Impact of comorbidities on overall survival of high-risk and advanced melanoma.

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 8536-8536
Author(s):  
Prashanth Peddi ◽  
Jeong Hoon Oh ◽  
Kevin B. Kim ◽  
Jeffrey E. Gershenwald ◽  
Shana L. Palla ◽  
...  

8536 Background: Comorbidities have been shown to adversely affect survival among patients with cancer. Currently, the most important prognostic factor in patients with melanoma is AJCC stage. However, little is known regarding the impact of comorbidities on melanoma prognosis. The aim of our study was to determine the impact of the severity of concurrent comorbidities on the overall survival of patients with high-risk and advanced melanoma (defined as AJCC stages IIc, III and IV). Methods: We conducted a retrospective cohort study of eligible adult melanoma patients available in the MelCore prospective database at MD Anderson Cancer Center (MDACC) from 01-2003 to 12-2006 who were diagnosed and completed staging within 3 months of presentation to MDACC. Patients with ocular melanoma were excluded. Demographic, AJCC staging, and survival data were collected. The Adult Comorbidity Evaluation-27 (ACE-27) was utilized to collect comorbidity information and grade its severity. A Cox proportional hazards model was used. Results: Of 444 patients that met enrollment criteria, 176 (39.6%) had grade 0 (no comorbidities), 222 (50%) had grade 1 or 2 (mild or moderate comorbidities) and 46 (10.4%) had grade 3 (severe comorbidities). The median age of the entire cohort was 56.4 years (19.7-98.9), 141 (32%) were female, and 406 (91.4%) were white. The median overall survival after presentation to MDACC was 5.0 years for the entire cohort. Adjusted hazard ratios are shown in the table. Comorbidity and AJCC Stage were significantly associated with survival. Age, gender and race did not have a significant impact on overall survival. Conclusions: In our study, the presence of comorbidities at presentation were independent predictors of decreased survival, even when adjusted for AJCC stage. Our findings suggest that comorbidity should be incorporated into prognostic models and therapeutic decision-making for patients with high-risk or advanced melanoma. [Table: see text]

2014 ◽  
Vol 32 (3_suppl) ◽  
pp. 303-303 ◽  
Author(s):  
Mairead Geraldine McNamara ◽  
Priya Aneja ◽  
Lisa W Le ◽  
Anne M Horgan ◽  
Elizabeth McKeever ◽  
...  

303 Background: BTCs include intrahepatic (IHC), hilar, distal bile duct (DBD), and gallbladder carcinoma (GBC). Statins, aspirin and metformin may have antineoplastic properties. The impact of their use on overall survival and the recurrence free survival of patients who had curative resection of BTC has not been evaluated. Methods: Baseline demographics and use of statins, aspirin or metformin at diagnosis were evaluated in 913 patients with BTC from 01/87 - 07/13 treated at Princess Margaret Cancer Center, Toronto. Their prognostic significance for recurrence free and overall survival was determined using a Cox proportional hazards model. Results: The median age at diagnosis for the entire cohort was 65.7 years (range 23.7-93.7). 795 patients had a performance status < 2 and 461 (50.5%) were male. The primary site was GBC in 310 (34%) patients, DBD in 212 (23%), IHC in 200 (22%) and hilar in 191 (21%). Curative surgical resection was performed in 355 (39%) patients. Among the entire cohort of 913 patients, 151 (16.5%) reported statin use at diagnosis. Atorvastatin was the statin used in 55% of patients. 146 (16%) reported aspirin use and 81 (9%) reported metformin use at diagnosis. Age (p=0.05, p<0.01), and stage (p<0.001, p<0.001) were prognostic on multivariable analysis for recurrence free and overall survival respectively. GBC (p=0.01), DBD (p<0.01) primary and performance status ≥ 2 (p < 0.0001) were also prognostic for overall survival. Recurrence free and overall survival among statin users and nonusers was similar (Hazard Ratio (HR) 1.07, 95% confidence interval (CI) 0.78-1.48, p=0.68) and (HR 0.84 (95% CI 0.67-1.05, p=0.12) respectively. Recurrence free and overall survival among aspirin users and nonusers was similar (HR 0.91, 95% CI 0.64-1.29, P=0.58) and (HR 0.98 (95% CI 0.80-1.22, P=0.88) respectively. Recurrence free and overall survival among metformin users and nonusers was also similar (HR 0.71, 95% CI 0.43-1.17, p=0.18) and (HR 0.81 (95% CI 0.60-1.08, p=0.14) respectively. Conclusions: In this large retrospective cohort of BTC patients, statin, aspirin or metformin use was not associated with improved recurrence free or overall survival.


2014 ◽  
Vol 32 (3_suppl) ◽  
pp. 528-528
Author(s):  
David Mitchell Marcus ◽  
Dana Nickleach ◽  
Bassel F. El-Rayes ◽  
Jerome Carl Landry

528 Background: The standard treatment for locally advanced rectal cancer is neoadjuvant chemoradiation followed by surgery, but many physicians question the benefit of multimodality therapy in patients with stage T3N0M0 disease. We aimed to determine the impact of radiation therapy (RT) on overall survival (OS) in this group of patients. Methods: We used the Surveillance, Epidemiology, and End Results database to identify patients undergoing surgery for T3N0M0 adenocarcinoma of the rectum from 2004 to 2010. The Kaplan-Meier method was used to compare OS for patients receiving RT vs. no RT, along with for pre-op vs. post-op RT among patients that received RT. Multivariable analysis (MVA) using a Cox proportional hazards model was performed to assess the association of RT with OS after adjusting for patient age, gender, race, tumor grade, carcinoembryonic antigen, type of surgery, and circumferential margin status. The analysis was repeated separately on patients that underwent total colectomy (TC) vs. sphincter-sparing surgery. Results: The cohort included 8,679 patients, including 4,705 who received RT and 3,974 who did not. Median age was 66 years. Five year OS was 76.5% in patients who received RT, compared to 60.0% in patients who did not receive RT (p <0.001). Five year OS was 76.9% for patients receiving pre-op RT vs. 75.7% in patients receiving post-op RT (p = 0.247). In patients undergoing TC, five year OS was 74.7% for patients receiving RT, compared to 47.5% in patients not receiving RT (p <0.001). In patients undergoing sphincter-sparing surgery, five year OS was 77.7% in patients receiving RT, compared to 62.9% in patients not receiving RT (p <0.001). Use of RT was significantly associated with OS on MVA, both in the entire cohort (HR 0.70 [95% CI 0.60-0.81]; p<0.001) and in subsets of patients undergoing TC (HR 0.55 [95% CI 0.38-0.79]; p=0.001) and sphincter-sparing surgery (HR 0.70 [95% CI 0.59-0.84]; p<0.001). Conclusions: The use of RT is associated with superior OS in patients undergoing surgery for T3N0M0 adenocarcinoma of the rectum. This benefit is demonstrated in both the pre-op and post-op settings and applies to patients undergoing both TC and sphincter-sparing surgery.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e16122-e16122
Author(s):  
Vanessa Wookey ◽  
Gabriella Bufalino ◽  
Gregory A. Vidal ◽  
Bradley G. Somer ◽  
Lee S. Schwartzberg ◽  
...  

e16122 Background: WCCRI, a comprehensive regional community oncology center in Memphis, Tennessee and the Mid-South region, serves a racially, geographically and socioeconomically diverse patient cohort. We sought to evaluate disparity of outcomes in survival by race and socioeconomic status, in addition to patient and tumor characteristics. Methods: All consecutive patients referred to and treated at WCCRI with colorectal adenocarcinoma from 2007-2013 were included. Individual chart review was performed to verify diagnosis, stage, and date and cause of death. Kaplan-Meier Overall Survival curves were generated for the entire cohort and by race, sex, tumor location and income derived from zip code. WCCRI survival data were compared to SEER data. Results: From 2007-2013, 1,176 patients were included in the analysis: 405 blacks, 757 whites, 14 others. Median age at diagnosis: Blacks 58 yrs, whites 61 yrs. Stage distribution at diagnosis: stage 1: 100, stage 2: 275, stage 3: 425, stage 4: 376. All stages combined, blacks trended towards shorter OS vs whites (5-year OS: 52.8% vs 58.3%; median survival 71.0 mos vs 98.6 mos; p= 0.095). Blacks presented at later stages (71.4% at stage 3 or 4 vs 66.3% for whites) but no statistically significant OS differences were seen when compared by stage. Patients at or below the median income of $39,590 for WCC had worse 5-year OS (51.6% vs. 61.1%; p= 0.006), as did patients without private insurance (5-year OS: uninsured: 48.0%, Medicare/Medicaid: 50.0%, private: 62.0%; p< 0.001). Adjusted for stage, 5-year OS was statistically significant for stage 4 (private: 18.0%, Medicare/Medicaid: 9.4%, uninsured: 8.3%; p= 0.020). A higher proportion of blacks were below the median income (69% vs 39%) but no statistically significant OS differences were seen when adjusted by race. Overall, cancer survival outcomes were similar to SEER results. Conclusions: At WCCRI, black patients with CRC presented at a later stage than whites, however, adjusted for stage, no significant racial difference in OS was found. Income and insurance status influenced survival outcomes. Overall, our results reveal racial and socioeconomic disparities in colorectal cancer in a diverse US population and further detailed multivariate data analyses are underway.


2020 ◽  
Vol 38 (4_suppl) ◽  
pp. 80-80
Author(s):  
Vanessa Wookey ◽  
Gabriella Bufalino ◽  
Gregory A. Vidal ◽  
Bradley G. Somer ◽  
Lee S. Schwartzberg ◽  
...  

80 Background: WCC, a comprehensive regional community oncology center in Memphis, Tennessee and the Mid-South region, serves a racially, geographically and socioeconomically diverse patient cohort. We sought to evaluate disparity of outcomes in survival by race and socioeconomic status, in addition to patient and tumor characteristics. Methods: All consecutive patients referred to and treated at WCC with colorectal adenocarcinoma from 2007-2013 were included. Individual chart review was performed to verify diagnosis, stage, and date and cause of death. Kaplan-Meier Overall Survival curves were generated for the entire cohort and by race, sex, tumor location and income derived from zip code. WCC survival data were compared to SEER data. Results: From 2007-2013, 1,176 patients were included in the analysis: 405 blacks, 757 whites, 14 others. Median age at diagnosis: Blacks 58 yrs, whites 61 yrs. Stage distribution at diagnosis: stage 1: 100, stage 2: 275, stage 3: 425, stage 4: 376. All stages combined, blacks trended towards shorter OS vs whites (5-year OS: 52.8% vs 58.3%; median survival 71.0 mos vs 98.6 mos; p= 0.095). Blacks presented at later stages (71.4% at stage 3 or 4 vs 66.3% for whites) but no statistically significant OS differences were seen when compared by stage. Patients at or below the median income of $39,590 for WCC had worse 5-year OS (51.6% vs. 61.1%; p= 0.006), as did patients without private insurance (5-year OS: uninsured: 48.0%, Medicare/Medicaid: 50.0%, private: 62.0%; p< 0.001). Adjusted for stage, 5-year OS was statistically significant for stage 4 (private: 18.0%, Medicare/Medicaid: 9.4%, uninsured: 8.3%; p= 0.020). A higher proportion of blacks were below the median income (69% vs 39%) but no statistically significant OS differences were seen when adjusted by race. Overall, cancer survival outcomes were similar to SEER results. Conclusions: At WCC, black patients with CRC presented at a later stage than whites, however, adjusted for stage, no significant racial difference in OS was found. Income and insurance status affected survival outcomes. Overall, our results reveal racial and socioeconomic disparities in colorectal cancer in a diverse US population.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3396-3396
Author(s):  
Victor H Jimenez Zepeda ◽  
Suzanne Trudel ◽  
Donna E. Reece ◽  
Christine I. Chen ◽  
Rodger E. Tiedemann ◽  
...  

Abstract Introduction The role of high-dose therapy (HDT) followed by autologous stem cell transplantation (ASCT) in the treatment of multiple myeloma (MM) continues to evolve in with the availability of effective novel agents. It has been shown that exposure to novel agents (NA) can improve the post-relapse survival (Venner CP, et al, 2011). However, the impact of early relapse (ER) after ASCT in the era of NA has not been fully defined. In this study, we aimed to assess the impact of ER after ASCT on overall survival (OS) for MM patients (pts) undergoing single ASCT who had received NA induction regimens Patient and Methods All consecutive pts with documented MM undergoing single ASCT at Princess Margaret Cancer Center from 01/02 to 09/12 who had novel induction therapy were evaluated. ER was defined as progression of myeloma within 12 months from the ASCT. The Cox proportional hazard model was used to perform multivariate analyses of possible prognostic variables for overall survival. A p value of <0.05 was considered statistically significant. Results The clinical and laboratory characteristics are of the 184 pts given induction therapy with NAs are listed in Table 1. The median age for this cohort of patients was 58 years (range 31-72). At day-100 post-ASCT, CR was achieved in 15.3% and VGPR in 57.1%. At the time of this analysis, 81% of pts are alive and 75 (40.7%) have already progressed. The median OS of the whole group was 93 months (95% CI, 70-116) (Fig.1a) and medianPFS was 25.43 months. Median time to relapse was 17.23 months (C I95%; 12-22). Early relapse was seen in 27/75 pts (36% of relapsed cases). Median OS was significantly shorter for the group of patients with ER (20 months, CI 95%; 15.56-24.51 versus 93 months, CI 95%; 75-111) (p=0.001) (Fig. 1b) Multivariate analysis showed the presence of ER as the major independent prognostic factor for OS; age > 60, B2M > 460 µmol/L, LDH > 350 IU/L, CRP > 20mg/L, albumin < 35g/L, and creatinine>200 µmol/L did not correlate with OS. In addition, FISH was available in only 78/184 cases. Only 1/27 pts in the ER group exhibited high-risk cytogenetics versus 0/48 in the non-ER group. In conclusion, not only the quality of response but also maintatenance of a deep response after ASCT, appear to be major prognostic variables in MM. Patients with ER post-ASCT should be biologically characterized to better understand the mechanisms of resistance associated with this particular entity and to develop new predictive models that can identify prospectively this myeloma subset. Also, patients with ER should be included in the definition of “high-risk” disease and priority candidates for the development of individualized therapeutic measures. Disclosures: Reece: Otsuka: Honoraria, Research Funding; BMS: Research Funding; Merck: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Millennium Pharmaceuticals: Research Funding; Celgene: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Onyx: Consultancy. Chen:Roche: Honoraria; Johnson & Johnson: Consultancy, Research Funding; Lundbeck: Consultancy; Celgene: Consultancy, Research Funding; GlaxoSmithKline: Research Funding. Tiedemann:Celgene: Honoraria; Janssen: Honoraria. Kukreti:Millennium Pharmaceuticals: Research Funding; Onyx: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1604-1604 ◽  
Author(s):  
Vikas Gupta ◽  
Claire N. Harrison ◽  
Elizabeth O. Hexner ◽  
Haifa Kathrin Al-Ali ◽  
Lynda Foltz ◽  
...  

Abstract Introduction: Anemia is a sign of disease progression and a key prognostic factor in myelofibrosis (MF). In the phase 3 COMFORT studies, ruxolitinib, a Janus kinase (JAK) 1/JAK2 inhibitor, demonstrated improved overall survival compared with placebo and best available therapy in patients with intermediate 2 or high-risk MF (per the International Prognostic Scoring System [IPSS]). Ruxolitinib treatment was associated with dose-dependent increases in cytopenias that occurred mostly in the first 12 weeks of therapy and were manageable with dose adjustments and red blood cell (RBC) transfusions. The objective of this analysis was to evaluate the differential impact of disease-related anemia (anemia at baseline) and anemia occurring after initiation of ruxolitinib therapy on overall survival (OS) in patients with MF. Methods: Data from the COMFORT-I and COMFORT-II 3-year data cuts were pooled for this exploratory analysis for patients who survived at least 12 weeks after day 1. Baseline anemia was defined as hemoglobin (Hgb) < 10 g/dL at baseline or receipt of RBC transfusions within 12 weeks prior to day 1. Postbaseline anemia was defined as new or worsening anemia within the first 12 weeks after initiation of ruxolitinib therapy that was grade ≥ 2 (per Common Terminology Criteria for Adverse Events v3.0) and a higher grade than at baseline; this time frame captures 85% of these events during ruxolitinib treatment. Patients were assessed for OS using a cox model stratified by study and baseline anemia status to evaluate the impact of anemia occurring after ruxolitinib initiation on OS. Results: Among patients treated with ruxolitinib (n = 296), 162 patients were not anemic at baseline and 134 patients were anemic at baseline. Of the 162 patients who were not anemic at baseline, 99 had postbaseline anemia; of the 134 with anemia at baseline, 93 also had postbaseline anemia. Consistent with published prognostic models, anemia at study baseline was prognostic of decreased survival in this study (Figure 1). A higher proportion of those who were anemic at baseline had PMF (58% vs 40%) and high-risk disease per the IPSS (69% vs 40%) vs those who were not anemic at baseline. As expected, mean platelet count (308.3 × 109/L vs 280.5 × 109/L), Hgb (12.2g/dL vs 9.1 g/dL), and leukocyte count (24.2 × 109/L vs 17.0 × 109/L) were higher among patients who were not anemic at baseline vs those who were. Within the subgroups of patients with and without baseline anemia, postbaseline anemia did not impact OS during ruxolitinib therapy (Figure 1) (overall hazard ratio for no postbaseline anemia vs postbaseline anemia = 1.030; 95% CI: 0.615-1.725). Patients treated with ruxolitinib had improved OS compared to control patients, both among those with anemia and without anemia at baseline (Figure 1) (overall hazard ratio for ruxolitinib vs control = 0.644; 95% CI: 0.459-0.903; P = .0102). OS probability at 3 years for ruxolitinib vs control was 0.66 vs 0.57 among patients with anemia at baseline, and 0.87 vs 0.66 among patients without anemia at baseline. Conclusions: Consistent with validated prognostic models, baseline anemia was associated with decreased OS in the COMFORT studies. Treatment with ruxolitinib improved OS as compared with the control arm regardless of baseline anemia status. Notably, postbaseline anemia that occurred on ruxolitinib therapy did not impact OS and was manageable with dose adjustments and RBC transfusions. We conclude that early onset ruxolitinib-related anemia does not have the same deleterious effect as disease-related anemia. Figure 1. Overall survival based on baseline and postbaseline anemia status and treatment received in the COMFORT studies. Rux, ruxolitinib; TI, therapy initiation. Figure 1. Overall survival based on baseline and postbaseline anemia status and treatment received in the COMFORT studies. Rux, ruxolitinib; TI, therapy initiation. Disclosures Gupta: Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Incyte: Honoraria, Research Funding. Harrison:Gilead: Honoraria; Shire: Speakers Bureau; Sanofi: Honoraria, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau; CTI Biopharma: Consultancy, Honoraria, Speakers Bureau. Al-Ali:Celgene: Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding. Foltz:Promedior: Research Funding; Novartis: Honoraria, Research Funding; Gilead: Research Funding. Montgomery:Incyte: Employment. Peng:Incyte Corporation: Employment, Equity Ownership. Gopalakrishna:Novartis Pharma AG: Employment. Verstovsek:Incyte Corporation: Research Funding; Astrazeneca: Research Funding; Lilly Oncology: Research Funding; Roche: Research Funding; Geron: Research Funding; NS Pharma: Research Funding; Bristol Myers Squibb: Research Funding; Gilead: Research Funding; Celgene: Research Funding; Seattle Genetics: Research Funding; Promedior: Research Funding; CTI BioPharma Corp.(formerly Cell Therapeutics, Inc.): Research Funding; Galena BioPharma: Research Funding; Pfizer: Research Funding.


2019 ◽  
Vol 4 ◽  
pp. 71 ◽  
Author(s):  
Priti Gupta ◽  
David Prieto-Merino ◽  
Vamadevan S. Ajay ◽  
Kalpana Singh ◽  
Ambuj Roy ◽  
...  

Introduction: Cardiovascular diseases (CVDs) are the leading cause of death in India. The CVD risk approach is a cost-effective way to identify those at high risk, especially in a low resource setting. As there is no validated prognostic model for an Indian urban population, we have re-calibrated the original Framingham model using data from two urban Indian studies. Methods: We have estimated three risk score equations using three different models. The first model was based on Framingham original model; the second and third are the recalibrated models using risk factor prevalence from CARRS (Centre for cArdiometabolic Risk Reduction in South-Asia) and ICMR (Indian Council of Medical Research) studies, and estimated survival from WHO 2012 data for India. We applied these three risk scores to the CARRS and ICMR participants and estimated the proportion of those at high-risk (>30% 10 years CVD risk) who would be eligible to receive preventive treatment such as statins. Results: In the CARRS study, the proportion of men with 10 years CVD risk > 30% (and therefore eligible for statin treatment) was 13.3%, 21%, and 13.6% using Framingham, CARRS and ICMR risk models, respectively. The corresponding proportions of women were 3.5%, 16.4%, and 11.6%. In the ICMR study the corresponding proportions of men were 16.3%, 24.2%, and 16.5% and for women, these were 5.6%, 20.5%, and 15.3%. Conclusion: Although the recalibrated model based on local population can improve the validity of CVD risk scores our study exemplifies the variation between recalibrated models using different data from the same country. Considering the growing burden of cardiovascular diseases in India, and the impact that the risk approach has on influencing cardiovascular prevention treatment, such as statins, it is essential to develop high quality and well powered local cohorts (with outcome data) to develop local prognostic models.


2020 ◽  
Author(s):  
Lijie Jiang ◽  
Tengjiao Lin ◽  
Yu Zhang ◽  
Wenxiang Gao ◽  
Jie Deng ◽  
...  

Abstract Background Increasing evidence indicates that the pathology and the modified Kadish system have some influence on the prognosis of esthesioneuroblastoma (ENB). However, an accurate system to combine pathology with a modified Kadish system has not been established. Methods This study aimed to set up and evaluate a model to predict overall survival (OS) accurately in ENB, including clinical characteristics, treatment and pathological variables. We screened the information of patients with ENB between January 1, 1976, and December 30, 2016 from the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) program as a training cohort. The validation cohort consisted of patients with ENB at Sun Yat-sen University Cancer Center and The First Affiliated Hospital of Sun Yat-sen University in the same period, and 87 patients were identified. The Pearson’s chi-squared test was used to assess significance of clinicopathological and demographic characteristics. We used the Cox proportional hazards model to examine univariate and multivariate analyses. The model coefficients were used to calculate the Hazard ratios (HR) with 95% confidence intervals (CI). Prognostic factors with a p- value < 0.05 in multivariate analysis were included in the nomogram. The concordance index (c-index) and calibration curve were used to evaluate the predictive power of the nomogram. Results The c-index of training cohort and validation cohort are 0.737 (95% CI, 0.709 to 0.765) and 0.791 (95% CI, 0.767 to 0.815) respectively. The calibration curves revealed a good agreement between the nomogram prediction and actual observation regarding the probability of 3-year and 5-year survival. We used a nomogram to calculate the 3-year and 5-year growth probability and stratified patients into three risk groups. Conclusions The nomogram provided the risk group information and identified mortality risk and can serve as a reference for designing a reasonable follow-up plan.


2021 ◽  
Vol 8 (2) ◽  
pp. 27-33
Author(s):  
Jiping Zeng ◽  
Ken Batai ◽  
Benjamin Lee

In this study, we aimed to evaluate the impact of surgical wait time (SWT) on outcomes of patients with renal cell carcinoma (RCC), and to investigate risk factors associated with prolonged SWT. Using the National Cancer Database, we retrospectively reviewed the records of patients with pT3 RCC treated with radical or partial nephrectomy between 2004 and 2014. The cohort was divided based on SWT. The primary out-come was 5-year overall survival (OS). Logistic regression analysis was used to investigate the risk factors associated with delayed surgery. Cox proportional hazards models were fitted to assess relations between SWT and 5-year OS after adjusting for confounding factors. A total of 22,653 patients were included in the analysis. Patients with SWT > 10 weeks had higher occurrence of upstaging. Using logistic regression, we found that female patients, African-American or Spanish origin patients, treatment in academic or integrated network cancer center, lack of insurance, median household income of <$38,000, and the Charlson–Deyo score of ≥1 were more likely to have prolonged SWT. SWT > 10 weeks was associated with decreased 5-year OS (hazard ratio [HR], 1.24; 95% confidence interval [CI], 1.15–1.33). This risk was not markedly attenuated after adjusting for confounding variables, including age, gender, race, insurance status, Charlson–Deyo score, tumor size, and surgical margin status (adjusted HR, 1.13; 95% CI, 1.04–1.24). In conclusion, the vast majority of patients underwent surgery within 10 weeks. There is a statistically significant trend of increasing SWT over the study period. SWT > 10 weeks is associated with decreased 5-year OS.


2021 ◽  
Vol 31 (7) ◽  
pp. 1075-1079
Author(s):  
Günter Emons ◽  
Jae-Weon Kim ◽  
Karin Weide ◽  
Nikolaus de Gregorio ◽  
Pauline Wimberger ◽  
...  

BackgroundThe impact of comprehensive pelvic and para-aortic lymphadenectomy on survival in patients with stage I or II endometrial cancer with a high risk of recurrence is not reliably documented. The side effects of this procedure, including lymphedema and lymph cysts, are evident.Primary ObjectiveEvaluation of the effect of comprehensive pelvic and para-aortic lymphadenectomy in the absence of bulky nodes on 5 year overall survival of patients with endometrial cancer (International Federation of Gynecology and Obstetrics (FIGO) stages I and II) and a high risk of recurrence.Study HypothesisComprehensive pelvic and para-aortic lymphadenectomy will increase 5 year overall survival from 75% (no lymphadenectomy) to 83%, corresponding to a hazard ratio of 0.65.Trial DesignOpen label, randomized, controlled trial. In arm A, a total hysterectomy plus bilateral salpingo-oophorectomy is performed. In arm B, in addition, a systematic pelvic and para-aortic lymphadenectomy up to the level of the left renal vein is performed. For all patients, vaginal brachytherapy and adjuvant chemotherapy (carboplatin/paclitaxel) are recommended.Major Inclusion CriteriaPatients with histologically confirmed endometrial cancer stages pT1b–pT2, all histological subtypes, and pT1a endometrioid G3, serous, clear cell, or carcinosarcomas can be included when bulky nodes are absent. When hysterectomy has already been performed (eg, for presumed low risk endometrial cancer), study participation is also possible.Exclusion CriteriaPatients with pT1a, G1 or 2 of type 1 histology or uterine sarcomas (except for carcinosarcomas), endometrial cancers of FIGO stage III or IV (except for microscopic lymph node metastases) or visual extrauterine disease.Primary EndpointOverall survival calculated from the date of randomization until death.Sample Size640 patients will be enrolled in the study.Estimated Dates for Completing Accrual and Presenting ResultsAt present, 252 patients have been recruited. Based on this, accrual should be completed in 2025. Results should be presented in 2031.Trial RegistrationNCT03438474.


Sign in / Sign up

Export Citation Format

Share Document