Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Gan To Kagaku Ryoho ; 50(13): 1892-1894, 2023 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-38303243

RESUMO

A 25-year-old male received palliative total gastrectomy plus D1 dissection plus Roux-en-Y reconstruction for hemorrhagic gastric cancer with left Virchow lymph node metastasis in 2013. The final diagnosis was Type 2, pT4a(se), pap>tub2 >hepatoid adenocarcinoma, pN3b, sM1, fStage Ⅳ. Because AFP was as high as 11,000 ng/mL, he was diagnosed with AFP-producing gastric cancer and started S-1 plus CDDP therapy. Left adrenal gland metastasis and #106pre, #16b1int lymph node metastasis were observed after 9 courses, and the therapy was changed to irinotecan plus CDDP therapy. After 17 courses, the patient was diagnosed with CR, and the drug was discontinued. Recurrence of the left adrenal gland and an increase in AFP were confirmed by CT after 8 months of suspension, and the drug was resumed. After 8 courses of resumption, PET-CT showed mediastinal and #16b1lat lymph node metastasis and changed to weekly PTX plus Ram therapy. After 2 courses, enlargement of lymph nodes and elevation of AFP was observed, and CapeOX therapy was changed. Diagnosis of left adrenal metastasis recurrence by PET-CT after 21 courses. Nivolumab was used, and radiotherapy(total 39 Gy)was performed locally. After the continuation of nivolumab for 3 years, no findings of recurrent metastasis were observed on imaging, and it was judged as CR, and nivolumab was terminated. As of June 2023, he is alive without recurrence. AFP-producing gastric cancer in the young is rare, and no cases with Virchow metastasis or para-aortic lymph node metastasis have been reported. We report a case of long-term survival in which CR was obtained with combined modality therapy.


Assuntos
Neoplasias Gástricas , Masculino , Humanos , Adulto , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , alfa-Fetoproteínas , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Nivolumabe/uso terapêutico , Metástase Linfática/patologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfonodos/patologia , Terapia Combinada , Gastrectomia
2.
Ann Surg Oncol ; 29(9): 5885-5891, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35763232

RESUMO

BACKGROUND: Prophylactic splenectomy for hilar lymph node (#10) dissection has shown no survival benefit for patients with proximal advanced gastric cancer that does not invade the greater curvature. However, the survival benefit of prophylactic splenectomy for proximal advanced gastric cancer invading the greater curvature side, particularly for clinically negative #10 lymph node metastasis (#10[-]) cases remains controversial. METHODS: This multi-institutional retrospective study enrolled 146 consecutive patients with proximal advanced gastric cancers invading the greater curvature side with clinical #10(-) who underwent R0 total gastrectomy. For 33 of these patients, splenectomy was performed, and the remaining 113 underwent spleen-preservation gastrectomy. Short- and long-term results were compared between the splenectomy and spleen-preservation groups, with the incidence of #10 metastasis in the splenectomy group and recurrence in the spleen-preservation group compared. RESULTS: In the splenectomy group, longer operative time, greater blood loss, more frequent postoperative abdominal infection, and longer hospital stay were observed than in the spleen-preservation group. The two groups exhibited no differences in median relapse-free survival time (31.1 vs 59.8 months; P = 0.684) or median overall survival time (64.9 vs 65.1 months; P = 0.765). The pathologic #10 lymph node metastasis rate was 3% in the splenectomy group, and the #10 lymph node recurrence rate was 2.7% in the spleen-preservation group. CONCLUSIONS: Prophylactic splenectomy showed more frequent postoperative morbidities and a longer hospital stay than spleen preservation, without any long-term survival benefits.


Assuntos
Neoplasias Gástricas , Estudos de Coortes , Gastrectomia , Humanos , Excisão de Linfonodo , Linfonodos/patologia , Metástase Linfática/patologia , Recidiva Local de Neoplasia/patologia , Complicações Pós-Operatórias/patologia , Estudos Retrospectivos , Esplenectomia , Neoplasias Gástricas/patologia
3.
Gan To Kagaku Ryoho ; 49(2): 208-210, 2022 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-35249063

RESUMO

A 75-year-old man was showed wall thickening just below esophagogastric junction(EGJ)by gastroscopy(GS). Biopsy indicated mucinous carcinoma. He was referred to our hospital. Computed tomography(CT), PET-CT showed EGJ cancer and splenic tumor. EGJ cancer was diagnosed GE, Siewert Type Ⅱ, GrePostAnt, Type 1, cT2, cN0, cM0, cStage Ⅰ. The patient underwent total gastrectomy, lower esophagectomy, D2+ #19, 20, 110, 111, 112 lymph nodes dissection, Rou-en- Y reconstruction, distal pancreatectomy, splenectomy, cholecystectomy, and enterostomy. Postoperative complication was pancreatic fistula(Grade Ⅱ). Pathological diagnosis was esophagogastric junction cancer, neuroendocrine carcinoma(NEC), GE, Siewert Type Ⅱ, GrePostAnt, Type 1, pT2(MP), pN1, pM0, pStage ⅡA. Splenic tumor was diagnosed splenic malignant lymphoma, large B-cell, diffuse(DLBCL), NOS, low-immediate risk. Patient was discharged 15 days after the operation and underwent adjuvant chemotherapy with S-1. In this case, he started taking S-1 because the prognosis of NEC is poorer than PSML. There was no evidence of recurrence after 5 months from gastrectomy. As a result of searching for"neuroendocrine tumor"and"malignant lymphoma"in the JAMAS, there was no report of NEC associated with malignant lymphoma. We experienced the rare case of primary splenic malignant lymphoma associated with EGJ NEC. In the case of gastric cancer with splenic tumor, malignant lymphoma of spleen should be concerned.


Assuntos
Adenocarcinoma , Carcinoma Neuroendócrino , Neoplasias Gástricas , Adenocarcinoma/cirurgia , Idoso , Carcinoma Neuroendócrino/tratamento farmacológico , Carcinoma Neuroendócrino/cirurgia , Junção Esofagogástrica/patologia , Junção Esofagogástrica/cirurgia , Gastrectomia , Humanos , Masculino , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia
4.
Anticancer Res ; 38(10): 6015-6021, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30275234

RESUMO

BACKGROUND/AIM: The aim of this phase I/II study was to determine the safety, and efficacy of combination of neoadjuvant chemotherapy (NAC) with biweekly docetaxel, cisplatin, and S-1 (DCS) in stage III gastric cancer patients. PATIENTS AND METHODS: In the phase I study, S1 was administered at doses of 80 mg/day to 120 mg/day depending on the body surface area and docetaxel was administered at 20 mg/m2, whereas cisplatin was initially administered at 25 mg/m2 and was escalated by 5 mg/m2 up to 50 mg/m2 In the phase II study, safety and therapeutic efficacy of DCS were evaluated using the recommended dose of cisplatin. RESULTS: In phase I, 21 patients were enrolled. In level II, perforation of gastric cancer occurred in one case although no dose limiting toxicities (DLTs) were noted in level III-VI. Recommended dose for cisplatin was 50 mg/m2/day. In phase II, among 47 patients, 14 experienced grade 3/4 adverse events. Clinically, response rate was 66.7% and disease control rate was 97.9%. The curative (R0) resection rate was 95.7%. Pathological response rate was 53.3%. Three-year overall survival and relapse-free survival rates were 78.5% and 65.3%, respectively. CONCLUSION: Biweekly DCS as NAC was efficient, safe, and acceptable; however, long-term survival should be evaluated to confirm the efficacy of biweekly DCS for stage III gastric cancer patients.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Adenocarcinoma/secundário , Idoso , Cisplatino/administração & dosagem , Docetaxel , Combinação de Medicamentos , Feminino , Seguimentos , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Ácido Oxônico/administração & dosagem , Prognóstico , Neoplasias Gástricas/patologia , Taxa de Sobrevida , Taxoides/administração & dosagem , Tegafur/administração & dosagem
5.
Cancer Chemother Pharmacol ; 80(5): 939-943, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28913549

RESUMO

PURPOSE: S-1 plus cisplatin therapy is the recommended standard first-line regimen for human epidermal growth factor receptor 2 (HER-2)-negative advanced unresectable or recurrent gastric cancer (AGC) in the Japanese Gastric Cancer Treatment Guidelines. By contrast, capecitabine plus cisplatin (XP) therapy has been second-line therapy for these patients. This prospective study aimed to evaluate the efficacy and safety of XP as a first-line regimen for HER2-negative patients with AGC. METHODS: In this multicenter, open-label, phase II study, patients received cisplatin (80 mg/m2 i.v. day 1) plus capecitabine (1000 mg/m2 orally, twice daily, days 1-14) at 3 week intervals until disease progression or non-continuation for various reasons. The primary endpoint was overall response rate; secondary endpoints included progression-free survival (PFS), overall survival (OS), and toxicity profiles. RESULTS: Thirty-six patients with HER2-negative AGC were enrolled in this study. Of these, 16 patients with evaluable lesions were assessable for efficacy and 36 were assessable for toxicity. One patient achieved a complete response and five partial responses. The overall response rate was 37.5% [95% confidence interval (CI) 13.7-61.2%] calculated on an intention-to-treat basis. The median PFS and median OS were 5.2 months (95% CI 4.2-6.2 months) and 16.9 months (95% CI 5.8-27.9 months), respectively. Treatment-related adverse events were generally mild; the most common grade 3/4 adverse event was neutropenia (27.8%), followed by anorexia (19.4%), leucopenia (16.7%), anemia (16.7%), and nausea (13.9%). CONCLUSION: XP as first-line therapy is effective and well tolerated by patients with HER2-negative AGC.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Capecitabina/uso terapêutico , Cisplatino/uso terapêutico , Adulto , Idoso , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/farmacologia , Capecitabina/administração & dosagem , Capecitabina/farmacologia , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia
6.
Anticancer Res ; 36(4): 1773-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27069158

RESUMO

BACKGROUND: Neoadjuvant chemotherapy (NAC) has been widely adopted for patients with advanced gastric cancer; however, the safety of gastrectomy with D2 lymphadenectomy followed by NAC has not yet been evaluated. We retrospectively analyzed the influence of NAC on morbidity and mortality after gastrectomy in patients with advanced gastric cancer. PATIENTS AND METHODS: A series of 364 patients with advanced gastric cancer who underwent gastrectomy without pancreatectomy between January 2008 and December 2010 at eight hospitals registered to the Yokohama Clinical Oncology Group were studied retrospectively. There were 330 patients who underwent surgical treatment immediately after diagnosis (surgery alone group) and 34 patients (NAC group) who first received NAC and then underwent surgical resection. RESULTS: Although there were no significant differences in the morbidity rate between the two groups, postoperative pancreatic fistula was more often observed in NAC patients than in patients of the group treated with surgery alone [5 cases (14.7%) vs. 11 cases (3.3%); p=0.011]. In the univariate analysis, NAC (p=0.029), bursectomy (p<0.001) and operative bleeding (≥300 ml, p=0.002), were significantly correlated with postoperative pancreatic fistula, and NAC [odds ratio (OR)=4.901, 95% confidence interval (CI)=1.455-16.67; p=0.010] and bursectomy (OR=11.2, 95% CI=3.460-37.04; p<0.001) were independent risk factors for postoperative pancreatic fistula by multivariate analysis. The incidence of postoperative pancreatic fistula was 40.0% among patients who underwent gastrectomy with bursectomy followed by NAC. CONCLUSION: The incidence of pancreatic fistula in patients treated with NAC and bursectomy was significantly higher than that in other patients. Bursectomy may be discouraged for the prevention of pancreatic fistula from gastrectomy following NAC.


Assuntos
Quimioterapia Adjuvante/efeitos adversos , Gastrectomia/efeitos adversos , Terapia Neoadjuvante/efeitos adversos , Fístula Pancreática/tratamento farmacológico , Fístula Pancreática/etiologia , Neoplasias Gástricas/cirurgia , Idoso , Quimioterapia Adjuvante/métodos , Feminino , Gastrectomia/métodos , Humanos , Incidência , Excisão de Linfonodo/efeitos adversos , Excisão de Linfonodo/métodos , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante/métodos , Fístula Pancreática/cirurgia , Complicações Pós-Operatórias/tratamento farmacológico , Estudos Retrospectivos , Estômago/cirurgia , Resultado do Tratamento
7.
Anticancer Res ; 35(10): 5201-10, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26408678

RESUMO

BACKGROUND: Long-term outcomes of patients with peritoneal dissemination of gastric cancer remain unsatisfactory despite advances in treatment modalities. Internal luminescence conditionally replicative adenovirus (CRAd) presents a novel approach for cancer treatment and imaging. MATERIALS AND METHODS: 3CL is a modified cyclooxygenase-2 (COX2) promoter-driven CRAd which contains the luciferase expression gene for bioluminescence imaging. The visualizing and therapeutic effect of 3CL was evaluated in a mouse model of peritoneal dissemination. RESULTS: Intraperitoneal injection of 3CL achieved the shrinkage and reduction of lesions of peritoneal dissemination. Six model mice treated with 3CL had a significantly longer mean survival time than 6 mock-treated mice (85.7 versus 34.3 days, p=0.0005). By whole-body bioluminescent imaging, the sensitivity and specificity of peritoneal dissemination detection through macroscopic inspection were 58.1% and 83.2%, respectively, whereas 3CL viral imaging modality yielded corresponding values of 78.8% and 99.3%. Peritoneal lesions detected by imaging histologically contained cancer cells and necrotic tissue, which originated from viral oncolytic effects. CONCLUSION: Cox2 CRAds with 5/3 chimeric-fiber modification, therefore, appear to be a promising imaging and therapeutic tools for peritoneal dissemination of gastric cancer.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dependovirus/fisiologia , Diagnóstico por Imagem/métodos , Luciferases/metabolismo , Neoplasias Peritoneais/terapia , Neoplasias Peritoneais/virologia , Neoplasias Gástricas/terapia , Neoplasias Gástricas/virologia , Animais , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Dependovirus/genética , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Luciferases/genética , Camundongos , Transplante de Neoplasias , Neoplasias Peritoneais/secundário , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sensibilidade e Especificidade , Análise de Sobrevida , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Oncology ; 88(5): 281-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25591954

RESUMO

OBJECTIVE: This retrospective study aimed to address the therapeutic outcome for scirrhous gastric cancer patients by evaluating the effect of neoadjuvant chemotherapy prior to gastrectomy. METHODS: Two cycles of a 3-week regimen of fluoropyrimidine S-1 (40 mg/m(2), orally, twice daily), together with cisplatin (60 mg/m(2), intravenously, day 8), were administered to patients, separated by a 2-week rest period. Surgery was performed 3 weeks later in the neoadjuvant group (n = 27). We retrospectively evaluated overall survival and prognostic factors in these patients. RESULTS: Univariate analysis showed that positive lavage cytology indicated significantly worse prognoses. In the 15 patients who also underwent curative gastrectomies after S-1 plus cisplatin chemotherapy, the pathological response grade was a significant prognostic factor for 5-year survival. Additionally, lymph node metastasis tended to be an adverse prognostic factor. CONCLUSION: After S-1 plus cisplatin neoadjuvant chemotherapy, a grade 2-3 pathological response may predict favorable outcomes in scirrhous gastric cancer patients receiving curative gastrectomy, but further studies are needed to confirm these results.


Assuntos
Adenocarcinoma Esquirroso/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Gastrectomia , Linfonodos/patologia , Terapia Neoadjuvante/métodos , Neoplasias Gástricas/tratamento farmacológico , Adenocarcinoma Esquirroso/patologia , Adenocarcinoma Esquirroso/cirurgia , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Quimioterapia Adjuvante/efeitos adversos , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Esquema de Medicação , Combinação de Medicamentos , Feminino , Humanos , Estimativa de Kaplan-Meier , Metástase Linfática/diagnóstico , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante/efeitos adversos , Invasividade Neoplásica , Estadiamento de Neoplasias , Ácido Oxônico/administração & dosagem , Ácido Oxônico/efeitos adversos , Prognóstico , Estudos Retrospectivos , Tamanho da Amostra , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia , Tegafur/administração & dosagem , Tegafur/efeitos adversos , Resultado do Tratamento
9.
Hepatogastroenterology ; 61(133): 1262-73, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25436294

RESUMO

BACKGROUND/AIMS: Here we investigated postoperative prognostic factors and surveillance in patients with esophageal cancer. METHODOLOGY Prognostic factors were evaluated at several different postoperative stages in 257 patients with curative (R0) esophagectomy. Cause of death and pattern of tumor recurrence were also analyzed. RESULTS: There was a significant difference in the distribution of cause of death according to the time after surgery (p<0.001). The pattern of recurrence also differed according to the time after surgery, although this was not statistically significant. A Cox proportional regression hazard model for disease-specific survival revealed that Tumor-Node-Metastasis (TNM) stage was an independent prognostic factor only from the time of initial surgery until the third postoperative year, and no postoperative prognostic factors were detected after the fourth and fifth years. There were significant differences in disease-specific survival among pathological TNM stages between the time of initial surgery and postoperative year five, but not between stages I and II at postoperative years two, three, or four. There were no significant differences between the stages at postoperative year five. Relapse-free survival differed between stages II and III at postoperative year five, although the other results were similar to those for disease-specific survival. CONCLUSIONS: Prognostic factors for esophageal cancer alter during the postoperative period. Although the pathological stage at the time of initial surgery has less prognostic power after 3 years, it remains important to monitor treatments for esophageal cancer continuously, as well as concomitant diseases and other malignancies.


Assuntos
Neoplasias Esofágicas/cirurgia , Esofagectomia , Adulto , Idoso , Idoso de 80 Anos ou mais , Causas de Morte , Quimioterapia Adjuvante , Distribuição de Qui-Quadrado , Intervalo Livre de Doença , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/secundário , Esofagectomia/efeitos adversos , Esofagectomia/mortalidade , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Cuidados Pós-Operatórios , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
10.
Cancer Sci ; 104(8): 1083-90, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23679574

RESUMO

Pancreatic cancer has a poor prognosis after complete macroscopic resection combined with chemotherapy. Even after neoadjuvant chemotherapy, R0 resection is often not possible. Moreover, current imaging techniques cannot reliably distinguish viable cancer cells from scar tissue at the resectional margin. We investigated the use of a conditionally replicative adenovirus (CRAd), Ad5/3Cox2CRAd-ΔE3ADP-Luc, for imaging the effects of chemotherapy. The CRAd infectivity of pancreatic cancer cells was enhanced by a chimeric Ad5/3 fiber, E1A expression was under the control of the Cox2 promoter, and the luciferase gene was inserted adjacent to the adenovirus death protein (ADP) gene. Subcutaneous xenografts of the pancreatic cancer cell line MiaPaCa-2 were established in 24 BALB/c nu/nu mice. When xenografts reached a diameter of 4-6 mm (day 1), the mice were injected i.p. with either PBS (group A; n = 12) or 1000 mg/kg gemcitabine (group B; n = 12), weekly. On days 19, 26, 33, and 40, CRAd were injected intratumorally into three mice in groups A and B. Bioluminescence was imaged 72 h after CRAd injection, and gross tumor volumes were measured then tumors were removed for ex vivo histopathology using H&E and Ki-67 staining. Correlations between gross tumor volume, pathological evaluation of the percentage of viable tumor area, and CRAd bioluminescence were analyzed. Bioluminescence correlated closely with the percentage of viable tumor area (R = 0.96), but not with gross tumor volume (R = 0.31). Therefore, CRAds might be reliable imaging tools for monitoring chemotherapy in pancreatic cancer, and could improve our ability to distinguish viable tumor cells from scar tissue.


Assuntos
Adenoviridae/fisiologia , Diagnóstico por Imagem/métodos , Vetores Genéticos/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/virologia , Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Proteínas E3 de Adenovirus/genética , Proteínas E3 de Adenovirus/metabolismo , Animais , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Feminino , Genes Reporter , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Anticancer Res ; 33(2): 697-704, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23393370

RESUMO

BACKGROUND/AIM: This study assessed the toxicity and activity of biweekly docetaxel and S-1 combination therapy in elderly patients with advanced gastric cancer. PATIENTS AND METHODS: One-hundred and thirteen patients were enrolled: 35 were 75 years old or more. The objective response rate, toxicity, progression-free survival (PFS), and overall survival (OS) were compared. RESULTS: Dose reduction was significantly frequent in the elderly group (24/35 versus 25/78, p<0.001). The overall response rate was 54.9%. Out of these, 18 (15.9%) underwent gastrectomy (13 R0 gastrectomy). The median OS was 17.3 months and the median PFS was 8.0 months. Neutropenia was the most frequently observed hematological toxicity at grade 3 and 4 (34.5%), followed by leukopenia (24.8%). Most non-hematological toxicities were of grade 1 or 2. There were no significant differences in overall response rate, median OS, median PFS, or toxicities between the two groups. CONCLUSION: This combination offers favourable survival benefits with controllable tolerance for therapy of AGC in the elderly.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Adenocarcinoma/mortalidade , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Intervalo Livre de Doença , Docetaxel , Combinação de Medicamentos , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Ácido Oxônico/administração & dosagem , Ácido Oxônico/efeitos adversos , Modelos de Riscos Proporcionais , Neoplasias Gástricas/mortalidade , Taxoides/administração & dosagem , Taxoides/efeitos adversos , Tegafur/administração & dosagem , Tegafur/efeitos adversos
12.
J Chemother ; 24(6): 364-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23174102

RESUMO

To establish a safe, long-term regimen of docetaxel (DOC) and cisplatin (CDDP) in an outpatient setting for gastric cancer refractory to S-1 adjuvant chemotherapy, a dose-escalating phase I study was conducted. Cohorts of patients were treated with escalating doses of DOC (starting at 20 mg/m² per week with 5 mg/m² increments) and a fixed dose of CDDP (25 mg/m²). Drugs were administered on days 1, 8, and 15. A cycle of this treatment was 28 days. In total, 52 courses were performed, and the mean number of courses was 5.3. Two of the four patients at dose level 3 showed dose-limiting toxicities (grade 4 neutropenia, and grade 3 anorexia and dehydration). The recommended dose (RD) of DOC was therefore defined as 25 mg/m². There is a need for a phase II clinical trial using this regimen in patients with S-1-refractory stage II/III gastric cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cisplatino/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Taxoides/uso terapêutico , Idoso , Anorexia/induzido quimicamente , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Quimioterapia Adjuvante , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Estudos de Coortes , Desidratação/induzido quimicamente , Docetaxel , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neutropenia/induzido quimicamente , Ácido Oxônico/uso terapêutico , Neoplasias Gástricas/patologia , Taxoides/administração & dosagem , Taxoides/efeitos adversos , Tegafur/uso terapêutico
13.
Dig Surg ; 29(3): 261-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22907557

RESUMO

BACKGROUND/AIMS: Single-port and reduced-port laparoscopic surgeries are performed as a less invasive form of surgery than conventional laparoscopy. In this study, short-term patient outcomes were compared between reduced-port laparoscopic distal gastrectomy (RPLDG) and conventional laparoscopy-assisted distal gastrectomy (LADG) to evaluate the feasibility of RPLDG for gastric cancer. METHODS: Between August 2010 and July 2011, 38 patients underwent LADGs that were performed by a single surgeon. Of these, 20 patients underwent RPLDG, and 18 patients underwent conventional LADG. Short-term outcomes were compared between the two groups. RESULTS: Surgical procedures, total operation time (278.8 versus 228.7 min, p = 0.0002) and time for lymph node dissection (181.3 versus 136.3 min, p = 0.0001) were significantly longer in the RPLDG group compared with the LADG group, while the volume of blood loss during reconstruction was reduced (17.5 versus 49.6 ml, p = 0.0019). Cosmetic satisfaction in the RPLDG group showed significant superiority over that in the conventional LADG group (p = 0.0252). CONCLUSION: RPLDG was shown to be an acceptable and satisfactory procedure for the treatment of gastric cancer. To confirm the feasibility of this surgical procedure, it is necessary to conduct a well-designed randomized controlled study comparing RPLDG and conventional LADG in many patients.


Assuntos
Adenocarcinoma/cirurgia , Gastrectomia/métodos , Laparoscopia/métodos , Excisão de Linfonodo/métodos , Neoplasias Gástricas/cirurgia , Adulto , Idoso , Idoso de 80 Anos ou mais , Perda Sanguínea Cirúrgica , Distribuição de Qui-Quadrado , Estética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Duração da Cirurgia , Projetos Piloto
14.
Oncology ; 83(4): 183-91, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22890015

RESUMO

OBJECTIVES: This study was conducted to determine the prognostic value of the Glasgow Prognostic Score (GPS), an inflammation-based prognostic score composed of C-reactive protein and albumin, for patients with advanced cancer. METHODS: A total of 83 advanced gastric cancer patients receiving biweekly docetaxel/S-1 treatment (DS) were included in the study. To identify the value of GPS as prognostic factor for disease-specific survival (DSS) and progression-free survival (PFS), univariate and multivariate analyses were performed. RESULTS: Unresectable tumors were observed in 78 patients and recurrent tumors were detected in 5 patients. Of these, 12 patients underwent gastrectomy. There were significant correlations between the GPS and the neutrophil/lymphocyte ratio. Univariate analysis revealed that the GPS, Eastern Cooperative Oncology Group performance status and gastrectomy after DS treatment significantly affected prognosis. Multivariate analysis showed that the GPS, age and gastrectomy independently influenced DSS, and that the GPS and gastrectomy also influenced PFS. Multivariate analysis restricted to patients without gastrectomy showed that the GPS and age independently affected DSS, and that the GPS influenced PFS. CONCLUSION: In the low GPS group, it may be possible to obtain favorable outcomes by chemotherapy in advanced gastric cancer patients. However, a well-designed prospective trial in a large patient cohort is required to corroborate the prognostic value of the GPS.


Assuntos
Albuminas/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína C-Reativa/metabolismo , Inflamação/diagnóstico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Ensaios Clínicos Fase II como Assunto , Docetaxel , Combinação de Medicamentos , Feminino , Seguimentos , Humanos , Inflamação/metabolismo , Inflamação/mortalidade , Masculino , Pessoa de Meia-Idade , Ácido Oxônico/administração & dosagem , Prognóstico , Estudos Retrospectivos , Neoplasias Gástricas/imunologia , Taxa de Sobrevida , Taxoides/administração & dosagem , Tegafur/administração & dosagem
15.
Surg Endosc ; 26(3): 804-10, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22002202

RESUMO

BACKGROUND: The aim of this study was to clarify the technical feasibility and oncological efficacy of laparoscopy-assisted gastrectomy (LAG) for gastric cancer compared with open gastrectomy (OG). METHODS: Between April 2002 and March 2008, a series of 623 patients with gastric cancer underwent R0 gastrectomy (314 LAG patients and 309 OG patients). Age, gender, lymph node dissection, and pathological stage were matched by propensity scoring, and 212 patients (106 LAG and 106 OG) were selected for analysis after the exclusion of 40 patients who had proximal gastrectomy. Intraoperative factors, postoperative morbidity, long-term quality of life (QOL), and survival were evaluated. Moreover, these outcomes were also compared between the laparoscopy-assisted total gastrectomy (LATG) and the open total gastrectomy (OTG). RESULTS: There was no significant difference in preoperative characteristics between the two patient groups. Regarding intraoperative characteristics, blood loss was significantly lower in the LAG group (143 ml) than in the OG group (288 ml), while operation time was significantly longer in the LAG group (273 min) than the OG group (231 min). The degree of lymph node dissection and number of retrieved lymph nodes did not differ between the two groups. There were no significant differences in postoperative courses or overall and disease-specific survival (89.8% vs. 83.6%, P = 0.0886; 100% vs. 95.2%, P = 0.1073) except time to first flatus and time to use of nonsteroidal anti-inflammatory derivatives between the two groups. Significantly fewer patients felt wound pain in the LAG group 1 year after surgery. Analyses between the LATG and OTG groups showed similar results. CONCLUSIONS: LAG for gastric cancer may be both feasible and safe. However, it will be necessary to conduct a well-designed randomized controlled trial comparing short-term and long-term outcomes between LAG and OG in a larger number of patients.


Assuntos
Gastrectomia/métodos , Laparoscopia/métodos , Neoplasias Gástricas/cirurgia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Estudos de Viabilidade , Feminino , Humanos , Tempo de Internação , Excisão de Linfonodo/métodos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Cuidados Pós-Operatórios/métodos , Cuidados Pré-Operatórios/métodos , Qualidade de Vida , Resultado do Tratamento
16.
J Gastrointest Surg ; 15(11): 1939-51, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21909843

RESUMO

BACKGROUND: This retrospective study evaluated the surgical learning curve and outcomes of thoracolaparoscopic esophagectomy. PATIENTS AND METHODS: The study group comprised a series of 92 patients with preoperatively diagnosed resectable thoracic esophageal cancer. Additionally, the surgical outcomes in 79 esophageal cancer patients receiving open esophagectomies were compared. All patients underwent thoracolaparoscopic esophagectomy in the lateral decubitus position. The short- and long-term outcomes were evaluated, and the surgical learning curve was assessed. RESULTS: The total operation time was 477.8 ± 102.2 min, the thoracoscopic time was 157.9 ± 61.3 min, the total blood loss was 554.4 ± 280.5 ml, and the number of retrieved lymph nodes was 34.3 ± 14.3. Postoperative morbidity was observed in 23 patients. After the surgeon's first 40 cases, the surgical technique and short-term outcomes were stable. The 5-year disease-specific survival was 66.6% and the 5-year overall survival was 64.6% in patients receiving R0 thoracolaparoscopic esophagectomy. Comparison of 5-year disease-specific survival rate according to tumor-node-metastasis stage between patients receiving R0 thoracolaparoscopic esophagectomy and conventional open esophagectomy showed that there were no significant differences in survival in any stage between the two groups. Loco-regional recurrence was observed in 6 patients, distant recurrence in seven, and combined recurrence in nine after R0 thoracolaparoscopic esophagectomy. There was no significant difference in the pattern of recurrence between the two groups. CONCLUSIONS: Thoracolaparoscopic esophagectomy for esophageal cancer was technically feasible and oncologically satisfactory, according to the surgical learning curve.


Assuntos
Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/cirurgia , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/cirurgia , Esofagectomia/métodos , Adulto , Idoso , Perda Sanguínea Cirúrgica/estatística & dados numéricos , Esofagectomia/efeitos adversos , Feminino , Laparoscopia Assistida com a Mão/métodos , Humanos , Estimativa de Kaplan-Meier , Curva de Aprendizado , Tempo de Internação/estatística & dados numéricos , Excisão de Linfonodo , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Recidiva Local de Neoplasia , Estudos Retrospectivos , Toracoscopia/métodos , Toracotomia , Fatores de Tempo , Resultado do Tratamento
17.
Cancer Chemother Pharmacol ; 67(6): 1363-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20803016

RESUMO

PURPOSE: We evaluated the efficacy and toxicity of biweekly S-1 and docetaxel combination therapy in patients with advanced gastric cancer. METHODS: Patients with histologically proven, unresectable advanced or recurrent gastric cancer, a performance status (PS) of 0-2 and no prior chemotherapy history were eligible for inclusion (n = 45). Patients received a total of 215 treatment courses (median, 4; range, 2-12) of S-1 oral administration twice daily for 1 week followed by a drug-free interval of 1 week. Docetaxel (40 mg/m(2)) was administered intravenously on days 1 and 15. RESULTS: We observed 25 partial responses (55.6%) and one complete response (2.2%), resulting in an overall response rate of 57.8%. Twenty-four patients (53.3%) received second-line chemotherapy. Five patients (11.1%) underwent R0 gastrectomy during the course of the study. The median overall survival time was 15.3 months, the median time to progression was 6.9 months, and the median duration of response in 26 patients was 8.0 months. Neutropenia was the most frequently observed (40.4%) haematological toxicity at grades 3 and 4 and leucopenia was the second most common (29.8%). There were no treatment-related deaths. CONCLUSIONS: S-1 plus docetaxel combination therapy in an outpatient setting provided promising activity with acceptable adverse toxicities.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Intervalo Livre de Doença , Docetaxel , Esquema de Medicação , Combinação de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Recidiva Local de Neoplasia , Ácido Oxônico/administração & dosagem , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia , Taxa de Sobrevida , Taxoides/administração & dosagem , Taxoides/farmacocinética , Tegafur/administração & dosagem
18.
Dig Surg ; 27(6): 502-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21079403

RESUMO

BACKGROUND/AIMS: The incidence of proximal gastric cancer is increasing, so proximal gastrectomies are often performed to preserve gastric function, but the optimal reconstruction method after surgery remains controversial. We therefore conducted a prospective pilot study comparing reconstructions using jejunal pouch interposition or jejunal interposition. METHODS: Thirty-eight patients with early proximal gastric cancer were included in this study. Equal numbers of patients were randomly assigned for reconstruction using jejunal interposition (the IP group) or jejunal pouch interposition (the PO group). Postoperative morbidity and patient symptoms were compared between the 2 groups. RESULTS: Postoperative morbidity was significantly more frequent in the IP than the PO group (p = 0.036). Moreover, the incidence of gastrointestinal complaints was more frequent in the IP group until 6 months after surgery. By contrast, the caloric intake was more favorable in the PO group until 1 year post-surgery. CONCLUSION: Short-term and mid-term outcomes were more favorable following jejunal pouch interposition compared with jejunal interposition after proximal gastrectomy.


Assuntos
Gastrectomia/métodos , Gastroplastia/métodos , Jejuno/cirurgia , Idoso , Ingestão de Energia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Complicações Pós-Operatórias , Estudos Prospectivos , Neoplasias Gástricas/cirurgia , Resultado do Tratamento
19.
Anticancer Res ; 30(7): 3063-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20683056

RESUMO

BACKGROUND: The prognosis for patients with N0 oesophageal cancer is favourable, but relevant prognostic factors and appropriate surveillance protocols have not been identified for these patients. PATIENTS AND METHODS: A total of 210 oesophageal cancer patients were included in this study. Of these, 92 (43.8%) had no lymph node metastasis. Predictive factors for lymph node metastasis were evaluated in N0 oesophageal cancer. Survival, prognostic factors, causes of death and pattern of recurrence were assessed between patients with and without lymph node metastasis. RESULTS: Logistic regression analysis revealed that depth of tumour invasion (T1) was an independent predictive factor for N0. The Cox proportional hazard regression model showed that venous invasion was an independent prognostic factor for disease-specific survival in N0 oesophageal cancer patients (hazard ratio=3.977, p=0.042). Locoregional recurrence was less frequent in patients with N0 oesophageal cancer (p=0.0319). CONCLUSION: Meticulous and long-term follow-up is necessary even for patients with N0 oesophageal cancer, particularly for those with adverse prognostic factors.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/cirurgia , Neoplasias Esofágicas/cirurgia , Feminino , Humanos , Modelos Logísticos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Prognóstico , Modelos de Riscos Proporcionais , Taxa de Sobrevida
20.
J Surg Oncol ; 102(2): 141-7, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20648584

RESUMO

BACKGROUND AND OBJECTIVES: This study compared surgical outcomes between patients undergoing laparoscopy-assisted distal gastrectomy (LADG) and those undergoing open distal gastrectomy (ODG) from the viewpoint of obesity. METHODS: Between June 2002 and May 2008, 146 patients with preoperatively diagnosed early gastric cancer who underwent LADG (n = 90) or ODG (n = 56) were enrolled in this study and compared in terms of clinicopathological findings and operative outcome. The visceral fat area (VFA) and subcutaneous fat area (SFA) were assessed as identifiers of obesity using FatScan software. The relationship between obesity and operative outcomes after LADG and ODG was evaluated. RESULTS: There were no significant correlations between intraoperative blood loss (IBL) and any obesity-related factors, or between operation time (OT) and any obesity-related factors in the LADG group. There was a significant correlation between IBL and BMI (r = 0.486, P = 0.0001), IBL and VFA (r = 0.456, P = 0.0003), IBL and SFA (r = 0.311, P = 0.0193), OT and BMI (r = 0.406, P = 0.0017), OT and VFA (r = 0.314, P = 0.0178), and between OT and SFA (r = 0.382, P = 0.0034) in the ODG group. CONCLUSIONS: LADG may be a useful operative manipulation that is not influenced by obesity, whereas ODG may be influenced by obesity even after reaching the surgical plateau.


Assuntos
Gastrectomia/métodos , Laparoscopia , Obesidade/complicações , Neoplasias Gástricas/cirurgia , Adenocarcinoma/cirurgia , Idoso , Perda Sanguínea Cirúrgica , Índice de Massa Corporal , Feminino , Humanos , Masculino , Gordura Subcutânea Abdominal , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA