Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Endocr Relat Cancer ; 30(12)2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37902083

RESUMEN

Ataxia telangiectasia and Rad3-related protein (ATR) is a critical component of the DNA damage response and a potential target in the treatment of cancers. An ATR inhibitor, BAY 1895344, was evaluated for its use in differentiated thyroid cancer (DTC) therapy. BAY 1895344 inhibited cell viability in four DTC cell lines (TPC1, K1, FTC-133, and FTC-238) in a dose-dependent manner. BAY 1895344 treatment arrested DTC cells in the G2/M phase, increased caspase-3 activity, and caused apoptosis. BAY 1895344 in combination with either sorafenib or lenvatinib showed mainly synergistic effects in four DTC cell lines. The combination of BAY 1895344 with dabrafenib plus trametinib revealed synergistic effects in K1 cells that harbor BRAFV600E. BAY 1895344 monotherapy retarded the growth of K1 and FTC-133 tumors in xenograft models. The combinations of BAY 1895344 plus lenvatinib and BAY 1895344 with dabrafenib plus trametinib were more effective than any single therapy in a K1 xenograft model. No appreciable toxicity appeared in animals treated with either a single therapy or a combination treatment. Our findings provide the rationale for the development of clinical trials of BAY 1895344 in the treatment of DTC.


Asunto(s)
Adenocarcinoma , Neoplasias de la Tiroides , Animales , Humanos , Neoplasias de la Tiroides/patología , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Sorafenib/farmacología , Adenocarcinoma/tratamiento farmacológico , Proteínas de la Ataxia Telangiectasia Mutada
2.
Cancers (Basel) ; 13(14)2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34298699

RESUMEN

Differentiated thyroid cancer (DTC) patients are usually known for their excellent prognoses. However, some patients with DTC develop refractory disease and require novel therapies with different therapeutic mechanisms. Targeting Wee1 with adavosertib has emerged as a novel strategy for cancer therapy. We determined the effects of adavosertib in four DTC cell lines. Adavosertib induces cell growth inhibition in a dose-dependent fashion. Cell cycle analyses revealed that cells were accumulated in the G2/M phase and apoptosis was induced by adavosertib in the four DTC tumor cell lines. The sensitivity of adavosertib correlated with baseline Wee1 expression. In vivo studies showed that adavosertib significantly inhibited the xenograft growth of papillary and follicular thyroid cancer tumor models. Adavosertib therapy, combined with dabrafenib and trametinib, had strong synergism in vitro, and revealed robust tumor growth suppression in vivo in a xenograft model of papillary thyroid cancer harboring mutant BRAFV600E, without appreciable toxicity. Furthermore, combination of adavosertib with lenvatinib was more effective than either agent alone in a xenograft model of follicular thyroid cancer. These results show that adavosertib has the potential in treating DTC.

3.
Endocr Relat Cancer ; 28(5): 311-324, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33769310

RESUMEN

Wee1 is a kinase that regulates the G2/M progression by the inhibition of CDK1, which is critical for ensuring DNA damage repair before initiation of mitotic entry. Targeting Wee1 may be a potential strategy in the treatment of anaplastic thyroid cancer, a rare but lethal disease. The therapeutic effects of adavosertib, a Wee1 inhibitor for anaplastic thyroid cancer was evaluated in this study. Adavosertib inhibited cell growth in three anaplastic thyroid cancer cell lines in a dose-dependent manner. Cell cycle analysis revealed cells were accumulated in the G2/M phase. Adavosertib induced caspase-3 activity and led to apoptosis. Adavosertib monotherapy showed significant retardation of the growth of two anaplastic thyroid cancer tumor models. The combination of adavosertib with dabrafenib and trametinib revealed strong synergism in vitro and demonstrated robust suppression of tumor growth in vivo in anaplastic thyroid cancer xenograft models with BRAFV600E mutation. The combination of adavosertib with either sorafenib or lenvatinib also demonstrated synergism in vitro and had strong inhibition of tumor growth in vivo in an anaplastic thyroid cancer xenograft model. No appreciable toxicity appeared in mice treated with either a single agent or combination treatment. Our findings suggest adavosertib holds the promise for the treatment of patients with anaplastic thyroid cancer.


Asunto(s)
Carcinoma Anaplásico de Tiroides , Neoplasias de la Tiroides , Animales , Línea Celular Tumoral , Humanos , Ratones , Pirazoles/efectos adversos , Pirimidinonas , Sorafenib , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/inducido químicamente , Neoplasias de la Tiroides/tratamiento farmacológico
4.
Cancer Sci ; 112(2): 803-814, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33306266

RESUMEN

Polo-like kinases (PLKs) are potent regulators of cell proliferation and cell survival. Polo-like kinases are potential targets in the treatment of anaplastic thyroid cancer (ATC), a rare but deadly disease. The therapeutic effects of volasertib, a PLK inhibitor, was evaluated for the treatment of ATC either alone or in combination with sorafenib. Volasertib decreased cell viability in three ATC cell lines (8505C, 8305C, and KAT18) in a dose-dependent manner. Volasertib caused ATC cells to accumulate in G2 /M phase, activated caspase-3 activity, and induced apoptosis. Combination therapy using volasertib and sorafenib in ATC cells showed mostly synergistic effects. In vivo studies revealed that combination therapy of volasertib and sorafenib was effective in the treatment of 8505C xenografts. Single-agent volasertib treatment was sufficient to retard 8305C tumor growth. No substantial morbidity was observed in animals that received either single-agent or combination treatment. These preclinical findings suggest that volasertib could be an effective drug in treating ATC.


Asunto(s)
Antineoplásicos/farmacología , Pteridinas/farmacología , Carcinoma Anaplásico de Tiroides/patología , Neoplasias de la Tiroides/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Ratones , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Endocr Relat Cancer ; 26(8): 727-738, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189135

RESUMEN

Polo-like kinases (PLKs) are pivotal regulators of cell proliferation and cell survival; therefore, PLKs may be potential targets in the treatment of malignancy. The therapeutic effects of volasertib, a PLKs inhibitor for papillary and follicular thyroid cancer (known as well-differentiated thyroid cancer (WDTC)), were evaluated in this study. Volasertib inhibited cell proliferation in two papillary and two follicular thyroid cancer cell lines in a dose-dependent manner. Volasertib treatment reduced cells in the S phase and increased cells in the G2/M phase. Volasertib activated caspase-3 activity and induced apoptosis. Drug combinations of volasertib and sorafenib showed mostly synergism in four well-differentiated thyroid carcinoma cell lines in vitro. Volasertib treatment in vivo retarded the growth of a papillary thyroid tumor model. Furthermore, the combination of volasertib with sorafenib was more effective than a single treatment of either in a follicular thyroid cancer xenograft model. Promising safety profiles appeared in animals treated with either volasertib alone or volasertib and sorafenib combination therapy. These findings support volasertib as a potential drug for the treatment of patients with WDTC.


Asunto(s)
Adenocarcinoma Folicular/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Cáncer Papilar Tiroideo/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/metabolismo , Adenocarcinoma Folicular/patología , Animales , Apoptosis/efectos de los fármacos , Bencimidazoles/administración & dosificación , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/administración & dosificación , Pteridinas/administración & dosificación , Sorafenib/administración & dosificación , Tiofenos/administración & dosificación , Cáncer Papilar Tiroideo/metabolismo , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
6.
Oncotarget ; 9(46): 28030-28041, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29963260

RESUMEN

Altered cyclin-dependent kinase activity is observed in many human malignancies. Cyclin-dependent kinases that promote cell cycle progression may be promising targets in the treatment of cancer. The therapeutic effects of roniciclib, a cyclin-dependent kinase inhibitor for medullary thyroid cancer were investigated in the present study. Roniciclib inhibited medullary thyroid cancer cell proliferation in a dose-dependent manner. Roniciclib induced caspase-3 activity and contributed to apoptosis. Cell cycle progression was arrested in the G2 phase. In vivo, roniciclib treatment retarded the growth of tumors of medullary thyroid cancer xenografts. In addition, roniciclib in combination with sorafenib was more effective than either single treatment in a xenograft model. No morbidity was observed in animals treated with single roniciclib therapy and combination treatment of roniciclib and sorafenib. These data provide a rationale for clinical assessment of using roniciclib in the treatment of patients with medullary thyroid cancer.

7.
Endocr Relat Cancer ; 25(10): 853-864, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29895526

RESUMEN

Activation of cyclin-dependent kinase activity is frequently observed in many human cancers; therefore, cyclin-dependent kinases that promote cell cycle transition and cell proliferation may be potential targets in the treatment of malignancy. The therapeutic effects of roniciclib, a cyclin-dependent kinase inhibitor for papillary and follicular thyroid cancer (designated as well-differentiated thyroid cancer), were investigated in this study. Roniciclib inhibited cell proliferation in two papillary and two follicular thyroid cancer cell lines in a dose-dependent manner. Roniciclib activated caspase-3 activity and induced apoptosis. Cell cycle progression was arrested in the G2/M phase. Roniciclib treatment in vivo retarded the growth of two well-differentiated thyroid tumors in xenograft models in a dose-dependent fashion. Furthermore, the combination of roniciclib with sorafenib was more effective than either single treatment in a follicular thyroid cancer xenograft model. Acceptable safety profiles appeared in animals treated with either roniciclib alone or roniciclib and sorafenib combination therapy. These findings support roniciclib as a potential drug for the treatment of patients with well-differentiated thyroid cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Pirimidinas/farmacología , Sorafenib/farmacología , Sulfóxidos/farmacología , Neoplasias de la Tiroides/patología , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Neoplasias de la Tiroides/metabolismo
8.
Oncotarget ; 8(40): 67990-68000, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978090

RESUMEN

Many human cancers have altered cyclin-dependent kinase activity. Inhibition of cyclin-dependent kinases may arrest cell cycle progression and represents an important strategy in the treatment of malignancies. We evaluated the therapeutic effects of roniciclib, a cyclin-dependent kinase inhibitor, as a treatment for anaplastic thyroid cancer. Roniciclib inhibited anaplastic thyroid cancer cell proliferation in a dose-dependent manner. Roniciclib activated caspase-3 activity and induced apoptosis. Cell cycle progression was arrested in G2/M phase. In vivo, the growth of anaplastic thyroid cancer xenograft tumors was retarded by roniciclib treatment without evidence of toxicity. These data provide a rationale for further clinical evaluation using roniciclib in the treatment of patients with anaplastic thyroid cancer.

9.
Oncotarget ; 8(25): 41294-41304, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28476040

RESUMEN

Heat shock protein 90 is a molecular chaperon that maintains the correct folding and function of multiple client proteins. The inhibition of heat shock protein 90, which leads to the simultaneous degradation of multiple proteins involved in oncogenic signaling pathways, has revealed an innovative strategy to treat a variety of cancer types. We evaluated the therapeutic effects of ganetespib, a heat shock protein 90 inhibitor, in treating thyroid cancer. Ganetespib effectively inhibited cell proliferation in a dose-dependent manner in eight cell lines originating from four major histologic types of thyroid cancer (papillary, follicular, anaplastic and medullary). Ganetespib decreased cyclin-dependent kinase 1 and arrested cell cycle progression in G2/M phase. The expression of proteins involved in RAS/RAF/ERK and PI3K/AKT/mTOR signaling pathways was also inhibited. The RET level was decreased in a medullary thyroid cancer cell line. Ganetespib increased Bim expression, activated caspase-3 and induced apoptosis. In vivo, ganetespib retarded the tumor growth of anaplastic and medullary thyroid cancer xenografts with acceptable safety profiles. These findings indicate that ganetespib has potential in the treatment of patients with thyroid cancer.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias de la Tiroides/tratamiento farmacológico , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Neoplasias de la Tiroides/metabolismo
10.
PLoS One ; 12(2): e0172315, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28207834

RESUMEN

BACKGROUND: We explored the therapeutic effects of dinaciclib, a cyclin-dependent kinase (CDK) inhibitor, in the treatment of thyroid cancer. MATERIALS AND METHODS: Seven cell lines originating from three pathologic types of thyroid cancer (papillary, follicular and anaplastic) were studied. The cytotoxicity of dinaciclib was measured using a lactate dehydrogenase assay. The expression of proteins associated with cell cycle and apoptosis was assessed using Western blot analysis and immunofluorescence microscopy. Cell cycle distribution was measured by flow cytometry and immunofluorescence microscopy. Apoptosis and caspase-3 activity were measured by flow cytometry and fluorometric assay. Mice bearing flank anaplastic thyroid cancer (ATC) were treated with intraperitoneal injections of dinaciclib. RESULTS: Dinaciclib inhibited thyroid cancer cell proliferation in a dose-dependent manner. Dinaciclib had a low median-effect dose (≤ 16.0 nM) to inhibit cell proliferation in seven thyroid cancer cell lines. Dinaciclib decreased CDK1, cyclin B1, and Aurora A expression, induced cell cycle arrest in the G2/M phase, and induced accumulation of prophase mitotic cells. Dinaciclib decreased Mcl-1, Bcl-xL and survivin expression, activated caspase-3 and induced apoptosis. In vivo, the growth of ATC xenograft tumors was retarded in a dose-dependent fashion with daily dinaciclib treatment. Higher-dose dinaciclib (50 mg/kg) caused slight, but significant weight loss, which was absent with lower-dose dinaciclib (40 mg/kg) treatment. CONCLUSIONS: Dinaciclib inhibited thyroid cancer proliferation both in vitro and in vivo. These findings support dinaciclib as a potential drug for further studies in clinical trials for the treatment of patients with refractory thyroid cancer.


Asunto(s)
Adenocarcinoma Folicular/tratamiento farmacológico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Carcinoma Papilar/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Compuestos de Piridinio/farmacología , Neoplasias de la Tiroides/tratamiento farmacológico , Adenocarcinoma Folicular/metabolismo , Adenocarcinoma Folicular/patología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma/metabolismo , Carcinoma/patología , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Óxidos N-Cíclicos , Femenino , Citometría de Flujo , Humanos , Indolizinas , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Arch Toxicol ; 91(7): 2677-2687, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27915442

RESUMEN

The global incidence of Fusarium head blight and attendant cereal grains multi-contamination by the trichothecene mycotoxins deoxynivalenol (DON) and nivalenol (NIV) are increasing as a possible result of climate change and inadequate agricultural practices. At the molecular level, these mycotoxins bind to the ribosome, activate the mitogen-activated protein kinase and induce a local and systemic inflammation. DON is of public health concern owing to the narrow margin between exposure and tolerable daily intake. The intestinal inflammatory response to DON, NIV and their mixture was analyzed to determine thresholds for their intestinal pro-inflammatory effects and characterize the type and magnitude of their interaction. Fully differentiated three-dimensional porcine jejunal explants were exposed to increasing doses of DON and NIV alone or in combination; the expression levels of IL-1α, IL-1ß, IL-8, IL-17A and IL-22 were measured by RT-PCR. Doses as low as 0.16 µM DON or 0.73 µM NIV significantly increase the intestinal expression levels of the tested inflammation-related genes. These doses are lower than those previously reported for other intestinal toxicity endpoints. The combined pro-inflammatory activity of DON and NIV was synergistic for all the tested genes with combination index value range of 0.23-0.8. Our results indicate that (1) inflammation is a very sensitive endpoint for the intestinal toxicity of the trichothecenes and (2) co-exposure to DON and NIV has a greater inflammatory effect than induced by mycotoxins alone. This synergy should be taken into account considering the frequent co-occurrence of DON and NIV in the diet.


Asunto(s)
Contaminación de Alimentos , Yeyuno/efectos de los fármacos , Tricotecenos/toxicidad , Animales , Citocinas/genética , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Enteritis/inducido químicamente , Enteritis/patología , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedades del Yeyuno/inducido químicamente , Enfermedades del Yeyuno/patología , Yeyuno/patología , Micotoxinas/administración & dosificación , Micotoxinas/toxicidad , Técnicas de Cultivo de Órganos/métodos , Porcinos , Pruebas de Toxicidad/métodos , Tricotecenos/administración & dosificación
12.
Am J Cancer Res ; 6(1): 97-104, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27073727

RESUMEN

This brief article focuses on two aims: i) To investigate the in vitro pharmaco-dynamic interactions of combining synthetic potent microtubule targeting anticancer agent, Fludelone (FD) with cyto-protective agent, Panaxytriol (PXT) derived from Panax ginseng, and ii) To illustrate step-by-step operation for conducting two-drug combination in vitro using the combination index method, in terms of experimental design, data acquisition, computerized simulation and data interpretation. The Chou-Talalay method for drug combination is based on the median-effect equation, which provides the theoretical basis for the combination index (CI)-isobologram equation that allows quantitative determination of drug interactions, where CI<1, =1, and >1 indicates synergism, additive effect and antagonism, respectively. Based on these algorithms, computer software, CompySyn, is used for determining synergism and antagonism at all doses or effect levels simulated automatically. The use of Chou-Talalay's CI method in quantifying synergism or antagonism is increasing steadily during the past two decades, however, confusing questions and pitfalls were still frequently raised by insufficient understanding of the theory, especially reflected when researchers trying to use the computerized software to design and conduct experiments. In order to specifically address the confusions and to illustrate the practical features of this method, in this paper, a selected example is given based on our unpublished data regarding the combinational pharmacologic interactions of FD and PXT against the growth of breast cancer cell line MX-1. The step-by-step operation from experimental design to the real data analysis is illustrated. The results indicated that FD and PXT combination in vitro exerted synergistic effect when cell growth inhibition was greater than 45%, with CI ranged 0.836-0.609 for the fractional inhibition of Fa=0.50~0.90, as shown by the Fa-CI plot and by the isobologram. Thus, quantitative conclusion of synergism is obtained using the Chou-Talalay CI method, under the well-defined simple conditions for the FD and PXT combinations in vitro.

13.
PLoS One ; 8(10): e77684, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24155971

RESUMEN

BACKGROUND: We evaluated the therapeutic effects of the histone deacetylase inhibitor PXD101 alone and in combination with conventional chemotherapy in treating thyroid cancer. METHODOLOGY/PRINCIPAL FINDINGS: We studied eight cell lines from four types of thyroid cancer (papillary, follicular, anaplastic and medullary). The cytotoxicity of PXD101 alone and in combination with three conventional chemotherapeutic agents (doxorubicin, paclitaxel and docetaxel) was measured using LDH assay. Western blot assessed expression of acetylation of histone H3, histone H4 and tubulin, proteins associated with apoptosis, RAS/RAF/ERK and PI3K/AKT/mTOR signaling pathways, DNA damage and repair. Apoptosis and intracellular reactive oxygen species (ROS) were measured by flow cytometry. Mice bearing flank anaplastic thyroid cancers (ATC) were daily treated with intraperitoneal injection of PXD101 for 5 days per week. PXD101 effectively inhibited thyroid cancer cell proliferation in a dose-dependent manner. PXD101 induced ROS accumulation and inhibited RAS/RAF/ERK and PI3K/mTOR pathways in sensitive cells. Double-stranded DNA damage and apoptosis were induced by PXD101 in both sensitive and resistant cell lines. PXD101 retarded growth of 8505C ATC xenograft tumors with promising safety. Combination therapy of PXD101with doxorubicin and paclitaxel demonstrated synergistic effects against four ATC lines in vitro. CONCLUSIONS: PXD101 represses thyroid cancer proliferation and has synergistic effects in combination with doxorubicin and paclitaxel in treating ATC. These findings support clinical trials using PXD101 for patients with this dismal disease.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Sulfonamidas/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Acetilación/efectos de los fármacos , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Reparación del ADN/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Desnudos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Carcinoma Anaplásico de Tiroides , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Tubulina (Proteína)/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Med Chem ; 56(4): 1544-63, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23360284

RESUMEN

A series of bis(hydroxymethyl)indolizino[6,7-b]indoles and their bis(alkylcarbamates) were synthesized for antitumor studies. These agents were designed as hybrid molecules of ß-carboline (topoisomerase inhibition moiety) and bis(hydroxymethyl)pyrrole (DNA cross-linking moiety). The preliminary antitumor studies indicated that these agents exhibited significant cytotoxicity against a variety of human tumor cells in vitro. Treatment of human breast carcinoma MX-1 xenograft-bearing nude mice with compounds 18b and 28c achieved more than 99% tumor remission. We also observed that 18a displayed potent therapeutic efficacy against human lung adenocarcinoma A549 and colon cancer HT-29 xenografts. These results revealed that compound 18a was more potent than irinotecan against HT-29 cells and was as potent as irinotecan against A549 cells in xenograft models. Furthermore, we demonstrated that these derivatives possess multiple modes of action, such as induction of DNA cross-linking, inhibition of topoisomerase I and II, and cell-cycle arrest at the S-phase.


Asunto(s)
Antineoplásicos/síntesis química , Reactivos de Enlaces Cruzados/síntesis química , ADN/metabolismo , Indoles/síntesis química , Indolizinas/síntesis química , Inhibidores de Topoisomerasa I/síntesis química , Inhibidores de Topoisomerasa II/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indoles/química , Indoles/farmacología , Indolizinas/química , Indolizinas/farmacología , Sustancias Intercalantes/síntesis química , Sustancias Intercalantes/química , Sustancias Intercalantes/farmacología , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Ratas , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Relación Estructura-Actividad , Inhibidores de Topoisomerasa I/química , Inhibidores de Topoisomerasa I/farmacología , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/farmacología , Trasplante Heterólogo
15.
PLoS One ; 7(10): e46726, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077520

RESUMEN

BACKGROUND: We assessed the utility of the dual PI3K/mTOR inhibitor NVP-BEZ235 (BEZ235) as single agent therapy and in combination with conventional chemotherapy for thyroid cancer. METHODOLOGY/PRINCIPAL FINDINGS: Eight cell lines from four types of thyroid cancer (papillary, follicular, anaplastic, medullary) were studied. The cytotoxicity of BEZ235 and five conventional chemotherapeutic agents alone and in combination was measured using LDH assay. Quantitative western blot assessed expression of proteins associated with cell cycle, apoptosis and signaling pathways. Cell cycle distribution and apoptosis were measured by flow cytometry. Murine flank anaplastic thyroid cancers (ATC) were treated with oral BEZ235 daily. We found that BEZ235 effectively inhibited cell proliferation of all cancer lines, with ATC exhibiting the greatest sensitivity. BEZ235 consistently inactivated signaling downstream of mTORC1. BEZ235 generally induced cell cycle arrest at G0/G1 phase, and also caused apoptosis in the most sensitive cell lines. Baseline levels of p-S6 ribosomal protein (Ser235/236) and p27 correlated with BEZ235 sensitivity. Growth of 8505C ATC xenograft tumors was inhibited with BEZ235, without any observed toxicity. Combination therapy of BEZ235 and paclitaxel consistently demonstrated synergistic effects against ATC in vitro. CONCLUSIONS: BEZ235 as a single therapeutic agent inhibits thyroid cancer proliferation and has synergistic effects in combination with paclitaxel in treating ATC. These findings encourage future clinical trials using BEZ235 for patients with this fatal disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Imidazoles/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Quinolinas/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Neoplasias de la Tiroides/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis , Western Blotting , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Humanos , Imidazoles/farmacología , Ratones , Quinolinas/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Tiroides/patología
16.
Eur J Med Chem ; 53: 28-40, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22507893

RESUMEN

A series of novel 2,3-bis(hydroxymethyl)benzo[d]pyrrolo[2,1-b]thiazoles and their bis(alkylcarbamate) derivatives were synthesized starting from benzothiazole via reaction with dimethyl acetylenedicarboxylate (DMAD)/tetra-fluoro boric acid, catalytic hydrogenation, and alkylcarbamoylation. The anti-proliferative activity of these agents against human leukemia and various solid tumor cell growth in vitro was studied. The structure-activity relationship studies revealed that the bis(alkylcarbamates) derivatives are generally more cytotoxic than the corresponding bis(hydroxymethyl) congeners in inhibiting human lymphoblastic leukemia CCRF-CEM and various human solid tumor cell growth in culture. These agents have no cross-resistance to taxol or vinblastine. Studies on the therapeutic effect against human breast carcinoma MX-1 xenograft showed that complete tumor remission (CR) were achieved by treating with C1-4'-F- or C1-4'-Cl-Ph-bis(i-propylcarbamates) derivatives (19b and 19c, respectively) and more than 99% tumor suppression by the corresponding bis(ethylcarbamates) 18b and 18c at the maximal tolerated dose. Alkaline agarose gel shifting assay revealed that the newly synthesized compounds are able to induce DNA interstrand cross-linking. The present studies generated a series of new potent DNA interstrand cross-linking agents, which have potential for further antitumor drug development.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Tiazoles/síntesis química , Tiazoles/farmacología , Animales , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , ADN/química , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Tiazoles/química , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Am J Cancer Res ; 1(7): 925-54, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22016837

RESUMEN

The mass-action law based system analysis via mathematical induction and deduction lead to the generalized theory and algorithm that allows computerized simulation of dose-effect dynamics with small size experiments using a small number of data points in vitro, in animals, and in humans. The median-effect equation of the mass-action law deduced from over 300 mechanism specific-equations has been shown to be the unified theory that serves as the common-link for complicated biomedical systems. After using the median-effect principle as the common denominator, its applications are mechanism-independent, drug unit-independent, and dynamic order-independent; and can be used generally for single drug analysis or for multiple drug combinations in constant-ratio or non-constant ratios. Since the "median" is the common link and universal reference point in biological systems, these general enabling lead to computerized quantitative bio-informatics for econo-green bio-research in broad disciplines. Specific applications of the theory, especially relevant to drug discovery, drug combination, and clinical trials, have been cited or illustrated in terms of algorithms, experimental design and computerized simulation for data analysis. Lessons learned from cancer research during the past fifty years provide a valuable opportunity to reflect, and to improve the conventional divergent approach and to introduce a new convergent avenue, based on the mass-action law principle, for the efficient cancer drug discovery and the low-cost drug development.

18.
Proc Natl Acad Sci U S A ; 108(34): 14336-41, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21844359

RESUMEN

We describe herein the discovery of a series of panaxytriol (PXT)-derived polyacetylene small molecules with promising cytoprotective activity. In mouse xenograft models, we have demonstrated the capacity of our synthetic analogs to mitigate a range of cancer therapeutic agent-induced toxicities, including body weight loss, lethality, neurotoxicity, and hematotoxicity. Our PXT analogs have also been found to reduce radiation-induced body weight loss and lethality in mouse models. Moreover, several PXT analogs appear to exhibit moderate in vivo antiinflammatory activity as well as in vitro immunoenhancing capabilities. These compounds appear to derive their activity through induction of cancer preventive phase 2 enzymes. The studies described herein suggest that coadministration of a PXT-derived agent with cancer chemotherapeutics or radiation therapy may serve to mitigate a range of therapy-associated toxicities.


Asunto(s)
Productos Biológicos/farmacología , Citoprotección/efectos de los fármacos , Enediinos/farmacología , Alcoholes Grasos/farmacología , Panax/química , Poliinos/farmacología , Animales , Productos Biológicos/química , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Citoprotección/efectos de la radiación , Enediinos/química , Alcoholes Grasos/química , Femenino , Fluorouracilo/farmacología , Humanos , Isomerismo , Longevidad/efectos de los fármacos , Longevidad/efectos de la radiación , Ratones , Ratones Desnudos , Paclitaxel/farmacología , Poliinos/química , Pérdida de Peso/efectos de los fármacos , Pérdida de Peso/efectos de la radiación , Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto
19.
ChemMedChem ; 6(6): 1119-29, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21542133

RESUMEN

A series of 2-substituted quinolin-4-yl-benzenesulfonate derivatives were synthesized for the purpose of evaluating antiproliferative activity. Structure-activity relationships of the newly synthesized compounds against human lymphoblastic leukemia and various solid tumor cell growths in culture are discussed. Of these derivatives, 2-phenyl-6-pyrrolidinyl-4-quinoline sulfonate analogues 10 f, 10 g, and 10 k, and 4'-nitrophenyl sulfonate 10 m exhibit superior cytotoxicity over other sulfonates. The antiproliferative activities of these compounds correlate well with their abilities to induce mitotic arrest and apoptosis. Mechanistic studies indicate that they target the vinblastine binding site of tubulin and inhibit cellular tubulin polymerization. Hence, these compounds induce the formation of aberrant mitotic spindles and mitotic arrest, resulting in intensive apoptosis. The tested compounds were shown to be poor substrates for membrane multidrug resistance transporters. The present studies suggest that these newly synthesized compounds are promising tubulin polymerization inhibitors and are worthy of further investigation as antitumor agents.


Asunto(s)
4-Quinolonas/farmacología , Antineoplásicos/farmacología , Bencenosulfonatos/farmacología , Moduladores de Tubulina/farmacología , Tubulina (Proteína)/metabolismo , 4-Quinolonas/química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Bencenosulfonatos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Mitosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Moduladores de Tubulina/química
20.
Bioorg Med Chem ; 19(6): 1987-98, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21356592

RESUMEN

A series of N-mustard-quinazoline conjugates was synthesized and subjected to antitumor studies. The N-mustard pharmacophore was attached at the C-6 of the 4-anilinoquinazolines via a urea linker. To study the structure-activity relationships of these conjugates, various substituents were introduced to the C-4 anilino moiety. The preliminary antitumor studies revealed that these agents exhibited significant antitumor activity in inhibiting various human tumor cell growths in vitro. Compounds 21b, 21g, and 21h were selected for further antitumor activity evaluation against human breast carcinoma MX-1 and prostate PC-3 xenograft in animal model. These agents showed 54-75% tumor suppression with low toxicity (5-7% body-weight changes). We also demonstrate that the newly synthesized compounds are able to induce DNA cross-linking through alkaline agarose gel shift assay and inhibited cell cycle arrest at G2/M phase.


Asunto(s)
Antineoplásicos/síntesis química , Quinazolinas/química , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Ratones , Ratones Desnudos , Planta de la Mostaza/química , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad , Urea/química , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA