Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Cancer ; 138(4): 1024-32, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26370161

RESUMEN

Erlotinib (ERL), an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, shows notable efficacy against non-small cell lung cancer (NSCLC) harboring EGFR mutations. Bevacizumab (BEV), a humanized monoclonal antibody to vascular endothelial cell growth factor (VEGF), in combination with ERL (BEV+ERL) significantly extended progression-free survival in patients with EGFR-mutated NSCLC compared with ERL alone. However, the efficacy of BEV+ERL against EGFR-mutated NSCLC harboring T790M mutation or MET amplification, is unclear. Here, we examined the antitumor activity of BEV+ERL in four xenograft models of EGFR-mutated NSCLC (three harboring ERL resistance mutations). In the HCC827 models (exon 19 deletion: DEL), ERL significantly inhibited tumor growth by blocking EGFR signal transduction. Although there was no difference between ERL and BEV+ERL in maximum tumor growth inhibition, BEV+ERL significantly suppressed tumor regrowth during a drug-cessation period. In the HCC827-EPR model (DEL+T790M) and HCC827-vTR model (DEL+MET amplification), ERL reduced EGFR signal transduction and showed less pronounced but still significant tumor growth inhibition than in the HCC827 model. In these models, tumor growth inhibition was significantly stronger with BEV+ERL than with each single agent. In the NCI-H1975 model (L858R+T790M), ERL did not inhibit growth or EGFR signal transduction, and BEV+ERL did not inhibit growth more than BEV. BEV alone significantly decreased microvessel density in each tumor. In conclusion, addition of BEV to ERL did not enhance antitumor activity in primarily ERL-resistant tumors with T790M mutation; however, BEV+ERL enhanced antitumor activity in T790M mutation- or MET amplification-positive tumors as long as their growth remained significantly suppressed by ERL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Amplificación de Genes , Genes erbB-1 , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-met/genética , Animales , Bevacizumab/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Receptores ErbB/genética , Clorhidrato de Erlotinib/administración & dosificación , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación , Reacción en Cadena de la Polimerasa , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Gastric Cancer ; 17(4): 638-47, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24414131

RESUMEN

BACKGROUND: Accurate and reliable assessment of human epidermal growth factor receptor type 2 (HER2) status is important for selecting patients with gastric cancer who may benefit from trastuzumab treatment. Here we examined the impact of formalin fixing conditions on HER2 immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) in xenografted tumor tissues. METHODS: Xenografted tumor tissues of the human gastric cancer cell lines NCI-N87, SCH, and SNU-16 were collected and kept at room temperature for 0, 6, or 24 h before being fixed with 10 % neutral buffered formalin (NBF) for 24 h or 5, 7, or 10 days and embedded in paraffin. Use of 10 % NBF, 20 % NBF, or nonbuffered formalin as fixative was investigated. RESULTS: The HER2 IHC scores for NCI-N87, SCH, and SNU-16 tumors were 3+, 2+, and 1+, respectively, when specimens were fixed with 10 % NBF for 24 h immediately after resection of the tumors. Specimens left for longer than 6 h before fixation had shrinkage of the tumor periphery and decreased immunostaining intensity in this region in all specimens. In SCH and SNU-16 specimens, starting fixation 24 h after tumor tissue collection induced autolysis and reduction of the number of stained cells, and 10-day-fixation lowered the HER2 score. Prolongation of fixation time did not affect FISH results, but if samples were left for more than 6 h before fixation, the FISH score was strongly reduced in SCH specimens (2.3 to 1.3). Reduced IHC staining intensity was observed with 20 % NBF and nonbuffered formalin compared to 10 % NBF. CONCLUSIONS: The time to and length of fixation of tumor specimens can affect HER2 IHC and FISH scores. The fixative used can affect IHC results.


Asunto(s)
Formaldehído , Inmunohistoquímica/métodos , Receptor ErbB-2/análisis , Neoplasias Gástricas/patología , Fijación del Tejido/métodos , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Hibridación Fluorescente in Situ/métodos , Masculino , Ratones Endogámicos BALB C , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/metabolismo , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int Immunopharmacol ; 8(1): 12-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18068095

RESUMEN

OK-432 (Picibanil), a Streptococcal immunotherapeutic agent, has been used for immunotherapy of various cancers as a biological response modifier (BRM). However, OK-432 contains multiple components consisting of immunotherapeutic ones and contaminants which may weaken the effects or exert side-effects. In this study, we investigated extraction of contaminants from OK-432 using Triton X-114 (TX-114)-water phase partitioning and examined an antitumor effect of the resulting preparation. OK-432 was subjected to TX-114 partitioning to give residual precipitate designated as OK-TX-ppt. OK-TX-ppt exerted no TLR2-mediated activity, but induced interleukin (IL)-6 in human PBMC. OK-TX-ppt also induced tumor necrosis factor (TNF)-alpha, IL-10, IL-12, and interferon (IFN)-gamma in PBMC. Moreover, IFN-gamma-inducing activity of OK-TX-ppt was significantly higher and IL-10 production was lower than that of OK-432. In tumor-bearing mice model, administration of OK-TX-ppt i.p. extended the survival time of Meth-A-bearing mice compared to OK-432. OK-TX-ppt also increased the levels of IL-12 and IFN-gamma in mouse spleen cells in vitro. These results indicated that TX-114 partitioning removed some contaminants, which attenuates the antitumor effect, from OK-432 and increase the immunotherapeutic effects of OK-432.


Asunto(s)
Adyuvantes Farmacéuticos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia , Picibanil/uso terapéutico , Polietilenglicoles , Adyuvantes Farmacéuticos/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Octoxinol , Picibanil/administración & dosificación
4.
Cancer Chemother Pharmacol ; 59(6): 795-805, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17031648

RESUMEN

PURPOSE: To clarify the antitumor activity of trastuzumab and its potential as an effective treatment for gastric cancer patients. METHODS: Levels of HER2 expression in tumor tissues of gastric cancer cell lines were examined using immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and mRNA quantification. Efficacy of trastuzumab was examined as a single agent or in combination with chemotherapeutic agents widely used clinically for gastric cancers in HER2-overexpressing human gastric cancer xenograft models. RESULTS: Two of nine human gastric cancer xenograft models, NCI-N87 and 4-1ST, showed overexpression of HER2 mRNA and protein by IHC (HercepTest) and HER2 gene amplification by FISH (Pathvysion). HER2 protein showed potent staining in peripheral membranes, similar to the staining pattern of breast cancer. FISH scores were also comparable to those of breast cancer models. Trastuzumab as a single agent inhibited the tumor growth in both of the HER2-overexpressing models but not in the HER2-negative models, GXF97 and MKN-45. In any combination with capecitabine, cisplatin, irinotecan, docetaxel, or paclitaxel, trastuzumab showed more potent antitumor activity than the anticancer agents alone. A three-drug combination of capecitabine, cisplatin, and trastuzumab showed remarkable tumor growth inhibition. In NCI-N87 in vitro, trastuzumab showed direct antiproliferative activity according to cell count or crystal violet dying, and showed indirect antitumor activity such as antibody-dependent cellular cytotoxicity. CONCLUSION: The antitumor activity of trastuzumab observed in human gastric cancer models warrants consideration of its use in clinical treatment regimens for human gastric cancer as a single agent or a combination drug with various chemotherapeutic agents.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias Gástricas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Ratones , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/metabolismo , Trastuzumab , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Oncol Lett ; 14(4): 4197-4205, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28959366

RESUMEN

In a Phase III trial for HER2-positive breast cancer (the CLEOPATRA study), the triple-drug combination arm of pertuzumab plus trastuzumab plus docetaxel showed significantly longer progression-free survival and overall survival than did the trastuzumab plus docetaxel arm. In this study, we investigated the mechanism of action of the triple-drug combination therapy in vivo. For this purpose, we established a mouse xenograft model using KPL-4, a HER2-positive human breast cancer cell line, in which the triple-drug combination treatment dramatically induced tumor regression compared with double-drug combinations (trastuzumab plus docetaxel, pertuzumab plus docetaxel, or pertuzumab plus trastuzumab). Four days after the triple-drug treatment was started, strong reduction in the phosphorylation of HER2, epidermal growth factor receptor (EGFR), HER3, extracellular signal-regulated kinase (ERK), and AKT in tumor tissues was seen, despite only weak suppression of phosphorylation seen with the single- or double-drug treatments. Histopathological analysis and flow cytometric analysis showed that the triple-drug treatment enhanced apoptosis after mitotic arrest induced by docetaxel. Furthermore, infiltration of mononuclear cells around the tumor cells was strongly induced by the triple-drug combination treatment. These results suggested that the mechanism underlying the synergistic efficacy of the triple-drug combination was attributable, at least in part, to the docetaxel-mediated apoptosis being promoted by enhanced inhibition of HER2-HER3-AKT signaling as well to the intratumor infiltration of mononuclear cells induced by anti-HER2 antibodies being enhanced by docetaxel.

6.
Int Immunopharmacol ; 6(5): 764-73, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16546707

RESUMEN

We examined the role of nitric oxide (NO) induced by OK-432, a streptococcal immunotherapeutic agent, in anti-tumor effects of the OK-432 by in vitro and in vivo experiments using an NO synthase inhibitor, N-monomethyl-l-arginine acetate (NMA). The in vitro treatment of mouse splenocytes with OK-432 increased the expression of inducible NO synthase (iNOS) gene and NO production in a dose-dependent manner. Although it is well known that OK-432 induces cytokines such as interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha, both of which are known to be potent NO inducers, we observed only a partial reduction of OK-432-induced NO production with the addition of anti-IFN-gamma and/or anti-TNF-alpha neutralizing antibodies. The cytotoxicity of the splenocytes increased by in vitro OK-432 stimulation was almost completely inhibited by the treatment with NMA. OK-432 administration resulted in a marked prolongation of survival and a significant inhibition of tumor growth in syngeneic tumor-bearing mice, whereas NMA significantly inhibited the anti-tumor effects of OK-432. Although the increased cytotoxicity of adherent splenocytes derived from OK-432-treated tumor-bearing mice was almost completely inhibited by NMA, only partial inhibition by NMA was observed in the cytotoxicity of the nonadherent splenocytes. These findings strongly suggest that the iNOS/NO induced by OK-432 is intimately involved in the anti-tumor effects of OK-432.


Asunto(s)
Antineoplásicos/farmacología , Óxido Nítrico/biosíntesis , Picibanil/farmacología , Animales , Arginina/análogos & derivados , Arginina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Inmunización , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Nitratos/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , ARN Mensajero/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Streptococcus pyogenes
7.
J Natl Cancer Inst ; 95(4): 316-26, 2003 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-12591988

RESUMEN

BACKGROUND: The streptococcal agent OK-432 has been used for immunotherapy of head and neck cancer, among other malignancies, but its mechanism of action is unknown. Because the Toll-like receptor 4 (TLR4)/MD-2 complex is important in enabling the mammalian immune system to recognize bacterial components, we investigated whether expression of the TLR4 and MD-2 genes is associated with OK-432-induced anticancer immunity. METHODS: Peripheral blood mononuclear cells (PBMCs) from 28 patients with head and neck cancer were analyzed for TLR4 and MD-2 mRNA expression by reverse transcription-polymerase chain reaction (RT-PCR) analysis. PBMCs were treated in vitro with OK-432 or with OK-PSA (a lipoteichoic-acid-related molecule that is an active component of OK-432), and interferon-gamma (IFN-gamma) mRNA expression, an immune response measure, was analyzed by RT-PCR. Patient sera collected 24 hours after OK-432 administration were examined for IFN-gamma protein using an enzyme-linked immunosorbent assay. Lewis lung carcinoma-bearing wild-type C57BL/6 and TLR4-deficient mice (four mice per group) received intraperitoneal injections of OK-432, and tumor volumes and sera IFN-gamma levels were measured over time. All statistical tests were two-sided. RESULTS: Twenty patients expressed both TLR4 and MD-2. Expression of TLR4 and MD-2 genes was associated with the in vivo IFN-gamma induction in 19 patients administered OK-432 (Fisher's exact test P<.001). Although both OK-432 and OK-PSA induced IFN-gamma expression from PBMCs in vitro, expression of TLR4 and MD-2 was associated only with IFN-gamma expression induced by OK-PSA (P<.001). In vivo intraperitoneal administration of OK-432 resulted in an increase of IFN-gamma in sera from wild-type mice but not in sera from TLR4-deficient mice. Tumors in wild-type mice treated with OK-432 were statistically significantly smaller than those in mice treated with saline (P =.007). By contrast, in TLR4-deficient mice, there was no difference in tumor volume between the two treatment groups. CONCLUSIONS: TLR4 and MD-2 may mediate OK-432-induced anticancer immunity.


Asunto(s)
Antígenos de Superficie/metabolismo , Antineoplásicos/farmacología , Proteínas de Drosophila , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/inmunología , Interferón gamma/biosíntesis , Glicoproteínas de Membrana/metabolismo , Picibanil/farmacología , Receptores de Superficie Celular/metabolismo , Anciano , Animales , Antibacterianos/farmacología , Antígenos de Superficie/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Neoplasias de Cabeza y Cuello/química , Humanos , Inyecciones Intraperitoneales , Interferón gamma/sangre , Interferón gamma/efectos de los fármacos , Luciferasas/análisis , Antígeno 96 de los Linfocitos , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Polimixina B/farmacología , ARN Mensajero/análisis , ARN Neoplásico/análisis , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4 , Receptores Toll-Like
8.
Cancer Res ; 64(15): 5461-70, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15289356

RESUMEN

A lipoteichoic acid-related molecule OK-PSA is an active component of OK-432, a Streptococcus-derived anticancer immunotherapeutic agent. In the present study, we first examined the effect of OK-PSA on the maturation of dendritic cells (DCs) in vitro by using the DCs derived from 5 healthy donors and 10 patients with head and neck cancer with or without expression of toll-like receptor 4 (TLR4) or MD-2 mRNA. OK-PSA treatment effectively increased the surface expression of MHC class II, CD80, CD83, and CD86. OK-PSA-stimulated DCs secreted the cytokines that can induce helper T-cell 1 (Th1)-type T-cell response, and stimulated allogeneic T cells to produce IFN-gamma and to elicit an allogeneic antigen-specific cytotoxicity. These activities almost depended on expression of TLR4 and MD-2 genes. We next investigated the in vivo anticancer effect of intratumoral administration of syngeneic DCs followed by OK-PSA against established tumors in mice. C57BL/6 mice, which express wild-type TLR4, and C57BL/6-derived TLR4-knockout (TLR4(-/-)) mice were used. Although OK-PSA accelerated the antitumor effect of intratumoral DC administration in wild-type mice bearing syngeneic tumors, the antitumor effect of OK-PSA as well as of the combination therapy with DCs and OK-PSA was not significant in TLR4(-/-) mice. Interestingly, an administration of wild-type-mouse-derived DCs followed by OK-PSA exhibited a marked antitumor effect even in the TLR4(-/-) mice. These findings suggest that OK-PSA may be a potent adjuvant for local DC therapy, and that DC therapy followed by OK-PSA is able to elicit anticancer activity even in a TLR4-deficient host when TLR4 is expressed only in DCs injected intratumorally.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Dendríticas/metabolismo , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/terapia , Inmunoterapia , Glicoproteínas de Membrana/fisiología , Picibanil/uso terapéutico , Receptores de Superficie Celular/fisiología , Adyuvantes Inmunológicos , Adulto , Anciano , Animales , Antígenos CD/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Quimiocinas/metabolismo , Cromo/metabolismo , Terapia Combinada , Citocinas/metabolismo , Células Dendríticas/inmunología , Femenino , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Interferón gamma/metabolismo , Antígeno 96 de los Linfocitos , Linfocitos Infiltrantes de Tumor , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Streptococcus/química , Linfocitos T Citotóxicos/metabolismo , Células TH1/inmunología , Receptor Toll-Like 4 , Receptores Toll-Like
9.
Mol Clin Oncol ; 3(5): 987-994, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26623038

RESUMEN

In the treatment of human epidermal growth factor receptor 2 (HER2)-positive advanced gastric or gastroesophageal junction cancer, it has been reported that the combination of trastuzumab with capecitabine plus cisplatin, or with 5-fluorouracil (5-FU) plus cisplatin, significantly increased overall survival compared with chemotherapy alone (ToGA trial). In addition, adjuvant therapy with capecitabine plus oxaliplatin (XELOX) improved the survival of patients who received curative D2 gastrectomy (CLASSIC trial). However, the efficacy of the combination of trastuzumab with XELOX for patients with HER2-positive gastric cancer remains unknown. The aim of this study, was to investigate the efficacy of the combination of trastuzumab with XELOX in a HER2-positive human gastric cancer xenograft model. Combination treatment with these three agents (trastuzumab 20 mg/kg, capecitabine 359 mg/kg and oxaliplatin 10 mg/kg), was found to exhibit a significantly stronger antitumor activity in NCI-N87 xenografts compared with either trastuzumab or XELOX alone. In this model, treatment with trastuzumab alone or trastuzumab plus oxaliplatin enhanced the expression of thymidine phosphorylase (TP), a key enzyme in the generation of 5-FU from capecitabine in tumor tissues. In in vitro experiments, trastuzumab induced TP mRNA expression in NCI-N87 cells. In addition, NCI-N87 cells co-cultured with the natural killer (NK) cell line CD16(158V)/NK-92 exhibited increased expression of TP mRNA. When NCI-N87 cells were cultured with CD16(158V)/NK-92 cells in the presence of trastuzumab, the mRNA expression of cytokines reported to have the ability to induce TP was upregulated in tumor cells. Furthermore, a medium conditioned by CD16(158V)/NK-92 cells also upregulated the expression of TP mRNA in NCI-N87 cells. These results suggest that trastuzumab promotes TP expression, either by acting directly on NCI-N87 cells, or indirectly via a mechanism that includes trastuzumab-mediated interactions between NK and NCI-N87 cells. Therefore, the combination of trastuzumab with XELOX may be a potent therapy for HER2-positive gastric cancer.

10.
Int Immunopharmacol ; 3(5): 643-55, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12757734

RESUMEN

We have previously isolated a lipoteichoic acid (LTA)-related molecule (OK-PSA) from OK-432, a streptococcal agent, by affinity chromatography on a CNBr-activated Sepharose 4B bound TS-2 monoclonal antibody (mAb) that neutralizes the interferon (IFN)-gamma-inducing activity of OK-432. In the current study, we compared the cytokine-inducing and anti-tumor activities of OK-PSA, a TS-2-binding fraction, with those of OK-PTF, a TS-2-unbinding fraction, in order to determine the efficacy of OK-PSA for clinical use in affinity chromatography using TS-2. In the in vitro experiments using human peripheral blood mononuclear cells (PBMCs), OK-PSA markedly induced Th1-type cytokines, while interleukin (IL)-6 and IL-10, Th2-type cytokines, were induced by OK-PTF. Th1-cytokine induction by OK-PTF was not dose-dependent and was suppressed when PBMCs were treated with a high concentration of OK-PTF. In a mouse model, Th1 cytokines were also induced by OK-PSA and Th2 cytokines were induced by OK-PTF. Th2 cytokine-inducing activity of OK-PTF was accelerated in tumor-bearing mice relative to that in healthy mice. Although the anti-tumor effect of OK-PTF was statistically significant, it was much weaker than that of OK-PSA. A significant difference between the anti-tumor effect of OK-PSA and that of OK-PTF was observed (P<0.05). Finally, OK-PSA elicited its cytokine-inducing effect via Toll-like receptor (TLR) 4, whereas OK-PTF-induced signaling was mediated by both TLR2 and TLR4. These findings strongly suggested that the affinity chromatography using TS-2 is a useful strategy to separate the effective component for cancer therapy (OK-PSA) from other components.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/biosíntesis , Inductores de Interferón/antagonistas & inhibidores , Interferón gamma/antagonistas & inhibidores , Interferón gamma/biosíntesis , Picibanil/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/metabolismo , Células Cultivadas , Cromatografía de Afinidad , Medios de Cultivo , Citocinas/biosíntesis , Femenino , Humanos , Hibridomas , Técnicas In Vitro , Inductores de Interferón/farmacología , Luciferasas/genética , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/fisiopatología , Picibanil/farmacología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Transducción de Señal/efectos de los fármacos , Células TH1/metabolismo , Células Th2/metabolismo , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like
11.
Anticancer Res ; 24(5C): 3295-301, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15515424

RESUMEN

OK-432 (Picibanil), a streptococcal preparation with potent biological response modifying activities, was approved in Japan as an anticancer agent in 1975. In the ensuing 30 years, since then, a significant amount of data, including clinical as well as experimental studies, has been accumulated. OK-432 has been reported to induce various cytokines, activate immunological cells and thus augment anticancer immunity. Recently, the interrelation between innate immunity and adaptive immunity has become clear and it was reported that OK-432 acts, at least in part, via Toll-like receptor (TLR) 4-MD2 signaling pathway. In addition, dendritic cells (DCs) are considered to play a pivotal role in immunological response and it is reported that OK-432 induced maturation of DCs both in vitro and in vivo. These results suggest that OK-432 is a useful adjuvant in DC-based anticancer immunotherapy. Clinical studies of DC therapy with OK-432 are under way.


Asunto(s)
Antineoplásicos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Picibanil/farmacología , Animales , Antineoplásicos/inmunología , Humanos , Picibanil/inmunología
12.
Anticancer Res ; 22(6A): 3229-39, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12530069

RESUMEN

BACKGROUND: OK-PSA, a lipoteichoic acid (LTA)-related molecule isolated from a streptococcal agent OK-432, enhances anti-tumor immunity as a potent inducer of Th1-type cytokines. Recently, we obtained the data suggesting that natural killer (NK) cells may play a significant role for OK-PSA-induced cytokine production in vitro. MATERIALS AND METHODS: We conducted the animal experiments using athymic nude mice bearing human salivary adenocarcinoma to examine the role of NK cells in OK-PSA-induced anti-tumor immunity. OK-PSA was peritumorally injected into the mice. Cytokines in the sera were analyzed by ELISA. mRNAs for cytokines were detected by RT-PCR. 51Cr release test was performed to measure killer cell activities. RESULTS: OK-PSA markedly increased the amounts of IFN-gamma, tumor necrosis factor (TNF)-alpha, interleukin (IL)-2, IL-12 and IL-18 that are generally called "Th1-type cytokines" in the sera derived from tumor-bearing nude mice, and also accelerated the killing activities of tumor-infiltrating lymphocytes as well as of draining lymph node cells. Furthermore, OK-PSA administration resulted in significant inhibition of tumor growth, but the effect of OK-PSA was almost completely inhibited by the deletion of NK cells using anti-asialo GM1 antibody. CONCLUSION: These findings strongly suggested that NK cells are closely involved in OK-PSA-mediated anti-tumor immunity.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/terapia , Células Asesinas Naturales/inmunología , Lipopolisacáridos/farmacología , Neoplasias de las Glándulas Salivales/inmunología , Neoplasias de las Glándulas Salivales/terapia , Ácidos Teicoicos/farmacología , Adenocarcinoma/radioterapia , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Movimiento Celular/efectos de la radiación , Terapia Combinada , Citocinas/biosíntesis , Citocinas/sangre , Citocinas/genética , Gangliósido G(M1)/inmunología , Humanos , Inmunoterapia/métodos , Lipopolisacáridos/inmunología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/efectos de la radiación , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Picibanil/química , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Neoplasias de las Glándulas Salivales/radioterapia , Ácidos Teicoicos/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Lung Cancer ; 83(1): 44-50, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24192512

RESUMEN

Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) show notable effects against non-small cell lung cancers (NSCLCs) harboring EGFR-activating mutations. However, almost all patients eventually acquire resistance to EGFR-TKIs. In this study, we established novel erlotinib resistant NSCLC cells and examined their resistant mechanisms. Resistant cells were established in 14, 3, and 0 wells exposed to 0.1, 1, and 10 µM erlotinib, respectively. The IC(50) values of these cells were 47- to 1209-fold higher than that of the parent cells. No secondary T790M mutation was detected in any resistant cells. However, in 13/17 resistant cells, EGFR copy number was reduced less than approximately one-eighth of parent cells, and in one resistant cell (B10), >99.99% of the population was EGFR-unamplified cells. Most (97.5%) parent cells showed EGFR amplification, but 2.5% of the population comprised EGFR-unamplified cells. An EGFR-unamplified clone (4D8) isolated from parent cells in erlotinib-free normal medium also showed erlotinib resistance comparable to the resistant B10 cells. Loss of an EGFR-amplified chromosome 7 (EGFR-ampch7) was observed in 4D8 and B10 cells. EGFR-unamplified cells were constantly maintained as a minor population of the parent cells under normal cell culture conditions. In conclusion, loss of an EGFR-ampch7 causes acquired resistance in EGFR-mutated HCC827 cells exposed to a relatively low concentration of erlotinib, but a high concentration prevents the emergence of resistance.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Quinazolinas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Aberraciones Cromosómicas , Cromosomas Humanos Par 7/genética , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Clorhidrato de Erlotinib , Amplificación de Genes , Humanos , Neoplasias Pulmonares/genética , Mutación/genética
14.
Anticancer Res ; 34(2): 613-21, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24510990

RESUMEN

Epidermal growth factor receptor (EGFR) and v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are common in non-small cell lung cancer (NSCLC) and colorectal cancer (CRC). The aim of the present study was to develop a simple and versatile tool to determine EGFR and KRAS mutations for pre-clinical research in the laboratory. We developed a melting curve analysis to detect exon 19 deletion, L858R mutation, and T790M mutation of EGFR, and codon 12/13 and codon 61 mutations of KRAS using LightCycler480 with mutation-specific sensor and anchor probes. The analytical method was applicable to determine the approximate rate of heterogeneity of mutation in the genomic DNA of cancer cell lines. In conclusion, our melting curve analysis is a rapid and semi-quantitative method to screen for exon 19 deletion, L858R or T790M mutations of EGFR and codon 12/13/61 mutations of KRAS in cancer cell lines.


Asunto(s)
Genes erbB-1 , Genes ras , Mutación , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Animales , Línea Celular Tumoral , Codón , Receptores ErbB/genética , Exones , Eliminación de Gen , Células HCT116 , Xenoinjertos , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Plásmidos/química , Plásmidos/genética , Proteínas Proto-Oncogénicas p21(ras) , Temperatura de Transición
15.
Oncol Rep ; 27(4): 923-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22209766

RESUMEN

Erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor, has been shown to have benefits for non-small cell lung cancer and pancreatic cancer patients; however, almost all patients develop progressive disease during the therapy. On the other hand, it has been reported that a tumor continues to express epidermal growth factor receptor even after developing progressive disease. To demonstrate the clinical relevance of erlotinib treatment after progressive disease, we investigated whether continuous administration of erlotinib in combination with chemotherapy has a useful effect on progressive disease development during erlotinib treatment. For this purpose, we examined the antitumor effect of a combination therapy of a chemotherapeutic agent with erlotinib using two types of erlotinib-resistant tumor xenograft models: a non-small cell lung cancer model, in which EBC-1, H1975 and HCC827TR3 tumors were implanted, and an HPAC pancreatic cancer cell xenograft which generates erlotinib-resistant tumors in vivo. As a result, the combination therapy showed a significantly higher antitumor activity compared with chemomonotherapy in all xenograft models except the H1975 xenografts. Furthermore, erlotinib alone suppressed the phosphorylation of epidermal growth factor receptor in HPAC tumors and the two non-small cell lung cancer cell lines other than H1975. Therefore, combination therapy which uses erlotinib can be considered effective if epidermal growth factor receptor phosphorylation is inhibited by erlotinib, even in erlotinib-resistant tumor xenograft models. Our results suggest that the continuous inhibition of epidermal growth factor receptor phosphorylation by erlotinib after progressive disease enhances the antitumor activity of chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Docetaxel , Relación Dosis-Respuesta a Droga , Receptores ErbB/genética , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Humanos , Irinotecán , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosforilación , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinazolinas/administración & dosificación , Taxoides/administración & dosificación , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
16.
Oncol Rep ; 27(2): 303-10, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22076074

RESUMEN

We examined the efficacy of chemoendocrine therapy using capecitabine as a chemotherapeutic agent in premenopausal and postmenopausal models with estrogen receptor (ER)-positive human breast cancer xenografts. Tamoxifen and letrozole were used as endocrine therapeutic agents for premenopausal and postmenopausal models, respectively. The antitumor activity of capecitabine in combination was significantly superior to either monotherapy treatment in both premenopausal (p<0.01) and postmenopausal (p<0.05) models. No increase in toxicity in terms of body weight loss was observed during treatment in either of the xenograft models. In the premenopausal model, the level of thymidine phosphorylase (TP), a key enzyme generating 5-FU from capecitabine, was upregulated (p<0.05) in tumors by tamoxifen but not by letrozole treatment in the postmenopausal model. The combination of 5'-deoxy-5-fluorouridine (5'-DFUR; an intermediate of capecitabine) with 4-hydroxytamoxifen (4-OHT; an active form of tamoxifen) or letrozole was also evaluated in vitro by using estrogen-responsive element (ERE) reporter gene assays aimed to model premenopausal and postmenopausal breast cancer. Both combinations decreased the number of estrogen-responding cells in a concentration-dependent manner and further analysis by isobolograms revealed a synergistic effect of the combination of 5'-DFUR with 4-OHT, and at least an additive effect of the combination of 5'-DFUR with letrozole. These results suggest that chemoendocrine therapy using capecitabine may be a useful treatment modality for patients with hormone-receptor-positive breast cancer, regardless of the menopausal status and should be explored in clinical trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Fluorouracilo/análogos & derivados , Posmenopausia , Premenopausia , Animales , Antineoplásicos Hormonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Aromatasa/genética , Aromatasa/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Capecitabina , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/uso terapéutico , Genes Reporteros , Humanos , Letrozol , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nitrilos/administración & dosificación , Tamoxifeno/administración & dosificación , Timidina Fosforilasa/metabolismo , Triazoles/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Clin Exp Metastasis ; 29(3): 207-16, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22170031

RESUMEN

Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) show dramatic antitumor activity in a subset of patients with non-small cell lung cancer who have an active mutation in the epidermal growth factor receptor (EGFR) gene. On the other hand, some lung cancer patients with wild type EGFR also respond to EGFR-TKIs, suggesting that EGFR-TKIs have an effect on host cells as well as tumor cells. However, the effect of EGFR-TKIs on host microenvironments is largely unknown. A multiple organ metastasis model was previously established in natural killer cell-depleted severe combined immunodeficient mice using human lung cancer cells. This model was used to investigate the therapeutic efficacy of erlotinib, an EGFR-TKI, on multiple organ metastases induced by human small cell lung cancer cells (SBC-5 cells) that did not express EGFR. Although erlotinib did not have any effect on the proliferation of SBC-5 cells in vitro, it significantly suppressed bone and lung metastases in vivo, but not liver metastases. An immunohistochemical analysis revealed that, erlotinib significantly suppressed the number of osteoclasts in bone metastases, whereas no difference was seen in microvessel density. Moreover, erlotinib inhibited EGF-induced receptor activator of nuclear factor kappa-B expression in an osteoblastic cell line (MC3T3-E1 cells). These results strongly suggested that erlotinib prevented bone metastases by affecting host microenvironments irrespective of its direct effect on tumor cells.


Asunto(s)
Carcinoma de Células Pequeñas/patología , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia/prevención & control , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Animales , Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Carcinoma de Células Pequeñas/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Receptores ErbB/análisis , Clorhidrato de Erlotinib , Humanos , Neoplasias Pulmonares/química , Masculino , Ratones , Neovascularización Patológica/prevención & control , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Ligando RANK/análisis
18.
Clin Exp Metastasis ; 28(7): 649-59, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21688034

RESUMEN

Previous preclinical and clinical findings have suggested a potential role of epidermal growth factor receptor (EGFR) in osteoclast differentiation and the pathogenesis of bone metastasis in cancer. In this study, we investigated the effect of erlotinib, an orally active EGFR tyrosine kinase inhibitor (TKI), on the bone invasion of human non-small-cell lung cancer (NSCLC) cell line NCI-H292. First, we established a novel osteolytic bone invasion model of NCI-H292 cells which was made by inoculating cancer cells into the tibia of scid mice. In this model, NCI-H292 cells markedly activated osteoclasts in tibia, which resulted in osteolytic bone destruction. Erlotinib treatment suppressed osteoclast activation to the basal level through suppressing receptor activator of NF-κB ligand (RANKL) expression in osteoblast/stromal cell at the bone metastatic sites, which leads to inhibition of osteolytic bone destruction caused by NCI-H292 cells. Erlotinib inhibited the proliferation of NCI-H292 cells in in vitro. Erlotinib suppressed the production of osteolytic factors, such as parathyroid hormone-related protein (PTHrP), IL-8, IL-11 and vascular endothelial growth factor (VEGF) in NCI-H292 cells. Furthermore, erlotinib also inhibited osteoblast/stromal cell proliferation in vitro and the development of osteoclasts induced by RANKL in vitro. In conclusion, erlotinib inhibits tumor-induced osteolytic invasion in bone metastasis by suppressing osteoclast activation through inhibiting tumor growth at the bone metastatic sites, osteolytic factor production in tumor cells, osteoblast/stromal cell proliferation and osteoclast differentiation from mouse bone marrow cells.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Modelos Animales de Enfermedad , Osteoblastos/efectos de los fármacos , Osteólisis/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Humanos , Masculino , Ratones , Invasividad Neoplásica/patología , Osteoblastos/metabolismo , Osteoblastos/patología , Osteólisis/metabolismo , Osteólisis/patología , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología
19.
Oncol Lett ; 1(2): 231-235, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22966286

RESUMEN

Erlotinib treatment in combination with gemcitabine is a standard therapy for patients with locally advanced pancreatic cancer in many countries, including the US and the EU. Since mutations of the K-ras oncogene (KRAS) occur in approximately 90% of pancreatic cancers, we examined the antitumor activity of erlotinib in combination with gemcitabine in KRAS-mutated pancreatic cancer cell lines, HPAC and Capan-1, which have the KRAS mutation G12D and G12V, respectively. We analyzed the mode of inhibition of in vitro tumor cell proliferation by means of a combination index and found that a combination treatment of erlotinib plus gemcitabine had an additive effect in the two cell lines. We then examined the effect of erlotinib and gemcitabine on the phosphorylation of epidermal growth factor receptor (EGFR). Erlotinib strongly suppressed, while gemcitabine augmented the phosphorylation of EGFR, which was completely blocked by erlotinib in the two cell lines. An in vivo tumor growth inhibition test was then performed using the HPAC tumor xenograft model. The combination therapy of erlotinib and gemcitabine resulted in a significant inhibition of tumor growth compared with erlotinib or gemcitabine monotherapy. To the best of our knowledge, this is the first study to show the combination effect of erlotinib and gemcitabine in vivo using a xenograft model of a KRAS-mutated pancreatic cancer cell line.

20.
Oncol Rep ; 24(5): 1141-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20878103

RESUMEN

Erlotinib is used as a standard treatment for recurrent advanced non-small cell lung cancer (NSCLC). Epidermal growth factor receptor (EGFR) mutations in NSCLC have been shown to be a predictive factor of erlotinib, although the relationship between K-ras oncogene (KRAS) mutations and erlotinib resistance is controversial. Recently, in vitro sequence-dependent interactions of erlotinib and docetaxel have been studied on as a novel therapeutic approach against NSCLC. The purpose of the present study was to determine the optimum novel regimen of erlotinib and docetaxel against NSCLC cells which have EGFR mutation (HCC827 cells), KRAS mutation (A549 cells) or both wild-type (NCI-H292 cells). First, we analyzed the effects of in vitro combination for cell proliferation-inhibition using a combination index. In all cell lines, docetaxel followed by erlotinib treatment showed nearly additive effects. On the other hand, erlotinib followed by docetaxel treatment showed remarkable antagonistic interactions. Second, we examined the effect of combinations on the in vitro apoptosis induction. Erlotinib followed by docetaxel treatment reduced apoptosis induction compared with docetaxel alone; in contrast, docetaxel followed by erlotinib treatment had no inhibitory effects on docetaxel-induced apoptosis in any of the cell lines. Finally, an in vivo tumor growth inhibition test was performed using xenograft models. Docetaxel followed by erlotinib administration resulted in significant tumor growth inhibition compared with erlotinib or docetaxel monotherapy in all models. In conclusion, we demonstrated that docetaxel followed by erlotinib therapy was a potentially optimum regimen against NSCLC regardless of the mutation status of EGFR and KRAS.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/genética , Genes ras , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Animales , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Docetaxel , Esquema de Medicación , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Erlotinib , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Quinazolinas/administración & dosificación , Taxoides/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA