Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Endocrinol (Lausanne) ; 12: 795724, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975766

RESUMO

We analyzed data from 466 patients with premature ovarian insufficiency (POI) who wished to have a biological child and were followed up while undergoing hormone replacement (HR) therapy with or without ovarian stimulation (OS) between April 2014 and December 2020. OS was conducted in 6891 cycles in 429 patients (Group OS), whereas only HR (Group HR) was conducted in 1117 cycles in 37 patients. The follicle growth rate was 48.3% (207/429) per patient in Group OS and 5.4% (2/37) in Group HR (p<0.01). There were 51 live births (LBs) in 50 patients during follow-up. In Group OS, the LB rate was 5.8% (47/807) in cycles where in vitro fertilization (IVF) and embryo transfer were attempted (Group IVF), and 1.3% (3/236) in cycles where intrauterine insemination/timed intercourse was attempted (p<0.01). No pregnancies occurred in Group HR. Among the patients in Group IVF, the LB rate was significantly higher in patients aged <35 years at the initiation of follow-up than in patients who started at later ages (p<0.01). Among the cases who achieved an LB, 39 were patients with idiopathic POI (Group IVF-1, n=297) and seven were patients who had undergone surgical treatment for benign ovarian tumors (Group IVF-2, n=50); however, no LBs occurred in patients who had undergone treatment for malignancy (n=17), and only one in patients with chromosomal abnormalities (n=22). The LB rate per case in the patients in Group IVF-1 and those aged <35 years at the start of follow-up (Group IVF-1-a) was 24.1% (26/108), which was higher than those of the other age groups. The LB rate per case in the patients in Group IVF-1-a with <4 years of amenorrhea was 37.3% (19/51), and that in the patients in Group IVF-2 with <4 years of amenorrhea was 21.2% (7/33). These results suggest that infertility treatment is possible in some patients with POI, especially those that can be classified in Group IVF-1-a and Group IVF-2 with <4 years of amenorrhea. Therefore, OS combined with HR therapy should be considered for such patients before attempts at oocyte donation.


Assuntos
Terapia de Reposição de Estrogênios/tendências , Infertilidade Feminina/terapia , Nascido Vivo , Indução da Ovulação/tendências , Insuficiência Ovariana Primária/terapia , Adulto , Estudos de Coortes , Terapia de Reposição de Estrogênios/métodos , Feminino , Seguimentos , Humanos , Infertilidade Feminina/sangue , Masculino , Indução da Ovulação/métodos , Gravidez , Insuficiência Ovariana Primária/sangue , Estudos Retrospectivos , Análise do Sêmen/métodos , Análise do Sêmen/tendências , Fatores de Tempo
2.
Retin Cases Brief Rep ; 13(2): 118-120, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-28248745

RESUMO

PURPOSE: To report a case of macular telangiectasia Type 2 that showed a resolution of a foveal cystic change accompanying the release of vitreomacular attachment. METHODS: Retrospective chart review. REPORT OF THE CASE: A 75-year-old man presented with a bilateral decrease in visual acuity and metamorphopsia. At the initial visit, the left eye demonstrated a hyporeflective foveal cyst and a perifoveal vitreous detachment on optical coherence tomography. The characteristic clinical findings were consistent with the diagnosis of macular telangiectasia Type 2. Fifteen months later, the patient returned with a remission of the metamorphopsia in the left eye. The left eye had a vitreous detachment from the macula, and the hyporeflective foveal cyst was completely resolved. CONCLUSION: At least in some cases, vitreous traction or attachment may play a role in the morphologic changes seen in macular telangiectasia Type 2.


Assuntos
Cistos/patologia , Fóvea Central/patologia , Telangiectasia Retiniana/patologia , Descolamento do Vítreo/patologia , Idoso , Humanos , Masculino , Remissão Espontânea , Estudos Retrospectivos , Transtornos da Visão/etiologia
3.
Reprod Med Biol ; 17(4): 493-499, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30377405

RESUMO

PURPOSE: High-mobility group N (HMGN) proteins are the only non-histone proteins that specifically bind within the nucleosome between core histones and DNA. Among them, HMGN5 is one of the candidates that could participate in mouse endometrial decidualization; however, the specific role of HMGN5 remains to be clarified in human endometrial stromal cells (HESCs). METHODS: Primary HESCs were isolated from hysterectomy specimens and incubated with or without 8-bromo-cyclic adenosine monophosphate (8-br-cAMP) and medroxyprogesterone acetate (MPA). RESULTS: We demonstrated that HMGN5 expression in decidualized HESCs stimulated by 8-br-cAMP and MPA decreased significantly. The inhibition of HMGN5 expression by small interfering RNA (siRNA) induced the major decidual marker genes expression, including IGFBP1 (insulin-like growth factor binding protein 1) and PRL (prolactin). In addition, microRNA-542-3p (miR-542-3p), which was identified as a regulatory miRNA of IGFBP1 during decidualization, was significantly suppressed by HMGN5 siRNA. However, the expression of HMGN5 was not alternated by miR-542-3p overexpression. CONCLUSIONS: These findings suggest that the down-regulation of HMGN5 plays a role in the promotion of human endometrial stromal decidualization and acts upstream of miR-542-3p.

4.
Reprod Med Biol ; 16(2): 170-178, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-29259466

RESUMO

Aim: Endometriosis is defined as the presence of endometrial glandular and stromal cells outside of the uterine cavity. A previous study reported that microRNA (miR)-542-3p plays a critical role in eutopic endometrial decidualization. This study aims to clarify the potential role of miR-542-3p and the target gene, IGFBP-1 (insulin-like growth factor-binding protein 1), in the impairment of the decidualizing capacity of human ectopic endometrial stromal cells (HEcESCs). Methods: In vitro analysis of primary undifferentiated and decidualizing human eutopic endometrial stromal cells (HEuESCs) and HEcESCs was conducted. The primary HEuESCs or HEcESCs were expanded in culture and decidualized with 8-bromo-cyclic adenosine monophosphate (8-bromo-cAMP) and medroxyprogesterone acetate (MPA). Results: The morphological and biological differentiating capacities of the HEcESCs were markedly impaired. In contrast to the HEuESCs, the HEcESCs that were treated with the decidual stimulus retained the mesenchymal phenotype and capacity for migration. The down-regulation of miR-542-3p in the HEcESCs treatment with 8-bromo-cAMP and MPA was much weaker than that of the HEuESCs. High expression of miR-542-3p led to a significant decrease in the expression of IGFBP1 in the HEcESCs. Conclusion: Impairment of the differentiating capacity by the overexpression of miR-542-3p could influence the capacity for migration and invasion of endometriotic cells in an ectopic environment.

5.
Oncol Rep ; 35(4): 2495, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26781072

RESUMO

Oncol Rep 17: [Related article:] 289­295, 2007; DOI: 10.3892/or.17.2.289 After the publication of the article, the authors noted that the relevance of the findings reported in this article on ZD6474 inhibition of growth of head and neck squamous cell carcinoma (HNSCC) is now in question. To examine the antitumor effect of ZD6474 in vitro and in vivo, this study was conducted using YCU-H891 cell line that the authors believed to be HNSCC cell line. Because different human leukocyte antigen (HLA) was detected in 2 cell lines (KCC-TCM901 and KCC-T873) which were derived from the same patient in an experiment after publication, 16 cell lines established in our institution were analyzed by short tandem repeat (STR) analysis. STR analysis revealed that genotype of KCC-TCM901, YCU-H891 and KCC-MS871 was identical to that of HeLa cells, and that genotype of YCU-T891 was identical to that of YCU-L891, which was considered to be cross-contamination.

6.
Clin Case Rep ; 1(2): 50-3, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25356211

RESUMO

KEY CLINICAL MESSAGE: Maternal serum screening-positive patient had prenatal diagnosis with amniotic fluid, which showed inconsistent results between interphase fluorescence in situ hybridization (three signals of 21q22.13-21q22.2) and G-banding analysis (46,XY). Further analyses proved that the fetus had extremely complex rearrangements of chromosome 21, including the interstitial duplication of Down syndrome critical region.

7.
Nagoya J Med Sci ; 73(3-4): 129-35, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21928694

RESUMO

Free fetal DNA (ffDNA) in maternal plasma has now become a valuable source for noninvasive prenatal diagnosis. Being able to accurately identify the size of ffDNA in maternal plasma is essential for a noninvasive prenatal diagnosis. Furthermore, it is important to investigate the molecular characteristics related to apoptosis which gives rise to ffDNA. We investigated the fragment size of ffDNA in each sample more precisely, using both Y-STR and SRY primers, in 20 maternal plasma samples from the 17th to 39th weeks of gestation. PCR was conducted with Y-STR and SRY primers which can be used to amplify 100-524 bp fragments. In samples from 10 pregnant women carrying male fetuses, the maximum fragment size detected by Y-STR and SRY primers ranged from 219 to 313 bp. As a result, the mean average maximum fragment size of free fetal DNA detected by Y-STR and SRY primers was 286 +/- 28 bp. The Y-STR alleles detected in each maternal plasma DNA sample were all in agreement with the results of their cord blood samples. We concluded that the fragment size of ffDNA comprises 2 nucleosomal complexes or less, but not exceeding 3.


Assuntos
Cromossomos Humanos Y/genética , DNA/sangue , Feto/citologia , Genes sry , Repetições de Microssatélites/genética , Diagnóstico Pré-Natal/métodos , Apoptose , Feminino , Humanos , Reação em Cadeia da Polimerase , Gravidez
8.
Auris Nasus Larynx ; 38(5): 589-99, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21362583

RESUMO

OBJECTIVE: Telomelysin (OBP-301) is a telomerase-specific replication-competent adenovirus with a human telomerase reverse transcriptase (hTERT) promoter. Telomelysin has a strong antitumor effect on a variety of cancers, including head and neck squamous cell carcinoma (HNSCC), and combining telomelysin treatment with paclitaxel or cisplatin enhances the antitumor effect on HNSCC. In the present study, we investigated the relationship between the antitumor activity of telomelysin and tumor cell doubling time(DT), S-phase fraction, and E1A expression. We also investigated whether the antitumor effects of OBP-301-resistant tumor cells are enhanced by cisplatin, paclitaxel, or streptolysin O. METHODS: The tumor cell DT of 17 human HNSCC cell lines was examined. Antitumor activities of telomelysin (OBP-301) for each HNSCC cell line were examined by MTT assay. Cell cycle analysis was conducted by flowcytometry. E1A gene expressions after infection with telomelysin, hTERT, CAR (Cocksackie Adenovirus Receptor), and c-Myc were examined by quantitative PCR, and E1A expressions were examined again after pretreatment with cisplatin, paclitaxel, or streptolysin O. Correlations were analyzed by Spearman's correlation coefficient. RESULTS: There was a significant relationship between telomelysin sensitivity and DT, S-phase fraction and early E1A expression, and pretreatment with cisplatin, paclitaxel, and streptolysin O increased infectivity of telomelysin-resistant HNSCC cell lines. CONCLUSION: These findings are useful for advancing clinical trials, and suggest that adjuvant telomelysin treatment would be effective even in telomelysin-resistant HNSCC cell lines.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias de Cabeça e Pescoço/patologia , Terapia Viral Oncolítica , Vírus Oncolíticos , Infecções por Adenoviridae , Proteínas E1A de Adenovirus/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Proteínas de Bactérias/farmacologia , Carcinoma/metabolismo , Carcinoma/patologia , Carcinoma de Células Escamosas , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes myc , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Neoplasias de Células Escamosas/metabolismo , Neoplasias de Células Escamosas/patologia , Terapia Viral Oncolítica/métodos , Paclitaxel/farmacologia , RNA Mensageiro/metabolismo , Receptores Virais/genética , Fase S/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Estreptolisinas/farmacologia , Telomerase/genética
9.
Oncol Rep ; 23(4): 957-63, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20204279

RESUMO

The epidermal growth factor receptor (EGFR) and a related family member, HER-2, are often overexpressed simultaneously in patients with a variety of malignant tumors, and the combination may cooperatively promote cancer cell growth and survival. Heterodimerization of EGFR and HER-2 has been known to create intense proliferative signals. Lapatinib (GW572016) is a small molecule that is administrated orally and functions as a reversible inhibitor of both EGFR and HER-2 tyrosine kinases. In the present study, we evaluated the antitumor effect of lapatinib on head and neck squamous cell carcinoma (HNSCC) cell lines in vitro and in vivo. In vivo we examined the antitumor effects of combined treatment with lapatinib and either cisplatin or paclitaxel. In vitro lapatinib displayed antiproliferative effects on HNSCC cells. The IC50 of lapatinib ranged between 13.6 and 60.2 microM after 24-h exposure to lapatinib. A correlation was not observed between results of in vitro proliferation assays for lapatinib and the expression of EGFR or HER-2. In vivo lapatinib displayed antitumor activity, and induced apoptosis in nude mice bearing an established xenograft of YCU-H891 cells. Lapatinib did not significantly inhibit angiogenesis. Combination treatment of lapatinib with cisplatin or paclitaxel enhanced antitumor activity mainly by inducing apoptosis. Inhibition of antiangiogenesis was observed only for combination treatment of lapatinib with paclitaxel (compared to vehicle control). These results suggest that: i) lapatinib has antitumor effects in vitro and in vivo; ii) lapatinib may be more effective in combination with cisplatin or paclitaxel; and iii) lapatinib might provide useful clinical benefits to HNSCC patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/administração & dosagem , Receptores ErbB/efeitos dos fármacos , Humanos , Marcação In Situ das Extremidades Cortadas , Concentração Inibidora 50 , Lapatinib , Camundongos , Paclitaxel/administração & dosagem , Quinazolinas/administração & dosagem , Receptor ErbB-2/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncol Rep ; 23(2): 355-63, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20043095

RESUMO

Telomelysin (OBP-301) is a telomerase-specific replication-component adenovirus. Telomelysin has a human telomerase reverse transcriptase (hTERT) promoter element which efficiently kills human cancer cells, but not normal cells. The present study investigated the correlation between the antitumor effect of telomelysin and mRNA expression of hTERT and coxsackievirus and adenovirus receptor (CAR) in head and neck squamous cell carcinoma (HNSCC) in vitro and whether telomelysin enhances the antitumor effect of paclitaxel or cisplatin, in vivo using a HNSCC xenograft model. We also determined the optimal order for combining telomelysin treatment and chemotherapy as concurrent treatment, telomelysin treatment first and chemotherapy later, chemotherapy first and telomelysin treatment later for achieving the best anticancer effect. The mRNA expression of hTERT and CAR genes was examined by quantitative RT-PCR in 17 HNSCC cell lines. There was no significant correlation between the growth inhibition of telomelysin (ID50 for day 3, 5 and 7) in vitro and mRNA expression levels of hTERT and CAR. Regarding the correlation between CAR expression and telomelysin ID50 for day 3, all cell lines that showed a relative amount of CAR/beta-actin mRNA >0.4 had a low telomelysin ID50. This may indicate that CAR expression contributes to the efficacy of adenovirus infection and the antitumor activity of telomelysin in early stages of treatment. In our in vivo study, combining telomelysin and paclitaxel had an additive effect regardless of treatment order. On the other hand, combining telomelysin and cisplatin had additive effect only when cisplatin treatment preceded telomelysin treatment. These results suggest that paclitaxel is considered innocuous for replication of telomelysin, however cisplatin may influence replication of telomelysin.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/terapia , Cisplatino/administração & dosagem , Neoplasias de Cabeça e Pescoço/terapia , Vírus Oncolíticos/fisiologia , Paclitaxel/administração & dosagem , Adenoviridae/química , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Terapia Combinada , Feminino , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia Viral Oncolítica , Regiões Promotoras Genéticas/genética , Telomerase/administração & dosagem , Telomerase/genética , Telomerase/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Int J Clin Oncol ; 14(4): 337-43, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19705245

RESUMO

BACKGROUND: Chemotherapy-induced nausea and vomiting (CINV) remains a significant problem for patients and is associated with a substantial deterioration in quality of life; appropriate use of antiemetic drugs is crucial in maintaining the quality of life in patients undergoing chemotherapy. METHODS: This randomized, crossover trial evaluated the antiemetic efficacy and safety of 8 mg per day (low-dose) and 16 mg per day (standard-dose) dexamethasone, in combination with the 5-HT(3) receptor antagonist granisetron, in 36 patients receiving cisplatin (CDDP)-containing chemotherapy for head and neck cancer. Following chemotherapy, the antinausea/vomiting inhibition rate for each dexamethasone dose was measured. RESULTS: During the 24-h period following administration of chemotherapy (acute phase), the antinausea/vomiting inhibition rates (no nausea and no episodes of vomiting) for 8 mg and 16 mg dexamethasone were comparably high (58.3% and 63.8%, respectively; P = 0.8092). Similar results were seen on days 2-5 following chemotherapy. Efficacy during the acute phase, based on the number of instances of vomiting and degree of nausea, was also comparably high for the two dexamethasone doses (overall efficacy rates were 94.4% and 88.8%, respectively, for 8 mg and 16 mg dexamethasone; P = 0.7637). Both doses maintained an 80% or higher response rate until day 3, and neither dose produced severe side effects. CONCLUSION: The results suggest that granisetron and dexamethasone combination therapy is useful in controlling acute and delayed nausea and vomiting induced by CDDP-containing chemotherapy for head and neck cancer. Furthermore, 8 mg and 16 mg dexamethasone have equivalent antiemetic efficacy.


Assuntos
Antieméticos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Dexametasona/administração & dosagem , Granisetron/administração & dosagem , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Náusea/prevenção & controle , Antagonistas do Receptor 5-HT3 de Serotonina , Antagonistas da Serotonina/administração & dosagem , Vômito/prevenção & controle , Idoso , Antieméticos/efeitos adversos , Apetite/efeitos dos fármacos , Cisplatino , Estudos Cross-Over , Dexametasona/efeitos adversos , Quimioterapia Combinada , Feminino , Granisetron/efeitos adversos , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Náusea/induzido quimicamente , Estadiamento de Neoplasias , Antagonistas da Serotonina/efeitos adversos , Fatores de Tempo , Resultado do Tratamento , Vômito/induzido quimicamente
12.
Int J Oncol ; 34(2): 321-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19148465

RESUMO

The ephB4-ephrinB2 system plays an important role in the interaction of tumor cells with endothelial cells (ECs). To assess the role of ephB4 in the in vivo growth of head and neck squamous cell carcinoma (HNSCC), we used ephrinB2-Fc, a fusion protein consisting of the extracellular domain of ephrin-B2 and the Fc portion of human IgG1, as the soluble ligand for ephB4. EphrinB2-Fc injection into HNSCC xenografted mice significantly suppressed xenograft growth, accompanied by a decrease in vessel cross-sectional area, but there was no change in vessel number. EphrinB2-Fc injection also induced the formation of mature blood vessels rich in alpha-smooth muscle actin positive pericytes in the xenograft tissue. In vitro assays revealed that ephrinB2-Fc inhibited the proliferation of human umbilical vein ECs (HUVECs) but not tumor cells. Furthermore, real-time quantitative RT-PCR showed that ephrinB2-Fc down-regulated matrix metalloproteinase-2 mRNA expression in HUVECs and vascular endothelial growth factor-A in tumor cells. These data suggest that treatment with ephrinB2-Fc, the soluble ligand of ephB4, inhibited the growth of HNSCC through vessel maturation/stabilization, preventing leakiness and endothelial sprout formation.


Assuntos
Carcinoma de Células Escamosas/patologia , Divisão Celular/efeitos dos fármacos , Efrina-B2/farmacologia , Neoplasias de Cabeça e Pescoço/patologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/irrigação sanguínea , Primers do DNA , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/patologia , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
13.
Oncol Rep ; 20(6): 1363-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19020715

RESUMO

Telomelysin is a telomerase-specific replication-competent adenovirus with telomerase reverse transcriptase (hTERT) promoter, which has shown strong anti-tumor effects on a variety of human cancer cells. Human head and neck squamous cell carcinoma (HNSCC) cell lines and a murine HNSCC (NR-S1) model were used to investigate whether telomelysin (OBP-301) had a therapeutic efficacy for HNSCC. We examined the cell killing effects of telomelysin and the induction of tumor cell apoptosis by telomelysin in vitro. Based on these data, we examined whether telomelysin therapy produced therapeutic benefits in vivo. The results demonstrated that the treatment of telomelysin led to significant tumor regression on the side with subcutaneous NR-S1 tumor. We first confirmed the direct anti-tumor effect of intratumoral telomelysin injections in a murine HNSCC model. Further analyses of the augmented anti-tumor effects revealed that telomelysin increased the source of tumor antigens for immune cells, resulting in the induction of CD4+ and CD8+ T cells responsible for the in vivo tumor regression of treated and untreated tumors. Subsequently, an elevated IFN-gamma production of spleen cells was observed in mice treated with telomelysin. These results raise the possibility that telomelysin enhances the immune response in addition to its direct tumor cell killing activity. These findings suggest that telomelysin is a potent agent for the treatment of HNSCC patients with multiple metastases.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Animais , Antígenos de Neoplasias/química , Apoptose , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C3H , Metástase Neoplásica , Transplante de Neoplasias
14.
Oncol Rep ; 20(2): 373-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18636200

RESUMO

The overexpression of EGFR and/or HER-2 is associated with tumor cell resistance to chemotherapy, radiotherapy, disease progression and poor prognosis in patients with a variety of malignant tumors. Treatment combining the EGFR-targeting drug, gefitinib (ZD1839, Iressa) with the HER-2-targeting drug, trastuzumab (Herceptin) has been reported to improve therapeutic efficacy in patients with breast cancer. The purpose of this study was to examine the antitumor effect of this combination on head and neck squamous cell carcinoma (HNSCC) in vitro. Cell proliferation was inhibited significantly in two cell lines. Although IC50 of gefitinib alone against some cell lines was not reached, it was achieved after being combined with trastuzumab. Furthermore, IC50 was lower for the combination than for gefitinib alone in several cell lines. These results suggest that the combination may improve efficacy against HNSCC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Gefitinibe , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Concentração Inibidora 50 , Quinazolinas/administração & dosagem , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Trastuzumab , Células Tumorais Cultivadas
15.
Transfusion ; 48(6): 1156-63, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18422850

RESUMO

BACKGROUND: Recently, a more accurate and reliable screening test has been investigated for noninvasive prenatal fetal RHD genotyping from D- women. The objective of this study was to perform the new method of noninvasive fetal RHD genotyping with maternal plasma from D- women by use of capillary electrophoresis. STUDY DESIGN AND METHODS: Blood samples were obtained from 8 D+ and 8 D- nonpregnant donors and mixed to make test plasma samples. DNA was extracted and the appropriate conditions relating to the initial sample volume as well as polymerase chain reaction cycle numbers were analyzed to detect the RHD gene with RHD exon 10 primer. Blood samples were also obtained from 13 D- pregnant women ranging from the 12th to 39th weeks of gestation. The presence of the RHD gene and Y-chromosome-specific STR (Y-STR) derived from the fetus was analyzed. The results were compared with the D status of newborns. RESULTS: In samples from 12 D- pregnant women, the RHD gene was detected. In one sample, the RHD gene was not detected but Y-STR loci were demonstrated in this sample, indicating a D- male baby. The results of fetal genotyping were all in concordance with the postpartum samples by serologic tests on D as well as with the sex of newborns. CONCLUSION: Capillary electrophoresis can be used for the determination of fetal RHD status in D- women. This diagnostic method is useful for the noninvasive prenatal diagnosis of the fetal RHD genotyping from D- women.


Assuntos
Eletroforese Capilar/métodos , Feto/metabolismo , Mães , Sistema do Grupo Sanguíneo Rh-Hr/sangue , Sistema do Grupo Sanguíneo Rh-Hr/genética , Feminino , Feto/imunologia , Genótipo , Humanos , Gravidez , Sistema do Grupo Sanguíneo Rh-Hr/imunologia
16.
Oncol Rep ; 18(1): 47-51, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17549344

RESUMO

Human tumors are dependent on angiogenesis for growth, and the vascular endothelial growth factor (VEGF) is a major regulator of this process. Bevacizumab (Avastin), a monoclonal antibody directed against VEGF, has shown promise in treating a variety of cancers. In this study, we first examined the anti-tumor effects of bevacizumab on head and neck squamous cell carcinoma (HNSCC). Then we examined the effects of bevacizumab combined with paclitaxel, a chemotherapeutic agent, in HNSCC. This is the first demonstration of the anti-tumor effects of bevacizumab on HNSCC. In vitro, bevacizumab did not show any antiproliferative effects against the HNSCC cell lines. However, in vivo, bevacizumab showed dramatic anti-tumor effects against HNSCC tumor xenografts in mice. In addition, treatment with a bevacizumab-paclitaxel combination resulted in a remarkable inhibition of the HNSCC tumor xenografts, compared to the effects of each agent separately. A decreased blood vessel density and an increased apoptotic index were seen in the shrunken tumors. These results suggest that bevacizumab in combination with paclitaxel could have useful clinical application in HNSCC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Bevacizumab , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/patologia , Feminino , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microcirculação , Neovascularização Patológica/patologia , Paclitaxel/administração & dosagem , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/transplante , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Auris Nasus Larynx ; 34(4): 487-91, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17337140

RESUMO

OBJECTIVE: Nafamostat mesilate (FUT-175), a synthetic serine protease inhibitor, has antitumor activities toward adenocarcinoma, e.g., colon cancer. However, its antitumor effects on other types of cancer have been less extensively studied. We investigated the biological activities of Nafamostat mesilate on cell proliferation, cell-invasive potential and growth factor production in head and neck squamous cell carcinoma (HNSCC). METHODS: Two human HNSCC cell lines established in our department, YCU-L891 and -H891, and a human vulvar squamous cell carcinoma cell line, A431, were examined for the effect of Nafamostat mesilate. The effects on cell growth were evaluated using the MTT assay. The effects on the relative expression levels of mRNA were measured by quantitative RT-PCR. Cytokine secretion was analyzed by enzyme-linked immunosorbent assay. RESULTS: Nafamostat mesilate inhibited the proliferation of two HNSCC cell lines, YCU-L891 and YCU-H891, and A431. In these cell lines, Nafamostat mesilate down-regulated both matrix metalloproteinase (MMP)-2 and -9. In addition, it reduced the productions of vascular endothelial growth factor (VEGF) and transforming growth factor beta1 (TGF-beta1) by the tumor cells. CONCLUSION: Our results suggest that Nafamostat mesilate has potential for use as a treatment against local growth, invasion and metastasis of squamous cell carcinoma.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Guanidinas/farmacologia , Neoplasias Hipofaríngeas/patologia , Neoplasias Laríngeas/patologia , Fator de Crescimento Transformador beta1/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco , Fator A de Crescimento do Endotélio Vascular/genética , Benzamidinas , Carcinoma de Células Escamosas/genética , Divisão Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hipofaríngeas/genética , Técnicas In Vitro , Neoplasias Laríngeas/genética , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Invasividade Neoplásica/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas/patologia , Neoplasias Vulvares/patologia
18.
Oncol Rep ; 17(2): 289-95, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17203163

RESUMO

Angiogenesis is required for tumor growth and metastasis and, therefore, represents a target for cancer treatment. While many factors have been implicated in promoting angiogenesis, vascular endothelial growth factor (VEGF) plays a key role in tumor angiogenesis. ZD6474 is a potent VEGF receptor-2 (VEGFR-2) tyrosine kinase inhibitor which also has activity against the epidermal growth factor receptor (EGFR) tyrosine kinase. The purpose of this study was to investigate the sensitivity of head and neck squamous cell carcinoma (HNSCC) cell lines to ZD6474, and to evaluate its antitumor efficacy on HNSCC xenografts. This is the first demonstration of antitumor effects of ZD6474 on HNSCC. In vitro ZD6474 displayed antiproliferative effects on HNSCC cells and inhibition of VEGFR-2 and EGFR pathways. In vivo ZD6474 displayed antitumor activity, induced apoptosis and antiangiogenic activity on nude mice bearing an established xenograft of YCU-H891 cells. These results suggest that ZD6474 has the potential to inhibit two key pathways in tumor growth via inhibition of VEGF-dependent tumor angiogenesis and via inhibition of EGFR-dependent tumor cell proliferation.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neovascularização Patológica , Piperidinas/farmacologia , Quinazolinas/farmacologia , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Nus , Modelos Químicos , Transplante de Neoplasias
19.
Nihon Jibiinkoka Gakkai Kaiho ; 108(2): 157-63, 2005 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-15765729

RESUMO

We evaluated the recommend dose and efficacy of chemotherapy (CTx) and concurrent chemoradiotherapy (ConcCRTx) with docetaxel (DOC), cisplatin (CDDP) and 5-FU (TPF) in patients with locally advanced squamous cell carcinoma of the head and neck (SCCHN). Patients underwent 2 cycles of chemotherapy with TPF. In ConcCRTx, radiation was targeted to begin on Day 1. We compared the efficacy of ConcCRTx and induction chemotherapy followed by radiation (CTx followed by RTx) with TPF. In CTx followed by RTx, radiation was targeted to begin 21 days after the end of CTx. The recommend dose of CTx with TPF was DOC 60 (Day 1), CDDP 70 (Day 4) and 5 FU 750 (Day 1-5) mg/ m2/ day and overall response rate of CTx with TPF was 95%. The recommended dose of ConcCRTx was DOC 50 (Day 1), CDDP 60 (Day 4) and 5-FU 600 (Day 1-5) mg/ m2/ day. Overall response rate of ConcCRTx and CTx followed by RTx with TPF were both 100%, and CR rate of them were 87% and 84% (p > 0.05). One-year survival rate of them were 69% and 95% (p < 0.05). More patients had distant metastasis in CTx followed by RTx than in ConcCRTx. Toxicity, such as mucositis, leukocytopenia and neutropenia, was higher in ConcCRTx than in CTx followed by RTx.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/terapia , Cisplatino/administração & dosagem , Terapia Combinada , Docetaxel , Fluoruracila/administração & dosagem , Humanos , Taxoides/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA