Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Chem Commun (Camb) ; 60(4): 436-439, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38086706

RESUMEN

Sulf-2 has been identified as a putative target for anticancer therapies. Here we report the design and synthesis of sulfated disaccharide inhibitors based on IdoA(2S)-GlcNS(6S). Trisulfated disaccharide inhibitor IdoA(2S)-GlcNS(6Sulfamate) demonstrated potent Sulf-2 inhibition. The IC50 value was determined to be 39.8 µM ± 18.3, which is comparable to a tetrasaccharide inhibitor of HSulf-1 reported in the literature. We propose that the disaccharide IdoA(2S)-GlcNS(6S) is the shortest fragment size required for effective inhibition of the Sulfs.


Asunto(s)
Heparitina Sulfato , Oligosacáridos , Heparitina Sulfato/farmacología , Oligosacáridos/farmacología , Disacáridos/farmacología , Sulfotransferasas
2.
J Med Chem ; 67(1): 110-137, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38146625

RESUMEN

Orphan G-protein-coupled receptor 84 (GPR84) is a receptor that has been linked to cancer, inflammatory, and fibrotic diseases. We have reported DL-175 as a biased agonist at GPR84 which showed differential signaling via Gαi/cAMP and ß-arrestin, but which is rapidly metabolized. Herein, we describe an optimization of DL-175 through a systematic structure-activity relationship (SAR) analysis. This reveals that the replacement of the naphthalene group improved metabolic stability and the addition of a 5-hydroxy substituent to the pyridine N-oxide group, yielding compounds 68 (OX04528) and 69 (OX04529), enhanced the potency for cAMP signaling by 3 orders of magnitude to low picomolar values. Neither compound showed detectable effects on ß-arrestin recruitment up to 80 µM. Thus, the new GPR84 agonists 68 and 69 displayed excellent potency, high G-protein signaling bias, and an appropriate in vivo pharmacokinetic profile that will allow investigation of GPR84 biased agonist activity in vivo.


Asunto(s)
Proteínas de Unión al GTP , Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Unión al GTP/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo , Relación Estructura-Actividad
3.
Br J Pharmacol ; 181(10): 1509-1523, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38148720

RESUMEN

GPR84 was first identified as an open reading frame encoding an orphan Class A G protein coupled receptor in 2001. Gpr84 mRNA is expressed in a limited number of cell types with the highest levels of expression being in innate immune cells, M1 polarised macrophages and neutrophils. The first reported ligands for this receptor were medium chain fatty acids with chain lengths between 9 and 12 carbons. Subsequently, a series of synthetic agonists that signal via the GPR84 receptor were identified. Radioligand binding assays and molecular modelling with site-directed mutagenesis suggest the presence of three ligand binding sites on the receptor, but the physiological agonist(s) of the receptor remain unidentified. Here, we review the effects of GPR84 agonists on innate immune cells following a series of chemical discoveries since 2001. The development of highly biased agonists has helped to probe receptor function in vitro, and the remaining challenge is to follow the effects of biased signalling to the physiological functions of innate immune cell types. LINKED ARTICLES: This article is part of a themed issue GPR84 Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.10/issuetoc.


Asunto(s)
Receptores Acoplados a Proteínas G , Transducción de Señal , Receptores Acoplados a Proteínas G/metabolismo , Macrófagos , Ligandos , Fagocitosis
4.
Eur J Med Chem ; 258: 115509, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37343464

RESUMEN

Acute myeloid leukaemia (AML) is an aggressive type of leukaemia with low rates of long-term survival. While the current standard of care is based on cytotoxic chemotherapy, a promising emerging approach is differentiation therapy. However, most current differentiating agents target specific mutations and are effective only in certain patient subtypes. To identify agents which may be effective in wider population cohorts, we performed a phenotypic screen with the myeloid marker CD11b and identified a compound series that was able to differentiate AML cell lines in vitro regardless of their mutation status. Structure-activity relationship studies revealed that replacing the formamide and catechol methyl ether groups with sulfonamide and indazole respectively improved the in vitro metabolic profile of the series while maintaining the differentiation profile in multiple cell lines. This optimisation exercise enabled progression of a lead compound to in vivo efficacy testing. Our work supports the promise of phenotypic screening to identify novel small molecules that induce differentiation in a wide range of AML subtypes.


Asunto(s)
Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular , Diferenciación Celular , Piridinas/farmacología
5.
J Histochem Cytochem ; 71(6): 301-320, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37350564

RESUMEN

Retinal astrocytes are vital for neuronal homeostasis in the retina. Together with Müller glia, they provide retinal cells with neurotrophic factors, antioxidative support, and defense mechanisms such as the formation of the blood-retinal barrier. Substantial heterogeneity of astrocyte morphology and function represents a challenge for identification of distinct subtypes which may be potential targets for therapeutic purposes. Hence, identification of novel markers of astrocyte subpopulations is highly relevant to better understand the molecular mechanisms involved in retinal development, homeostasis, and pathology. In this study, we observed that the cell cycle regulator, p16INK4a, is expressed in immature astrocytes in the mouse retina. Immunohistochemical analysis showed p16INK4a expression in the optic nerve of wild-type mice from 3 days to 3 months of age and in the nerve fiber layer of the adult mouse retina. Colocalization of p16INK4a expression and glial fibrillary acidic protein (immature/mature astrocyte marker) tends to decrease with age. However, colocalization of p16INK4a expression and vimentin (immature astrocyte marker) remains high in the optic nerve from the early postnatal period to adulthood. The observations from this study provide a valuable tool for further investigations of ocular astrocytes in the developing retina as well as in degenerative retinopathies.


Asunto(s)
Astrocitos , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Ratones , Animales , Astrocitos/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Neuroglía , Retina/metabolismo , Proteína Ácida Fibrilar de la Glía/análisis , Ciclo Celular
6.
iScience ; 25(8): 104787, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35992086

RESUMEN

Despite much progress in developing better drugs, many patients with acute myeloid leukemia (AML) still die within a year of diagnosis. This is partly because it is difficult to identify therapeutic targets that are effective across multiple AML subtypes. One common factor across AML subtypes is the presence of a block in differentiation. Overcoming this block should allow for the identification of therapies that are not dependent on a specific mutation for their efficacy. Here, we used a phenotypic screen to identify compounds that stimulate differentiation in genetically diverse AML cell lines. Lead compounds were shown to decrease tumor burden and to increase survival in vivo. Using multiple complementary target deconvolution approaches, these compounds were revealed to be anti-mitotic tubulin disruptors that cause differentiation by inducing a G2-M mitotic arrest. Together, these results reveal a function for tubulin disruptors in causing differentiation of AML cells.

7.
Molecules ; 26(21)2021 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-34771052

RESUMEN

Acute myeloid leukemia (AML) is the most aggressive type of blood cancer, and there is a continued need for new treatments that are well tolerated and improve long-term survival rates in patients. Induction of differentiation has emerged as a promising alternative to conventional cytotoxic chemotherapy, but known agents lack efficacy in genetically distinct patient populations. Previously, we established a phenotypic screen to identify small molecules that could stimulate differentiation in a range of AML cell lines. Utilising this strategy, a 1,5-dihydrobenzo[e][1,4]oxazepin-2(3H)-one hit compound was identified. Herein, we report the hit validation in vitro, structure-activity relationship (SAR) studies and the pharmacokinetic profiles for selected compounds.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Antineoplásicos/síntesis química , Línea Celular Tumoral , Células Cultivadas , Técnicas de Química Sintética , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Mieloide Aguda , Estructura Molecular , Relación Estructura-Actividad
8.
J Med Chem ; 64(21): 15608-15628, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34672555

RESUMEN

Induction of differentiation is a promising therapeutic strategy against acute myeloid leukemia. However, current differentiation therapies are effective only to specific patient populations. To identify novel differentiation agents with wider efficacy, we developed a phenotypic high-throughput screen with a range of genetically diverse cell lines. From the resulting hits, one chemical scaffold was optimized in terms of activity and physicochemical properties to yield OXS007417, a proof-of-concept tool compound, which was also able to decrease tumor volume in a murine in vivo xenograft model.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Fenotipo , Relación Estructura-Actividad , Células Tumorales Cultivadas
9.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34413188

RESUMEN

TMEM16A Ca2+-activated chloride channels are involved in multiple cellular functions and are proposed targets for diseases such as hypertension, stroke, and cystic fibrosis. This therapeutic endeavor, however, suffers from paucity of selective and potent modulators. Here, exploiting a synthetic small molecule with a biphasic effect on the TMEM16A channel, anthracene-9-carboxylic acid (A9C), we shed light on sites of the channel amenable for pharmacological intervention. Mutant channels with the intracellular gate constitutively open were generated. These channels were entirely insensitive to extracellular A9C when intracellular Ca2+ was omitted. However, when physiological Ca2+ levels were reestablished, the mutants regained sensitivity to A9C. Thus, intracellular Ca2+ is mandatory for the channel response to an extracellular modulator. The underlying mechanism is a conformational change in the outer pore that enables A9C to enter the pore to reach its binding site. The explanation of this structural rearrangement highlights a critical site for pharmacological intervention and reveals an aspect of Ca2+ gating in the TMEM16A channel.


Asunto(s)
Anoctamina-1/metabolismo , Antracenos/farmacología , Calcio/farmacología , Cloruros/farmacología , Animales , Anoctamina-1/genética , Estimulación Eléctrica , Fenómenos Electrofisiológicos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Simulación de Dinámica Molecular , Farmacología en Red , Técnicas de Placa-Clamp , Mutación Puntual
10.
DNA Cell Biol ; 39(11): 1926-1937, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33001759

RESUMEN

GPR84 is an inflammation-induced receptor highly expressed on immune cells, yet its endogenous ligand is still unknown. This makes any interpretation of its physiological activity in vivo difficult. However, experiments with potent synthetic agonists have highlighted what the receptor can do, namely, enhance proinflammatory signaling and macrophage effector functions such as phagocytosis. Developing drugs to block these effects has attracted interest from the scientific community with the aim of decreasing disease activity in inflammatory disorders or enhancing inflammation resolution. In this review, we critically reassess the widely held belief that the major role of GPR84 is that of being a medium-chain fatty acid (MCFA) receptor. While MCFAs have been shown to activate GPR84, it remains to be demonstrated that they are present in relevant tissues at appropriate concentrations. In contrast to four other "full-time" free fatty acid receptor subtypes, GPR84 is not expressed by enteroendocrine cells and has limited expression in the gastrointestinal tract. Across multiple tissues and cell types, the highest expression levels of GPR84 are observed hours after exposure to an inflammatory stimulus. These factors obscure the relationship between ligand and receptor in the human body and do not support the exclusive physiological pairing of MCFAs with GPR84. To maximize the chances of developing efficacious drugs for inflammatory diseases, we must advance our understanding of GPR84 and what it does in vivo.


Asunto(s)
Ácidos Grasos/genética , Inflamación/genética , Receptores Acoplados a Proteínas G/genética , Ácidos Grasos/metabolismo , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Ligandos , Macrófagos/metabolismo , Fagocitosis/genética , Transducción de Señal/genética
11.
Bioorg Med Chem ; 28(22): 115724, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33128909

RESUMEN

We have previously reported the discovery of a series of rhodanine-based inhibitors of the PIM family of serine/threonine kinases. Here we described the optimisation of those compounds to improve their physicochemical and ADME properties as well as reducing their off-targets activities against other kinases. Through molecular modeling and systematic structure activity relationship (SAR) studies, advanced molecules with high inhibitory potency, reduced off-target activity and minimal efflux were identified as new pan-PIM inhibitors. One example of an early lead, OX01401, was found to inhibit PIMs with nanomolar potency (15 nM for PIM1), inhibit proliferation of two PIM-expressing leukaemic cancer cell lines, MV4-11 and K562, and to reduce intracellular phosphorylation of a PIM substrate in a concentration dependent manner.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Tiazoles/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/química
12.
Blood ; 136(21): 2410-2415, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32599615

RESUMEN

Although cytokine-mediated expansion of human hematopoietic stem cells (HSCs) can result in high yields of hematopoietic progenitor cells, this generally occurs at the expense of reduced bone marrow HSC repopulating ability, thereby limiting potential therapeutic applications. Because bromodomain-containing proteins (BCPs) have been demonstrated to regulate mouse HSC self-renewal and stemness, we screened small molecules targeting various BCPs as potential agents for ex vivo expansion of human HSCs. Of 10 compounds tested, only the bromodomain and extra-terminal motif inhibitor CPI203 enhanced the expansion of human cord blood HSCs without losing cell viability in vitro. The expanded cells also demonstrated improved engraftment and repopulation in serial transplantation assays. Transcriptomic and functional studies showed that the expansion of long-term repopulating HSCs was accompanied by synchronized expansion and maturation of megakaryocytes consistent with CPI203-mediated reprogramming of cord blood hematopoietic stem and progenitor cells. This approach may therefore prove beneficial for ex vivo gene editing, for enhanced platelet production, and for the improved usage of cord blood for transplantation research and therapy.


Asunto(s)
Acetamidas/farmacología , Azepinas/farmacología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Proteínas/antagonistas & inhibidores , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Supervivencia de Injerto/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos NOD , Transcriptoma/efectos de los fármacos
13.
J Ocul Pharmacol Ther ; 36(4): 257-266, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32027217

RESUMEN

Purpose: Age-related macular degeneration leads to retinal pigment epithelium (RPE) cell death and loss of central vision. In vivo studies have shown that the RPE layer has an innate, but limited, ability to repopulate atrophic areas. We aimed to establish a semiautomated, in vitro, wound healing assay workflow for targeted screening of compounds able to influence RPE wound healing. Methods: The ARPE-19 phenotype was evaluated using bright-field microscopy, immunocytochemistry, and quantitative real-time polymerase chain reaction. ARPE-19 monolayers were simultaneously scratched in a 96-well format and treated with Hoechst-33342 and an array of compounds. Initial wound dimensions and wound healing were subsequently evaluated using the EVOS FL Auto 2.0 imaging platform combined with automated image analyses. Results: Long-term cultured ARPE-19 cells displayed a more in vivo RPE-like phenotype compared with recently seeded or short-term cultured cells. No statistical difference of initial scratch width was observed between short-term and long-term cultured cells, but more wells were excluded from analyses in total in the latter case due to scratch width, scratch smoothness, and imaging errors. Furthermore, the previous time spent in continuous culture had an effect on the observation of an altered wound healing response to different treatment conditions. Conclusions: We have established a semiautomated, 96-well format, in vitro wound healing assay with a reproducible workflow. This would enable screening of a significant number of compounds and greatly advances the potential of identifying novel therapeutics that may enhance the innate ability of RPE cells to repopulate atrophic areas.


Asunto(s)
Células Epiteliales/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Pigmentos Retinianos/metabolismo , Cicatrización de Heridas/genética , Animales , Células Cultivadas/metabolismo , Células Epiteliales/fisiología , Células Epiteliales/ultraestructura , Humanos , Inmunohistoquímica , Técnicas In Vitro/métodos , Degeneración Macular/complicaciones , Degeneración Macular/patología , Ratones , Microscopía/métodos , Modelos Animales , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/fisiología , Epitelio Pigmentado de la Retina/ultraestructura , Cicatrización de Heridas/inmunología , Cicatrización de Heridas/fisiología
14.
Angew Chem Int Ed Engl ; 59(6): 2420-2428, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31755636

RESUMEN

Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease arising from mutations in the dystrophin gene. Upregulation of utrophin to compensate for the missing dystrophin offers a potential therapy independent of patient genotype. The first-in-class utrophin modulator ezutromid/SMT C1100 was developed from a phenotypic screen through to a Phase 2 clinical trial. Promising efficacy and evidence of target engagement was observed in DMD patients after 24 weeks of treatment, however trial endpoints were not met after 48 weeks. The objective of this study was to understand the mechanism of action of ezutromid which could explain the lack of sustained efficacy and help development of new generations of utrophin modulators. Using chemical proteomics and phenotypic profiling we show that the aryl hydrocarbon receptor (AhR) is a target of ezutromid. Several lines of evidence demonstrate that ezutromid binds AhR with an apparent KD of 50 nm and behaves as an AhR antagonist. Furthermore, other reported AhR antagonists also upregulate utrophin, showing that this pathway, which is currently being explored in other clinical applications including oncology and rheumatoid arthritis, could also be exploited in future DMD therapies.


Asunto(s)
Benzoxazoles/química , Naftalenos/química , Proteómica/métodos , Receptores de Hidrocarburo de Aril/metabolismo , Utrofina/metabolismo , Animales , Benzoxazoles/metabolismo , Benzoxazoles/farmacología , Benzoxazoles/uso terapéutico , Reacción de Cicloadición , Diseño de Fármacos , Humanos , Cinética , Ratones , Sondas Moleculares/química , Distrofia Muscular de Duchenne/tratamiento farmacológico , Mioblastos/citología , Mioblastos/metabolismo , Naftalenos/metabolismo , Naftalenos/farmacología , Naftalenos/uso terapéutico , Unión Proteica , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Regulación hacia Arriba/efectos de los fármacos , Utrofina/agonistas , Utrofina/genética
15.
ACS Chem Biol ; 14(9): 2055-2064, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31465201

RESUMEN

GPR84 is an orphan G-protein-coupled receptor that is expressed on immune cells and implicated in several inflammatory diseases. The validation of GPR84 as a therapeutic target is hindered by the narrow range of available chemical tools and consequent poor understanding of GPR84 pathophysiology. Here we describe the discovery and characterization of DL-175, a potent, selective, and structurally novel GPR84 agonist and the first to display significantly biased signaling across GPR84-overexpressing cells, primary murine macrophages, and human U937 cells. By comparing DL-175 with reported GPR84 ligands, we show for the first time that biased GPR84 agonists have markedly different abilities to induce chemotaxis in human myeloid cells, while causing similar levels of phagocytosis enhancement. This work demonstrates that biased agonism at GPR84 enables the selective activation of functional responses in immune cells and delivers a high-quality chemical probe for further investigation.


Asunto(s)
Factores Quimiotácticos/farmacología , Óxidos N-Cíclicos/farmacología , Macrófagos/efectos de los fármacos , Piridinas/farmacología , Receptores Acoplados a Proteínas G/agonistas , Animales , Células CHO , Línea Celular Tumoral , Factores Quimiotácticos/química , Cricetulus , Óxidos N-Cíclicos/química , Humanos , Ratones , Estructura Molecular , Fagocitosis/efectos de los fármacos , Piridinas/química , Relación Estructura-Actividad Cuantitativa , Transducción de Señal/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 116(7): 2545-2550, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30683716

RESUMEN

The RAS gene family is frequently mutated in human cancers, and the quest for compounds that bind to mutant RAS remains a major goal, as it also does for inhibitors of protein-protein interactions. We have refined crystallization conditions for KRAS169Q61H-yielding crystals suitable for soaking with compounds and exploited this to assess new RAS-binding compounds selected by screening a protein-protein interaction-focused compound library using surface plasmon resonance. Two compounds, referred to as PPIN-1 and PPIN-2, with related structures from 30 initial RAS binders showed binding to a pocket where compounds had been previously developed, including RAS effector protein-protein interaction inhibitors selected using an intracellular antibody fragment (called Abd compounds). Unlike the Abd series of RAS binders, PPIN-1 and PPIN-2 compounds were not competed by the inhibitory anti-RAS intracellular antibody fragment and did not show any RAS-effector inhibition properties. By fusing the common, anchoring part from the two new compounds with the inhibitory substituents of the Abd series, we have created a set of compounds that inhibit RAS-effector interactions with increased potency. These fused compounds add to the growing catalog of RAS protein-protein inhibitors and show that building a chemical series by crossing over two chemical series is a strategy to create RAS-binding small molecules.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Proteína Oncogénica p21(ras)/antagonistas & inhibidores , Cristalografía por Rayos X , Desarrollo de Medicamentos , Estructura Molecular , Proteína Oncogénica p21(ras)/metabolismo , Unión Proteica , Resonancia por Plasmón de Superficie
17.
ACS Chem Biol ; 13(10): 2849-2854, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30216042

RESUMEN

Targeting the protein-protein interaction between p53 and MDM2/MDMX (MDM4) represents an attractive anticancer strategy for the treatment of p53-competent tumors. Several selective and potent MDM2 inhibitors have been developed and entered the clinic; however, the repertoire of MDMX antagonists is still limited. The arylmethylidenepyrazolinone SJ-172550 has been reported as a selective MDMX antagonist; yet, uncertainties about its mechanism of action have raised doubts about its use as a chemical probe. Here, we show that, in addition to its unclear mode of action, SJ-172550 is unstable in aqueous buffers, giving rise to side products of unknown biological activity. Using an SJ-172550-derived affinity probe, we observed promiscuous binding to cellular proteins whereas cellular thermal shift assays did not reveal a stabilizing effect on MDMX. Overall, our results raise further questions about the interpretation of data using SJ-172550 and related compounds to investigate cellular phenotypes.


Asunto(s)
Acetatos/metabolismo , Inhibidores Enzimáticos/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Pirazoles/metabolismo , Acetatos/química , Marcadores de Afinidad/química , Alquinos/química , Sitios de Unión , Carbocianinas/química , Proteínas de Ciclo Celular , Línea Celular Tumoral , Química Clic , Estabilidad de Medicamentos , Inhibidores Enzimáticos/química , Humanos , Proteínas Nucleares/química , Unión Proteica , Estabilidad Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/química , Pirazoles/química
18.
Nat Commun ; 9(1): 3169, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30093669

RESUMEN

Targeting specific protein-protein interactions (PPIs) is an attractive concept for drug development, but hard to implement since intracellular antibodies do not penetrate cells and most small-molecule drugs are considered unsuitable for PPI inhibition. A potential solution to these problems is to select intracellular antibody fragments to block PPIs, use these antibody fragments for target validation in disease models and finally derive small molecules overlapping the antibody-binding site. Here, we explore this strategy using an anti-mutant RAS antibody fragment as a competitor in a small-molecule library screen for identifying RAS-binding compounds. The initial hits are optimized by structure-based design, resulting in potent RAS-binding compounds that interact with RAS inside the cells, prevent RAS-effector interactions and inhibit endogenous RAS-dependent signalling. Our results may aid RAS-dependent cancer drug development and demonstrate a general concept for developing small compounds to replace intracellular antibody fragments, enabling rational drug development to target validated PPIs.


Asunto(s)
Sitios de Unión de Anticuerpos , Fragmentos de Inmunoglobulinas/química , Transducción de Señal , Anticuerpos/química , Biomarcadores/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Cristalografía por Rayos X , Células HEK293 , Humanos , Mutación , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes/química , Bibliotecas de Moléculas Pequeñas , Resonancia por Plasmón de Superficie , Proteínas ras/química
19.
J Pharmacol Exp Ther ; 361(3): 417-428, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28360334

RESUMEN

Hypercholesterolemia remains one of the leading risk factors for the development of cardiovascular disease. Many large double-blind studies have demonstrated that lowering low-density lipoprotein (LDL) cholesterol using a statin can reduce the risk of having a cardiovascular event by approximately 30%. However, despite the success of statins, some patient populations are unable to lower their LDL cholesterol to meet the targeted lipid levels, due to compliance or potency issues. This is especially true for patients with heterozygous familial hypercholesterolemia who may require additional upregulation of the low-density lipoprotein receptor (LDLR) to reduce LDL cholesterol levels below those achievable with maximal dosing of statins. Here we identify a series of small molecules from a genomic DNA reporter screen that upregulate the LDLR in mouse and human liver cell lines at nanomolar potencies (EC50 = 39 nM). Structure-activity relationship studies carried out on the lead compound, OX03771 [(E)-N,N-dimethyl-3-(4-styrylphenoxy)propan-1-amine], led to the identification of compound OX03050 [(E)-3-(4-styrylphenoxy)propan-1-ol], which had similar potency (EC50 = 26 nM) but a much-improved pharmacokinetic profile and showed in vivo efficacy. Compounds OX03050 and OX03771 were found to inhibit squalene synthase, the first committed step in cholesterol biosynthesis. These squalene synthase inhibitors were shown to act cooperatively with statins to increase LDLR expression in vitro. Overall, we demonstrated here a novel series of small molecules with the potential to be further developed to treat patients either alone or in combination with statins.


Asunto(s)
Farnesil Difosfato Farnesil Transferasa/antagonistas & inhibidores , Pruebas Genéticas/métodos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Receptores de LDL/biosíntesis , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Regulación hacia Arriba/fisiología , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Inhibidores Enzimáticos , Farnesil Difosfato Farnesil Transferasa/metabolismo , Humanos , Masculino , Ratones , Bibliotecas de Moléculas Pequeñas/farmacología , Regulación hacia Arriba/efectos de los fármacos
20.
Bioorg Med Chem ; 25(9): 2657-2665, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28341403

RESUMEN

The PIM family of serine/threonine kinases have become an attractive target for anti-cancer drug development, particularly for certain hematological malignancies. Here, we describe the discovery of a series of inhibitors of the PIM kinase family using a high throughput screening strategy. Through a combination of molecular modeling and optimization studies, the intrinsic potencies and molecular properties of this series of compounds was significantly improved. An excellent pan-PIM isoform inhibition profile was observed across the series, while optimized examples show good selectivity over other kinases. Two PIM-expressing leukemic cancer cell lines, MV4-11 and K562, were employed to evaluate the in vitro anti-proliferative effects of selected inhibitors. Encouraging activities were observed for many examples, with the best example (44) giving an IC50 of 0.75µM against the K562 cell line. These data provide a promising starting point for further development of this series as a new cancer therapy through PIM kinase inhibition.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Rodanina/análogos & derivados , Sulfonamidas/farmacología , Tiazolidinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Humanos , Isoenzimas/antagonistas & inhibidores , Células K562 , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Rodanina/síntesis química , Rodanina/farmacocinética , Rodanina/farmacología , Solubilidad , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética , Tiazolidinas/síntesis química , Tiazolidinas/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA