Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Radiother Oncol ; 196: 110316, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38679201

RESUMO

BACKGROUND AND PURPOSE: This phase Ib study was designed to assess the safety/tolerability and preliminary antitumor activity of neoadjuvant low-dose radiotherapy (LDRT) plus durvalumab and chemotherapy for potentially resectable stage III non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS: Eligible patients received dose-escalated radiotherapy (10 Gy in 5 fractions [cohort 1], 20 Gy in 10 fractions [cohort 2], and 30 Gy in 15 fractions [cohort 3]) according to a 3 + 3 design, with concurrent durvalumab plus standard chemotherapy for two cycles. Primary objective was safety/tolerability. Secondary objectives included major pathological response (MPR), pathological complete response (pCR), event-free survival (EFS), and exploratory biomarker analysis. RESULTS: Nine patients were enrolled and completed the planned neoadjuvant therapy. No dose-limiting toxicity was recorded. Grade 3-4 treatment-related adverse events were observed in three (33.3 %) patients. Seven (77.8 %) patients successfully converted to resectable cases with R0 resection. No treatment-related surgical delay or death was reported. The MPR and pCR rates were both 33.3 % % (1/3) for cohort 1, 66.7 % (2/3) and 0.0 % for cohort 2, and 100.0 % (3/3), and 66.7 % (2/3) for cohort 3. At data cutoff, the 12 month-EFS rates were 33.3 %, 66.7 %, and 100 % for three cohorts, respectively. By biomarker analysis, TMB values were higher in either pathologically or radiologically responders than in others (all p > 0.05). CONCLUSION: Neoadjuvant LDRT plus durvalumab and chemotherapy was well-tolerated in potentially resectable stage III NSCLC. The preliminary efficacy supports this combined regimen's potential, the optimal radiotherapy dosage was determined to be 30 Gy in 15 fractions, warranting further clinical investigation.


Assuntos
Anticorpos Monoclonais , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Terapia Neoadjuvante , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Masculino , Feminino , Pessoa de Meia-Idade , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Idoso , Estadiamento de Neoplasias , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimiorradioterapia , Dosagem Radioterapêutica
2.
Sci Rep ; 13(1): 6955, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37117226

RESUMO

Previous studies indicate differences in short-term postoperative outcomes depending on the surgical starting time of the day, but long-term data are lacking. The aim of this study was to clarify if surgical starting time of the day influences long-term survival in gastric cancer patients. This cohort study consecutively included 2728 patients who underwent curatively intended gastrectomy for gastric cancer in 2011-2015 at a high-volume hospital in China, with follow-up until June 2019. Cox regression provided hazard ratios (HRs) with 95% confidence intervals (CIs) for 3-year all-cause mortality, adjusted for age, sex, health insurance, pathological tumor stage, surgical approach, neoadjuvant therapy, and weekday of surgery. Compared with patients with early starting time of gastrectomy (08:00-09:29), the point estimates for 3-year all-cause mortality were modestly increased in patients with a starting time in the middle of day (09:30-13:29; HR 1.15, 95% CI 0.97 to 1.37) and later (13:30-21:25; HR 1.10, 0.91 to 1.32). The corresponding HRs were increased particularly in patients who underwent laparoscopic gastrectomy (HR 1.54, 1.10 to 2.14 and HR 1.59, 1.12 to 2.25, respectively) and in those with stage II tumors (HR 1.74, 1.11 to 2.73 and HR 1.60, 1.00 to 2.58, respectively). Our study indicated that in patients who underwent laparoscopic gastrectomy and in those who with stage II tumors, starting surgery in the early morning might be associated with better long-term survival.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Estudos de Coortes , Estudos Retrospectivos , Modelos de Riscos Proporcionais , Gastrectomia , Resultado do Tratamento
3.
Cancer Epidemiol Biomarkers Prev ; 32(7): 936-946, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37104672

RESUMO

BACKGROUND: Sex hormones may influence the development of gastrointestinal cancer, but evidence is inconsistent. METHODS: We systematically searched MEDLINE and Embase databases to identify prospective studies examining associations between prediagnostic circulating levels of sex hormones and risk of five gastrointestinal cancers: esophageal, gastric, liver, pancreatic, and colorectal cancer. Pooled ORs and 95% confidence intervals (95% CI) were calculated using random-effects models. RESULTS: Among 16,879 identified studies, 29 were included (11 cohort, 15 nested case-control, and three case-cohort studies). Comparing the highest versus lowest tertiles, levels of most sex hormones were not associated with the studied tumors. Higher levels of sex hormone binding globulin (SHBG) were associated with increased risk of gastric cancer (OR = 1.35; 95% CI, 1.06-1.72), but such associations were restricted in men only (OR = 1.43; 95% CI, 1.10-1.85) when stratified by sex. Higher SHBG levels were associated with increased risk of liver cancer (OR = 2.07; 95% CI, 1.40-3.06). Higher testosterone levels were associated with increased risk of liver cancer overall (OR = 2.10; 95% CI, 1.48-2.96), particularly in men (OR = 2.63; 95% CI, 1.65-4.18), Asian populations (OR = 3.27; 95% CI, 1.57-6.83), and in hepatitis B surface antigen-positive individuals (OR = 3.90; 95% CI, 1.43-10.64). Higher levels of SHBG and testosterone were associated with decreased risk of colorectal cancer in men (OR = 0.89; 95% CI, 0.80-0.98 and OR = 0.88; 95% CI, 0.80-0.97, respectively) but not in women. CONCLUSIONS: Circulating levels of SHBG and testosterone may influence the risk of gastric, liver, and colorectal cancer. IMPACT: Further clarifying the role of sex hormones in the development of gastrointestinal cancer may unravel future novel targets for prevention and treatment.


Assuntos
Neoplasias Colorretais , Neoplasias Gastrointestinais , Neoplasias Hepáticas , Masculino , Humanos , Feminino , Estudos Prospectivos , Fatores de Risco , Hormônios Esteroides Gonadais , Testosterona , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Globulina de Ligação a Hormônio Sexual/análise , Estradiol
4.
Int J Surg ; 109(5): 1141-1148, 2023 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-36999825

RESUMO

BACKGROUND: The authors aimed to produce a prediction model for survival at any given date after surgery for esophageal cancer (conditional survival), which has not been done previously. MATERIALS AND METHODS: Using joint density functions, the authors developed and validated a prediction model for all-cause and disease-specific mortality after surgery with esophagectomy, for esophageal cancer, conditional on postsurgery survival time. The model performance was assessed by the area under the receiver operating characteristic curve (AUC) and risk calibration, with internal cross-validation. The derivation cohort was a nationwide Swedish population-based cohort of 1027 patients treated in 1987-2010, with follow-up throughout 2016. This validation cohort was another Swedish population-based cohort of 558 patients treated in 2011-2013, with follow-up throughout 2018. RESULTS: The model predictors were age, sex, education, tumor histology, chemo(radio)therapy, tumor stage, resection margin status, and reoperation. The medians of AUC after internal cross-validation in the derivation cohort were 0.74 (95% CI: 0.69-0.78) for 3-year all-cause mortality, 0.76 (95% CI: 0.72-0.79) for 5-year all-cause mortality, 0.74 (95% CI: 0.70-0.78) for 3-year disease-specific mortality, and 0.75 (95% CI: 0.72-0.79) for 5-year disease-specific mortality. The corresponding AUC values in the validation cohort ranged from 0.71 to 0.73. The model showed good agreement between observed and predicted risks. Complete results for conditional survival any given date between 1 and 5 years of surgery are available from an interactive web-tool: https://sites.google.com/view/pcsec/home . CONCLUSION: This novel prediction model provided accurate estimates of conditional survival any time after esophageal cancer surgery. The web-tool may help guide postoperative treatment and follow-up.


Assuntos
Neoplasias Esofágicas , Humanos , Estudos de Coortes , Reoperação , Neoplasias Esofágicas/cirurgia , Neoplasias Esofágicas/patologia , Período Pós-Operatório
5.
Br J Cancer ; 127(5): 892-897, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35715630

RESUMO

BACKGROUND: To investigate if anti-androgenic medications 5α-reductase inhibitors (5-ARIs) decrease the risk of developing oesophageal and gastric tumours, analysed by histological type and anatomical sub-site. METHODS: A Swedish population-based cohort study between 2005 and 2018 where men using 5-ARIs were considered exposed. For each exposed participant, ten male age-matched non-users of 5-ARIs (non-exposed) were included. Multivariable Cox regression provided hazard ratios (HR) with 95% confidence intervals (CI) adjusted for age, calendar year, smoking, non-steroidal anti-inflammatory drugs/aspirin use, and statins use. Further adjustments were made depending on the tumour analysed. RESULTS: The cohort included 191,156 users of 5-ARIs and 1,911,560 non-users. Overall, the use of 5-ARIs was not associated with any statistically significantly reduced risk of oesophageal or cardia adenocarcinoma (adjusted HR 0.92, 95% CI 0.82-1.02) or gastric non-cardia adenocarcinoma (adjusted HR 0.90, 95% CI 0.80-1.02). However, the use of 5-ARIs indicated a decreased risk of oesophageal or cardia adenocarcinoma among obese or diabetic participants (adjusted HR 0.55, 95% CI 0.39-0.80) and a reduced risk of oesophageal squamous cell carcinoma (adjusted HR 0.49, 95% CI 0.37-0.65). CONCLUSION: Users of 5-ARIs may have a decreased risk of developing oesophageal or cardia adenocarcinoma among those obese or diabetic, and a decreased risk of oesophageal squamous cell carcinoma.


Assuntos
Adenocarcinoma , Diabetes Mellitus , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Neoplasias Gástricas , Inibidores de 5-alfa Redutase/uso terapêutico , Antagonistas de Androgênios/efeitos adversos , Estudos de Coortes , Neoplasias Esofágicas/epidemiologia , Neoplasias Esofágicas/etiologia , Humanos , Masculino , Obesidade , Oxirredutases , Fatores de Risco , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/etiologia
7.
Gastric Cancer ; 25(3): 652-658, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35166957

RESUMO

BACKGROUND: Low-dose aspirin use may reduce cancer incidence and mortality, but its influence on gastric adenocarcinoma survival is unclear. This study aimed to assess whether aspirin use improves long-term survival following gastrectomy for gastric adenocarcinoma. METHODS: This population-based cohort study included almost all patients who underwent gastrectomy for gastric adenocarcinoma in Sweden from 2006 to 2015, with follow-up throughout 2020. Preoperative exposure to a daily low-dose (75-160 mg) aspirin for 1 (main exposure), 2 and 3 years and for 1 year after gastrectomy was examined in relation to 5-year all-cause mortality (primary outcome) and disease-specific mortality. Multivariable Cox regression provided hazard ratios (HR) with 95% confidence intervals (CI), adjusted for age, sex, education, calendar year, comorbidity, statin use, tumour location, tumour stage, neoadjuvant chemotherapy, surgeon volume of gastrectomy and surgical radicality. RESULTS: Among 2025 patients, 545 (26.9%) used aspirin at the date of gastrectomy. Aspirin use within 1 year before surgery did not decrease the adjusted risk of 5-year all-cause mortality (HR = 0.98, 95% CI 0.85-1.13) or disease-specific mortality (HR = 1.00, 95% CI 0.86-1.17). Preoperative aspirin use for 2 years (HR = 0.98, 95% CI 0.84-1.15) or 3 years (HR = 0.94, 95% CI 0.79-1.12) did not decrease the risk of 5-year all-cause mortality. Patients remaining on aspirin during the first year after gastrectomy had a similar 5-year all-cause mortality as non-users of aspirin (HR = 1.01, 95% CI 0.82-1.25). CONCLUSIONS: Low-dose aspirin use might not improve long-term survival after gastrectomy for gastric adenocarcinoma and may thus not be a target for adjuvant therapy in this group of patients.


Assuntos
Adenocarcinoma , Neoplasias Gástricas , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Aspirina/uso terapêutico , Estudos de Coortes , Gastrectomia , Humanos , Estadiamento de Neoplasias , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia
8.
Br J Cancer ; 126(7): 1100-1107, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35027671

RESUMO

BACKGROUND: This systematic review and meta-analysis examined associations between serum levels of haemoglobin A1c (HbA1c) and glucose and the risk of gastric cancer. METHODS: MEDLINE, Embase, and Cochrane Library were searched for studies examining associations between serum levels of HbA1c or glucose and the risk of gastric cancer. Inclusion of studies, quality assessment, and data extraction were conducted independently by two authors. Pooled hazard ratios (HR) with 95% confidence intervals (CI) were synthesised using random-effects models. Cochran's Q test and I2 statistic were used to assess heterogeneity. RESULTS: Among 3473 identified studies, 12 were included. Of these, 5 studies examined HbA1c levels and 7 studies examined serum glucose levels. Serum HbA1c levels >6% were associated with an increased risk of gastric cancer (HR 1.36, 95% CI 1.06-1.74). When compared with the lowest glucose categories, the highest glucose categories were associated with a borderline increased risk of gastric cancer (HR 1.11, 95% CI 0.98-1.26). In subgroup analyses, studies that adjusted for Helicobacter pylori infection indicated stronger associations between elevated HbA1c levels and gastric cancer (HR 2.08, 95% CI 1.46-2.98) than those without such adjustment (HR 1.10, 95% CI 0.91-1.32). CONCLUSIONS: Long-standing poor glycaemic control may increase the risk of gastric cancer. REGISTRATION NUMBER: PROSPERO CRD42020157453.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Glucose , Hemoglobinas Glicadas , Infecções por Helicobacter/complicações , Humanos , Neoplasias Gástricas/epidemiologia
9.
Br J Cancer ; 126(1): 129-133, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34671128

RESUMO

BACKGROUND: Oesophageal adenocarcinoma is characterised by a strong male predominance. We aimed to test the hypothesis that menopausal hormonal therapy decreases the risk of oesophageal adenocarcinoma. METHODS: This population-based cohort study included all women who used systemic menopausal hormonal therapy (exposed) in Sweden between 2005 and 2018. For each exposed participant, five randomly selected female age-matched non-users of menopausal hormonal therapy (unexposed) were included. Cox regression provided hazard ratios (HR) with 95% confidence intervals (CI) adjusted for age, smoking-related diagnoses, Helicobacter pylori eradication, use of non-steroidal anti-inflammatory drugs/aspirin, use of statins and hysterectomy. RESULTS: The study included 296,964 users of menopausal hormonal therapy and 1,484,820 non-users. Ever-users of menopausal hormonal therapy had an overall decreased risk of oesophageal adenocarcinoma (HR 0.78, 95% CI 0.63-0.97), which remained unchanged after further adjustment for gastro-oesophageal reflux disease (HR 0.78, 95% CI 0.63-0.97) and obesity/diabetes (HR 0.79, 95% CI 0.63-0.98). Decreased HRs were indicated both in users of oestrogen only (HR 0.82, 95% CI 0.60-1.12) and oestrogen combined with progestogen (HR 0.75, 95% CI 0.56-1.00). The risk reduction was more pronounced in users younger than 60 years (HR 0.57, 95% CI 0.38-0.86). CONCLUSIONS: Menopausal hormone therapy in women may decrease the risk of oesophageal adenocarcinoma.


Assuntos
Adenocarcinoma/patologia , Aspirina/uso terapêutico , Neoplasias Esofágicas/patologia , Terapia de Reposição Hormonal/efeitos adversos , Menopausa , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/epidemiologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Estudos de Coortes , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/epidemiologia , Feminino , Humanos , Pessoa de Meia-Idade , Fatores de Risco , Suécia/epidemiologia
10.
Br J Cancer ; 125(2): 277-283, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33972744

RESUMO

BACKGROUND: Metformin may improve the prognosis in gastric adenocarcinoma, but the existing literature is limited and contradictory. METHODS: This was a Swedish population-based cohort study of diabetes patients who were diagnosed with gastric adenocarcinoma in 2005-2018 and followed up until December 2019. The data were retrieved from four national health data registries: Prescribed Drug Registry, Cancer Registry, Patient Registry and Cause of Death Registry. Associations between metformin use before the gastric adenocarcinoma diagnosis and the risk of disease-specific and all-cause mortality were assessed using multivariable Cox proportional hazard regression. The hazard ratios (HRs) and 95% confidence intervals (CIs) were adjusted for sex, age, calendar year, comorbidity, use of non-steroidal anti-inflammatory drugs or aspirin, and use of statins. RESULTS: Compared with non-users, metformin users had a decreased risk of disease-specific mortality (HR 0.79, 95% CI 0.67-0.93) and all-cause mortality (HR 0.78, 95% CI 0.68-0.90). The associations were seemingly stronger among patients of female sex (HR 0.66, 95% CI 0.49-0.89), patients with tumour stage III or IV (HR 0.71, 95% CI 0.58-0.88), and those with the least comorbidity (HR 0.71, 95% CI 0.57-0.89). CONCLUSIONS: Metformin use may improve survival in gastric adenocarcinoma among diabetes patients.


Assuntos
Adenocarcinoma/mortalidade , Metformina/uso terapêutico , Neoplasias Gástricas/mortalidade , Adenocarcinoma/patologia , Idoso , Idoso de 80 Anos ou mais , Causas de Morte , Estudos de Coortes , Feminino , Humanos , Masculino , Estadiamento de Neoplasias , Prognóstico , Sistema de Registros , Caracteres Sexuais , Neoplasias Gástricas/patologia , Análise de Sobrevida , Suécia/epidemiologia
11.
BMJ Open ; 11(4): e042762, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33931407

RESUMO

OBJECTIVE: The substantial differences in socioeconomic and lifestyle exposures between urban and rural areas in China may lead to urban-rural disparity in cancer risk. This study aimed to assess the urban-rural disparity in cancer incidence in China. METHODS: Using data from 36 regional cancer registries in China in 2008-2012, we compared the age-standardised incidence rates of cancer by sex and anatomic site between rural and urban areas. We calculated the rate difference and rate ratio comparing rates in rural versus urban areas by sex and cancer type. RESULTS: The incidence rate of all cancers in women was slightly lower in rural areas than in urban areas, but the total cancer rate in men was higher in rural areas than in urban areas. The incidence rates in women were higher in rural areas than in urban areas for cancers of the oesophagus, stomach, and liver and biliary passages, but lower for cancers of thyroid and breast. Men residing in rural areas had higher incidence rates for cancers of the oesophagus, stomach, and liver and biliary passages, but lower rates for prostate cancer, lip, oral cavity and pharynx cancer, and colorectal cancer. CONCLUSIONS: Our findings suggest substantial urban-rural disparity in cancer incidence in China, which varies across cancer types and the sexes. Cancer prevention strategies should be tailored for common cancers in rural and urban areas.


Assuntos
Neoplasias , População Rural , China/epidemiologia , Estudos Transversais , Feminino , Humanos , Incidência , Masculino , Neoplasias/epidemiologia , Sistema de Registros , População Urbana
12.
Am J Gastroenterol ; 116(4): 683-691, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33982937

RESUMO

INTRODUCTION: Esophageal squamous cell carcinoma (ESCC) carries a poor prognosis, but earlier tumor detection would improve survival. We aimed to develop and externally validate a risk prediction model based on exposure to readily available risk factors to identify high-risk individuals of ESCC. METHODS: Competing risk regression modeling was used to develop a risk prediction model. Individuals' absolute risk of ESCC during follow-up was computed with the cumulative incidence function. We used prospectively collected data from the Nord-Trøndelag Health Study (HUNT) for model derivation and the UK Biobank cohort for validation. Candidate predictors were age, sex, tobacco smoking, alcohol consumption, body mass index (BMI), education, cohabitation, physical exercise, and employment. Model performance was validated internally and externally by evaluating model discrimination using the area under the receiver-operating characteristic curve (AUC) and model calibration. RESULTS: The developed risk prediction model included age, sex, smoking, alcohol, and BMI. The AUC for 5-year risk of ESCC was 0.76 (95% confidence interval [CI], 0.58-0.93) in the derivation cohort and 0.70 (95% CI, 0.64-0.75) in the validation cohort. The calibration showed close agreement between the predicted cumulative risk and observed probabilities of developing ESCC. Higher net benefit was observed when applying the risk prediction model than considering all participants as being at high risk, indicating good clinical usefulness. A web tool for risk calculation was developed: https://sites.google.com/view/escc-ugis-ki. DISCUSSION: This ESCC risk prediction model showed good discrimination and calibration and validated well in an independent cohort. This readily available model can help select high-risk individuals for preventive interventions.


Assuntos
Neoplasias Esofágicas/epidemiologia , Carcinoma de Células Escamosas do Esôfago/epidemiologia , Medição de Risco/métodos , Adulto , Progressão da Doença , Neoplasias Esofágicas/diagnóstico , Carcinoma de Células Escamosas do Esôfago/diagnóstico , Feminino , Seguimentos , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Noruega/epidemiologia , Prognóstico , Estudos Prospectivos , Curva ROC , Fatores de Risco , Reino Unido/epidemiologia
13.
Ann Surg ; 273(5): 933-939, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33824250

RESUMO

OBJECTIVE: We aimed to develop prediction models for estimating the long-term survival in patients who have undergone surgery for esophageal cancer. BACKGROUND: Few prediction models have been developed for the long-term survival in esophageal cancer patients. METHODS: This nationwide Swedish population-based cohort study included 1542 patients who survived for ≥90 days after esophageal cancer surgery between 1987 and 2010, with follow-up until 2016. Risk prediction models for 1-, 3-, and 5-year all-cause mortality and 3- and 5-year disease-specific mortality were developed using logistic regression. Candidate predictors were established and readily identifiable prognostic factors. The performance of the models was assessed by the area under receiver-operating characteristic curve (AUC) with interquartile range (IQR) using bootstrap cross-validation and risk calibration. RESULTS: Predictors included in all models were age, sex, pathological tumor stage, tumor histology, and resection margin status. The models also included various additional predictors depending on the outcome, that is, education level, neoadjuvant therapy, reoperation (within 30 d of primary surgery) and comorbidity (Charlson comorbidity index). The AUC statistics after cross-validation were 0.71 (IQR 0.69-0.74) for 1-year, 0.77 (IQR 0.75-0.80) for 3-year, and 0.78 (IQR 0.76-0.81) for 5-year all-cause mortality. The corresponding values were 0.76 (IQR 0.74-0.79) for 3-year and 0.77 (IQR 0.71-0.83) for 5-year disease-specific mortality. All models showed good agreement between the observed and predicted risks. CONCLUSIONS: These models showed good performance for predicting long-term survival after esophageal cancer surgery and may thus be useful for patients in planning their lives and to guide the postoperative treatment and follow-up.


Assuntos
Neoplasias Esofágicas/mortalidade , Esofagectomia/métodos , Vigilância da População/métodos , Medição de Risco/métodos , Idoso , Neoplasias Esofágicas/cirurgia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Período Pós-Operatório , Estudos Retrospectivos , Fatores de Risco , Taxa de Sobrevida/tendências , Suécia/epidemiologia , Fatores de Tempo
14.
Cancers (Basel) ; 13(1)2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33466239

RESUMO

This study aimed to examine the global burden, risk factors, and trends of esophageal cancer based on age, sex, and histological subtype. The data were retrieved from cancer registries database from 48 countries in the period 1980-2017. Temporal patterns of incidence and mortality were evaluated by average annual percent change (AAPC) using joinpoint regression. Associations with risk factors were examined by linear regression. The highest incidence of esophageal cancer was observed in Eastern Asia. The highest incidence of adenocarcinoma (AC) was found in the Netherlands, the United Kingdom, and Ireland. A higher AC/squamous cell carcinoma (SCC) incidence ratio was associated with a higher prevalence of obesity and elevated cholesterol. We observed an incidence increase (including AC and SCC) in some countries, with the Czech Republic (female: AAPC 4.66), Spain (female: 3.41), Norway (male: 3.10), Japan (female: 2.18), Thailand (male: 2.17), the Netherlands (male: 2.11; female: 1.88), and Canada (male: 1.51) showing the most significant increase. Countries with increasing mortality included Thailand (male: 5.24), Austria (female: 3.67), Latvia (male: 2.33), and Portugal (male: 1.12). Although the incidence of esophageal cancer showed an overall decreasing trend, an increasing trend was observed in some countries with high AC/SCC incidence ratios. More preventive measures are needed for these countries.

15.
Int J Cancer ; 148(4): 905-913, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-32895915

RESUMO

Sex hormonal differences may contribute to the strong male predominance in esophageal adenocarcinoma (EAC), but whether sex hormone levels influence survival in EAC is unstudied. Our study aimed to assess associations between prediagnostic sex hormone levels and survival in EAC. In a population-based cohort study, 244 male EAC patients from the Janus Serum Bank Cohort in Norway were followed up through 2018. Associations between prediagnostic serum levels of 12 sex hormone measures and disease-specific mortality were assessed using multivariable Cox regression, providing hazard ratios (HR) with 95% confidence intervals (CI) adjusted for age, calendar year, body mass index, tobacco smoking, physical activity and surgical resection. Higher levels of sex hormone-binding globulin (SHBG) indicated decreased disease-specific mortality (HR 0.68, 95% CI 0.44-1.07, highest vs lowest tertile). In stratified analyses by surgery, such associations remained in nonoperated patients (HR 0.58, 95% CI 0.35-0.96, highest vs lowest tertile), but not in operated patients. Higher levels of follicle-stimulating hormone (FSH) were associated with increased disease-specific mortality in an exposure-response pattern; HRs for the middle and highest tertiles vs the lowest tertile were 1.35 (95% CI 0.89-2.05) and 1.61 (95% CI 1.06-2.43), respectively. No clear associations were observed with serum levels of dehydroepiandrosterone sulfate, luteinizing hormone, prolactin, testosterone, 17-OH-progesterone, progesterone, estradiol, androstenedione, testosterone:estradiol ratio or free testosterone index. These findings suggest that higher endogenous levels of SHBG and lower levels of FSH may increase the survival in EAC. The other 10 examined sex hormone measures may not influence the survival.


Assuntos
Adenocarcinoma/sangue , Neoplasias Esofágicas/sangue , Hormônios Esteroides Gonadais/sangue , Adenocarcinoma/diagnóstico , Adenocarcinoma/mortalidade , Adulto , Idoso , Estudos de Coortes , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/mortalidade , Hormônio Foliculoestimulante/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prolactina/sangue , Globulina de Ligação a Hormônio Sexual/metabolismo , Análise de Sobrevida , Taxa de Sobrevida
17.
BMC Gastroenterol ; 20(1): 418, 2020 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-33308167

RESUMO

BACKGROUND: Gastric cancer is more common in men than in women, but underlying reasons have not been completely understood. This study aimed to assess patterns of the sex difference in the incidence of gastric cancer in the United States. METHODS: Using data from 13 cancer registries in the Surveillance, Epidemiology, and End Results Program, we analyzed the age-specific sex difference in the incidence of gastric cancer by ethnicity, anatomic site and histological type in the United States during 1992-2014. We assessed the temporal trends in the sex differences in the incidence of gastric cancer during the study period. RESULTS: The male-to-female incidence ratio of cardia cancer increased with age until peaking at ages 55-69 years and decreased thereafter, while the ratio for non-cardia gastric cancer increased with age before ages < 60 years and remained stable onwards. The age-specific patterns in the sex difference of gastric cancer incidence varied between intestinal and diffuse histological types. The sex difference in the incidence of cardia cancer remained relatively stable except for that the absolute difference between the sexes in whites decreased on average by 0.8% per year from 1992 to 2014. The absolute incidence difference between the sexes in non-cardia gastric cancer decreased over time in whites, blacks, and Asian and Pacific islanders by approximately 4% per year. The male-to-female incidence ratio of non-cardia gastric cancer decreased over time in whites and blacks, but remained relatively stable in Asian and Pacific islanders. CONCLUSIONS: Both extrinsic and intrinsic factors may have contributed to the sex difference in gastric cancer. Sex hormones may play a role in the development of cardia cancer and intestinal type of gastric cancer.


Assuntos
Caracteres Sexuais , Neoplasias Gástricas , Idoso , Cárdia , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Programa de SEER , Neoplasias Gástricas/epidemiologia , Estados Unidos/epidemiologia
18.
Cancer Epidemiol Biomarkers Prev ; 29(11): 2109-2118, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32855267

RESUMO

Associations between circulating levels of obesity-related biomarkers and risk of esophageal adenocarcinoma and Barrett esophagus have been reported, but the results are inconsistent. A literature search until October 2018 in MEDLINE and EMBASE was performed. Pooled ORs with 95% confidence intervals (CI) were estimated for associations between 13 obesity-related inflammatory and metabolic biomarkers and risk of esophageal adenocarcinoma or Barrett esophagus using random effect meta-analyses. Among 7,641 studies, 19 were eligible for inclusion (12 cross-sectional, two nested case-control, and five cohort studies). Comparing the highest versus lowest categories of circulating biomarker levels, the pooled ORs were increased for leptin (OR, 1.68; 95% CI, 0.95-2.97 for Barrett esophagus), glucose (OR, 1.12; 95% CI, 1.03-1.22 for esophageal adenocarcinoma), insulin (OR, 1.47; 95% CI, 1.06-2.00 for Barrett esophagus), C-reactive protein (CRP; OR, 2.06; 95% CI, 1.28-3.31 for esophageal adenocarcinoma), IL6 (OR, 1.50; 95% CI, 1.03-2.19 for esophageal adenocarcinoma), and soluble TNF receptor 2 (sTNFR-2; OR, 3.16; 95% CI, 1.76-5.65 for esophageal adenocarcinoma). No associations were identified for adiponectin, ghrelin, insulin-like growth factor 1, insulin-like growth factor-binding protein 3, triglycerides, IL8, or TNFα. Higher circulating levels of leptin, glucose, insulin, CRP, IL6, and sTNFR-2 may be associated with an increased risk of esophageal adenocarcinoma or Barrett esophagus. More prospective studies are required to identify biomarkers that can help select high-risk individuals for targeted prevention and early detection.


Assuntos
Adenocarcinoma/imunologia , Esôfago de Barrett/imunologia , Biomarcadores Tumorais/metabolismo , Neoplasias Esofágicas/imunologia , Humanos
19.
Cancer Sci ; 111(7): 2451-2459, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32359209

RESUMO

The causes of death in patients with gastric adenocarcinoma have not been well characterized. This nationwide population-based cohort study included 56 240 patients diagnosed with gastric adenocarcinoma in 1970-2014 in Sweden. We used competing-risks regression to compare cause-specific risks of death in patients with different characteristics and a multiple-cause approach to assess proportions of deaths attributable to each cause. Among 53 049 deaths, gastric cancer was the main (77.7% of all deaths) underlying cause. Other major underlying causes were nongastric malignancies (8.0%), ischemic heart disease or cerebrovascular disease (6.5%), and respiratory diseases (1.4%). Risk of death from gastric cancer steadily decreased in patients with cardia adenocarcinoma over the study period, but remained relatively stable in patients with noncardia adenocarcinoma since the 1980s. Risk of death from other malignancies increased during later calendar periods (subhazard ratio [SHR] = 2.16, 95% confidence interval [CI] 1.97-2.38, comparing 2001-2014 with 1970-1980). Compared with men, the risk of death in women with cardia adenocarcinoma was higher from gastric cancer (SHR = 1.18, 95% CI 1.10-1.27), but lower from other malignancies (SHR = 0.80, 95% CI 0.71-0.91). In multiple-cause models, 60.4%-71.2% of all deaths were attributable to gastric cancer and 9.5%-12.1% to other malignancies. The temporal trends of cause-specific risks from multiple-cause models were similar to those of underlying causes. Our findings suggest that although most deaths in patients with gastric adenocarcinoma are due to gastric cancer, other causes of death are common. Patients with cardia adenocarcinoma face considerable increasing risk of death from other causes over time, particularly from other malignancies.


Assuntos
Adenocarcinoma/epidemiologia , Causas de Morte , Neoplasias Gástricas/epidemiologia , Adenocarcinoma/diagnóstico , Adenocarcinoma/história , Adenocarcinoma/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , História do Século XX , História do Século XXI , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Vigilância da População , Sistema de Registros , Suécia/epidemiologia
20.
Scand J Gastroenterol ; 55(3): 258-264, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32045532

RESUMO

Background: Geographical variations in the incidence and tumour stage distribution of oesophageal cancer in Sweden are not well characterised.Methods: Using data from the Swedish Cancer Registry over 45 years (1972-2016), we compared the age-standardised incidence rates of oesophageal cancer by histological type across all seven national areas (in five-year periods) and 21 counties (in 15-year periods) in Sweden, and assessed the geographical distribution of tumour stage at diagnosis since 2004.Results: The incidence rate of oesophageal adenocarcinoma increased in all national areas and counties and in both sexes over time, while the rate of oesophageal squamous cell carcinoma decreased from the 1980s onwards. In the latest period (2012- 2016), the incidence rate of adenocarcinoma in men ranged from 3.5/100,000 person-years in West Sweden to 6.2/100,000 person-years in North Middle Sweden. At the county level, the rate of adenocarcinoma in men was lowest in Jämtland (2.7/100,000 person-years) and highest in Gotland (6.2/100 000 person-years) in 2002-2016. The incidence rates of both adenocarcinoma and squamous cell carcinoma in women were below 2/100,000 person-years in all national areas and counties in the latest calendar periods, i.e., 2012-2016 and 2002-2016, respectively. The proportion of patents with tumour stage IV ranged from 22% in Stockholm area to 31% in Middle Norrland, while at the healthcare region level it was lowest in Stockholm healthcare region (23%) and highest in North (30%) and Uppsala-Örebro (29%) healthcare regions.Conclusion: There are considerable geographical variations in the incidence and tumour stage distribution of oesophageal cancer in Sweden.


Assuntos
Adenocarcinoma/epidemiologia , Carcinoma de Células Escamosas/epidemiologia , Neoplasias Esofágicas/epidemiologia , Geografia Médica , Disparidades em Assistência à Saúde , Adenocarcinoma/patologia , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/patologia , Feminino , Humanos , Incidência , Masculino , Estadiamento de Neoplasias , Sistema de Registros , Fatores de Risco , Distribuição por Sexo , Suécia/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA