Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Ann Surg Oncol ; 31(7): 4250-4260, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38334847

RESUMO

BACKGROUND: The prognosis of limited-stage small cell lung cancer (LS-SCLC) after surgery usually is estimated at diagnosis, but how the prognosis actually evolves over time for patients who survived for a predefined time is unknown. METHODS: Data on patients with a diagnosis of LS-SCLC after surgery between 2004 and 2015 were retrieved from the Surveillance, Epidemiology, and End Results (SEER) database. The 5-year conditional cancer-specific survival (CCSS) and conditional overall survival (COS) were calculated. RESULTS: This study analyzed 997 patients (555 women, 55.7%) with a median age, of 67 years (interquartile range [IQR], 60-73 years). The 5-year CCSS and COS increased from 44.7% and 38.3%, respectively, at diagnosis to 83.7% and 67.9% at 5 years after diagnosis. Although there were large differences with different stages (stages I, II, and III) at diagnosis (respectively 59.5%, 28.4%; 28.1% for CCSS and 50.6%, 24.8%, and 23.6% for COS), the gap decreased with time, and the rates were similar after 5 years (respectively 85.0%, 80.3%, and 79.4% for CCSS; 65.6%, 56.9%, and 61.3% for COS). The 5-year conditional survival for the patients who received lobectomy was better than for those who received sublobectomy or pneumonectomy. Multivariable analyses showed that only age and resection type were independent predictors for CCSS and COS, respectively, throughout the period. CONCLUSION: Conditional survival estimates for LS-SCLC generally increased over time, with the most significant improvement in patients with advanced stage of disease. Resection type and old age represented extremely important determinants of prognosis after a lengthy event-free follow-up period.


Assuntos
Neoplasias Pulmonares , Estadiamento de Neoplasias , Programa de SEER , Carcinoma de Pequenas Células do Pulmão , Humanos , Neoplasias Pulmonares/cirurgia , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Feminino , Carcinoma de Pequenas Células do Pulmão/cirurgia , Carcinoma de Pequenas Células do Pulmão/mortalidade , Carcinoma de Pequenas Células do Pulmão/patologia , Pessoa de Meia-Idade , Masculino , Taxa de Sobrevida , Idoso , Prognóstico , Seguimentos , Pneumonectomia/mortalidade , Estudos de Coortes
2.
Surg Endosc ; 38(2): 640-647, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38012439

RESUMO

BACKGROUND: Lymph node status is an important factor in determining preoperative treatment strategies for stage T1b-T2 esophageal cancer (EC). Thus, the aim of this study was to investigate the risk factors for lymph node metastasis (LNM) in T1b-T2 EC and to establish and validate a risk-scoring model to guide the selection of optimal treatment options. METHODS: Patients who underwent upfront surgery for pT1b-T2 EC between January 2016 and December 2022 were analyzed. On the basis of the independent risk factors determined by multivariate logistic regression analysis, a risk-scoring model for the prediction of LNM was constructed and then validated. The area under the receiver operating characteristic curve (AUC) was used to assess the discriminant ability of the model. RESULTS: The incidence of LNM was 33.5% (214/638) in our cohort, 33.4% (169/506) in the primary cohort and 34.1% (45/132) in the validation cohort. Multivariate analysis confirmed that primary site, tumor grade, tumor size, depth, and lymphovascular invasion were independent risk factors for LNM (all P < 0.05), and patients were grouped based on these factors. A 7-point risk-scoring model based on these variables had good predictive accuracy in both the primary cohort (AUC, 0.749; 95% confidence interval 0.709-0.786) and the validation cohort (AUC, 0.738; 95% confidence interval 0.655-0.811). CONCLUSION: A novel risk-scoring model for lymph node metastasis was established to guide the optimal treatment of patients with T1b-T2 EC.


Assuntos
Neoplasias Esofágicas , Humanos , Metástase Linfática/patologia , Estudos Retrospectivos , Fatores de Risco , Neoplasias Esofágicas/cirurgia , Neoplasias Esofágicas/patologia , Excisão de Linfonodo , Linfonodos/cirurgia , Linfonodos/patologia
3.
Ann Surg Oncol ; 30(5): 2942-2953, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36352297

RESUMO

BACKGROUND: An accurate recurrence risk assessment system and surveillance strategy for hepatoid adenocarcinoma of the stomach (HAS) remain poorly defined. This study aimed to develop a nomogram to predict postoperative recurrence of HAS and guide individually tailored surveillance strategies. METHODS: The study enrolled all patients with primary HAS who had undergone curative-intent resection at 14 institutions from 2004 to 2019. Clinicopathologic variables with statistical significance in the multivariate Cox regression were incorporated into a nomogram to build a recurrence predictive model. RESULTS: The nomogram of recurrence-free survival (RFS) based on independent prognostic factors, including age, preoperative carcinoembryonic antigen, number of examined lymph nodes, perineural invasion, and lymph node ratio, achieved a C-index of 0.723 (95% confidence interval [CI], 0.674-0.772) in the whole cohort, which was significantly higher than those of the eighth American Joint Committed on Cancer (AJCC) staging system (C-index, 0.629; 95% CI, 0.573-0.685; P < 0.001). The nomogram accurately stratified patients into low-, middle-, and high-risk groups of postoperative recurrence. The postoperative recurrence risk rates for patients in the middle- and high-risk groups were respectively 3 and 10 times higher than for the low-risk group. The patients in the middle- and high-risk groups showed more recurrence and metastasis, particularly multiple site metastasis, within 36 months after the operation than those in the low-risk group (low, 2.2%; middle, 8.6%; high, 24.0%; P = 0.003). CONCLUSIONS: The nomogram achieved good prediction of postoperative recurrence for the patients with HAS after radical resection. For the middle- and high-risk patients, more active surveillance and targeted examination methods should be adopted within 36 months after the operation, particularly for liver and multiple metastases.


Assuntos
Adenocarcinoma , Neoplasias Gástricas , Humanos , Nomogramas , Prognóstico , Adenocarcinoma/cirurgia , Adenocarcinoma/patologia , Estadiamento de Neoplasias , Estudos Retrospectivos , Neoplasias Gástricas/patologia , Recidiva Local de Neoplasia/patologia
4.
Ann Surg Oncol ; 29(8): 5022-5033, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35294651

RESUMO

BACKGROUND: The tumor immunosuppressive microenvironment can influence treatment response and outcomes. A previously validated immunosuppression scoring system (ISS) assesses multiple immune checkpoints in gastric cancer (GC) using tissue-based assays. We aimed to develop a radiological signature for non-invasive assessment of ISS and treatment outcomes. METHODS: A total of 642 patients with resectable GC from three centers were divided into four cohorts. Radiomic features were extracted from portal venous-phase CT images of GC. A radiomic signature for predicting ISS (RISS) was constructed using the least absolute shrinkage and selection operator (LASSO) regression method. Moreover, we investigated the value of the RISS in predicting survival and chemotherapy response. RESULTS: The RISS, which consisted of 10 selected features, showed good discrimination of immunosuppressive status in three independent cohorts (area under the curve = 0.840, 0.809, and 0.843, respectively). Multivariate analysis revealed that the RISS was an independent prognostic factor for both disease-free survival (DFS) and overall survival (OS) in all cohorts (all p < 0.05). Further analysis revealed that stage II and III GC patients with low RISS exhibited a favorable response to adjuvant chemotherapy (OS: hazard ratio [HR] 0.407, 95% confidence interval [CI] 0.284-0.584); DFS: HR 0.395, 95% CI 0.275-0.568). Furthermore, the RISS could predict prognosis and select stage II and III GC patients who could benefit from adjuvant chemotherapy independent of microsatellite instability status and Epstein-Barr virus status. CONCLUSION: The new, non-invasive radiomic signature could effectively predict the immunosuppressive status and prognosis of GC. Moreover, the RISS could help identify stage II and III GC patients most likely to benefit from adjuvant chemotherapy and avoid overtreatment.


Assuntos
Infecções por Vírus Epstein-Barr , Neoplasias Gástricas , Infecções por Vírus Epstein-Barr/patologia , Herpesvirus Humano 4 , Humanos , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos , Neoplasias Gástricas/diagnóstico por imagem , Neoplasias Gástricas/tratamento farmacológico , Resultado do Tratamento , Microambiente Tumoral
5.
Br J Cancer ; 123(3): 418-425, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32451469

RESUMO

BACKGROUND: In clinical practice, carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9 are the most common markers measured before and after surgery for gastric cancer (GC). However, which pre- or post-operative combined tumour markers (CEA and CA19-9) have more prognostic value remains unclear. METHODS: Consecutive patients undergoing a resection for GC at the Fujian Medical University Union Hospital were included as a discovery database between January 2011 and December 2014. The prognostic impact of pre- and post-operative tumour markers was evaluated using Kaplan-Meier log-rank survival analysis and multivariable Cox regression analysis. The results were then externally validated. RESULTS: A total of 735 and 400 patients were identified in the discovery cohort and in the validation cohort, respectively. Overall survival rates decreased in a stepwise manner in association with the number of pre- and post-operative positive tumour markers (both P < 0.001). Multivariable analysis revealed that the number of pre-operative positive tumour markers was an independent prognostic factor (P < 0.05). For patients with abnormal pre-operative tumour markers, normalisation of tumour markers after surgery is an independent prognostic protective factor (hazard ratio (HR) = 0.618; 95% confidence interval (CI) = 0.414-0.921), and patients with both positive post-operative tumour markers had double the risk of overall death (HR = 2.338; 95% CI = 1.071-5.101). Similar results were observed in the internal validation and external validation cohorts. CONCLUSION: Pre-operative tumour markers have a better discriminatory ability for post-operative survival in GC patients than post-operative tumour markers, and the normalisation of tumour markers after surgery was associated with better survival.


Assuntos
Antígenos Glicosídicos Associados a Tumores/metabolismo , Biomarcadores Tumorais/metabolismo , Antígeno Carcinoembrionário/metabolismo , Neoplasias Gástricas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Feminino , Gastrectomia , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Período Pós-Operatório , Período Pré-Operatório , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/cirurgia
6.
BMC Cancer ; 20(1): 11, 2020 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-31906893

RESUMO

BACKGROUND: We sought to investigate the prognostic value of complete blood count (CBC)-based biomarkers for patients with resectable gastric cancer (GC). METHODS: Patients with GC who underwent primary surgical resection between December 2008 and December 2013 were included. The estimated area under the curve (AUC) and multivariate Cox regression models were used to identify the best CBC-based biomarker. Time-dependent receiver operating characteristic (t-ROC) curve analysis was used to predict overall survival and compare the prognostic impact. RESULTS: In the 1810 patients analyzed, the median follow-up period was 51.0 months (range 1-101 months). Based on multivariate analysis, the lymphocyte-monocyte ratio (LMR) and hemoglobin (Hb) level were independent prognostic factors (both P < 0.05). Based on the LMR and Hb level, we established the CBC-based inflammatory score (CBCS). A higher CBCS was associated with older age, female sex, higher American Society of Anesthesiologists (ASA) score, proximal tumor location, larger tumor size, later stage and vascular involvement (all P < 0.05). Univariate analyses showed that a higher CBCS was also associated with worse overall survival (OS), which was consistent in each stage (all P < 0.05). Multivariate analysis revealed that the CBCS was a significant independent biomarker (P < 0.05). The AUC for the CBCS (0.627) was significantly higher than the AUCs for the LMR (0.573) and Hb level (0.605) (both P < 0.05). Furthermore, the t-ROC curve of the CBCS was superior to that of the prognostic nutritional index (PNI), systemic immune-inflammation index (SII), modified Glasgow prognostic score (mGPS) and C-reactive protein/albumin ratio (CRP/Alb) throughout the observation period. CONCLUSION: The preoperative LMR and Hb level were optimal CBC-based biomarkers for predicting OS in GC patients after curative resection. Based on the LMR and Hb, we developed a novel and easily obtainable prognostic score called the CBCS, which may improve the prediction of clinical outcomes.


Assuntos
Contagem de Células Sanguíneas , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/cirurgia , Idoso , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/imunologia , Proteína C-Reativa/metabolismo , Feminino , Gastrectomia , Hemoglobinas/metabolismo , Humanos , Inflamação/patologia , Estimativa de Kaplan-Meier , Linfócitos/patologia , Masculino , Pessoa de Meia-Idade , Monócitos/patologia , Análise Multivariada , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Risco , Neoplasias Gástricas/sangue , Neoplasias Gástricas/patologia
7.
Oncologist ; 24(11): e1091-e1101, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30910865

RESUMO

OBJECTIVE: The aim of this study was to investigate the prognostic value of preoperative sarcopenia and systemic inflammation for patients with resectable gastric cancer (GC) and develop a novel and powerful prognostic score based on these factors. MATERIALS AND METHODS: Patients with GC who underwent radical gastrectomy between December 2009 and December 2013 were included. A multivariate Cox regression analysis was performed to identify the prognostic factors. A novel prognostic score (SLMR) was developed based on preoperative sarcopenia and the lymphocyte-monocyte ratio (LMR), and its prognostic value was evaluated. RESULTS: In total, 1,167 patients with resectable GC were included in the study. On multivariate analysis, preoperative sarcopenia and the LMR were shown to be independent prognostic factors (both p < .001). A low LMR was an independent predictor from sarcopenia (p < .001). Based on preoperative sarcopenia and the LMR, we established the SLMR. An elevated SLMR was associated with older age, higher ASA scores, larger tumor size, advanced stages, and vascular invasion (all p < .05). Multivariate analysis revealed that the SLMR was a significant independent predictor (p < .001). We incorporated the SLMR into a prognostic model that included tumor size and TNM stage and generated a nomogram, which accurately predicted 3- and 5-year survival for GC patients. CONCLUSION: Preoperative systemic inflammation is significantly associated with sarcopenia. The LMR combined with sarcopenia could enhance prognostication for patients with GC who underwent radical gastrectomy. IMPLICATIONS FOR PRACTICE: Increasing evidence shows that sarcopenia and systemic inflammation are closely associated with the prognosis of malignant tumors, and it is essential for clinicians to understand the relationship and combined prognostic effects of these factors for gastric cancer (GC). Based on a large data set, this study found that preoperative systemic inflammation was significantly associated with sarcopenia in GC, and combining these two predictors could effectively predict the prognosis and complement the prognostic value of the TNM staging system. These findings may lead to the development of new therapeutic avenues to improve cancer outcomes.


Assuntos
Sarcopenia/patologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia , Síndrome de Resposta Inflamatória Sistêmica/patologia , Idoso , Feminino , Gastrectomia , Humanos , Contagem de Leucócitos , Linfócitos/patologia , Masculino , Pessoa de Meia-Idade , Monócitos/patologia , Nomogramas , Estudos Retrospectivos , Análise de Sobrevida
9.
Ann Surg Oncol ; 26(12): 4027-4036, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31463693

RESUMO

BACKGROUND: Previous studies have suggested that preoperative anemia negatively influences survival in patients with gastric cancer (GC). We sought to investigate which anemic markers can better predict the prognosis of patients with resectable GC. METHODS: The study involved 2277 GC patients who underwent curative resection between December 2008 and December 2014. Cox regression models were used to identify the best anemic markers associated with prognosis. Time-dependent receiver operating characteristics analysis (t-ROC) and the estimated area under the curve (AUC) were used to compare the prognostic values. RESULTS: Of all patients, 1709 (75.1%) were male, and the median age was 61 years. Univariate analyses showed that preoperative hematocrit (HCT), hemoglobin, and mean corpuscular volume were associated with OS (all P < 0.05). However, in a separate analysis of individual stages, only HCT was shown to be significantly prognostic across all tumor stages (all P < 0.05). In the multivariate analysis, preoperative HCT remained an independent prognostic factor for GC. Low HCT was significantly associated with older age, female sex, lower body mass index, higher American Society of Anesthesiologists score, higher preoperative transfusion rate, 90-day mortality, adjuvant chemotherapy, larger tumor size, lymph node metastasis, later stage, and vascular involvement. The t-ROC curve and AUC for HCT were similar to those for the controlling nutritional status and prognostic nutritional index throughout the observation period. CONCLUSIONS: The preoperative HCT is a novel, simple, and powerful prognostic indicator of poor outcome in patients with GC and can be used as a part of the preoperative risk stratification process.


Assuntos
Biomarcadores Tumorais/sangue , Gastrectomia/mortalidade , Cuidados Pré-Operatórios , Neoplasias Gástricas/sangue , Neoplasias Gástricas/cirurgia , Idoso , Feminino , Seguimentos , Hematócrito , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Estudos Retrospectivos , Neoplasias Gástricas/patologia , Taxa de Sobrevida
10.
Gastric Cancer ; 22(2): 403-412, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29982861

RESUMO

BACKGROUND: The systemic inflammation score (SIS), based on preoperative serum albumin (Alb) level and lymphocyte-to-monocyte ratio (LMR), has been shown to be a novel prognostic score for some tumors. We investigate the prognostic value of the SIS in patients with resectable gastric cancer (GC). METHODS: Patients with GC who underwent curative resection between December 2008 and December 2013 were included. Time-dependent receiver operating characteristics analysis (t-ROC), concordance index (C-index) and AUC were used to compare the prognostic impact. RESULTS: Totally, 1786 patients with resectable GC were included in the study. By multivariate analysis, the SIS was not an independent prognostic factor. However, the normal Alb level (≥ 40 g/l) and LMR ≥ 3.4 both remained independent protective factors for GC (both P < 0.05). Due to the similar survival of patients with LMR ≥ 3.4 and LMR < 3.4 in the normal Alb group, we combined the two subgroups to establish the modified SIS (mSIS). Multivariate analysis revealed that the mSIS was the only significant independent biomarker (P < 0.05). The t-ROC curve and C-index for the mSIS were superior to those of the SIS throughout the observation period. Furthermore, the AUC of the mSIS was significantly greater than that of the SIS at 3 and 5 years after operation (both P < 0.05). CONCLUSION: The preoperative mSIS is a novel, simple and useful prognostic factor for postoperative survival in patients with GC and can be used as a part of the preoperative risk stratification process to improve the prediction of clinical outcomes.


Assuntos
Biomarcadores Tumorais/sangue , Inflamação/sangue , Contagem de Linfócitos , Neutrófilos , Albumina Sérica/análise , Neoplasias Gástricas/sangue , Adulto , Idoso , Feminino , Gastrectomia/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Período Pré-Operatório , Prognóstico , Estudos Retrospectivos , Sensibilidade e Especificidade , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia
12.
Ann Surg Oncol ; 25(9): 2703-2712, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29971670

RESUMO

OBJECTIVE: The aim of this study was to determine the prognostic significance of preoperative carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9 levels in patients with resectable gastric cancer (GC). PATIENTS AND METHODS: Patients who underwent a radical resection for GC at the Fujian Medical University Union Hospital between 2007 and 2014 were included in this study. The estimated area under the curve (AUC) was compared to evaluate the discriminatory ability of tumor makers. Additional external validation was performed using a dataset from Sun Yat-sen University Cancer Center. RESULTS: Preoperative CEA/CA19-9 levels were identified as an independent predictor of overall survival (OS) and disease-specific survival (DSS) (both p < 0.05) in the development group. In a subgroup analysis based on TNM stage, preoperative CEA/CA19-9 levels clearly stratified the survival rates for stage III GC (p < 0.05). A multivariate analysis revealed that preoperative CEA/CA19-9 levels were an independent prognostic factor (p < 0.05) in stage III; the AUC of the preoperative CEA/CA19-9 was equivalent to that of T stage. A prediction model (TNMC) for stage III GC was developed by incorporating preoperative CEA/CA19-9 levels into the American Joint Committee on Cancer (AJCC) staging system. The AUC of the TNMC was significantly higher than that of the TNM staging system at 1, 3, and 5 years postoperatively (all p < 0.05), with similar results also being obtained in the external validation set. CONCLUSION: Preoperative CEA/CA19-9 levels are an independent predictor of OS and DSS in stage III GC patients. The inclusion of preoperative CEA/CA19-9 levels in AJCC TNM staging provided an optimal prognosis in stage III GC.


Assuntos
Adenocarcinoma/sangue , Adenocarcinoma/patologia , Antígeno CA-19-9/sangue , Antígeno Carcinoembrionário/sangue , Neoplasias Gástricas/sangue , Neoplasias Gástricas/patologia , Adenocarcinoma/cirurgia , Idoso , Intervalo Livre de Doença , Feminino , Gastrectomia , Humanos , Estimativa de Kaplan-Meier , Excisão de Linfonodo , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Período Pré-Operatório , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Neoplasias Gástricas/cirurgia , Taxa de Sobrevida
14.
Eur J Cancer Prev ; 33(2): 152-160, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-37991237

RESUMO

BACKGROUND: There is still a lack of high-level clinical evidence and uniform conclusions on whether there are differences in lymph node metastasis (LNM) and prognosis between early esophageal adenocarcinoma (EAC) and squamous cell carcinoma (ESCC). METHODS: Patients with surgically resected, histologically diagnosed, pT1 EAC or ESCC in the Surveillance, Epidemiology and End Results registries database from 2004 to 2015 were included. Multivariable logistic regression, Cox regression, multivariate competing risk model, and propensity score matching were used to analyze association the histology and LNM or prognosis. RESULTS: A total of 570 early esophageal cancer patients were included. The LNM rates were 13.8% and 15.1% for EAC and ESCC ( P  = 0.757), respectively. Multivariate logistic regression analysis showed no significant association between histological type and LNM (odds ratio [OR], 1.209; 95% CI, 0.538-2.715; P  = 0.646). Moreover, the prognosis of early EAC and ESCC was shown to be comparable in both multivariate Cox regression (hazard ratio [HR], 1.483; 95% CI, 0.699-3.150; P  = 0.305) and the multivariate competing risk model (subdistribution HR, 1.451; 95% CI, 0.628-3.354; P  = 0.383). After propensity score matching, there were no significant differences between early EAC and ESCC in terms of LNM (10.6% vs.18.2%, P  = 0.215), 5-year CSS (89.8% [95% CI, 81.0%-98.6%] vs. 79.1% [95% CI, 67.9%-90.3%], P  = 0.102) and 5-year cumulative incidence of CSS (10.2% [95% CI, 1.4%-19.0%] vs. 79.1% [95% CI, 9.7%-32.1%], P  = 0.124). CONCLUSION: The risk of LNM and prognosis of early ESCC and EAC are comparable, so the treatment choice for early esophageal cancer does not depend on the histologic type.


Assuntos
Adenocarcinoma , Neoplasias Esofágicas , Esofagectomia , Humanos , Estadiamento de Neoplasias , Metástase Linfática , Prognóstico , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/epidemiologia , Neoplasias Esofágicas/patologia
15.
16.
JAMA Surg ; 158(1): 10-18, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36383362

RESUMO

Importance: The survival benefit of laparoscopic total gastrectomy combined with spleen-preserving splenic hilar lymphadenectomy (LSTG) for locally advanced proximal gastric cancer (APGC) without invasion into the greater curvature remains uncertain. Objective: To compare the long-term and short-term efficacy of LSTG (D2 + No. 10 group) and conventional laparoscopic total gastrectomy (D2 group) for patients with APGC that has not invaded the greater curvature. Design, Setting, and Participants: In this open-label, prospective randomized clinical trial, a total of 536 patients with clinical stage cT2 to 4a/N0 to 3/M0 APGC without invasion into the greater curvature were enrolled from January 2015 to October 2018. The final follow-up was on October 31, 2021. Data were analyzed from December 2021 to February 2022. Interventions: Eligible patients were randomized to the D2 + No. 10 group or the D2 group. Main Outcomes and Measures: The primary outcome was 3-year disease-free survival (DFS). The secondary outcomes were 3-year overall survival (OS) and morbidity and mortality within 30 days after surgery. Results: Of 526 included patients, 392 (74.5%) were men, and the mean (SD) age was 60.6 (9.6) years. A total of 263 patients were included in the D2 + No. 10 group, and 263 were included in the D2 group. The 3-year DFS was 70.3% (95% CI, 64.8-75.8) for the D2 + No. 10 group and 64.3% (95% CI, 58.4-70.2; P = .11) for the D2 group, and the 3-year OS in the D2 + No. 10 group was better than that in the D2 group (75.7% [95% CI, 70.6-80.8] vs 66.5% [95% CI, 60.8-72.2]; P = .02). Multivariate analysis revealed that splenic hilar lymphadenectomy was not an independent protective factor for DFS (hazard ratio [HR], 0.86; 95% CI, 0.63-1.16) or OS (HR, 0.81; 95% CI, 0.59-1.12). Stratification analysis showed that patients with advanced posterior gastric cancer in the D2 + No. 10 group had better 3-year DFS (92.9% vs 39.3%; P < .001) and OS (92.9% vs 42.9%; P < .001) than those in the D2 group. Multivariate analysis confirmed that patients with advanced posterior gastric cancer could have the survival benefit from No. 10 lymph node dissection (DFS: HR, 0.10; 95% CI, 0.02-0.46; OS: HR, 0.12; 95% CI, 0.03-0.52). Conclusions and Relevance: Although LSTG could not significantly improve the 3-year DFS of patients with APGC without invasion into the greater curvature, patients with APGC located posterior gastric wall may benefit from LSTG. Trial Registration: ClinicalTrials.gov Identifier: NCT02333721.


Assuntos
Laparoscopia , Neoplasias Gástricas , Masculino , Humanos , Pessoa de Meia-Idade , Feminino , Neoplasias Gástricas/patologia , Baço , Estudos Prospectivos , Excisão de Linfonodo/mortalidade , Gastrectomia/mortalidade
17.
Front Immunol ; 12: 783243, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35116023

RESUMO

Objective: To investigate the safety and efficacy of camrelizumab in combination with nab-paclitaxel plus S-1 for the treatment of gastric cancer with serosal invasion. Method: Two hundred patients with gastric cancer with serosal invasion who received neoadjuvant therapy from January 2012 to December 2020 were retrospectively analyzed. According to the different neoadjuvant therapy regimens, the patients were divided into the following three groups: the SOX group (S-1 + oxaliplatin) (72 patients), SAP group (S-1 + nab-paclitaxel) (95 patients) and C-SAP group (camrelizumab + S-1 + nab-paclitaxel) (33 patients). Result: The pathological response (TRG 1a/1b) in the C-SAP group (39.4%) was not significantly different from that in the SAP group (26.3%) and was significantly higher than that in the SOX group (18.1%). The rate of ypT0 in the C-SAP group (24.2%) was higher than that in the SAP group (6.3%) and the SOX group (5.6%). The rate of ypN0 in the C-SAP group (66.7%) was also higher than that in the SAP group (38.9%) and the SOX group (36.1%). The rate of pCR in the C-SAP group (21.2%) was higher than that in the SAP group (5.3%) and the SOX group (2.8%). The use of an anti-PD-1 monoclonal antibody was an independent protective factor for TRG grade (1a/1b). The use of camrelizumab did not increase postoperative complications or the adverse effects of neoadjuvant therapy. Conclusion: Camrelizumab combined with nab-paclitaxel plus S-1 could significantly improve the rate of tumor regression grade (TRG 1a/1b) and the rate of pCR in gastric cancer with serosal invasion.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Adulto , Idoso , Albuminas/administração & dosagem , Albuminas/efeitos adversos , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Combinação de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante/métodos , Ácido Oxônico/administração & dosagem , Ácido Oxônico/efeitos adversos , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Estudos Retrospectivos , Tegafur/administração & dosagem , Tegafur/efeitos adversos , Resultado do Tratamento
18.
J Cancer ; 11(12): 3483-3491, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32284744

RESUMO

Objective: To evaluate the prognostic significance of the eighth edition of the American Joint Committee on Cancer (AJCC) TNM staging classification for gastric cancer. Methods: Prospective databases were reviewed to identify patients who underwent radical gastrectomy at two specialized eastern centers. The prognostic value of the eighth edition TNM classification was estimated and compared with that of the seventh edition. Additional external validation was performed using a dataset from a Western population. Results: Significant differences in 5-year overall survival (OS) rates were observed for each TNM stage when using the eighth edition system, and smaller Akaike information criteria (AIC) values and a higher c-statistic were observed relative to those of the seventh edition. However, the OS rates in each subgroup of stage III patients based on the eighth edition were significantly different. Patients with the same pN stage, namely, the pT4a and pT4b groups, showed similar 5-year OS (P>0.05). Based on the survival data, we propose a simplified staging system. In the improved TNM (iTNM) staging system, the subgroups of a given TNM stage do not show statistically significant differences in OS. The iTNM staging exhibits superior prognostic stratification, with lower AIC values and a higher c-statistic than the eighth edition TNM classification. Similar results were obtained with the external validation dataset from the IMIGASTRIC database. Conclusion: The prognostic prediction of the eighth edition of the AJCC TNM classification is superior to that of the seventh edition. However, it remains associated with some stage migration. The iTNM staging system permits simplification and slightly better prognostic prediction.

19.
J Cancer ; 10(17): 4106-4113, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31417655

RESUMO

Purpose: To compare the clinicopathologic data and short-term surgical outcomes of laparoscopic gastrectomy (LG) for gastric cancer (GC) between the east and west. Methods: Patient demographics, surgical procedures, pathological information, and postoperative recovery were compared among gastric cancer patients who underwent LG in the clinical trial of IMIGASTRIC (NCT02325453) between 2009 and 2016. Results: More younger males, higher BMI, lower ASA score and less neoadjvant chemotherapy were evident in east patient cohort. Eastern patients had a higher proportion of proximal, differentiated and advanced gastric cancers. More total gastrectomies, larger extent of lymph node (LN) dissection, and higher number of retrieved LNs were found in the eastern patients. However, more Roux-en-Y anastomosis procedures during distal gastrectomy and intra-corporeal anastomosis were performed in the western patients. The west patients showed faster postoperative recovery than the eastern patients. The mortality rates of the western patients were comparable to those of the eastern patients. However, fewer III-IV complications were evident in the eastern centers. Multivariate analyses revealed that an elderly age, higher ASA score, and more blood loss were the significant independent risk factors of postoperative complications for eastern patients. However, for the western patients, the independent risk factors were neoadjuvant therapy, more retrieval LNs, and pT3-4 stage. Conclusions: The selections and short-term surgical outcomes of LG for GC were widely different between East and West. To obtain more objective and accurate results, these differences should be considered in future international prospective studies.

20.
World J Clin Cases ; 7(2): 145-155, 2019 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-30705892

RESUMO

AIM: To establish an appropriate N classification system for early gastric cancer (EGC). METHODS: Data from 10714 patients who underwent radical gastrectomy between 1988 and 2011 were retrieved from the National Cancer Institute's Surveillance, Epidemiology, and End Result database. The overall survival (OS) based on the eighth edition and new tumor lymph node metastasis (TNM) staging systems were compared, and the analysis was repeated in an external validation set from the Fujian Medical University Union Hospital database. RESULTS: There were no significant differences in OS between N1 and N2 cancers or between N3a and N3b cancers in cases of EGC. The X-tile program identified that the new staging system for EGC consisted of T1N0, T1N1' [1-6 metastatic lymph nodes (LNs)], and T1N2' ( ≥ 7 metastatic LNs). Compared with the eighth edition of the TNM staging system, the OS of patients in T1N1' stage was similar to that of patients with stage IIA disease, whereas the OS of patients in T1N2' stage was similar to that of patients with stage IIB disease. The new TNM staging system exhibited a slightly lower Akaike Information Criterion value and higher χ 2 and c-statistic compared with the eighth edition of the TNM classification system. Similar results were found in the external validation dataset from the external validation set. CONCLUSION: We have developed an optional new TNM staging system with a better predictive ability that can be used to accurately predict the 5-year OS of patients with EGC.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa