Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Intervalo de año de publicación
1.
Commun Biol ; 7(1): 563, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740899

RESUMEN

Targeting the estrogen receptor alpha (ERα) pathway is validated in the clinic as an effective means to treat ER+ breast cancers. Here we present the development of a VHL-targeting and orally bioavailable proteolysis-targeting chimera (PROTAC) degrader of ERα. In vitro studies with this PROTAC demonstrate excellent ERα degradation and ER antagonism in ER+ breast cancer cell lines. However, upon dosing the compound in vivo we observe an in vitro-in vivo disconnect. ERα degradation is lower in vivo than expected based on the in vitro data. Investigation into potential causes for the reduced maximal degradation reveals that metabolic instability of the PROTAC linker generates metabolites that compete for binding to ERα with the full PROTAC, limiting degradation. This observation highlights the requirement for metabolically stable PROTACs to ensure maximal efficacy and thus optimisation of the linker should be a key consideration when designing PROTACs.


Asunto(s)
Receptor alfa de Estrógeno , Proteolisis , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau , Humanos , Receptor alfa de Estrógeno/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Femenino , Proteolisis/efectos de los fármacos , Animales , Administración Oral , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ratones , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación
2.
J Med Chem ; 67(6): 4541-4559, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38466661

RESUMEN

The optimization of an allosteric fragment, discovered by differential scanning fluorimetry, to an in vivo MAT2a tool inhibitor is discussed. The structure-based drug discovery approach, aided by relative binding free energy calculations, resulted in AZ'9567 (21), a potent inhibitor in vitro with excellent preclinical pharmacokinetic properties. This tool showed a selective antiproliferative effect on methylthioadenosine phosphorylase (MTAP) KO cells, both in vitro and in vivo, providing further evidence to support the utility of MAT2a inhibitors as potential anticancer therapies for MTAP-deficient tumors.


Asunto(s)
Neoplasias , Humanos , Entropía , Metionina Adenosiltransferasa/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(21): e2221967120, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37186857

RESUMEN

The structure-based design of small-molecule inhibitors targeting protein-protein interactions (PPIs) remains a huge challenge as the drug must bind typically wide and shallow protein sites. A PPI target of high interest for hematological cancer therapy is myeloid cell leukemia 1 (Mcl-1), a prosurvival guardian protein from the Bcl-2 family. Despite being previously considered undruggable, seven small-molecule Mcl-1 inhibitors have recently entered clinical trials. Here, we report the crystal structure of the clinical-stage inhibitor AMG-176 bound to Mcl-1 and analyze its interaction along with clinical inhibitors AZD5991 and S64315. Our X-ray data reveal high plasticity of Mcl-1 and a remarkable ligand-induced pocket deepening. Nuclear Magnetic Resonance (NMR)-based free ligand conformer analysis demonstrates that such unprecedented induced fit is uniquely achieved by designing highly rigid inhibitors, preorganized in their bioactive conformation. By elucidating key chemistry design principles, this work provides a roadmap for targeting the largely untapped PPI class more successfully.


Asunto(s)
Apoptosis , Naftalenos , Modelos Moleculares , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Ligandos
4.
J Med Chem ; 66(4): 2918-2945, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36727211

RESUMEN

Herein, we report the optimization of a meta-substituted series of selective estrogen receptor degrader (SERD) antagonists for the treatment of ER+ breast cancer. Structure-based design together with the use of modeling and NMR to favor the bioactive conformation led to a highly potent series of basic SERDs with promising physicochemical properties. Issues with hERG activity resulted in a strategy of zwitterion formation and ultimately in the identification of 38. This compound was shown to be a highly potent SERD capable of effectively degrading ERα in both MCF-7 and CAMA-1 cell lines. The low lipophilicity and zwitterionic nature led to a SERD with a clean secondary pharmacology profile and no hERG activity. Favorable physicochemical properties resulted in good oral bioavailability in preclinical species and potent in vivo activity in a mouse xenograft model.


Asunto(s)
Neoplasias de la Mama , Receptores de Estrógenos , Ratones , Humanos , Animales , Femenino , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Antagonistas de Estrógenos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Receptor alfa de Estrógeno/metabolismo , Línea Celular
5.
J Med Chem ; 65(4): 3306-3331, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35133824

RESUMEN

ATAD2 is an epigenetic bromodomain-containing target which is overexpressed in many cancers and has been suggested as a potential oncology target. While several small molecule inhibitors have been described in the literature, their cellular activity has proved to be underwhelming. In this work, we describe the identification of a novel series of ATAD2 inhibitors by high throughput screening, confirmation of the bromodomain region as the site of action, and the optimization campaign undertaken to improve the potency, selectivity, and permeability of the initial hit. The result is compound 5 (AZ13824374), a highly potent and selective ATAD2 inhibitor which shows cellular target engagement and antiproliferative activity in a range of breast cancer models.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de Unión al ADN/antagonistas & inhibidores , Línea Celular Tumoral , Cristalografía por Rayos X , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Modelos Moleculares , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Especificidad por Sustrato , Ensayo de Tumor de Célula Madre
6.
Commun Biol ; 4(1): 1241, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725463

RESUMEN

Next generation modified antisense oligonucleotides (ASOs) are commercially approved new therapeutic modalities, yet poor productive uptake and endosomal entrapment in tumour cells limit their broad application. Here we compare intracellular traffic of anti KRAS antisense oligonucleotide (AZD4785) in tumour cell lines PC9 and LK2, with good and poor productive uptake, respectively. We find that the majority of AZD4785 is rapidly delivered to CD63+late endosomes (LE) in both cell lines. Importantly, lysobisphosphatidic acid (LBPA) that triggers ASO LE escape is presented in CD63+LE in PC9 but not in LK2 cells. Moreover, both cell lines recycle AZD4785 in extracellular vesicles (EVs); however, AZD4785 quantification by advanced mass spectrometry and proteomic analysis reveals that LK2 recycles more AZD4785 and RNA-binding proteins. Finally, stimulating LBPA intracellular production or blocking EV recycling enhances AZD4785 activity in LK2 but not in PC9 cells thus offering a possible strategy to enhance ASO potency in tumour cells with poor productive uptake of ASOs.


Asunto(s)
Antineoplásicos/farmacología , Vesículas Extracelulares/fisiología , Lisofosfolípidos/metabolismo , Monoglicéridos/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Línea Celular Tumoral , Humanos
7.
J Med Chem ; 64(10): 6814-6826, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33900758

RESUMEN

MAT2a is a methionine adenosyltransferase that synthesizes the essential metabolite S-adenosylmethionine (SAM) from methionine and ATP. Tumors bearing the co-deletion of p16 and MTAP genes have been shown to be sensitive to MAT2a inhibition, making it an attractive target for treatment of MTAP-deleted cancers. A fragment-based lead generation campaign identified weak but efficient hits binding in a known allosteric site. By use of structure-guided design and systematic SAR exploration, the hits were elaborated through a merging and growing strategy into an arylquinazolinone series of potent MAT2a inhibitors. The selected in vivo tool compound 28 reduced SAM-dependent methylation events in cells and inhibited proliferation of MTAP-null cells in vitro. In vivo studies showed that 28 was able to induce antitumor response in an MTAP knockout HCT116 xenograft model.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/química , Metionina Adenosiltransferasa/antagonistas & inhibidores , Sitio Alostérico , Animales , Proliferación Celular , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Técnicas de Inactivación de Genes , Células HCT116 , Semivida , Humanos , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Ratones , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Quinazolinas/química , Quinazolinas/metabolismo , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Ratas , S-Adenosilmetionina/metabolismo , Relación Estructura-Actividad , Trasplante Heterólogo
8.
Angew Chem Int Ed Engl ; 60(25): 13937-13944, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33783110

RESUMEN

Protein complexes are defined by the three-dimensional structure of participating binding partners. Knowledge about these structures can facilitate the design of peptidomimetics which have been applied for example, as inhibitors of protein-protein interactions (PPIs). Even though ß-sheets participate widely in PPIs, they have only rarely served as the basis for peptidomimetic PPI inhibitors, in particular when addressing intracellular targets. Here, we present the structure-based design of ß-sheet mimetics targeting the intracellular protein ß-catenin, a central component of the Wnt signaling pathway. Based on a protein binding partner of ß-catenin, a macrocyclic peptide was designed and its crystal structure in complex with ß-catenin obtained. Using this structure, we designed a library of bicyclic ß-sheet mimetics employing a late-stage diversification strategy. Several mimetics were identified that compete with transcription factor binding to ß-catenin and inhibit Wnt signaling in cells. The presented design strategy can support the development of inhibitors for other ß-sheet-mediated PPIs.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Péptidos/farmacología , beta Catenina/antagonistas & inhibidores , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Modelos Moleculares , Péptidos/química , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
9.
J Med Chem ; 63(23): 14530-14559, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-32910656

RESUMEN

Herein we report the optimization of a series of tricyclic indazoles as selective estrogen receptor degraders (SERD) and antagonists for the treatment of ER+ breast cancer. Structure based design together with systematic investigation of each region of the molecular architecture led to the identification of N-[1-(3-fluoropropyl)azetidin-3-yl]-6-[(6S,8R)-8-methyl-7-(2,2,2-trifluoroethyl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl]pyridin-3-amine (28). This compound was demonstrated to be a highly potent SERD that showed a pharmacological profile comparable to fulvestrant in its ability to degrade ERα in both MCF-7 and CAMA-1 cell lines. A stringent control of lipophilicity ensured that 28 had favorable physicochemical and preclinical pharmacokinetic properties for oral administration. This, combined with demonstration of potent in vivo activity in mouse xenograft models, resulted in progression of this compound, also known as AZD9833, into clinical trials.


Asunto(s)
Antineoplásicos/administración & dosificación , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Administración Oral , Antineoplásicos/química , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Ciclización , Descubrimiento de Drogas , Femenino , Humanos , Lípidos/química , Estructura Molecular , Moduladores Selectivos de los Receptores de Estrógeno/química , Moduladores Selectivos de los Receptores de Estrógeno/farmacocinética , Relación Estructura-Actividad
10.
Cell Chem Biol ; 27(1): 41-46.e17, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31786184

RESUMEN

Deregulation of the PRC2 complex, comprised of the core subunits EZH2, SUZ12, and EED, drives aberrant hypermethylation of H3K27 and tumorigenicity of many cancers. Although inhibitors of EZH2 have shown promising clinical activity, preclinical data suggest that resistance can be acquired through secondary mutations in EZH2 that abrogate drug target engagement. To address these limitations, we have designed several hetero-bifunctional PROTACs (proteolysis-targeting chimera) to efficiently target EED for elimination. Our PROTACs bind to EED (pKD ∼ 9.0) and promote ternary complex formation with the E3 ubiquitin ligase. The PROTACs potently inhibit PRC2 enzyme activity (pIC50 ∼ 8.1) and induce rapid degradation of not only EED but also EZH2 and SUZ12 within the PRC2 complex. Furthermore, the PROTACs selectively inhibit proliferation of PRC2-dependent cancer cells (half maximal growth inhibition [GI50] = 49-58 nM). In summary, our data demonstrate a therapeutic modality to target PRC2-dependent cancer through a PROTAC-mediated degradation mechanism.


Asunto(s)
Complejo Represivo Polycomb 2/metabolismo , Proteolisis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Estructura Molecular , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Relación Estructura-Actividad
11.
Chembiochem ; 20(24): 2987-2990, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31680402

RESUMEN

The acyl-binding UNC119 proteins mediate the activation and transport of various N-myristoylated proteins. In particular, UNC119a plays a crucial role in the completion of cytokinesis. Herein, we report the use of a lipidated peptide originating from the UNC119 binding partner Gnat1 as the basis for the design of lipidated, stabilized α-helical peptides that target UNC119a. By using the hydrocarbon peptide-stapling approach, cell-permeable binders of UNC119a were generated that induced the accumulation of cytokinetic and binucleated cells; this suggests UNC119a as a potential target for the inhibition of cytokinesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Metabolismo de los Lípidos , Péptidos/metabolismo , Péptidos/farmacología , Proteínas Adaptadoras Transductoras de Señales/química , Secuencia de Aminoácidos , Células HeLa , Humanos , Modelos Moleculares , Terapia Molecular Dirigida , Péptidos/química , Unión Proteica , Conformación Proteica en Hélice alfa
12.
J Med Chem ; 62(21): 9418-9437, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31361481

RESUMEN

The three-dimensional conformations adopted by a free ligand in solution impact bioactivity and physicochemical properties. Solution 1D NMR spectra inherently contain information on ligand conformational flexibility and three-dimensional shape, as well as the propensity of the free ligand to fully preorganize into the bioactive conformation. Herein we discuss some key learnings, distilled from our experience developing potent and selective synthetic macrocyclic inhibitors, including Mcl-1 clinical candidate AZD5991. Case studies have been selected from recent oncology research projects, demonstrating how 1D NMR conformational signatures can complement X-ray protein-ligand structural information to guide medicinal chemistry optimization. Learning to extract free ligand conformational information from routinely available 1D NMR signatures has proven to be fast enough to guide medicinal chemistry decisions within design cycles for compound optimization.


Asunto(s)
Diseño de Fármacos , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Cinética , Ligandos , Compuestos Macrocíclicos/síntesis química , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Conformación Proteica , Relación Estructura-Actividad
13.
Nucleic Acids Res ; 47(9): 4375-4392, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30927008

RESUMEN

Antisense oligonucleotides (ASOs) modulate cellular target gene expression through direct binding to complementary RNA. Advances in ASO chemistry have led to the development of phosphorothioate (PS) ASOs with constrained-ethyl modifications (cEt). These next-generation cEt-ASOs can enter cells without transfection reagents. Factors involved in intracellular uptake and trafficking of cEt-ASOs leading to successful target knockdown are highly complex and not yet fully understood. AZD4785 is a potent and selective therapeutic KRAS cEt-ASO currently under clinical development for the treatment of cancer. Therefore, we used this to investigate mechanisms of cEt-ASO trafficking across a panel of cancer cells. We found that the extent of ASO-mediated KRAS mRNA knockdown varied significantly between cells and that this did not correlate with bulk levels of intracellular accumulation. We showed that in cells with good productive uptake, distribution of ASO was perinuclear and in those with poor productive uptake distribution was peripheral. Furthermore, ASO rapidly trafficked to the late endosome/lysosome in poor productive uptake cells compared to those with more robust knockdown. An siRNA screen identified several factors mechanistically involved in productive ASO uptake, including the endosomal GTPase Rab5C. This work provides novel insights into the trafficking of cEt-ASOs and mechanisms that may determine their cellular fate.


Asunto(s)
Neoplasias/genética , Oligonucleótidos Antisentido/genética , Oligonucleótidos Fosforotioatos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al GTP rab5/genética , Endosomas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Neoplasias/patología , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Fosforotioatos/química , Oligonucleótidos Fosforotioatos/farmacología , ARN Mensajero/genética , ARN Interferente Pequeño/genética
14.
Nat Commun ; 9(1): 5341, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30559424

RESUMEN

Mcl-1 is a member of the Bcl-2 family of proteins that promotes cell survival by preventing induction of apoptosis in many cancers. High expression of Mcl-1 causes tumorigenesis and resistance to anticancer therapies highlighting the potential of Mcl-1 inhibitors as anticancer drugs. Here, we describe AZD5991, a rationally designed macrocyclic molecule with high selectivity and affinity for Mcl-1 currently in clinical development. Our studies demonstrate that AZD5991 binds directly to Mcl-1 and induces rapid apoptosis in cancer cells, most notably myeloma and acute myeloid leukemia, by activating the Bak-dependent mitochondrial apoptotic pathway. AZD5991 shows potent antitumor activity in vivo with complete tumor regression in several models of multiple myeloma and acute myeloid leukemia after a single tolerated dose as monotherapy or in combination with bortezomib or venetoclax. Based on these promising data, a Phase I clinical trial has been launched for evaluation of AZD5991 in patients with hematological malignancies (NCT03218683).


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Mieloma Múltiple/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Animales , Bortezomib/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Mieloma Múltiple/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Ratas , Ratas Desnudas , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Med Chem ; 61(23): 10602-10618, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30411895

RESUMEN

Fluorination is commonly employed to optimize bioactivity and pharmaco-kinetic properties of drug candidates. Aliphatic fluorination often reduces the lipophilicity (log P), but polyfluoroalkylation typically increases lipophilicity. Hence, identification of polyfluorinated motifs that nonetheless lead to similar or even reduced lipophilicities is of interest to expand the arsenal of medicinal chemistry tools in tackling properties such as compound metabolic stability or off-target selectivity. We show that changing a CF3-group of a perfluoroalkyl chain to a methyl group leads to a drastic reduction in lipophilicity. We also show that changing a C-F bond of a trifluoromethyl group, including when incorporated as part of a perfluoroalkyl group, to a C-Me group, leads to a reduction in log P, despite the resulting chain elongation. The observed lipophilicity trends were identified in fluorinated alkanol models and reproduced when incorporated in analogues of a drug candidate, and the metabolic stability of these motifs was demonstrated.


Asunto(s)
Carbono/química , Hidrocarburos Fluorados/química , Interacciones Hidrofóbicas e Hidrofílicas , Animales , Antineoplásicos/química , Ensayos Clínicos como Asunto , Estabilidad de Medicamentos , Humanos , Modelos Moleculares , Conformación Molecular , Ratas
16.
ACS Chem Biol ; 13(11): 3131-3141, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30335946

RESUMEN

B-cell lymphoma 6 (BCL6) inhibition is a promising mechanism for treating hematological cancers but high quality chemical probes are necessary to evaluate its therapeutic potential. Here we report potent BCL6 inhibitors that demonstrate cellular target engagement and exhibit exquisite selectivity for BCL6 based on mass spectrometry analyses following chemical proteomic pull down. Importantly, a proteolysis-targeting chimera (PROTAC) was also developed and shown to significantly degrade BCL6 in a number of diffuse large B-cell lymphoma (DLBCL) cell lines, but neither BCL6 inhibition nor degradation selectively induced marked phenotypic response. To investigate, we monitored PROTAC directed BCL6 degradation in DLBCL OCI-Ly1 cells by immunofluorescence and discovered a residual BCL6 population. Analysis of subcellular fractions also showed incomplete BCL6 degradation in all fractions despite having measurable PROTAC concentrations, together providing a rationale for the weak antiproliferative response seen with both BCL6 inhibitor and degrader. In summary, we have developed potent and selective BCL6 inhibitors and a BCL6 PROTAC that effectively degraded BCL6, but both modalities failed to induce a significant phenotypic response in DLBCL despite achieving cellular concentrations.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Quinolonas/farmacología , Talidomida/análogos & derivados , Talidomida/farmacología , Proteínas Adaptadoras Transductoras de Señales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Línea Celular Tumoral , Células HEK293 , Humanos , Ligandos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Péptido Hidrolasas/metabolismo , Unión Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-bcl-6/química , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Quinolonas/síntesis química , Quinolonas/metabolismo , Talidomida/síntesis química , Talidomida/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
17.
J Med Chem ; 61(16): 7314-7329, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30091600

RESUMEN

Inhibitor of apoptosis proteins (IAPs) are promising anticancer targets, given their roles in the evasion of apoptosis. Several peptidomimetic IAP antagonists, with inherent selectivity for cellular IAP (cIAP) over X-linked IAP (XIAP), have been tested in the clinic. A fragment screening approach followed by structure-based optimization has previously been reported that resulted in a low-nanomolar cIAP1 and XIAP antagonist lead molecule with a more balanced cIAP-XIAP profile. We now report the further structure-guided optimization of the lead, with a view to improving the metabolic stability and cardiac safety profile, to give the nonpeptidomimetic antagonist clinical candidate 27 (ASTX660), currently being tested in a phase 1/2 clinical trial (NCT02503423).


Asunto(s)
Antineoplásicos/farmacología , Compuestos Heterocíclicos con 2 Anillos/farmacología , Piperazinas/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/antagonistas & inhibidores , Administración Oral , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Cristalografía por Rayos X , Canal de Potasio ERG1/antagonistas & inhibidores , Compuestos Heterocíclicos con 2 Anillos/química , Compuestos Heterocíclicos con 2 Anillos/farmacocinética , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Macaca fascicularis , Masculino , Ratones Endogámicos BALB C , Piperazinas/química , Piperazinas/farmacocinética , Ratas Sprague-Dawley , Relación Estructura-Actividad , Proteína Inhibidora de la Apoptosis Ligada a X/química , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Med Chem ; 60(11): 4611-4625, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28492317

RESUMEN

XIAP and cIAP1 are members of the inhibitor of apoptosis protein (IAP) family and are key regulators of anti-apoptotic and pro-survival signaling pathways. Overexpression of IAPs occurs in various cancers and has been associated with tumor progression and resistance to treatment. Structure-based drug design (SBDD) guided by structural information from X-ray crystallography, computational studies, and NMR solution conformational analysis was successfully applied to a fragment-derived lead resulting in AT-IAP, a potent, orally bioavailable, dual antagonist of XIAP and cIAP1 and a structurally novel chemical probe for IAP biology.


Asunto(s)
Compuestos Heterocíclicos con 2 Anillos/química , Compuestos Heterocíclicos con 2 Anillos/farmacología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Piperazinas/química , Piperazinas/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Cristalografía por Rayos X , Descubrimiento de Drogas , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Peptidomiméticos , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
19.
ACS Chem Biol ; 11(11): 3093-3105, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27571355

RESUMEN

The members of the NSD subfamily of lysine methyl transferases are compelling oncology targets due to the recent characterization of gain-of-function mutations and translocations in several hematological cancers. To date, these proteins have proven intractable to small molecule inhibition. Here, we present initial efforts to identify inhibitors of MMSET (aka NSD2 or WHSC1) using solution phase and crystal structural methods. On the basis of 2D NMR experiments comparing NSD1 and MMSET structural mobility, we designed an MMSET construct with five point mutations in the N-terminal helix of its SET domain for crystallization experiments and elucidated the structure of the mutant MMSET SET domain at 2.1 Å resolution. Both NSD1 and MMSET crystal systems proved resistant to soaking or cocrystallography with inhibitors. However, use of the close homologue SETD2 as a structural surrogate supported the design and characterization of N-alkyl sinefungin derivatives, which showed low micromolar inhibition against both SETD2 and MMSET.


Asunto(s)
Adenosina/análogos & derivados , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Oncogenes , Proteínas Represoras/antagonistas & inhibidores , Adenosina/química , Adenosina/farmacología , Sitios de Unión , Calorimetría , Cromatografía Liquida , Cristalografía por Rayos X , Diseño de Fármacos , N-Metiltransferasa de Histona-Lisina/genética , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Conformación Proteica , Proteínas Represoras/genética
20.
Biomol NMR Assign ; 10(2): 315-20, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27356987

RESUMEN

Proteins of the NSD family are histone-methyl transferases with critical functions in the regulation of chromatin structure and function. NSD1 and NSD2 are homologous proteins that function as epigenetic regulators of transcription through their abilities to catalyse histone methylation. Misregulation of NSD1 and NSD2 expression or mutations in their genes are linked to a number of human diseases such as Sotos syndrome, and cancers including acute myeloid leukemia, multiple myeloma, and lung cancer. The catalytic domain of both proteins contains a conserved SET domain which is involved in histone methylation. Here we report the backbone resonance assignments and secondary structure information of the catalytic domains of human NSD1 and NSD2.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/química , Péptidos y Proteínas de Señalización Intracelular/química , Resonancia Magnética Nuclear Biomolecular , Proteínas Nucleares/química , Proteínas Represoras/química , Secuencia de Aminoácidos , Histona Metiltransferasas , Humanos , Dominios Proteicos , Estructura Secundaria de Proteína , Soluciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA