Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cureus ; 16(8): e67626, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39310449

RESUMEN

Patients with esophageal cancer who have severe complications such as diabetes sometimes require two-stage surgery. Herein, we describe two cases of gastric emphysema that were treated at our facility after the patients had previously undergone minimally invasive esophagectomy as the first-stage surgical treatment of esophageal cancer. Case 1: A 72-year-old man with a history of diabetes mellitus (DM) was diagnosed with esophageal cancer and an esophageal obstruction and subsequently underwent percutaneous endoscopic gastrostomy (PEG) placement followed by neoadjuvant chemoradiotherapy. The treatment efficacy was good; once the tumor was deemed resectable, the patient underwent robot-assisted minimally invasive esophagectomy and cervical esophagostomy placement as the first stage of surgical treatment. The patient had a good postoperative course and was discharged on postoperative day (POD) 10. However, on POD 16, he returned to the hospital with abdominal distension. Computed tomography (CT) revealed gastric emphysema and hepatic portal vein gas. Conservative treatment was initiated as there were no signs of peritoneal irritation. An upper gastrointestinal (GI) series revealed delayed gastric emptying (DGE); therefore, replacement of the PEG with a percutaneous endoscopic gastrojejunostomy (PEG-J) was necessary. On POD 42, the patient underwent reconstructive surgery as the second-stage surgical treatment of esophageal cancer. Case 2: A 74-year-old man had a history of DM, chronic renal failure, and PEG placement for dysphagia caused by left recurrent nerve palsy after thoracic aortic aneurysm surgery. The patient underwent a thoracoscopic esophagectomy with cervical esophagostomy placement as the first-stage surgical treatment of esophageal cancer. On POD 6, the patient developed abdominal distension, his CT showed gastric emphysema. An upper GI series was performed, which showed DGE. After conservative treatment and improvement in his general condition, the patient underwent a jejunostomy placement on POD 30. Both patients developed gastric emphysema related to PEG placement after undergoing esophagectomy as the first-stage surgical treatment of esophageal cancer. Additionally, both patients had a history of DM. Gastric emphysema, which is thought to be caused by increased intragastric pressure due to postoperative DGE, developed within 30 days of undergoing minimally invasive esophagectomy in both patients. Therefore, the rate of nutrient administration and symptoms should be carefully monitored during the postoperative management of patients with these characteristics.

2.
Am J Case Rep ; 25: e944094, 2024 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-39342423

RESUMEN

BACKGROUND Gas in the portal venous system, or hepatic portal venous gas, is a rare occurrence associated with ischemic colitis, inflammatory bowel disease, or any cause of bowel perforation, including from a necrotic tumor. This report presents the case of a 72-year-old man with diabetes who had carcinoma of the ileocecal region, sepsis due to Klebsiella pneumoniae, and hepatic portal venous gas. CASE REPORT A 72-year-old man with ileocecal cancer was admitted to our hospital for preoperative diabetes control. He developed a fever and septic shock, without abdominal symptoms or signs of peritoneal irritation. Klebsiella pneumoniae was detected in blood cultures. Abdominal ultrasonography showed hepatic portal venous gas, and a simple computed tomography scan revealed gas in the vasculature and hepatic portal vein in the lateral segment, which led us to believe that the ileocecal mass was the source of infection, and emergency surgery was performed. The patient was discharged from the hospital on postoperative day 34 with good progress despite dehydration due to high-output syndrome. CONCLUSIONS Sepsis due to necrosis of ileocecal cancer is often difficult to diagnose because it is not accompanied by abdominal symptoms, as in our case. However, abdominal ultrasound is useful because it allows for a broad evaluation. This report has demonstrated and highlighted that the findings of hepatic portal venous gas on imaging should be regarded seriously, requiring urgent investigation to identify the cause and commence treatment in cases of infection or sepsis.


Asunto(s)
Infecciones por Klebsiella , Klebsiella pneumoniae , Vena Porta , Sepsis , Humanos , Masculino , Anciano , Vena Porta/diagnóstico por imagen , Sepsis/microbiología , Sepsis/complicaciones , Infecciones por Klebsiella/complicaciones , Infecciones por Klebsiella/diagnóstico , Neoplasias del Íleon/complicaciones , Embolia Aérea/diagnóstico por imagen , Embolia Aérea/etiología , Tomografía Computarizada por Rayos X
3.
Ann Gastroenterol Surg ; 8(3): 374-382, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38707222

RESUMEN

Background: Double-flap technique (DFT) is a reconstruction procedure after proximal gastrectomy (PG). We previously reported a multi-center, retrospective study in which the incidence of reflux esophagitis (RE) (Los Angeles Classification ≥Grade B [LA-B]) 1 year after surgery was 6.0%. There have been many reports, but all of them were retrospective. Thus, a multi-center, prospective study was conducted. Methods: Laparoscopic PG + DFT was performed for cT1N0 upper gastric cancer patients. The primary endpoint was the incidence of RE (≥LA-B) 1 year after surgery. The planned sample size was 40, based on an estimated incidence of 6.0% and an upper threshold of 20%. Results: Forty patients were recruited, and 39, excluding one with conversion to total gastrectomy, received protocol treatment. Anastomotic leakage (Clavien-Dindo ≥Grade III) was observed in one patient (2.6%). In 38 patients, excluding one case of postoperative mortality, RE (≥LA-B) was observed in two patients (5.3%) 1 year after surgery, and the upper limit of the 95% confidence interval was 17.3%, lower than the 20% threshold. Anastomotic stricture requiring dilatation was observed in two patients (5.3%). One year after surgery, body weight change was 88.9 ± 7.0%, and PNI <40 and CONUT ≥5, indicating malnutrition, were observed in only one patient (2.6%) each. In the quality of life survey using the PGSAS-45 questionnaire, the esophageal reflux subscale score was 1.4 ± 0.6, significantly better than the public data (2.0 ± 1.0; p = 0.001). Conclusion: Laparoscopic DFT showed anti-reflux efficacy. Taken together with the acceptable incidence of anastomotic stricture, DFT can be an option for reconstruction procedure after PG.

4.
J Gastrointest Cancer ; 55(3): 1125-1133, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38703333

RESUMEN

PURPOSE: Post-operative infectious complication (IC) is a well-known negative prognostic factor, while showing neoadjuvant chemotherapy (NAC) may cancel out the negative influence of IC. This analysis compared the clinical impacts of IC according to the presence or absence of NAC in gastric cancer patients enrolled in the phase III clinical trial (JCOG0501) which compared upfront surgery (arm A) and NAC followed by surgery (arm B) in type 4 and large type 3 gastric cancer. METHODS: The subjects were 224 patients who underwent R0 resection out of 316 patients enrolled in JCOG0501. The prognoses of the patients with or without ICs in each arm were investigated by univariable and multivariable Cox regression analyses. RESULTS: There were 21 (20.0%) IC occurrences in arm A and 15 (12.6%) in arm B. In arm A, the overall survival (OS) of patients with ICs was slightly worse than those without IC (3-year OS, 57.1% in patients with ICs, 79.8% in those without ICs; adjusted hazard ratio (95% confidence interval), 1.292 (0.655-2.546)). In arm B, patients with ICs showed a trend of better survival than those without ICs (3-year OS, 80.0% in patients with IC, 74.0% in those without IC; adjusted hazard ratio, 0.573 (0.226-1.456)). CONCLUSION: This study could not indicate the negative prognostic influence of ICs in gastric cancer patients receiving NAC, which might be canceled by NAC. To build exact evidence, further investigation with prospective and large numbers of data might be expected.


Asunto(s)
Terapia Neoadyuvante , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/cirugía , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/patología , Masculino , Femenino , Terapia Neoadyuvante/métodos , Terapia Neoadyuvante/efectos adversos , Persona de Mediana Edad , Pronóstico , Anciano , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Gastrectomía/efectos adversos , Quimioterapia Adyuvante/métodos , Quimioterapia Adyuvante/efectos adversos , Adulto
5.
Eur J Surg Oncol ; 50(3): 107982, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38290246

RESUMEN

BACKGROUND: Abdominal surgical infectious complications (ASIC) after gastrectomy for gastric cancer impair patients' survival and quality of life. JCOG0912 was conducted to compare laparoscopy-assisted distal gastrectomy with open distal gastrectomy for clinical stage IA or IB gastric cancer. The present study aimed to identify risk factors for ASIC using prospectively collected data. METHODS: We performed a post-hoc analysis of the risk factors for ASIC using the dataset from JCOG0912. All complications were evaluated according to the Clavien-Dindo classification (CD). ASIC was defined as CD grade I or higher anastomotic leakage, pancreatic fistula, abdominal abscess, and wound infection. Analyses were performed using the logistic regression model for univariable and multivariable analyses. RESULTS: A total of 910 patients were included (median age, 63 years; male sex, 61 %). Among them, ASIC occurred in 5.8 % of patients. In the univariable analysis, male sex (odds ratio [OR] 2.855, P = 0.003), diabetes (OR 2.565, P = 0.029), and Roux-en-Y (R-Y) reconstruction (vs. Billroth Ⅰ, OR 2.707, P = 0.002) were significant risk factors for ASIC. In the multivariable analysis, male sex (OR 2.364, P = 0.028) and R-Y reconstruction (vs. Billroth Ⅰ, OR 2.310, P = 0.015) were independent risk factors for ASIC. CONCLUSIONS: Male sex and R-Y reconstruction were risk factors for ASIC after distal gastrectomy. Therefore, when performing surgery on male patients or when R-Y reconstruction is selected after gastrectomy for gastric cancer, surgeons should pay special attention to prevent ASIC.


Asunto(s)
Laparoscopía , Neoplasias Gástricas , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/cirugía , Neoplasias Gástricas/complicaciones , Calidad de Vida , Gastroenterostomía/efectos adversos , Factores de Riesgo , Laparoscopía/efectos adversos , Gastrectomía/efectos adversos , Complicaciones Posoperatorias/etiología , Resultado del Tratamiento
6.
Gastric Cancer ; 27(1): 155-163, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37989806

RESUMEN

BACKGROUND: Postoperative adjuvant chemotherapy with S-1 for 1 year (corresponding to eight courses) is the standard treatment for pathological stage II gastric cancer. The phase III trial (JCOG1104) investigating the non-inferiority of four courses of S-1 to eight courses was terminated due to futility at the first interim analysis. To confirm the primary results, we reported the results after a 5-years follow-up in JCOG1104. METHODS: Patients histologically diagnosed with stage II gastric cancer after radical gastrectomy were randomly assigned to receive S-1 for eight or four courses. In detail, 80 mg/m2/day S-1 was administered for 4 weeks followed by a 2-week rest as a single course. RESULTS: Between February 16, 2012, and March 19, 2017, 590 patients were enrolled and randomly assigned to 8-course (295 patients) and 4-course (295 patients) regimens. After a 5-years follow-up, the relapse-free survival at 3 years was 92.2% for the 8-course arm and 90.1% for the 4-course arm, and that at 5 years was 87.7% for the 8-course arm and 85.6% for the 4-course arm (hazard ratio 1.265, 95% CI 0.846-1.892). The overall survival at 3 years was 94.9% for the 8-course arm, 93.2% for the 4-course arm, and that at 5 years was 89.7% for the 8-course arm, and 88.6% for the 4-course arm (HR 1.121, 95% CI 0.719-1.749). CONCLUSIONS: The survival of the four-course arm was slightly but consistently inferior to that of the eight-course arm. Eight-course S-1 should thus remain the standard adjuvant chemotherapy for pathological stage II gastric cancer.


Asunto(s)
Neoplasias Gástricas , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioterapia Adyuvante , Estudios de Seguimiento , Estadificación de Neoplasias , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/cirugía , Neoplasias Gástricas/patología
7.
Surg Oncol ; 50: 101990, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37717376

RESUMEN

PURPOSE: Although proximal gastrectomy (PG) is commonly used in patients with upper gastric cancer (GC) and esophagogastric junction (EGJ) cancer, long-term prognostic factors in these patients are poorly understood. The double-flap technique (DFT) is an esophagogastrostomy with anti-reflux mechanism after PG; we previously conducted a multicenter retrospective study (rD-FLAP) to evaluate the short-term outcomes of DFT reconstruction. Here, we evaluated the long-term prognostic factors in patients with upper GC and EGJ cancer. METHODS: The study was conducted as a secondary analysis of the rD-FLAP Study, which enrolled patients who underwent PG with DFT reconstruction, irrespective of disease type, between January 1996 and December 2015. RESULTS: A total of 509 GC and EGJ cancer patients were enrolled. Univariate and multivariate analyses of overall survival demonstrated that a preoperative prognostic nutritional index (PNI) < 45 (p < 0.001, hazard ratio [HR]: 3.59, 95% confidential interval [CI]: 1.93-6.67) was an independent poor prognostic factor alongside pathological T factor ([pT] ≥2) (p = 0.010, HR: 2.29, 95% CI: 1.22-4.30) and pathological N factor ([pN] ≥1) (p = 0.001, HR: 3.27, 95% CI: 1.66-6.46). In patients with preoperative PNI ≥45, PNI change (<90%) at 1-year follow-up (p = 0.019, HR: 2.54, 95%CI: 1.16-5.54) was an independent poor prognostic factor, for which operation time (≥300 min) and blood loss (≥200 mL) were independent risk factors. No independent prognostic factors were identified in patients with preoperative PNI <45. CONCLUSIONS: PNI is a prognostic factor in upper GC and EGJ cancer patients. Preoperative nutritional enhancement and postoperative nutritional maintenance are important for prognostic improvement in these patients.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/cirugía , Evaluación Nutricional , Pronóstico , Estudios Retrospectivos , Gastrectomía , Unión Esofagogástrica/cirugía
8.
Dig Surg ; 40(3-4): 114-120, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37459840

RESUMEN

INTRODUCTION: Splenectomy for proximal gastric cancer was found to offer no survival benefit in a randomized trial clarifying the role of splenectomy (JCOG0110 study). Although many studies have explored risk factors for morbidities following total gastrectomy, none have assessed the risk factors for postoperative complications in spleen-preserving total gastrectomy. METHODS: Using data from 505 patients enrolled in a previous randomized trial, risk factors for postoperative complications were identified by multivariable logistic regression analysis. Then, the risk factors were assessed separately between splenectomy and spleen-preserving total gastrectomy. RESULTS: Postoperative complications were identified in 119 patients (23.6%) and were more common following splenectomy than following spleen-preserving surgery (30.7% and 16.1%, respectively, p < 0.01). Multivariable analysis revealed that age ≥65 years (p = 0.032), body mass index ≥25 (p = 0.003), and blood loss ≥350 (p = 0.019) were independent risk factors for postoperative complications in the entire cohort. Among them, only body mass index was a significant independent risk factor for complications in both spleen preservation (p = 0.047) and splenectomy groups (p = 0.017). CONCLUSION: Risk factors for postoperative complications were essentially the same between splenectomy and spleen preservation. Being overweight increased the risk of postoperative complications.


Asunto(s)
Esplenectomía , Neoplasias Gástricas , Humanos , Anciano , Esplenectomía/efectos adversos , Bazo/cirugía , Neoplasias Gástricas/cirugía , Gastrectomía/efectos adversos , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/cirugía , Escisión del Ganglio Linfático , Factores de Riesgo , Estudios Retrospectivos
10.
Ann Surg Oncol ; 30(4): 2307-2316, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36692611

RESUMEN

BACKGROUND: Although proximal gastrectomy (PG) with the double-flap technique (DFT) is a function-preserving surgery that prevents esophagogastric reflux, there is a risk of developing metachronous remnant gastric cancer (MRGC). Moreover, details of MRGC and appropriate postoperative follow-up after PG with DFT are unclear. METHODS: We reviewed the medical records of 471 patients who underwent PG with DFT for cancer in a preceding, multicenter, retrospective study (rD-FLAP Study). We investigated the incidence of MRGC, frequency of follow-up endoscopy, and eradication of Helicobacter pylori (H. pylori) infection. RESULTS: MRGC was diagnosed in 42 (8.9%) of the 471 patients, and 56 lesions of MRGC were observed. The cumulative 5- and 10-year incidence rates were 5.7 and 11.4%, respectively. There was no clinicopathological difference at the time of primary PG between patients with and without MRGC. Curative resection for MRGC was performed for 49 (88%) lesions. All patients with a 1-year, follow-up, endoscopy interval were diagnosed with early-stage MRGC, and none of them died due to MRGC. Overall and disease-specific survival rates did not significantly differ between patients with and without MRGC. The incidence rate of MRGC in the eradicated group after PG was 10.8% and that in the uneradicated group was 19.6%, which was significantly higher than that in patients without H. pylori infection at primary PG (7.6%) (p = 0.049). CONCLUSIONS: The incidence rate of MRGC after PG with DFT was 8.9%. Early detection of MRGC with annual endoscopy provides survival benefits. Eradicating H. pylori infection can reduce the incidence of MRGC.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Primarias Secundarias , Neoplasias Gástricas , Humanos , Incidencia , Estudios Retrospectivos , Neoplasias Gástricas/epidemiología , Neoplasias Gástricas/cirugía , Neoplasias Gástricas/patología , Neoplasias Primarias Secundarias/epidemiología , Neoplasias Primarias Secundarias/cirugía , Neoplasias Primarias Secundarias/patología , Gastrectomía/efectos adversos , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/epidemiología , Infecciones por Helicobacter/diagnóstico , Estudios Multicéntricos como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA