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











Base de datos
Intervalo de año de publicación
1.
Clin Case Rep ; 9(9): e04759, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34484781

RESUMEN

Legionella is a rare cause of mild encephalitis/encephalopathy with reversible splenial lesion, which should be considered in patients with risk factors. Brain magnetic resonance imaging (MRI) and legionella urinary antigen test can help the diagnosis since cerebrospinal fluid (CSF) can be normal.

2.
Mol Cancer Ther ; 8(1): 119-26, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19139120

RESUMEN

Bone metastases occur in more than one-third of patients with advanced lung cancer and are difficult to treat. We showed previously the therapeutic effect of a third-generation bisphosphonate, minodronate, and anti-parathyroid hormone-related protein (PTHrP) neutralizing antibody on bone metastases induced by the human small cell lung cancer cell line, SBC-5, in natural killer cell-depleted severe combined immunodeficient mice. The purpose of our current study was to examine the effect of the combination of PTHrP antibody and zoledronic acid, which has been approved to treat bone metastases, against bone metastases produced by SBC-5 cells expressing PTHrP. Treatment with PTHrP antibody and/or zoledronic acid did not affect the proliferation of SBC-5 cells in vitro. Repeated treatments with either PTHrP antibody or zoledronic acid inhibited the formation of osteolytic bone metastases of SBC-5 cells but had no effect on metastases to visceral organs. Importantly, combined treatment with PTHrP antibody and zoledronic acid further inhibited the formation of bone metastases. Histologic assays showed that, compared with either PTHrP antibody or zoledronic acid alone, their combination decreased the number of tumor-associated osteoclasts and increased the number of apoptotic tumor cells. These findings suggest that this novel dual-targeting therapy may be useful for controlling bone metastases in a subpopulation of small cell lung cancer patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Difosfonatos/uso terapéutico , Imidazoles/uso terapéutico , Inmunoterapia , Neoplasias Pulmonares/tratamiento farmacológico , Proteína Relacionada con la Hormona Paratiroidea/inmunología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Animales , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones SCID , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Radiografía , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Zoledrónico
3.
Gan To Kagaku Ryoho ; 35(10): 1795-8, 2008 Oct.
Artículo en Japonés | MEDLINE | ID: mdl-18931592

RESUMEN

Platinum is the key drug in current treatment of ovarian cancer. The overall survival rate of patients with advanced ovarian cancer has improved. Recently, therapy-related myelodysplastic syndrome(T-MDS)and therapy-related leukemia( TRL)have been reported with increasing frequency in the literature. In this paper, we report two cases of TMDS followed by TRL after chemotherapy(TC therapy consisting of carboplatin and paclitaxel)with ovarian cancer stageIIIc. The interval from the primary chemotherapy to TMDS was 23 months or 90 months. The abnormal karyotype was observed in chromosomal analysis. Neither cases were given chemotherapy for TMDS, and died of the immunodeficiency. TRL and T-MDS are the most serious complications of chemotherapy. If pancytopenia is detected, bone marrow and cytogenetic examinations should be performed to rule out TMDS.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino/uso terapéutico , Leucemia/inducido químicamente , Síndromes Mielodisplásicos/inducido químicamente , Neoplasias Primarias Secundarias/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/uso terapéutico , Anciano , Femenino , Humanos , Persona de Mediana Edad , Insuficiencia del Tratamiento
4.
Clin Cancer Res ; 14(3): 660-7, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18245525

RESUMEN

PURPOSE: Follistatin (FST), an inhibitor of activin, regulates a variety of biological functions, including cell proliferation, differentiation, and apoptosis. However, the role of FST in cancer metastasis is still unknown. Previous research established a multiple-organ metastasis model of human small cell lung cancer in natural killer cell-depleted SCID mice. In this model, i.v. inoculated tumor cells produced metastatic colonies in multiple organs including the lung, liver, and bone. The purpose of this study is to determine the role of FST in multiple-organ metastasis using this model. EXPERIMENTAL DESIGN: A human FST gene was transfected into the small cell lung cancer cell lines SBC-3 and SBC-5 and established transfectants secreting biologically active FST. The metastatic potential of the transfectants was evaluated using the metastasis model. RESULTS: FST-gene transfection did not affect the cell proliferation, motility, invasion, or adhesion to endothelial cells in vitro. I.v. inoculated SBC-3 or SBC-5 cells produced metastatic colonies into multiple organs, including the lung, liver, and bone in the natural killer cell-depleted SCID mice. FST transfectants produced significantly fewer metastatic colonies in these organs when compared with their parental cells or vector control clones. Immunohistochemical analyses of the liver metastases revealed that the number of proliferating tumor cells and the tumor-associated microvessel density were significantly less in the lesions produced by FST transfectants. CONCLUSIONS: These results suggest that FST plays a critical role in the production of multiple-organ metastasis, predominantly by inhibiting the angiogenesis. This is the first report to show the role of FST in metastases.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Folistatina/fisiología , Células Asesinas Naturales/inmunología , Depleción Linfocítica , Metástasis de la Neoplasia/prevención & control , Animales , Carcinoma de Pulmón de Células no Pequeñas/inmunología , División Celular , Folistatina/genética , Humanos , Masculino , Ratones , Ratones SCID , Ratones Transgénicos , Transfección , Cicatrización de Heridas
5.
J Med Invest ; 54(1-2): 146-53, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17380026

RESUMEN

PURPOSE: Vascular endothelial growth factor (VEGF) plays a critical role in the production of malignant pleural effusions. In the present study, we examined the levels of soluble VEGF receptor-1 (sVEGFR-1) and angiopoietin-2 (Ang-2), as possible regulators of VEGF activity, in transudative and exudative pleural effusions. METHODS: Forty-two patients were included in this study: 4 with transudative pleural effusions due to heart failure (HF), 38 with exudative pleural effusions (lung cancer [LC], 22; other malignant diseases [MD], 10; tuberculosis [TB], 6). The levels of VEGF, Ang-2, and sVEGFR-1 in the pleural effusions were measured by an enzyme-linked immunosorbent assay. RESULTS: The levels of VEGF, Ang-2, and sVEGFR-1 in exudative effusions were higher than those in transudative effusions. Interestingly, the levels of VEGF and Ang-2 in bloody effusions were significantly higher than those in non-bloody effusions (p < 0.05), but the level of sVEGFR-1 in bloody effusions was lower than that in non-bloody effusions. The levels of VEGF and Ang-2 were significantly higher in the malignant effusions, compared with effusion from HF and TB (p < 0.05). In addition, sVEGFR-1 was significantly higher in the effusion from LC, MD, and TB compared with effusion from HF (p < 0.05). In the malignant effusions, direct correlations were observed among VEGF, sVEGFR-1, and Ang-2. CONCLUSIONS: The sVEGFR-1 levels were elevated in exudative pleural effusions, and were lower in bloody effusions than in non-bloody effusions, thus suggesting the regulatory role of sVEGFR-1 in the exudative pleural effusions.


Asunto(s)
Exudados y Transudados/química , Derrame Pleural/química , Receptor 1 de Factores de Crecimiento Endotelial Vascular/análisis , Adulto , Anciano , Angiopoyetina 1/análisis , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Derrame Pleural Maligno/química , Factor A de Crecimiento Endotelial Vascular/análisis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología
6.
Cancer Sci ; 97(3): 183-91, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16542214

RESUMEN

Malignant pleural mesothelioma (MPM) is closely related to exposure to asbestos, and a rapid increase in the number of MPM patients in Japan is estimated in the years 2010-2050. The purpose of the present study was to establish a clinically relevant animal model that shows human patient-like progression of MPM. Here, we demonstrate that a human MPM cell line (EHMES-10) inoculated orthotopically (thoracic cavity) into severe combined immunodeficiency (SCID) mice produces highly vascularized thoracic tumors with pleural dissemination and bloody pleural effusions by 5 weeks, suggesting a patient-like progression of this cell line after orthotopic inoculation. EHMES-10 cells overexpressed vascular endothelial growth factor (VEGF), a molecule responsible for malignant effusions, and its receptor. Treatment with cisplatin, but not gemcitabine, significantly inhibited the production of pleural effusions, but it was not effective for thoracic tumors, consistent with chemotherapy refractory characteristics of MPM in patients. Our patient-like orthotopic model using EHMES-10 cells overexpressing VEGF and its receptor may be useful for examining the molecular pathogenesis of MPM and may contribute to the development of novel treatment strategies for MPM.


Asunto(s)
Modelos Animales de Enfermedad , Mesotelioma/metabolismo , Neoplasias Pleurales/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cisplatino/uso terapéutico , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Humanos , Inmunohistoquímica , Masculino , Mesotelioma/tratamiento farmacológico , Mesotelioma/patología , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Gemcitabina
7.
Oncol Res ; 16(5): 235-43, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17294804

RESUMEN

Malignant pleural mesothelioma (MPM) is closely related to exposure to asbestos, and a rapid increase in the number of MPM patients is therefore estimated to occur from 2010 to 2040 in Japan. Because MPM is refractory to conventional chemotherapy and radiotherapy, the prognosis of MPM patients is extremely poor. MYO18B, a novel member of the myosin family, is a tumor suppressor gene isolated from a homozygously deleted region at 22q12.1 in a lung cancer cell line. The inactivation of the MYO18B gene plays an important role in several malignant diseases. However, the role of MYO18B in the progression of MPM is still unknown. Six different human MPM cell lines were used in this study. Western blot revealed that none of the cell lines expressed a detectable level of MYO18B protein. One of the MPM cell lines, EHMES-10, was transfected with the MYO18B gene. We found that a restored expression of the MYO18B protein in EHMES-10 cells resulted in the inhibition of their anchorage-independent growth and motility in vitro. In addition, it also inhibited their ectopic (subcutaneous space) and orthotopic (thoracic cavity) growth in SCID mice, in association with an increased degree of cell apoptosis. Furthermore, it also suppressed the production of bloody pleural effusion after orthotopic injection. These findings suggest that the restored expression of MYO18B may be a useful therapeutic strategy for the treatment of locally advanced MPM in humans.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Pulmonares/genética , Mesotelioma/genética , Miosinas/genética , Miosinas/farmacología , Derrame Pleural/genética , Neoplasias Pleurales/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/farmacología , Animales , Apoptosis/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Mesotelioma/tratamiento farmacológico , Mesotelioma/metabolismo , Ratones , Ratones SCID , Miosinas/metabolismo , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Derrame Pleural/metabolismo , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Clin Cancer Res ; 11(24 Pt 1): 8789-98, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16361567

RESUMEN

BACKGROUND: Small-cell lung cancer is often characterized by rapid growth and metastatic spread. Because tumor growth and metastasis are angiogenesis dependent, there is great interest in therapeutic strategies that aim to inhibit tumor angiogenesis. METHODS: The effect of ZD6474, an orally available inhibitor of vascular endothelial growth factor receptor-2 (VEGFR-2) and epidermal growth factor tyrosine kinases, was studied in experimental multiple-organ metastasis models with human small-cell lung cancer cell lines (SBC-3 or SBC-5) in natural killer cell-depleted severe combined immunodeficient mice. RESULTS: Intravenously inoculated SBC-5 cells produced experimental metastases in the liver, lung, and bone whereas SBC-3 cells produced the metastases in the liver, systemic lymph nodes, and kidneys. Daily oral treatment with ZD6474 (50 mg/kg), started on day 14 (after the establishment of micrometastases), significantly reduced the frequency of large (>3 mm) metastatic colonies (in the liver and lymph nodes) and osteolytic bone lesions. ZD6474 treatment did not significantly reduce the frequency of small (<2-3 mm) metastatic lesions found in the lung (SBC-5) or kidney (SBC-3), consistent with an antiangiogenic mechanism of action. Immunohistochemical analysis of SBC-5 metastatic deposits in the liver showed that ZD6474 treatment inhibited VEGFR-2 activation and induced apoptosis of tumor-associated endothelial cells, resulting in decreasing tumor microvessel density. ZD6474 treatment was also associated with a decrease in tumor cell proliferation and an increase in tumor cell apoptosis. The antitumor effects of ZD6474 were considered likely to be due to inhibition of VEGFR-2 tyrosine kinase because gefitinib, a small-molecule inhibitor of epidermal growth factor receptor tyrosine kinase, was inactive in these models. CONCLUSIONS: These results suggest that ZD6474 may be of potential therapeutic value in inhibiting the growth of metastatic small-cell lung cancer in humans. Phase II trials with ZD6474 are currently ongoing in a range of solid tumors.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma de Células Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Piperidinas/uso terapéutico , Quinazolinas/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Carcinoma de Células Pequeñas/inmunología , Carcinoma de Células Pequeñas/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Depleción Linfocítica , Ratones , Ratones SCID , Neovascularización Patológica/inmunología , Piperidinas/farmacología , Quinazolinas/farmacología , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Clin Cancer Res ; 11(24 Pt 1): 8822-8, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16361571

RESUMEN

PURPOSE: The purpose of this study was to determine therapeutic effect of a novel antibiotic, reveromycin A, against osteolytic bone metastasis of human small cell lung cancer (SBC-5) cells. RESULTS: Reveromycin A induced apoptosis specifically in osteoclasts in vitro. Although reveromycin A did not inhibit SBC-5 cell proliferation, it suppressed the expression of parathyroid hormone-related peptide. Intravenous inoculation of SBC-5 cells in natural killer cell-depleted severe combined immunodeficient mice produced experimental metastases in multiple organs, including the bone. Daily administration of reveromycin A inhibited the bone metastasis, but not visceral metastasis, in a dose-dependent manner. Histologic analyses revealed that although treatment with reveromycin A did not affect the number of proliferating tumor cells, it decreased the number of osteoclasts and increased apoptotic cells in bone lesions. CONCLUSIONS: These findings suggest that reveromycin A may inhibit osteolytic bone metastasis through suppression of osteoclast activity by directly inducing apoptosis and indirectly inhibiting tumor cell-derived parathyroid hormone-related peptide production. Therefore, reveromycin A may be a novel, potent therapeutic agent against osteolytic bone metastasis of lung cancer in humans.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Carcinoma de Células Pequeñas/tratamiento farmacológico , Carcinoma de Células Pequeñas/secundario , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Piranos/uso terapéutico , Compuestos de Espiro/uso terapéutico , Animales , Carcinoma de Células Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones SCID , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Piranos/farmacología , Compuestos de Espiro/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Clin Exp Metastasis ; 22(3): 195-204, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16158247

RESUMEN

Src, a proto-oncogene, has been strongly implicated in the growth, progression and metastasis of a number of human cancers. Its role in lung cancer is, however, still unknown. In the present study, we assessed the expression of Src in three different human lung adenocarcinoma cell lines (PC-9, PC14PE6, A549), and explored the effect of a novel Src kinase inhibitor, M475271, on the behavior of the cell lines. The three cell lines expressed various levels of auto-phosphorylated Src. While M475271 reduced Src-phosphorylation and invasiveness of all three cell lines, it inhibited the proliferation of PC-9 and A549 cells with highly phosphorylated Src, but not PC14PE6 cells. We further examined the effect of M475271 on subcutaneous tumors and lung metastasis caused by PC-9 and/or A549 cells in NK-cell depleted SCID mice. Daily oral treatment with M475271 inhibited the growth of subcutaneous tumors with PC-9 and A549 cells via inhibition of tumor cells proliferation, VEGF production and/or vascularization in the mice in a dose-dependent manner. In the metastasis model with A549 cells, the lung weight in the M475271 (50 mg/kg)-treated group was less than that of the control group, despite no difference in the number of metastatic nodules. Our results suggest that inhibition of tyrosine kinase Src by M475271 could reduce the growth, invasion and VEGF-mediated neovascularization of lung adenocarcinoma cells, resulting in inhibition of growth of subcutaneous tumors and lung metastasis. Therefore, a novel Src tyrosine kinase inhibitor, M475271, might be helpful for controlling the progression of human lung adenocarcinoma.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Piperidinas/farmacología , Quinazolinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adenocarcinoma/patología , Adenocarcinoma/fisiopatología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Humanos , Técnicas In Vitro , Inyecciones Subcutáneas , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/fisiopatología , Masculino , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Piperidinas/administración & dosificación , Proto-Oncogenes Mas , Quinazolinas/administración & dosificación , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Familia-src Quinasas/biosíntesis
11.
Oncol Res ; 15(2): 107-11, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16119008

RESUMEN

Gefitinib is a selective inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinases, and shows favorable antitumor activity against chemorefractory non-small cell lung cancer (NSCLC). The majority of responders (patients who are sensitive to gefitinib), however, relapse within 1.5 years, indicating an acquired resistance to gefitinib. Here we report three chemotherapy refractory NSCLC patients who were retreated with gefitinib. All three cases were nonsmokers and showed an adenocarcinoma histology. While they had experienced successful control from their initial treatment with gefitinib for more than 12 months, gefitinib therapy was terminated because two cases (cases 1 and 3) relapsed during the therapy and case 2 suffered alveolar hemorrhage. After more than 7 months from the time of discontinuation of the initial gefitinib treatment, they were retreated with gefitinib, as further tumor progression was observed. Of the three cases, cases 1 and 2 were well controlled by retreatment with gefitinib monotherapy for more than 7 months, suggesting sensitivity to retreatment. Case 3 also showed a regression in size of several tumors, while some other lesions progressively enlarged and developed a malignant pleural effusion after 4 months. These observations suggest the possibility that retreatment with gefitinib might be useful when 1) initial treatment shows a favorable clinical response, and 2) there has been a period of time following the termination of the initial gefitinib treatment.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Quinazolinas/farmacología , Anciano , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Gefitinib , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Derrame Pleural , Recurrencia , Retratamiento , Factores de Tiempo
13.
Int J Cancer ; 108(4): 511-5, 2004 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-14696114

RESUMEN

We previously established an osteolytic bone metastasis model with multiorgan dissemination in natural killer (NK) cell-depleted severe combined immunodeficient (SCID) mice using human small cell lung cancer cells (SBC-5), which highly express the parathyroid hormone-related protein (PTHrP). In our present study, we evaluated the role of PTHrP on bone metastasis by SBC-5 cells using anti-PTHrP neutralizing antibody (Ab). Anti-PTHrP Ab did not affect the proliferation or cytokine production of SBC-5 cells in vitro. Repeated intravenous injection with anti-PTHrP Ab inhibited the formation of bone metastasis in a dose-dependent manner, while the same treatment had no significant effect on the metastasis to visceral organs (lung, liver, kidney and lymph node). In addition, treatment with anti-PTHrP Ab improved the elevated serum calcium level, associated with inhibition of osteolytic bone metastasis, suggesting that anti-PTHrP Ab inhibited bone metastasis via suppression of bone resorption probably by neutralizing PTHrP. These findings suggest that PTHrP is essential for bone metastasis, but not visceral metastasis, by small cell lung cancer SBC-5 cells.


Asunto(s)
Neoplasias Óseas/secundario , Carcinoma de Células Pequeñas/secundario , Células Asesinas Naturales/fisiología , Neoplasias Pulmonares/patología , Depleción Linfocítica , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Animales , Anticuerpos Bloqueadores , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Óseas/metabolismo , Calcio/metabolismo , Carcinoma de Células Pequeñas/metabolismo , División Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/secundario , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/metabolismo , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos ICR , Ratones SCID , Células Tumorales Cultivadas
15.
Clin Exp Metastasis ; 20(2): 153-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12705636

RESUMEN

In the present study, we examined the effects of a newly developed bisphosphonate, minodronate (YM529), on osteolytic bone metastasis caused by lung cancer. Human small-cell lung cancer (SBC-5) cells, injected intravenously into natural killer cell-depleted SCID mice, produced radiologically detectable bone metastasis by day 18 and macroscopically visible visceral metastases (lung, liver, kidney, systemic lymph node) by day 35. Prophylactic treatment with YM529 on day 1 significantly inhibited the formation of osteolytic bone metastasis evaluated on X-ray photographs in a dose-dependent manner. In addition, treatment with YM529 after establishment of bone metastasis (on day 21) also inhibited bone metastasis, although the treatment was more effective when started earlier. Single administration was as effective as repeated treatment, suggesting a sustained inhibitory effect of YM529 on bone metastasis. YM529 reduced the number of osteoclasts in the bone metastatic lesions in vivo, but had no effect on the proliferation or cytokine production of SBC-5 cells in vitro. These results suggest that YM529 is a potent inhibitor of bone metastasis of human lung cancer, probably by suppressing osteoclastic bone resorption. In contrast, treatment with YM529 had no effect on visceral metastasis, even if started on day 1, and did not prolong the survival of the mice. Therefore, development of a combined modality is necessary for prolonging the survival of small-cell lung cancer patients with multiple-organ metastasis.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Carcinoma de Células Pequeñas/tratamiento farmacológico , Difosfonatos/uso terapéutico , Imidazoles/uso terapéutico , Células Asesinas Naturales/fisiología , Neoplasias Pulmonares/tratamiento farmacológico , Depleción Linfocítica , Animales , Antineoplásicos/uso terapéutico , Neoplasias Óseas/inmunología , Resorción Ósea/tratamiento farmacológico , Carcinoma de Células Pequeñas/inmunología , Carcinoma de Células Pequeñas/secundario , División Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Factores de Crecimiento Endotelial/metabolismo , Humanos , Técnicas para Inmunoenzimas , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/secundario , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Metástasis Linfática/prevención & control , Linfocinas/metabolismo , Masculino , Ratones , Ratones SCID , Osteólisis , Proteína Relacionada con la Hormona Paratiroidea , Hormonas Peptídicas/metabolismo , Inmunodeficiencia Combinada Grave/inmunología , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA