARP inhibitor discovery arena. To reduce nonspecific interactions or potential toxicities, and to understand the role of other minimally explored PARP enzymes, exciting new findings have emerged toward the development of selective inhibitors and targeted chemical biology probes. Importantly, the conventional PARP inhibitor design has evolved in a way that could potentially lead to multienzyme-targeting - a polypharmacological approach against aggressive cancers. AREAS COVERED: This review comprises recent progress made in the development of PARP inhibitors, primarily focused on human cancers. Discovery of novel PARP inhibitors with pan, selective, and multi-target inhibition using in vitro and in vivo cancer models is summarized and critically evaluated. Emphasis is given to patents published during 2016-2020, excluding TNKS 1/2 inhibitors. EXPERT OPINION: The outstanding success demonstrated by the FDA approved PARP inhibitors has fueled further clinical evaluations for expansion of their clinical utilities. The current clinical investigations include new candidates as well as marketed PARP-targeted drugs, both as single agents and in combination with other chemotherapeutics. Recent advances have also unveiled critical roles of other PARPs in oncogenic signal transduction, in addition to those of the well-documented PARP1/2 and TNKS1/2 enzymes. Further studies on lesser-known PARP members are urgently needed for functional annotations and for understanding their roles in cancer progression and other human diseases.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Diseño de Fármacos , Desarrollo de Medicamentos , Descubrimiento de Drogas , Humanos , Neoplasias/patología , Patentes como Asunto , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores
9.
J Med Chem ; 63(22): 13291-13315, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-32805118

RESUMEN

A quaternary carbon bears four other carbon substituents or combination of four non-hydrogen substituents at four vertices of a tetrahedron. The spirocyclic quaternary carbon positioned at the center of a bioactive molecule offers conformational rigidity, which in turn reduces the penalty for conformational entropy. The quaternary carbon is a predominant feature of natural product structures and has been associated with more effective and selective binding to target proteins compared to planar compounds with a high sp2 count. The presence of a quaternary carbon stereocenter allows the exploration of novel chemical space to obtain new molecules with enhanced three-dimensionality. These characteristics, coupled to an increasing awareness to develop sp3-rich molecules, boosted utility of quaternary carbon stereocenters in bioactive compounds. It is hoped that this Perspective will inspire the chemist to utilize quaternary carbon stereocenters to enhance potency, selectivity, and other drug-like properties.


Asunto(s)
Carbono/química , Diseño de Fármacos , Descubrimiento de Drogas/métodos , Conformación Molecular , Animales , Descubrimiento de Drogas/tendencias , Humanos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/química , Estereoisomerismo
10.
Bioorg Chem ; 102: 104075, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32777641

RESUMEN

Poly(ADP-ribose) polymerase 1 (PARP1), a widely explored anticancer drug target, plays an important role in single-strand DNA break repair processes. High-throughput virtual screening (HTVS) of a Maybridge small molecule library using the PARP1-benzimidazole-4-carboxamide co-crystal structure and pharmacophore model led to the identification of eleven compounds. These compounds were evaluated using recombinant PARP1 enzyme assay that resulted in the acquisition of three PARP1 inhibitors: 3 (IC50 = 12 µM), 4 (IC50 = 5.8 µM), and 10 (IC50 = 0.88 µM). Compound 4 (2,3-dihydro-1,4-benzodioxine-5-carboxamide) was selected as a lead and was subjected to further chemical modifications, involving analogue synthesis and scaffold hopping. These efforts led to the identification of (Z)-2-(4-hydroxybenzylidene)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-8-carboxamide (49, IC50 = 0.082 µM) as the most potent inhibitor of PARP1 from the series.


Asunto(s)
Dioxinas/síntesis química , Dioxinas/uso terapéutico , Ensayos Analíticos de Alto Rendimiento/métodos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/síntesis química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Dioxinas/farmacología , Humanos , Simulación del Acoplamiento Molecular , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Relación Estructura-Actividad
11.
Science ; 368(6486)2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32241924

RESUMEN

The success of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors (PARPi) to treat cancer relates to their ability to trap PARP-1 at the site of a DNA break. Although different forms of PARPi all target the catalytic center of the enzyme, they have variable abilities to trap PARP-1. We found that several structurally distinct PARPi drive PARP-1 allostery to promote release from a DNA break. Other inhibitors drive allostery to retain PARP-1 on a DNA break. Further, we generated a new PARPi compound, converting an allosteric pro-release compound to a pro-retention compound and increasing its ability to kill cancer cells. These developments are pertinent to clinical applications where PARP-1 trapping is either desirable or undesirable.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Roturas del ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Neoplasias/enzimología , Poli(ADP-Ribosa) Polimerasa-1/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Bencimidazoles/química , Bencimidazoles/farmacología , Línea Celular Tumoral , Humanos , Isoindoles/química , Isoindoles/farmacología , Piperazinas/química , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Dominios Proteicos
12.
J Med Chem ; 63(11): 5625-5663, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32031378

RESUMEN

The use of an acetylene (ethynyl) group in medicinal chemistry coincides with the launch of the Journal of Medicinal Chemistry in 1959. Since then, the acetylene group has been broadly exploited in drug discovery and development. As a result, it has become recognized as a privileged structural feature for targeting a wide range of therapeutic target proteins, including MAO, tyrosine kinases, BACE1, steroid receptors, mGlu5 receptors, FFA1/GPR40, and HIV-1 RT. Furthermore, a terminal alkyne functionality is frequently introduced in chemical biology probes as a click handle to identify molecular targets and to assess target engagement. This Perspective is divided into three parts encompassing: (1) the physicochemical properties of the ethynyl group, (2) the advantages and disadvantages of the ethynyl group in medicinal chemistry, and (3) the impact of the ethynyl group on chemical biology approaches.


Asunto(s)
Acetileno/química , Química Farmacéutica , Alquinos/química , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Química Clic , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Transcriptasa Inversa del VIH/metabolismo , VIH-1/enzimología , Semivida , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Ratas , Inhibidores de la Transcriptasa Inversa/química
13.
Biochimie ; 170: 65-72, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31862235

RESUMEN

We propose a new model for prochirality that satisfies all known examples: the prochiral plane. This plane contains the prochiral carbon and defines two separate faces for chemical modification. We extend this to enzyme catalysis, replacing the "three point attachment" hypothesis and its variants. Once a prochiral substrate is fixed on an enzyme surface, the asymmetry of the enzyme provides reactants exclusively on one side of the prochiral plane, producing an enantiomerically pure chiral product. The aconitase reaction is detailed as an example, using molecular modeling and its known enzymatic mechanism. We show that the prochiral substrate for this enzyme is not citrate, but rather cis-aconitate. The number of interaction points of cis-aconitate is not relevant to prochirality, but rather to substrate specificity. A second detailed example is the enzyme fumarase; here the substrate fumarate has only two binding sites, but is nonetheless fixed onto the enzyme and has a defined prochiral plane. We also provide a literature survey of more prochiral substrates, all of which have sp2 hybridized carbon and contain a prochiral plane. An example of a prochiral unnatural substrate for sphingosine kinase 2, fingolimod, has an sp3 hybridized prochiral carbon and also contains a prochiral plane. Finally, we provide an intuitive example of a prochiral physical object, a coffee cup, interacting with one hand and lip.


Asunto(s)
Aconitato Hidratasa/química , Aconitato Hidratasa/metabolismo , Ácido Aconítico/metabolismo , Citratos/metabolismo , Fumarato Hidratasa/química , Fumarato Hidratasa/metabolismo , Mycobacterium tuberculosis/enzimología , Sitios de Unión , Catálisis , Modelos Moleculares , Estereoisomerismo , Especificidad por Sustrato
14.
J Med Chem ; 62(21): 9772-9791, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31584822

RESUMEN

Clostridium difficile infection (CDI) is the leading cause of healthcare-associated infection in the United States. Therefore, development of novel treatments for CDI is a high priority. Toward this goal, we began in vitro screening of a structurally diverse in-house library of 67 compounds against two pathogenic C. difficile strains (ATCC BAA 1870 and ATCC 43255), which yielded a hit compound, 2-methyl-8-nitroquinazolin-4(3H)-one (2) with moderate potency (MIC = 312/156 µM). Optimization of 2 gave lead compound 6a (2-methyl-7-nitrothieno[3,2-d]pyrimidin-4(3H)-one) with improved potency (MIC = 19/38 µM), selectivity over normal gut microflora, CC50s > 606 µM against mammalian cell lines, and acceptable stability in simulated gastric and intestinal fluid. Further optimization of 6a at C2-, N3-, C4-, and C7-positions resulted in a library of >50 compounds with MICs ranging from 3 to 800 µM against clinical isolates of C. difficile. Compound 8f (MIC = 3/6 µM) was identified as a promising lead for further optimization.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Clostridioides difficile/efectos de los fármacos , Pirimidinas/química , Pirimidinas/farmacología , Animales , Antibacterianos/toxicidad , Células CACO-2 , Chlorocebus aethiops , Diseño de Fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación Molecular , Pirimidinas/toxicidad , Estereoisomerismo , Relación Estructura-Actividad , Células Vero
15.
J Med Chem ; 62(11): 5330-5357, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31042381

RESUMEN

Poly(adenosine 5'-diphosphate-ribose) polymerase (PARP) inhibitors are a class of anticancer drugs that block the catalytic activity of PARP proteins. Optimization of our lead compound 1 (( Z)-2-benzylidene-3-oxo-2,3-dihydrobenzofuran-7-carboxamide; PARP-1 IC50 = 434 nM) led to a tetrazolyl analogue (51, IC50 = 35 nM) with improved inhibition. Isosteric replacement of the tetrazole ring with a carboxyl group (60, IC50 = 68 nM) gave a promising new lead, which was subsequently optimized to obtain analogues with potent PARP-1 IC50 values (4-197 nM). PARP enzyme profiling revealed that the majority of compounds are selective toward PARP-2 with IC50 values comparable to clinical inhibitors. X-ray crystal structures of the key inhibitors bound to PARP-1 illustrated the mode of interaction with analogue appendages extending toward the PARP-1 adenosine-binding pocket. Compound 81, an isoform-selective PARP-1/-2 (IC50 = 30 nM/2 nM) inhibitor, demonstrated selective cytotoxic effect toward breast cancer gene 1 ( BRCA1)-deficient cells compared to isogenic BRCA1-proficient cells.


Asunto(s)
Adenosina/metabolismo , Benzofuranos/síntesis química , Benzofuranos/farmacología , Diseño de Fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/síntesis química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Secuencias de Aminoácidos , Benzofuranos/química , Benzofuranos/metabolismo , Biocatálisis , Línea Celular Tumoral , Técnicas de Química Sintética , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/química , Relación Estructura-Actividad
16.
J Med Chem ; 61(6): 2166-2210, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28850227

RESUMEN

The gem-dimethyl moiety is a structural feature frequently found in many natural products of clinical interest, including, but not limited to, taxanes, epothilones, statins, retinoids, di-/triterpenes, noviose deoxysugar, and antibiotics derived from ß-lactams, macrolides, and aminocoumarins. Inspired by this time-tested moiety, medicinal chemists have widely explored its use in developing bioactive molecules because of the possibility to (1) increase target engagement, potency, and selectivity through van der Waals interactions and entropically favorable restriction to a bioactive conformation, (2) mitigate toxicity, (3) obtain superior DMPK profile, (4) modulate the p Ka of nearby functionality, (5) induce symmetry into a monomethyl substituted chiral center, and (6) apply the Thorpe-Ingold conformational effect in an o-hydroxydihydrocinnamic acid based prodrug design. The aim of this Perspective is to illustrate how medicinal chemists have elegantly employed the gem-dimethyl group to obtain clinically useful drugs and to provide synthetic methods to install a gem-dimethyl group.


Asunto(s)
Productos Biológicos/química , Productos Biológicos/farmacología , Química Farmacéutica , Animales , Humanos
17.
J Med Chem ; 61(3): 834-864, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29251928

RESUMEN

A novel set of 64 analogues based on our lead compound 1 was designed and synthesized with an initial objective of understanding the structural requirements of ligands binding to a highly perplexing substrate-binding site of P-glycoprotein (P-gp) and their effect on modulating the ATPase function of the efflux pump. Compound 1, a stimulator of P-gp ATPase activity, was transformed to ATPase inhibitory compounds 39, 53, and 109. The ATPase inhibition by these compounds was predominantly contributed by the presence of a cyclohexyl group in lieu of the 2-aminobenzophenone moiety of 1. The 4,4-difluorocyclohexyl analogues, 53 and 109, inhibited the photolabeling by [125I]-IAAP, with IC50 values of 0.1 and 0.76 µM, respectively. Selected compounds were shown to reverse paclitaxel resistance in HEK293 cells overexpressing P-gp and were selective toward P-gp over CYP3A4. Induced-fit docking highlighted a plausible binding pattern of inhibitory compounds in the putative-binding pocket of P-gp. The current study underscores the stringent requirement by P-gp to bind to chemically similar molecules.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Aminoácidos/química , Peptidomiméticos/síntesis química , Peptidomiméticos/farmacología , Tiazoles/química , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Sitios de Unión/efectos de los fármacos , Técnicas de Química Sintética , Humanos , Simulación del Acoplamiento Molecular , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Conformación Proteica , Relación Estructura-Actividad
18.
Oncotarget ; 8(55): 93785-93799, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29212189

RESUMEN

Previous reports have shown that some tyrosine kinase inhibitors (TKIs) could inhibit the ATP-binding cassette (ABC) transporters involved in multidrug resistance (MDR). Quizartinib (AC220), a potent class III receptor tyrosine kinase inhibitor (TKI), was synthesized to selectively inhibit FMS-like tyrosine kinase-3 (FLT3), a target in the treatment of acute myeloid leukemia (AML). Quizartinib is currently under clinical trials for FLT3 ITD and wild-type AML and is tested in combination with chemotherapy. While non-toxic to cell lines, quizartinib at 3 µM showed significant reversal effect on wild-type and mutant ABCG2 (R482T)-mediated MDR, and only a moderate reversal effect on mutant ABCG2 (R482G)-mediated MDR. Results also showed that quizartinib reversed MDR not by reducing the expression of ABCG2 protein, but by antagonizing the drug efflux function and increasing the intracellular accumulation of substrate anticancer drugs in ABCG2-overexpressing cells. Importantly, quizartinib at 30 mg/kg strongly enhanced the effect of topotecan (3 mg/kg) in ABCG2-overexpressing (H460/MX20) xenografts in athymic nude mice. These results demonstrated that quizartinib potentiates the antineoplastic activity of wild-type and R482T mutant ABCG2 substrates. These findings may be useful in clinical practice for cancer combination therapy with quizartinib.

19.
Virus Genes ; 53(4): 522-531, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28425034

RESUMEN

The molluscum contagiosum virus (MCV) uses a variety of immune evasion strategies to antagonize host immune responses. Two MCV proteins, MC159 and MC160, contain tandem death effector domains (DEDs). They are reported to inhibit innate immune signaling events such as NF-κB and IRF3 activation, and apoptosis. The RxDL motif of MC159 is required for inhibition of both apoptosis and NF-κB activation. However, the role of the conserved RxDL motif in the MC160 DEDs remained unknown. To answer this question, we performed alanine mutations to neutralize the arginine and aspartate residues present in the MC160 RxDL in both DED1 and DED2. These mutations were further modeled against the structure of the MC159 protein. Surprisingly, the RxDL motif was not required for MC160's ability to inhibit MAVS-induced IFNß activation. Further, unlike previous results with the MC159 protein, mutations within the RxDL motif of MC160 had no effect on the ability of MC160 to dampen TNF-α-induced NF-κB activation. Molecular modeling predictions revealed no overall changes to the structure in the MC160 protein when the amino acids of both RxDL motifs were mutated to alanine (DED1 = R67A D69A; DED2 = R160A D162A). Taken together, our results demonstrate that the RxDL motifs present in the MC160 DEDs are not required for known functions of the viral protein.


Asunto(s)
Evasión Inmune , Molusco Contagioso/virología , Virus del Molusco Contagioso/inmunología , Proteínas Virales/química , Proteínas Virales/inmunología , Secuencias de Aminoácidos , Apoptosis , Humanos , Interferón beta/genética , Interferón beta/inmunología , Molusco Contagioso/genética , Molusco Contagioso/inmunología , Molusco Contagioso/fisiopatología , Virus del Molusco Contagioso/química , Virus del Molusco Contagioso/genética , Dominios Proteicos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Virales/genética
20.
Sci Rep ; 7: 42106, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28181548

RESUMEN

Multidrug resistance (MDR) attenuates the chemotherapy efficacy and increases the probability of cancer recurrence. The accelerated drug efflux mediated by ATP-binding cassette (ABC) transporters is one of the major MDR mechanisms. This study investigated if TTT-28, a newly synthesized thiazole-valine peptidomimetic, could reverse ABCB1-mediated MDR in vitro and in vivo. TTT-28 reversed the ABCB1-mediated MDR and increased the accumulation of [3H]-paclitaxel in ABCB1 overexpressing cells by selectively blocking the efflux function of ABCB1, but not interfering with the expression level and localization of ABCB1. Animal study revealed that TTT-28 enhanced the intratumoral concentration of paclitaxel and promoted apoptosis, thereby potently inhibiting the growth of ABCB1 overexpressing tumors. But TTT-28 did not induce the toxicity (cardiotoxicity/myelosuppression) of paclitaxel in mice. In this study, we synthesized and evaluated a novel selective inhibitor of ABCB1 (TTT-28) with high efficacy and low toxicity. The identification and characterization of this new thiazole-valine peptidomimetic will facilitate design and synthesis of a new generation of ABCB1 inhibitors, leading to further research on multidrug resistance and combination chemotherapy. Furthermore, the strategy that co-administer MDR-ABCB1 inhibitor to overcome the resistance of one FDA approved, widely used chemotherapeutic paclitaxel, may be promising direction for the field of adjuvant chemotherapy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Peptidomiméticos/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Xenoinjertos , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Paclitaxel/administración & dosificación , Paclitaxel/metabolismo , Paclitaxel/farmacocinética , Tiazoles/metabolismo , Resultado del Tratamiento , Valina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA