Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Med Chem ; 240: 114565, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-35797901

RESUMEN

Triple-negative breast cancer (TNBC) with the absence of estrogen receptor (ER), progesterone receptor (PR) and HER2 ptotein, is the highly aggressive subtype of breast cancer that exhibits poor prognosis and high tumor recurrence. It is vital to develop effective agents regulating the core molecular pathway of TNBC. Through a medium throughput screening and iterative medicinal chemistry optimization, we identified compound 7h as an autophagic flux inhibitor, which showed potent activities against human TNBC (MDA-MB-231 and MDA-MB-468) cell lines with IC50 values of 8.3 µM, and 6.0 µM, respectively, which are comparable to the potency of 5-FU and Cisplatin, the first line therapies for TNBC. Extensive investigation of mechanisms of action indicated that 7h inhibits autophagic flux and sequential accumulation of p62, leading to DNA damage and disrepair in TNBC cells. Importantly, nuclear p62 accumulation induced by compound 7h results in the inhibition of RNF168-mediated chromatin ubiquitination and the degradation of HR-related proteins in regulating the DNA damage response (DDR) process. In in vivo studies, compound 7h completely suppressed tumor growth in the MDA-MB-231 xenograft model at a dose of 15 mg/kg/q.d. Our findings indicate that compound 7h is an autophagic flux inhibitor and induced the degradation of HR-related proteins. Compound 7h could be potentially developed as an anti-cancer therapeutics for TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Autofagia , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Humanos , Imidazoles/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Ubiquitina-Proteína Ligasas
2.
J Med Chem ; 65(2): 1536-1551, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35081714

RESUMEN

Mutations of the rearranged during transfection (RET) kinase are frequently reported in cancer, which make it as an attractive therapeutic target. Herein, we discovered a series of N-trisubstituted pyrimidine derivatives as potent inhibitors for both wild-type (wt) RET and RETV804M, which is a resistant mutant for several FDA-approved inhibitors. The X-ray structure of a representative inhibitor with RET revealed that the compound binds in a unique pose that bifurcates beneath the P-loop and confirmed the compound as a type I inhibitor. Through the structure-activity relationship (SAR) study, compound 20 was identified as a lead compound, showing potent inhibition of both RET and RETV804M. Additionally, compound 20 displayed potent antiproliferative activity of CCDC6-RET-driven LC-2/ad cells. Analysis of RET phosphorylation indicated that biological activity was mediated by RET inhibition. Collectively, N-trisubstituted pyrimidine derivatives could serve as scaffolds for the discovery and development of potent inhibitors of type I RET and its gatekeeper mutant for the treatment of RET-driven cancers.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-ret/antagonistas & inhibidores , Pirimidinas/química , Adenocarcinoma del Pulmón/patología , Apoptosis , Proliferación Celular , Humanos , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-ret/genética , Relación Estructura-Actividad , Células Tumorales Cultivadas , Cicatrización de Heridas
3.
Eur J Med Chem ; 225: 113776, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34479037

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) with an internal tandem duplication (ITD) mutation has been validated as a driver lesion and a therapeutic target for acute myeloid leukemia (AML). Currently, several potent small-molecule FLT3 kinase inhibitors are being evaluated or have completed evaluation in clinical trials. However, many of these inhibitors are challenged by the secondary mutations on kinase domain, especially the point mutations at the activation loop (D835) and gatekeeper residue (F691). To overcome the resistance challenge, we identified a novel series of imidazo[1,2-a]pyridine-thiophene derivatives from a NIMA-related kinase 2 (NEK2) kinase inhibitor CMP3a, which retained inhibitory activities on FTL3-ITDD835V and FLT3-ITDF691L. Through this study, we identified the imidazo[1,2-a]pyridine-thiophene derivatives as type-I inhibitors of FLT3. Moreover, we observed compound 5o as an inhibitor displaying equal anti-proliferative activities against FLT3-ITD, FTL3-ITDD835Y and FLT3-ITDF691L driven acute myeloid leukemia (AML) cell lines. Meanwhile, the apoptotic effects of compound supported its mechanism of anti-proliferative action. These results indicate that the imidazo[1,2-a]pyridine-thiophene scaffold is promising for targeting acquired resistance caused by FLT3 secondary mutations and compound 5o is an interesting lead in this direction.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Tiofenos/farmacología , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Estructura Molecular , Mutación , Quinasas Relacionadas con NIMA/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Piridinas/síntesis química , Piridinas/química , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/química , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
4.
Sci Rep ; 11(1): 16103, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34373541

RESUMEN

We have recently described Pz-1, a benzimidazole-based type-2 RET and VEGFR2 inhibitor. Based on a kinome scan, here we show that Pz-1 is also a potent (IC50 < 1 nM) TRKA/B/C inhibitor. Pz-1 potently inhibited proliferation of human cancer cells carrying either RET- or TRKA oncoproteins (IC50 ~ 1 nM), with a negligible effect against RET- and TRKA-negative cells. By testing mutations, known to mediate resistance to other compounds, RET G810R/S, but not L730I/V, E732K, V738A and Y806N, showed some degree of resistance to Pz-1. In the case of TRKA, G595R and F589L, but not G667C, showed some degree of resistance. In xenograft models, orally administered Pz-1 almost completely inhibited RET- and TRKA-mutant tumours at 1-3 mg/kg/day but showed a reduced effect on RET/TRKA-negative cancer models. The activity, albeit reduced, on RET/TRKA-negative tumours may be justified by VEGFR2 inhibition. Tumours induced by NIH3T3 cells transfected by RET G810R and TRKA G595R featured resistance to Pz-1, demonstrating that RET or TRKA inhibition is critical for its anti-tumourigenic effect. In conclusion, Pz-1 represents a new powerful kinase inhibitor with distinct activity towards cancers induced by oncogenic RET and TRKA variants, including some mutants displaying resistance to other drugs.


Asunto(s)
Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptor trkA/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Neoplasias/metabolismo
5.
Eur J Med Chem ; 225: 113763, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34419892

RESUMEN

The tumor microenvironment contains high concentrations of TGFß, a crucial immunosuppressive cytokine. TGFß stimulates immune escape by promoting peripheral immune tolerance to avoid tumoricidal attack. Small-molecule inhibitors of TGFßR1 are a prospective method for next-generation immunotherapies. In the present study, we identified selective 4-aminoquinoline-based inhibitors of TGFßR1 through structural and rational-based design strategies. This led to the identification of compound 4i, which was found to be selective for TGFßR1 with the exception of MAP4K4 in the kinase profiling assay. The compound was then further optimized to remove MAP4K4 activity, since MAP4K4 is vital for proper T-cell function and its inhibition could exacerbate tumor immunosuppression. Optimization efforts led to compound 4s that inhibited TGFßR1 at an IC50 of 0.79 ± 0.19 nM with 2000-fold selectivity against MAP4K4. Compound 4s represents a highly selective TGFßR1 inhibitor that has potential applications in immuno-oncology.


Asunto(s)
Aminoquinolinas/farmacología , Descubrimiento de Drogas , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Aminoquinolinas/síntesis química , Aminoquinolinas/química , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/inmunología , Estructura Molecular , Proteínas Serina-Treonina Quinasas/inmunología , Receptor Tipo I de Factor de Crecimiento Transformador beta/inmunología , Relación Estructura-Actividad
6.
Eur J Med Chem ; 223: 113660, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34246853

RESUMEN

TGFß is crucial for the homeostasis of epithelial and neural tissues, wound repair, and regulating immune responses. Its dysregulation is associated with a vast number of diseases, of which modifying the tumor microenvironment is one of vital clinical interest. Despite various attempts, there is still no FDA-approved therapy to inhibit the TGFß pathway. Major mainstream approaches involve impairment of the TGFß pathway via inhibition of the TGFßRI kinase. With the purpose to identify non-receptor kinase-based inhibitors to impair TGFß signaling, an in-house chemical library was enriched, through a computational study, to eliminate TGFßRI kinase activity. Selected compounds were screened against a cell line engineered with a firefly luciferase gene under TGFß-Smad-dependent transcriptional control. Results indicated moderate potency for a molecule with phthalazine core against TGFß-Smad signaling. A series of phthalazine compounds were synthesized and evaluated for potency. The most promising compound (10p) exhibited an IC50 of 0.11 ± 0.02 µM and was confirmed to be non-cytotoxic up to 12 µM, with a selectivity index of approximately 112-fold. Simultaneously, 10p was confirmed to reduce the Smad phosphorylation using Western blot without exhibiting inhibition on the TGFßRI enzyme. This study identified a novel small-molecule scaffold that targets the TGFß pathway via a non-receptor-kinase mechanism.


Asunto(s)
Ftalazinas/química , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Fosforilación/efectos de los fármacos , Ftalazinas/metabolismo , Ftalazinas/farmacología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/química , Proteínas Smad/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Factor de Crecimiento Transformador beta/metabolismo
7.
Eur J Med Chem ; 216: 113265, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33652352

RESUMEN

Tropomyosin receptor kinase (TRK) represents an attractive oncology target for cancer therapy related to its critical role in cancer formation and progression. NTRK fusions are found to occur in 3.3% of lung cancers, 2.2% of colorectal cancers, 16.7% of thyroid cancers, 2.5% of glioblastomas, and 7.1% of pediatric gliomas. In this paper, we described the discovery of the type-II pan-TRK inhibitor 4c through the structure-based drug design strategy from the original hits 1b and 2b. Compound 4c exhibited excellent in vitro TRKA, TRKB, and TRKC kinase inhibitory activity and anti-proliferative activity against human colorectal carcinoma derived cell line KM12. In the NCI-60 human cancer cell lines screen, compound 4g demonstrated nearly 80% of growth inhibition for KM12, while only minimal inhibitory activity was observed for the remaining 59 cancer cell lines. Western blot analysis demonstrated that 4c and its urea cousin 4k suppressed the TPM3-TRKA autophosphorylation at the concentrations of 100 nM and 10 nM, respectively. The work presented that 2-(4-(thieno[3,2-d]pyrimidin-4-ylamino)phenyl)acetamides could serve as a novel scaffold for the discovery and development of type-II pan-TRK inhibitors for the treatment of TRK driven cancers.


Asunto(s)
Acetamidas/química , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/síntesis química , Receptor trkA/antagonistas & inhibidores , Receptor trkB/antagonistas & inhibidores , Acetamidas/metabolismo , Acetamidas/farmacología , Sitios de Unión , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/química , Pirimidinas/química , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Relación Estructura-Actividad
8.
Eur J Med Chem ; 206: 112691, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32823007

RESUMEN

Gene fusions and point mutations of RET kinase are crucial for driving thoracic cancers, including thyroid cancer and non-small cell lung cancer. Various scaffolds based on different heterocycles have been synthesized and evaluated as RET inhibitors. In this work, we investigate pyrrolo[2,3-d]pyrimidine derivatives for inhibition of RET-wt, drug resistant mutant RET V804M and RET gene fusion driven cell lines. Several compounds were synthesized and the structure activity relationship was extensively studied to optimize the scaffold. Thieno[2,3-d]pyrimidine, a bioisostere of pyrrolo[2,3-d]pyrimidine, was also explored for the effect on RET inhibition. We identified a lead compound, 59, which shows low nanomolar potency against RET-wt and RET V804M. Further 59 shows growth inhibition of LC-2/ad cells which RET-CCDC6 driven. We also determined that 59 is a type 2 inhibitor of RET and demonstrated its ability to inhibit migration of tumor cells. Based on computational studies, we proposed a binding pose of 59 in RET pocket and have quantified the contributions of individual residues for its binding. Together, 59 is an important lead compound which needs further evaluation in biological studies.


Asunto(s)
Diseño de Fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-ret/antagonistas & inhibidores , Pirimidinas/química , Pirimidinas/farmacología , Pirroles/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Humanos , Inhibidores de Proteínas Quinasas/síntesis química , Pirimidinas/síntesis química
9.
Eur J Med Chem ; 203: 112589, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32717530

RESUMEN

Aurora Kinase B is a serine-threonine kinase known to be overexpressed in several cancers, with no inhibitors approved for clinical use. Herein, we present the discovery and optimization of a series of novel quinazoline-based Aurora Kinase B inhibitors. The lead inhibitor SP-96 shows sub-nanomolar potency in Aurora B enzymatic assays (IC50 = 0.316 ± 0.031 nM). We identified the important pharmacophore features resulting in selectivity against receptor tyrosine kinases. Particularly, SP-96 shows >2000 fold selectivity against FLT3 and KIT which is important for normal hematopoiesis. This could diminish the adverse effect of neutropenia reported in the clinical trials of the Aurora B inhibitor Barasertib, which inhibits FLT3 and KIT in addition to Aurora B. Enzyme kinetics of SP-96 shows non-ATP-competitive inhibition which makes it a first-in-class inhibitor. Further, SP-96 shows selective growth inhibition in NCI60 screening, including inhibition of MDA-MD-468, a Triple Negative Breast Cancer cell line.


Asunto(s)
Adenosina Trifosfato/metabolismo , Aurora Quinasa B/antagonistas & inhibidores , Médula Ósea/efectos de los fármacos , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Unión Competitiva , Médula Ósea/inmunología , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Hematopoyesis/efectos de los fármacos , Humanos , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Quinazolinas/efectos adversos , Quinazolinas/química , Quinazolinas/metabolismo , Relación Estructura-Actividad
10.
Arch Anim Breed ; 63(1): 155-164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32490151

RESUMEN

Inner Mongolian Cashmere goat is an excellent local breed selected for the dual-purpose of cashmere and meat. There are three lines of Inner Mongolian Cashmere goat: Erlangshan, Alashan and Aerbasi. Cashmere is a kind of precious textile raw material with a high price. Cashmere is derived from secondary hair follicle (SHF), while hair is derived from primary hair follicle (PHF). The growth cycle of SHF of cashmere goat is 1 year, and it can be divided into three different stages: anagen, catagen and telogen. In this study, we tried to find some important influence factors of SHF growth cycle in skin tissue from Inner Mongolian Cashmere goats by RNA sequencing (RNA-Seq). Three female Aerbasi Inner Mongolian Cashmere goats (2 years old) were used as experimental samples in this study. Skin samples were collected in September (anagen), December (catagen) and March (telogen) at dorsal side from cashmere goats. Results showed that over 511 396 044 raw reads and 487 729 890 clean reads were obtained from sequence data. In total, 51 different expression genes (DEGs) including 29 downregulated genes and 22 upregulated genes were enriched in anagen-catagen comparing group. The 443 DEGs contained 117 downregulated genes and 326 upregulated genes that were enriched in catagen-telogen comparing group. In telogen-anagen comparing group, 779 DEGs were enriched including 582 downregulated genes and 197 upregulated genes. The result of gene ontology (GO) annotation showed that DEGs are in different growth cycle periods, and enriched GO items are mostly related to the transformation of cell and protein. The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment result indicated that metabolic process has a great impact on SHF growth cycle. Based on the results of a comprehensive analysis of differentially expressed genes, GO enrichment and KEGG enrichment, we found that FGF5, FGFR1 and RRAS had an effect on the hair follicle growth cycle. The results of this study may provide a theoretical basis for further research on the growth and development of SHF in Inner Mongolian Cashmere goats.

11.
J Med Chem ; 63(9): 4506-4516, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32298114

RESUMEN

RET receptor tyrosine kinase is a driver oncogene in human cancer. We recently identified the clinical drug candidate Pz-1, which targets RET and VEGFR2. A key in vivo metabolite of Pz-1 is its less active demethylated pyrazole analogue. Using bioisosteric substitution methods, here, we report the identification of NPA101.3, lacking the structural liability for demethylation. NPA101.3 showed a selective inhibitory profile and an inhibitory concentration 50 (IC50) of <0.003 µM for both RET and VEGFR2. NPA101.3 inhibited phosphorylation of all tested RET oncoproteins as well as VEGFR2 and proliferation of cells transformed by RET. Oral administration of NPA101.3 (10 mg/kg/day) completely prevented formation of tumors induced by RET/C634Y-transformed cells, while it weakened, but did not abrogate, formation of tumors induced by a control oncogene (HRAS/G12V). The balanced synchronous inhibition of both RET and VEGFR2, as well the resistance to demethylation, renders NPA101.3 a potential clinical candidate for RET-driven cancers.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-ret/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Descubrimiento de Drogas , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Mutación , Células 3T3 NIH , Polifarmacología , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
Yi Chuan ; 41(10): 928-938, 2019 Oct 20.
Artículo en Chino | MEDLINE | ID: mdl-31624055

RESUMEN

The goat genome is the research basis for the protection and utilization of goat resources, which is important for breeding and improving goat breeds. At present, with the continuous improvement of goat reference genome, various important research progress in goat origin, evolution and adaptability has been achieved. In this review, we summarize the research progress in the goat genome in detail, encompassing goat genome structure, genome map (genetic, physical and comparative maps), goat high throughput sequencing and SNP chip development. We aim to provide a theoretical foundation for the development of goat genome selection.


Asunto(s)
Mapeo Cromosómico , Genoma , Cabras/genética , Animales , Cruzamiento
13.
Chem Commun (Camb) ; 54(92): 12954-12957, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30375586

RESUMEN

Multicomponent reactions (MCRs) are robust tools for the rapid synthesis of complex, small molecule libraries for use in drug discovery and development. By utilizing MCR chemistry, we developed a protocol to functionalize the C-3 position of imidazo[1,2-a]pyridine through a three component, decarboxylation reaction involving imidazo[1,2-a]pyridine, glyoxalic acid, and boronic acid.


Asunto(s)
Descubrimiento de Drogas , Imidazoles/síntesis química , Piridinas/síntesis química , Antineoplásicos/síntesis química , Ácidos Borónicos/química , Línea Celular Tumoral , Descarboxilación , Humanos , Modelos Químicos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química
14.
Sci Rep ; 8(1): 3722, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29487300

RESUMEN

FMS-like Tyrosine Kinase 3 (FLT3) is a clinically validated target for acute myeloid leukemia (AML). Inhibitors targeting FLT3 have been evaluated in clinical studies and have exhibited potential to treat FLT3-driven AML. A frequent, clinical limitation is FLT3 selectivity, as concomitant inhibition of FLT3 and c-KIT is thought to cause dose-limiting myelosuppression. Through a rational design approach, novel FLT3 inhibitors were synthesized employing a pyridine/pyrimidine warhead. The most potent compound identified from the studies is compound 13a, which exhibited an IC50 value of 13.9 ± 6.5 nM against the FLT3 kinase with high selectivity over c-KIT. Mechanism of action studies suggested that 13a is a Type-II kinase inhibitor, which was also supported through computer aided drug discovery (CADD) efforts. Cell-based assays identified that 13a was potent on a variety of FLT3-driven cell lines with clinical relevance. We report herein the discovery and therapeutic evaluation of 4,6-diamino pyrimidine-based Type-II FLT3 inhibitors, which can serve as a FLT3-selective scaffold for further clinical development.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/química , Tirosina Quinasa 3 Similar a fms/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad
15.
J Appl Toxicol ; 36(9): 1090-4, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26663444

RESUMEN

The aim of the present study was to identify human cytosolic sulfotransferases (SULTs) that are capable of sulfating benzyl alcohol and to examine whether benzyl alcohol sulfation may occur in cultured human cells as well as in human organ homogenates. A systematic analysis revealed that of the 13 known human SULTs, SULT1A1 SULT1A2, SULTA3, and SULT1B1 are capable of mediating the sulfation of benzyl alcohol. The kinetic parameters of SULT1A1 that showed the strongest benzyl alcohol-sulfating activity were determined. HepG2 human hepatoma cells were used to demonstrate the generation and release of sulfated benzyl alcohol under the metabolic settings. Moreover, the cytosol or S9 fractions of human liver, lung, kidney and small intestine were examined to verify the presence of benzyl alcohol sulfating activity in vivo. Copyright © 2015 John Wiley & Sons, Ltd.


Asunto(s)
Arilsulfotransferasa/metabolismo , Alcohol Bencilo/toxicidad , Citosol/efectos de los fármacos , Células CACO-2 , Citosol/metabolismo , Células Hep G2 , Humanos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Sulfatos/metabolismo
16.
J Pharmacol Sci ; 128(3): 144-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26169578

RESUMEN

Previous studies demonstrated that sulfate conjugation is involved in the metabolism of three commonly used breast cancer drugs, tamoxifen, raloxifene and fulvestrant. The current study was designed to systematically identify the human cytosolic sulfotransferases (SULTs) that are capable of sulfating raloxifene, fulvestrant, and two active metabolites of tamoxifen, afimoxifene and endoxifen. A systematic analysis using 13 known human SULTs revealed SULT1A1 and SULT1C4 as the major SULTs responsible for the sulfation of afimoxifene, endoxifen, raloxifene and fulvestrant. Kinetic parameters of these two human SULTs in catalyzing the sulfation of these drug compounds were determined. Sulfation of afimoxifene, endoxifen, raloxifene and fulvestrant under metabolic conditions was examined using HepG2 human hepatoma cells and MCF-7 breast cancer cells. Moreover, human intestine, kidney, liver, and lung cytosols were examined to verify the presence of afimoxifene/endoxifen/raloxifene/fulvestrant-sulfating activity.


Asunto(s)
Citosol/enzimología , Estradiol/análogos & derivados , Clorhidrato de Raloxifeno/metabolismo , Sulfotransferasas/fisiología , Tamoxifeno/análogos & derivados , Catálisis , Estradiol/metabolismo , Fulvestrant , Células Hep G2 , Humanos , Células MCF-7 , Sulfatos , Tamoxifeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...