Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ACS Med Chem Lett ; 14(4): 396-404, 2023 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-37077386

RESUMEN

Deregulating fibroblast growth factor receptor (FGFR) signaling is a promising strategy for cancer therapy. Herein, we report the discovery of compound 5 (TAS-120, futibatinib), a potent and selective covalent inhibitor of FGFR1-4, starting from a unique dual inhibitor of mutant epidermal growth factor receptor and FGFR (compound 1). Compound 5 inhibited all four families of FGFRs in the single-digit nanomolar range and showed high selectivity for over 387 kinases. Binding site analysis revealed that compound 5 covalently bound to the cysteine 491 highly flexible glycine-rich loop region of the FGFR2 adenosine triphosphate pocket. Futibatinib is currently in Phase I-III trials for patients with oncogenically driven FGFR genomic aberrations. In September 2022, the U.S. Food & Drug Administration granted accelerated approval for futibatinib in the treatment of previously treated, unresectable, locally advanced, or metastatic intrahepatic cholangiocarcinoma harboring an FGFR2 gene fusion or other rearrangement.

2.
Int J Cancer ; 152(12): 2580-2593, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36752576

RESUMEN

Despite the effectiveness of imatinib, most gastrointestinal stromal tumors (GISTs) develop resistance to the treatment, mainly due to the reactivation of KIT tyrosine kinase activity. Sunitinib, which inhibits the phosphorylation of KIT and vascular endothelial growth factor (VEGF) receptor, has been established as second-line therapy for GISTs. The recently-developed heat shock protein 90 (HSP90) inhibitor pimitespib (PIM; TAS-116) demonstrated clinical benefits in some clinical trials; however, the effects were limited. The aim of our study was therefore to clarify the effectiveness and mechanism of the combination of PIM with sunitinib for imatinib-resistant GISTs. We evaluated the efficacy and mechanism of the combination of PIM with sunitinib against imatinib-resistant GIST using imatinib-resistant GIST cell lines and murine xenograft models. In vitro analysis demonstrated that PIM and sunitinib combination therapy strongly inhibited growth and induced apoptosis in imatinib-resistant GIST cell lines by inhibiting KIT signaling and decreasing auto-phosphorylated KIT in the Golgi apparatus. In addition, PIM and sunitinib combination therapy enhanced antitumor responses in the murine xenograft models compared to individual therapies. Further analysis of the xenograft models showed that the combination therapy not only downregulated the KIT signaling pathway but also decreased the tumor microvessel density. Furthermore, we found that PIM suppressed VEGF expression in GIST cells by suppressing protein kinase D2 and hypoxia-inducible factor-1 alpha, which are both HSP90 client proteins. In conclusion, the combination of PIM and sunitinib is effective against imatinib-resistant GIST via the downregulation of KIT signaling and angiogenic signaling pathways.


Asunto(s)
Antineoplásicos , Tumores del Estroma Gastrointestinal , Humanos , Animales , Ratones , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Sunitinib/farmacología , Sunitinib/uso terapéutico , Tumores del Estroma Gastrointestinal/patología , Factor A de Crecimiento Endotelial Vascular , Piperazinas/farmacología , Pirimidinas , Resistencia a Antineoplásicos , Antineoplásicos/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
3.
Mol Cancer Ther ; 18(7): 1205-1216, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31092565

RESUMEN

NEDD8-activating enzyme (NAE) is an essential E1 enzyme of the NEDD8 conjugation (neddylation) pathway, which controls cancer cell growth and survival through activation of cullin-RING ubiquitin ligase complexes (CRL). In this study, we describe the preclinical profile of a novel, highly potent, and selective NAE inhibitor, TAS4464. TAS4464 selectively inhibited NAE relative to the other E1s UAE and SAE. TAS4464 treatment inhibited cullin neddylation and subsequently induced the accumulation of CRL substrates such as CDT1, p27, and phosphorylated IκBα in human cancer cell lines. TAS4464 showed greater inhibitory effects than those of the known NAE inhibitor MLN4924 both in enzyme assay and in cells. Cytotoxicity profiling revealed that TAS4464 is highly potent with widespread antiproliferative activity not only for cancer cell lines, but also patient-derived tumor cells. TAS4464 showed prolonged target inhibition in human tumor xenograft mouse models; weekly or twice a week TAS4464 administration led to prominent antitumor activity in multiple human tumor xenograft mouse models including both hematologic and solid tumors without marked weight loss. As a conclusion, TAS4464 is the most potent and highly selective NAE inhibitor reported to date, showing superior antitumor activity with prolonged target inhibition. It is, therefore, a promising agent for the treatment of a variety of tumors including both hematologic and solid tumors. These results support the clinical evaluation of TAS4464 in hematologic and solid tumors.


Asunto(s)
Proteína NEDD8/genética , Neoplasias/tratamiento farmacológico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones SCID , Pirimidinas/farmacología , Pirroles/farmacología
4.
Int J Oncol ; 46(6): 2327-34, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25901475

RESUMEN

Trifluridine (FTD) and 2'-deoxy-5-fluorouridine (FdUrd), a derivative of 5-fluorouracil (5-FU), are antitumor agents that inhibit thymidylate synthase activity and their nucleotides are incorporated into DNA. However, it is evident that several differences occur in the underlying antitumor mechanisms associated with these nucleoside analogues. Recently, TAS-102 (composed of FTD and tipiracil hydrochloride, TPI) was shown to prolong the survival of patients with colorectal cancer who received a median of 2 prior therapies, including 5-FU. TAS-102 was recently approved for clinical use in Japan. These data suggest that the antitumor activities of TAS-102 and 5-FU proceed via different mechanisms. Thus, we analyzed their properties in terms of thymidine salvage pathway utilization, involving membrane transporters, a nucleoside kinase, a nucleotide-dephosphorylating enzyme, and DNA polymerase α. FTD incorporated into DNA with higher efficiency than FdUrd did. Both FTD and FdUrd were transported into cells by ENT1 and ENT2 and were phosphorylated by thymidine kinase 1, which showed a higher catalytic activity for FTD than for FdUrd. deoxyUTPase (DUT) did not recognize dTTP and FTD-triphosphate (F3dTTP), whereas deoxyuridine-triphosphate (dUTP) and FdUrd-triphosphate (FdUTP) were efficiently degraded by DUT. DNA polymerase α incorporated both F3dTTP and FdUTP into DNA at sites aligned with adenine on the opposite strand. FTD-treated cells showed differing nuclear morphologies compared to FdUrd-treated cells. These findings indicate that FTD and FdUrd are incorporated into DNA with different efficiencies due to differences in the substrate specificities of TK1 and DUT, causing abundant FTD incorporation into DNA.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Neoplasias Colorrectales/genética , ADN de Neoplasias/química , Fluorouracilo/farmacología , Timidina Quinasa/metabolismo , Trifluridina/farmacología , Uracilo/análogos & derivados , Antimetabolitos Antineoplásicos/farmacocinética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Combinación de Medicamentos , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Transportador Equilibrativo 2 de Nucleósido/metabolismo , Fluorouracilo/farmacocinética , Células HCT116 , Humanos , Masculino , Pirofosfatasas/metabolismo , Pirrolidinas , Especificidad por Sustrato , Timina , Trifluridina/farmacocinética , Uracilo/farmacocinética , Uracilo/farmacología
5.
Oncol Rep ; 32(6): 2319-26, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25230742

RESUMEN

TAS-102 is a novel oral nucleoside antitumor agent containing trifluridine (FTD) and tipiracil hydrochloride (TPI). The compound improves overall survival of colorectal cancer (CRC) patients who are insensitive to standard chemotherapies. FTD possesses direct antitumor activity since it inhibits thymidylate synthase (TS) and is itself incorporated into DNA. However, the precise mechanisms underlying the incorporation into DNA and the inhibition of TS remain unclear. We found that FTD-dependent inhibition of TS was similar to that elicited by fluorodeoxyuridine (FdUrd), another clinically used nucleoside analog. However, washout experiments revealed that FTD-dependent inhibition of TS declined rapidly, whereas FdUrd activity persisted. The incorporation of FTD into DNA was significantly higher than that of other antitumor nucleosides. Additionally, orally administered FTD had increased antitumor activity and was incorporated into DNA more effectively than continuously infused FTD. When TAS-102 was administered, FTD gradually accumulated in tumor cell DNA, in a TPI-independent manner, and significantly delayed tumor growth and prolonged survival, compared to treatment with 5-FU derivatives. TAS-102 reduced the Ki-67-positive cell fraction, and swollen nuclei were observed in treated tumor tissue. The amount of FTD incorporation in DNA and the antitumor activity of TAS-102 in xenograft models were positively and significantly correlated. These results suggest that TAS-102 exerts its antitumor activity predominantly due to its DNA incorporation, rather than as a result of TS inhibition. The persistence of FTD in the DNA of tumor cells treated with TAS-102 may underlie its ability to prolong survival in cancer patients.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , ADN de Neoplasias/genética , Trifluridina/administración & dosificación , Uracilo/análogos & derivados , Administración Oral , Animales , Antineoplásicos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Combinación de Medicamentos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Pirrolidinas , Timina , Trifluridina/metabolismo , Uracilo/administración & dosificación , Uracilo/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Int J Oncol ; 34(5): 1373-80, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19360349

RESUMEN

1-(3-C-Ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd, TAS-106) is a novel antitumor ribonucleoside that inhibits RNA polymerase. In the present study, we investigated the cellular and molecular interactions between TAS-106 and cisplatin (CDDP) in vitro using A549 human lung cancer cells and the in vivo antitumor effect of combined treatment using OCC-1 and LX-1 human tumor xenografts. The treatment effects were determined by evaluating cytotoxicity, the cell cycle distribution, apoptosis induction and the expression of checkpoint-associated proteins. In vitro, the combination of TAS-106 and CDDP synergistically inhibited the growth of A549 cells, as determined using isobologram analysis. TAS-106 potently inhibited the expression of Chk1 protein and the phosphorylation of Chk1 and Chk2. Moreover, based on the inhibition of checkpoint-associated protein, TAS-106 abrogated the CDDP-induced S- and G2M-checkpoints and induced apoptosis in A549 cells. In vivo, TAS-106 alone showed antitumor activity; however, its combination with CDDP significantly enhanced the growth inhibition of OCC-1 and LX-1 tumors. Moreover, combination therapy with TAS-106 and CDDP in the OCC-1 xenograft model resulted in significant life-prolongation. These findings provide a rationale for combination chemotherapy using TAS-106 and CDDP in clinical settings.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Cisplatino/administración & dosificación , Citidina/análogos & derivados , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Animales , Cisplatino/farmacología , Citidina/administración & dosificación , Citidina/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Neoplasias/patología , Ratas , Ratas Desnudas , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncol Rep ; 17(6): 1453-60, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487404

RESUMEN

1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd, TAS-106), is a new antitumor cytidine analogue, inhibiting RNA synthesis. In this study we investigated the cellular growth inhibition, intracellular metabolism, cell cycle phase specificity, and RNA synthesis of TAS-106 compared with those of 5-fluorouracil (5-FU), known to possess both DNA- (inhibition of thymidylate synthase activity) and RNA-synthesis-inhibiting activity (inhibition of RNA function). The IC50 values of TAS-106 and 5-FU ranged from 0.0173 to 3.11 microM, and from 6.80 to >1,000 microM, respectively, in a panel of 10 human tumor cells, indicating that TAS-106 possesses greater cytotoxicity than 5-FU. Using excess thymidine-synchronized cells, TAS-106 and 5-FU appeared to exert their cytotoxic effects independently of the cell cycle. The intracellular metabolism and the effect on pre-rRNA processing of TAS-106 differed from those of 5-FU. More than 50% of 5-FU incorporated into the cells was in the unchanged form, while 5-FU incorporated into RNA was approximately 20%. On the other hand, TAS-106 was incorporated in a time-dependent manner into the cells and rapidly converted to its mono-, di- and tri-phosphate form, however, the amount incorporated into RNA fraction was very small. 5-FU incorporated into RNA was confirmed to impair the normal processing of ribosomal RNA (formation of 34/32S RNA from 45S RNA), however, TAS-106 did not affect pre-rRNA processing and may be involved in the inhibition of the synthesis of ribosomal RNA. We concluded that intracellular accumulation and retention of the active metabolite of TAS-106, 3'-ethynylcytidine 5'-triphosphate (ECTP), may contribute to its potent cytotoxicity. The unique mechanism of antitumor activity and intensive cellular metabolism of TAS-106 could contribute to cancer chemotherapy through the pathways different from those of 5-FU or other antitumor nucleosides.


Asunto(s)
Antineoplásicos/farmacología , Citidina/análogos & derivados , ARN Ribosómico/antagonistas & inhibidores , Ribonucleósidos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citidina/análisis , Citidina/metabolismo , Citidina/farmacología , Fluorouracilo/análisis , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Humanos , Concentración 50 Inhibidora , Precursores del ARN/antagonistas & inhibidores , Precursores del ARN/química , Precursores del ARN/metabolismo , ARN Ribosómico/química , ARN Ribosómico/metabolismo
8.
Cancer Sci ; 96(5): 295-302, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15904471

RESUMEN

We established a variant of MIAPaCa-2 human pancreatic cancer cells that is resistant to 2',2'-difluorodeoxycytidine (gemcitabine, dFdCyd), MIAPaCa-2/dFdCyd, and elucidated the biochemical characteristics and mechanism of dFdCyd-resistance in these cells. We also evaluated 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd, TAS-106, RNA polymerase inhibitor), a new anticancer ribonucleoside, for antitumor activity against the resistant cells in vitro and in vivo. MIAPaCa-2/dFdCyd cells were 2541-fold more resistant to dFdCyd than parental MIAPaCa-2 cells, and the major mechanism of the dFdCyd-resistance was found to be a decrease in the intracellular pool of dFdCyd and its active metabolites, which would result in a decrease in incorporation of dFdCyd triphosphate into DNA. This finding was confirmed by the discovery of decreased deoxycytidine kinase activity, increased cytidine deaminase and ribonucleotide reductase activity, and increased 5'-nucleotidase mRNA expression in the MIAPaCa-2/dFdCyd cells. The cytotoxicity of TAS-106 as an antitumor nucleoside analog was similar in both parental and dFdCyd-resistant cells, with IC(50) values of 6.25 and 6.27 nM, respectively, and this finding was supported by similar intracellular uptake and metabolism of TAS-106 in both cell lines. We also evaluated the in vivo antitumor activity of TAS-106 against MIAPaCa-2 and dFdCyd-resistant MIAPaCa-2/dFdCyd tumors implanted into nude mice. The tumor growth inhibition rate of weekly additions of TAS-106 (7 mg/kg, iv) against parental and dFdCyd-resistant tumors was 73% and 76%, respectively, while that of dFdCyd administered twice a week (240 mg/kg, iv) was 84% and 34%, respectively. These results suggest that TAS-106 would contribute to the treatment of patients with advanced pancreatic carcinomas in whom dFdCyd-based chemotherapy has failed.


Asunto(s)
Antineoplásicos/farmacología , Citidina/análogos & derivados , Citidina/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos , Neoplasias Pancreáticas/patología , Animales , Línea Celular Tumoral , Desoxirribonucleótidos/metabolismo , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleótidos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
9.
Jpn J Cancer Res ; 93(7): 825-33, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12149149

RESUMEN

TAS-106 [1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine] is a new anticancer ribo-nucleoside with promising antitumor activity. We have previously presented evidence suggesting that the TAS-106 sensitivity of cells is correlated with intracellular accumulation of the triphosphate of TAS-106, which may be affected both by cellular membrane transport mechanisms and uridine-cytidine kinase (UCK) activity. Since the presence of a UCK family consisting of two members, UCK1 and UCK2, has recently been reported in human cells, we investigated the relation between expression of UCK1 and UCK2 at both the mRNA and protein levels and UCK activity (TAS-106 phosphorylation activity) in a panel of 10 human cancer cell lines. Measurement of UCK activity in these cell lines revealed that it was well correlated with the cells' sensitivity to TAS-106. In addition, the mRNA or protein expression level of UCK2 was closely correlated with UCK activity in these cell lines, but neither the level of expression of UCK1 mRNA nor that of protein was correlated with enzyme activity. We therefore compared the protein expression level of UCK2 in several human tumor tissues and the corresponding normal tissues. Expression of UCK2 protein was barely detectable in 4 of the 5 human tumor tissues, but tended to be high in the pancreatic tumor tissue. It could not be detected at all in any of the normal tissues. Thus, expression of UCK2 appeared to be correlated with cellular sensitivity to TAS-106, and it may contribute to the tumor-selective cytotoxicity of TAS-106.


Asunto(s)
Antineoplásicos/farmacología , Citidina/análogos & derivados , Citidina/farmacología , Western Blotting , Citosol/metabolismo , Humanos , Modelos Biológicos , Nucleósido-Fosfato Quinasa/metabolismo , Fosforilación , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Uridina Quinasa/metabolismo
10.
Jpn J Cancer Res ; 93(4): 445-52, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11985795

RESUMEN

We have established variants of DLD-1 human colon carcinoma and HT-1080 human fibrosarcoma cells resistant to the new anticancer ribo-nucleosides, 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)-cytosine (ECyd, TAS-106) and 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)uracil (EUrd). Both variants were shown to have decreased (3- to 24-fold decrease) uridine-cytidine kinase (UCK) activity, and exhibited cross-resistance to EUrd and TAS-106. Based on the IC(50) values determined by chemosensitivity testing, a 41- to 1102-fold resistance to TAS-106 was observed in the resistant cells. TAS-106 concentration-dependently inhibited RNA synthesis, while its effect on DNA synthesis was negligible. The degree of resistance (14- to 3628-fold resistance) calculated from the inhibition of RNA synthesis tended to be close to the degree of chemoresistance of tested cells to TAS-106. The experiments on the intracellular metabolism of TAS-106 in the parental cells revealed a rapid phosphorylation to its nucleotides, particularly the triphosphate (ECTP), its major active metabolite. The amount of TAS-106 transported into the resistant cells was markedly reduced and the intracellular level of ECTP was decreased from 1/19 to below the limit of detection; however, the unmetabolized TAS-106 as a percentage of the total metabolite level was high as compared with the parental cells. The ratio of the intracellular level of ECTP between parental and resistant cells tended to approximate to the degree of resistance calculated from the inhibitory effect on RNA synthesis. These results indicate that the TAS-106 sensitivity of cells is correlated with the intracellular accumulation of ECTP, which may be affected by both the cellular membrane transport mechanism and UCK activity.


Asunto(s)
Antineoplásicos/farmacología , Citidina/análogos & derivados , Citidina/farmacología , Resistencia a Antineoplásicos , Uridina Quinasa/metabolismo , Transporte Biológico , Línea Celular Tumoral , Membrana Celular/metabolismo , Cromatografía Líquida de Alta Presión , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Modelos Biológicos , Modelos Químicos , Nucleósido-Fosfato Quinasa/metabolismo , Fosforilación , ARN/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...