Pub Date : 2024-11-06DOI: 10.1007/s12094-024-03768-y
Jiao Jiang, Jieyu Peng, Shu Huang, Xiaomin Shi, Bei Luo, Jia Xu, Wei Zhang, Lei Shi, Muhan Lü, Xiaowei Tang
Background and aims: Primary digestive system lymphoma (PDSL) is an important entity of extranodal lymphoma, yet updated epidemiologic and survival data are lacking.
Methods: Patients diagnosed with PDSL between 1975 and 2020 were identified from the Surveillance, Epidemiology, and End Results database. Kaplan-Meier analysis estimated survival outcomes. Multivariable Cox regression identified independent risk factors, and nomograms were developed to predict 1-, 3-, and 5-year overall survival (OS) and cancer-specific survival (CSS).
Results: A total of 30,568 patients with PDSL were identified, with 57.9% being male and 80.4% white. The most frequent tumor site was the stomach (48.7%) and diffuse large B-cell lymphoma (DLBCL) was the predominant histologic subtype (45.0%). The overall incidence from 2016 to 2020 was 11.11 per 1,000,000 persons, with a decrease observed in lymphoma rates for the stomach, small intestine, large intestine, and pancreas. Long-term trends showed an initial rise in PDSL incidence, followed by a decline since the 1990s. The median OS across all patients was 103 months, with appendiceal lymphoma showing the highest median OS of 253 months. Factors including diagnosis year, age, sex, race, primary tumor site, histologic subtype, stage, and treatment modalities were significantly associated with OS and CSS. Nomograms achieved C-indices of 0.720 for OS and 0.723 for CSS in the training cohort.
Conclusion: The incidence of PDSL initially increased but has recently declined. Survival for all PDSL patients has improved over time. Nomograms to predict survival for patients with DLBCL exhibited good predictive and discriminating abilities.
{"title":"Epidemiologic trends and survival outcomes in patients with primary digestive system lymphoma in the United States.","authors":"Jiao Jiang, Jieyu Peng, Shu Huang, Xiaomin Shi, Bei Luo, Jia Xu, Wei Zhang, Lei Shi, Muhan Lü, Xiaowei Tang","doi":"10.1007/s12094-024-03768-y","DOIUrl":"https://doi.org/10.1007/s12094-024-03768-y","url":null,"abstract":"<p><strong>Background and aims: </strong>Primary digestive system lymphoma (PDSL) is an important entity of extranodal lymphoma, yet updated epidemiologic and survival data are lacking.</p><p><strong>Methods: </strong>Patients diagnosed with PDSL between 1975 and 2020 were identified from the Surveillance, Epidemiology, and End Results database. Kaplan-Meier analysis estimated survival outcomes. Multivariable Cox regression identified independent risk factors, and nomograms were developed to predict 1-, 3-, and 5-year overall survival (OS) and cancer-specific survival (CSS).</p><p><strong>Results: </strong>A total of 30,568 patients with PDSL were identified, with 57.9% being male and 80.4% white. The most frequent tumor site was the stomach (48.7%) and diffuse large B-cell lymphoma (DLBCL) was the predominant histologic subtype (45.0%). The overall incidence from 2016 to 2020 was 11.11 per 1,000,000 persons, with a decrease observed in lymphoma rates for the stomach, small intestine, large intestine, and pancreas. Long-term trends showed an initial rise in PDSL incidence, followed by a decline since the 1990s. The median OS across all patients was 103 months, with appendiceal lymphoma showing the highest median OS of 253 months. Factors including diagnosis year, age, sex, race, primary tumor site, histologic subtype, stage, and treatment modalities were significantly associated with OS and CSS. Nomograms achieved C-indices of 0.720 for OS and 0.723 for CSS in the training cohort.</p><p><strong>Conclusion: </strong>The incidence of PDSL initially increased but has recently declined. Survival for all PDSL patients has improved over time. Nomograms to predict survival for patients with DLBCL exhibited good predictive and discriminating abilities.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142584846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-05DOI: 10.1007/s12094-024-03776-y
Guillermo Suay, Paloma Martín-Martorell, Francisco Aparisi, María Arnal, María Guirado, Aitor Azkárate, Javier Garde-Noguera, José David Cumplido-Burón, Amelia Insa, José Francisco González-Muñoz, Sarai Palanca, María Díaz, Alfredo Sánchez-Hernández, Óscar Juan-Vidal
Objectives: EGFR exon 20 insertion (EGFRex20ins) mutations are found in up to 4% of all patients with non-small cell lung cancer (NSCLC). These patients are often insensitive to EGFR-tyrosine kinase inhibitors (TKIs) and have worse prognosis than patients with more common EGFR mutations. In this multicenter, retrospective, real-world study, we sought to determine whether the administration of recently approved treatments that specifically target EGFRex20ins mutations could significantly improve outcomes in this patient population.
Materials and methods: We evaluated the clinical features of 41 patients diagnosed with NSCLC and EGFRex20ins mutations, their evolution, and response to treatments received across 7 hospitals in the Valencian Community, Spain, between 31st December 2012 and 31st December 2022.
Results: 32 patients (72%) developed metastatic disease, and 29 (71%) of them received oncological treatment. We found that administering a targeted therapy against EGFRex20ins mutations (amivantamab, mobocertinib and/or sunvozertinib) at some point during the course of treatment, significantly increased the median OS of metastatic patients from 8 months (95% CI 0-21.7) to 30 months (95% CI 11.1-48.8; Hazard ratio = 0.297, p = 0.02).
Conclusion: Our findings contribute to the evolving standard of care for this specific population and highlight the clinical benefits of targeted cancer therapies.
{"title":"A real‑world study of clinical characteristics, treatment sequence and outcomes of patients with non-small cell lung cancer and EGFR exon 20 insertion mutations.","authors":"Guillermo Suay, Paloma Martín-Martorell, Francisco Aparisi, María Arnal, María Guirado, Aitor Azkárate, Javier Garde-Noguera, José David Cumplido-Burón, Amelia Insa, José Francisco González-Muñoz, Sarai Palanca, María Díaz, Alfredo Sánchez-Hernández, Óscar Juan-Vidal","doi":"10.1007/s12094-024-03776-y","DOIUrl":"https://doi.org/10.1007/s12094-024-03776-y","url":null,"abstract":"<p><strong>Objectives: </strong>EGFR exon 20 insertion (EGFRex20ins) mutations are found in up to 4% of all patients with non-small cell lung cancer (NSCLC). These patients are often insensitive to EGFR-tyrosine kinase inhibitors (TKIs) and have worse prognosis than patients with more common EGFR mutations. In this multicenter, retrospective, real-world study, we sought to determine whether the administration of recently approved treatments that specifically target EGFRex20ins mutations could significantly improve outcomes in this patient population.</p><p><strong>Materials and methods: </strong>We evaluated the clinical features of 41 patients diagnosed with NSCLC and EGFRex20ins mutations, their evolution, and response to treatments received across 7 hospitals in the Valencian Community, Spain, between 31st December 2012 and 31st December 2022.</p><p><strong>Results: </strong>32 patients (72%) developed metastatic disease, and 29 (71%) of them received oncological treatment. We found that administering a targeted therapy against EGFRex20ins mutations (amivantamab, mobocertinib and/or sunvozertinib) at some point during the course of treatment, significantly increased the median OS of metastatic patients from 8 months (95% CI 0-21.7) to 30 months (95% CI 11.1-48.8; Hazard ratio = 0.297, p = 0.02).</p><p><strong>Conclusion: </strong>Our findings contribute to the evolving standard of care for this specific population and highlight the clinical benefits of targeted cancer therapies.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142584830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-05DOI: 10.1007/s12094-024-03772-2
Francisco Cezar Aquino de Moraes, Vitor Kendi Tsuchiya Sano, Clara Rocha Dantas, Nathália Hoffmeister, Francinny Alves Kelly, Rommel Mario Rodríguez Burbano
Background: Degarelix is a third-generation GnRH receptor antagonist approved for the treatment of prostate cancer, however, the decision to use a GnRH agonist or an antagonist depends on several factors. We aimed to perform a meta-analysis comparing the cardiovascular disease risk between degarelix and gonadotropin-releasing hormone agonists in patients with all stages of prostate cancer.
Methods: Databases were searched for randomized control trials (RCTs) and observational studies that compared the risk of cardiovascular disease between degarelix and GnRH agonists in patients with prostate cancer. We computed for binary endpoints risk ratio (RR) or hazard ratio (HR) with 95% confidence intervals (CI) which were analyzed using a random-effects model.
Results: A total of 15 studies were included with 123,969 patients and follow-up ranging from 3 to 13 months. Degarelix was associated with a significantly lower incidence of major adverse cardiovascular events (RR 0.59; 95% CI 0.41-0.84; p = 0.003; I2 = 84%). Incidence of stroke (RR 0.89; 95% CI 0.56-1.42; p = 0.62; I2 = 0%), all-cause mortality (RR 0.64; 95% CI 0.37-1.13; p = 0.12; I2 = 41%), hypertension (RR 0.71; 95% CI 0.48, 1.04; p = 0.08; I2 = 0%), myocardial infarction (HR 1.04; 95% CI 0·59-1·84; p = 0·86; I2 = 66%), heart failure (HR 0.79; 95% CI 0.38-1.62; p = 0.52; I2 = 79%) and arrhythmia (RR 0.63; 95% CI 0.28-1.41; p = 0.86; I2 = 37%), did not reach a statistically significant difference between groups.
Conclusion: In patients with prostate cancer, degarelix is associated with a significantly lower incidence of major adverse cardiovascular events.
背景:地加瑞克是第三代促性腺激素释放激素受体拮抗剂,已被批准用于治疗前列腺癌,然而,使用促性腺激素释放激素激动剂还是拮抗剂取决于多种因素。我们的目的是进行一项荟萃分析,比较地加瑞克和促性腺激素释放激素激动剂对各期前列腺癌患者的心血管疾病风险:我们在数据库中搜索了比较地加瑞克和促性腺激素释放激素激动剂在前列腺癌患者中心血管疾病风险的随机对照试验(RCT)和观察性研究。我们计算了二元终点的风险比(RR)或危险比(HR)及95%置信区间(CI),并采用随机效应模型进行分析:结果:共纳入15项研究,患者人数为123969人,随访时间为3至13个月。地加瑞克可显著降低主要不良心血管事件的发生率(RR 0.59;95% CI 0.41-0.84;P = 0.003;I2 = 84%)。中风(RR 0.89;95% CI 0.56-1.42;p = 0.62;I2 = 0%)、全因死亡率(RR 0.64;95% CI 0.37-1.13;p = 0.12;I2 = 41%)、高血压(RR 0.71;95% CI 0.48,1.04;p = 0.08;I2 = 0%)、心肌梗死(HR 1.04;95% CI 0-59-1-84;p = 0-86;I2 = 66%)、心力衰竭(HR 0.79;95% CI 0.38-1.62;p = 0.52;I2 = 79%)和心律失常(RR 0.63;95% CI 0.28-1.41;p = 0.86;I2 = 37%),组间差异无统计学意义:结论:在前列腺癌患者中,地加瑞克可显著降低主要不良心血管事件的发生率。
{"title":"A systematic review and meta-analysis of cardiovascular disease risk with degarelix and GnRH agonists in prostate cancer.","authors":"Francisco Cezar Aquino de Moraes, Vitor Kendi Tsuchiya Sano, Clara Rocha Dantas, Nathália Hoffmeister, Francinny Alves Kelly, Rommel Mario Rodríguez Burbano","doi":"10.1007/s12094-024-03772-2","DOIUrl":"https://doi.org/10.1007/s12094-024-03772-2","url":null,"abstract":"<p><strong>Background: </strong>Degarelix is a third-generation GnRH receptor antagonist approved for the treatment of prostate cancer, however, the decision to use a GnRH agonist or an antagonist depends on several factors. We aimed to perform a meta-analysis comparing the cardiovascular disease risk between degarelix and gonadotropin-releasing hormone agonists in patients with all stages of prostate cancer.</p><p><strong>Methods: </strong>Databases were searched for randomized control trials (RCTs) and observational studies that compared the risk of cardiovascular disease between degarelix and GnRH agonists in patients with prostate cancer. We computed for binary endpoints risk ratio (RR) or hazard ratio (HR) with 95% confidence intervals (CI) which were analyzed using a random-effects model.</p><p><strong>Results: </strong>A total of 15 studies were included with 123,969 patients and follow-up ranging from 3 to 13 months. Degarelix was associated with a significantly lower incidence of major adverse cardiovascular events (RR 0.59; 95% CI 0.41-0.84; p = 0.003; I<sup>2</sup> = 84%). Incidence of stroke (RR 0.89; 95% CI 0.56-1.42; p = 0.62; I<sup>2</sup> = 0%), all-cause mortality (RR 0.64; 95% CI 0.37-1.13; p = 0.12; I<sup>2</sup> = 41%), hypertension (RR 0.71; 95% CI 0.48, 1.04; p = 0.08; I<sup>2</sup> = 0%), myocardial infarction (HR 1.04; 95% CI 0·59-1·84; p = 0·86; I<sup>2</sup> = 66%), heart failure (HR 0.79; 95% CI 0.38-1.62; p = 0.52; I2 = 79%) and arrhythmia (RR 0.63; 95% CI 0.28-1.41; p = 0.86; I<sup>2</sup> = 37%), did not reach a statistically significant difference between groups.</p><p><strong>Conclusion: </strong>In patients with prostate cancer, degarelix is associated with a significantly lower incidence of major adverse cardiovascular events.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142584831","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: This study aimed to investigate the impact of oligometastasis and the M descriptor on survival in small cell lung cancer (SCLC).
Methods: This multicenter, retrospective study included patients with newly diagnosed extensive-stage SCLC(ES-SCLC) from 2010 to 2020. Subgroups: Group 1: single metastasis in a single organ, Group 2: 2-5 metastases in a single organ, Group 3: 6 or more metastases in a single organ, and Group 4: metastases in two or more organs. This classification was based on the 9th Staging-M descriptor. Three-year progression-free survival (PFS) and overall survival (OS) analyses were conducted.
Results: The mean age of the 439 patients was 62 ± 10 years, and 89.5% of them were male. The mean PFS for Groups 1, 2, 3, 4 was 10.7 months (95% CI 8.9-12.5), 7.5 months (95% CI 5.6-9.4), 4.3 months (95% CI 2.9-5.7), and 5.4 months (95% CI 4.7-6.1), respectively. PFS in Group 2 was significantly higher. The mean OS for Groups 1, 2, 3, 4 was 13.3 months (95% CI 11.2-15.3), 9.5 months (95% CI 7.1-11.9), 7.1 months (95% CI 4.5-9.7), and 6.9 months (95% CI 6.0-7.9), respectively. OS in Group 1 was significantly higher. OS and PFS in the M1b group were significantly higher than in the M1c1 and M1c2 groups (p < 0.05) with no statistical difference between the M1c1 and M1c2 groups.
Conclusion: There is no significant difference in survival between the M1c1 and M1c2 groups. In ES-SCLC, the number of metastases may be a more predictive factor for prognosis than the number of metastatic organs.
目的:本研究旨在探讨寡转移和M描述因子对小细胞肺癌(SCLC)生存期的影响:这项多中心回顾性研究纳入了2010年至2020年新诊断的广泛期SCLC(ES-SCLC)患者。分组:第1组:单个器官有转移;第2组:单个器官有2-5个转移;第3组:单个器官有6个或更多转移;第4组:两个或更多器官有转移。这一分类是根据第 9 次分期-M 描述法进行的。进行了三年无进展生存期(PFS)和总生存期(OS)分析:439名患者的平均年龄为(62±10)岁,89.5%为男性。第1、2、3、4组的平均生存期分别为10.7个月(95% CI 8.9-12.5)、7.5个月(95% CI 5.6-9.4)、4.3个月(95% CI 2.9-5.7)和5.4个月(95% CI 4.7-6.1)。第 2 组的 PFS 明显更高。第1、2、3、4组的平均OS分别为13.3个月(95% CI 11.2-15.3)、9.5个月(95% CI 7.1-11.9)、7.1个月(95% CI 4.5-9.7)和6.9个月(95% CI 6.0-7.9)。第一组的 OS 明显更高。M1b组的OS和PFS明显高于M1c1组和M1c2组(P 结论:M1b组的OS和PFS明显高于M1c1组和M1c2组:M1c1组和M1c2组的生存率无明显差异。在 ES-SCLC 中,转移灶的数量可能比转移器官的数量更能预测预后。
{"title":"Different approach to M descriptor for future staging of oligometastatic disease in SCLC: A cross-sectional survival analysis.","authors":"Melahat Uzel Şener, Pınar Akın Kabalak, Suna Kavurgacı, Nilgün Yılmaz Demirci, Derya Kızılgöz, Fazlı Yanık, Sinem Ermin, Yasemin Söyler, Yekta Altemur Karamustafaoğlu, Demet Türkay Pakna, Ahmet Dumanlı, Ülkü Yılmaz","doi":"10.1007/s12094-024-03778-w","DOIUrl":"https://doi.org/10.1007/s12094-024-03778-w","url":null,"abstract":"<p><strong>Purpose: </strong>This study aimed to investigate the impact of oligometastasis and the M descriptor on survival in small cell lung cancer (SCLC).</p><p><strong>Methods: </strong>This multicenter, retrospective study included patients with newly diagnosed extensive-stage SCLC(ES-SCLC) from 2010 to 2020. Subgroups: Group 1: single metastasis in a single organ, Group 2: 2-5 metastases in a single organ, Group 3: 6 or more metastases in a single organ, and Group 4: metastases in two or more organs. This classification was based on the 9th Staging-M descriptor. Three-year progression-free survival (PFS) and overall survival (OS) analyses were conducted.</p><p><strong>Results: </strong>The mean age of the 439 patients was 62 ± 10 years, and 89.5% of them were male. The mean PFS for Groups 1, 2, 3, 4 was 10.7 months (95% CI 8.9-12.5), 7.5 months (95% CI 5.6-9.4), 4.3 months (95% CI 2.9-5.7), and 5.4 months (95% CI 4.7-6.1), respectively. PFS in Group 2 was significantly higher. The mean OS for Groups 1, 2, 3, 4 was 13.3 months (95% CI 11.2-15.3), 9.5 months (95% CI 7.1-11.9), 7.1 months (95% CI 4.5-9.7), and 6.9 months (95% CI 6.0-7.9), respectively. OS in Group 1 was significantly higher. OS and PFS in the M1b group were significantly higher than in the M1c1 and M1c2 groups (p < 0.05) with no statistical difference between the M1c1 and M1c2 groups.</p><p><strong>Conclusion: </strong>There is no significant difference in survival between the M1c1 and M1c2 groups. In ES-SCLC, the number of metastases may be a more predictive factor for prognosis than the number of metastatic organs.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142577121","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-05DOI: 10.1007/s12094-024-03775-z
Christina Jensen, Simone Maarup, Hans Skovgaard Poulsen, Benedikte Hasselbalch, Morten Karsdal, Inge Marie Svane, Ulrik Lassen, Nicholas Willumsen
Purpose: There is an unmet need for new treatment options and biomarkers for patients with glioblastoma (GBM). Here we investigated three non-invasive biomarkers: type VI collagen degraded by granzyme B (C4G) and matrix metalloproteases (C4M), respectively, and ADAM10-degraded Tau (Tau-A).
Methods: Biomarker levels in pre- and on-treatment serum samples from patients with recurrent GBM (n = 39) treated with nivolumab and bevacizumab (NCT03890952) were compared to healthy levels (n = 22) and associated with overall survival (OS) outcome (median cutpoint). Longitudinal changes in biomarkers were investigated by a Mixed-effects analysis.
Results: Tau-A (p < 0.0001) and C4G (p = 0.005), but not C4M (p = 0.106), were increased in patients. High Tau-A and C4G associated with improved OS (Tau-A: HR = 0.41, 95%CI = 0.20-0.86, C4G: HR = 0.47, 95%CI = 0.24-0.94). Only C4G increased with treatment (p = 0.024-0.005).
Conclusions: Tau-A and C4G are elevated in serum from patients with recurrent GBM and prognostic for OS. If validated, these biomarkers could be applied to clinical trials.
目的:胶质母细胞瘤(GBM)患者对新的治疗方案和生物标志物的需求尚未得到满足。在此,我们研究了三种非侵入性生物标志物:分别由颗粒酶 B(C4G)和基质金属蛋白酶(C4M)降解的 VI 型胶原,以及 ADAM10 降解的 Tau(Tau-A):将接受尼伐单抗和贝伐单抗(NCT03890952)治疗的复发性GBM患者(39人)治疗前和治疗中血清样本中的生物标志物水平与健康人(22人)的水平进行比较,并将其与总生存期(OS)结果(中位数切点)相关联。通过混合效应分析研究了生物标志物的纵向变化:结果:Tau-A(p复发性 GBM 患者血清中的 Tau-A 和 C4G 升高,是 OS 的预后指标。如果得到验证,这些生物标记物可应用于临床试验。
{"title":"Degradation fragments of Tau and type IV collagen as serum biomarkers in patients with recurrent glioblastoma treated with nivolumab and bevacizumab.","authors":"Christina Jensen, Simone Maarup, Hans Skovgaard Poulsen, Benedikte Hasselbalch, Morten Karsdal, Inge Marie Svane, Ulrik Lassen, Nicholas Willumsen","doi":"10.1007/s12094-024-03775-z","DOIUrl":"https://doi.org/10.1007/s12094-024-03775-z","url":null,"abstract":"<p><strong>Purpose: </strong>There is an unmet need for new treatment options and biomarkers for patients with glioblastoma (GBM). Here we investigated three non-invasive biomarkers: type VI collagen degraded by granzyme B (C4G) and matrix metalloproteases (C4M), respectively, and ADAM10-degraded Tau (Tau-A).</p><p><strong>Methods: </strong>Biomarker levels in pre- and on-treatment serum samples from patients with recurrent GBM (n = 39) treated with nivolumab and bevacizumab (NCT03890952) were compared to healthy levels (n = 22) and associated with overall survival (OS) outcome (median cutpoint). Longitudinal changes in biomarkers were investigated by a Mixed-effects analysis.</p><p><strong>Results: </strong>Tau-A (p < 0.0001) and C4G (p = 0.005), but not C4M (p = 0.106), were increased in patients. High Tau-A and C4G associated with improved OS (Tau-A: HR = 0.41, 95%CI = 0.20-0.86, C4G: HR = 0.47, 95%CI = 0.24-0.94). Only C4G increased with treatment (p = 0.024-0.005).</p><p><strong>Conclusions: </strong>Tau-A and C4G are elevated in serum from patients with recurrent GBM and prognostic for OS. If validated, these biomarkers could be applied to clinical trials.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142584844","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-04DOI: 10.1007/s12094-024-03777-x
Caglar Berkel
Background: Breast cancer has distinct causes and prognoses in patients with premenopausal and postmenopausal status. The expression status of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are analyzed by immunohistochemistry (IHC) to classify molecular subtypes of breast cancer among which large differences in prognosis exist.
Methods: The mRNA expression of ESR1 (encoding ERα), ESR2 (encoding ERβ), PGR (encoding PR), and ERBB2 (encoding HER2) was analyzed based on menopausal status (pre- vs post-menopausal) in tumors from breast cancer patients with different receptor status, in R programming environment, using transcriptomics data from TCGA-BRCA project.
Results: In ER-positive or PR-positive or HER2-negative breast tumors, ESR1 transcript levels were found to be higher in tumors from postmenopausal women than those from premenopausal women; in contrast, ESR2 transcript levels were lower in tumors from postmenopausal women than those from premenopausal women. Furthermore, PGR mRNA expression was lower in breast tumors from postmenopausal women than those from premenopausal women, only in those with ER + or PR + status. The expression of these genes between tumors from pre- and post-menopausal patients with breast cancer was also analyzed based on the combination of status of three receptors.
Conclusion: Together, the results suggest that mRNA expression of ESR1, ESR2, and PGR might differ depending on menopausal status in breast tumors with certain receptor status. More importantly, the change in the expression of ESR1 and ESR2 following menopause is in the opposite directions in breast cancer patients showing the need to identify particular molecular mechanisms regulating the expression of ER isoforms post-menopause in different directions in breast cancer patients, considering the high clinical importance of these receptors in terms of the prognosis of patients with breast cancer.
{"title":"Menopausal status-dependent alterations in the transcript levels of genes encoding ERα, ERβ, PR and HER2 in breast tumors with different receptor status.","authors":"Caglar Berkel","doi":"10.1007/s12094-024-03777-x","DOIUrl":"https://doi.org/10.1007/s12094-024-03777-x","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer has distinct causes and prognoses in patients with premenopausal and postmenopausal status. The expression status of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are analyzed by immunohistochemistry (IHC) to classify molecular subtypes of breast cancer among which large differences in prognosis exist.</p><p><strong>Methods: </strong>The mRNA expression of ESR1 (encoding ERα), ESR2 (encoding ERβ), PGR (encoding PR), and ERBB2 (encoding HER2) was analyzed based on menopausal status (pre- vs post-menopausal) in tumors from breast cancer patients with different receptor status, in R programming environment, using transcriptomics data from TCGA-BRCA project.</p><p><strong>Results: </strong>In ER-positive or PR-positive or HER2-negative breast tumors, ESR1 transcript levels were found to be higher in tumors from postmenopausal women than those from premenopausal women; in contrast, ESR2 transcript levels were lower in tumors from postmenopausal women than those from premenopausal women. Furthermore, PGR mRNA expression was lower in breast tumors from postmenopausal women than those from premenopausal women, only in those with ER + or PR + status. The expression of these genes between tumors from pre- and post-menopausal patients with breast cancer was also analyzed based on the combination of status of three receptors.</p><p><strong>Conclusion: </strong>Together, the results suggest that mRNA expression of ESR1, ESR2, and PGR might differ depending on menopausal status in breast tumors with certain receptor status. More importantly, the change in the expression of ESR1 and ESR2 following menopause is in the opposite directions in breast cancer patients showing the need to identify particular molecular mechanisms regulating the expression of ER isoforms post-menopause in different directions in breast cancer patients, considering the high clinical importance of these receptors in terms of the prognosis of patients with breast cancer.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142570348","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-02DOI: 10.1007/s12094-024-03769-x
Yin He, Xiaosheng Wang
Background: Regulated cell death (RCD) pathways play significant roles in tumorigenesis. However, systematic investigation into correlations between RCD and various molecular and clinical features, particularly anti-tumor immunity and immunotherapy response in pan-cancer remains lacking.
Methods: Using the single-sample gene set enrichment analysis, we quantified the activities of six RCD pathways (apoptosis, autophagy, ferroptosis, cuproptosis, necroptosis, and pyroptosis) in each cancer specimen. Then, we explored associations of these six RCD pathways with tumor immunity, genomic instability, tumor phenotypes and clinical features, and responses to immunotherapy and targeted therapies in pan-cancer by statistical analyses.
Results: Our results showed that the RCD (except autophagy) activities were oncogenic signatures, as evidenced by their hyperactivation in late stage or metastatic cancer patients, positive correlations with tumor proliferation, stemness, genomic instability and intratumor heterogeneity, and correlation with worse survival outcomes in cancer. In contrast, autophagy was a tumor suppressive signature as its associations with molecular and clinical features in cancer shows an opposite pattern compared to the other RCD pathways. Furthermore, heightened RCD (except cuproptosis) activities were correlated with increased sensitivity to immune checkpoint inhibitors. Additionally, elevated activities of pyroptosis, autophagy, cuproptosis and necroptosis were associated with increased drug sensitivity in a broad spectrum of anti-tumor targeted therapies, while the elevated activity of ferroptosis was correlated with decreased sensitivity to numerous targeted therapies.
Conclusion: RCD (except autophagy) activities correlate with unfavorable cancer prognosis, while the autophagy activity correlate with favorable clinical outcomes. RCD (except cuproptosis) activities are positive biomarkers for anti-tumor immunity and immunotherapy response.
{"title":"A comprehensive investigation of associations between cell death pathways and molecular and clinical features in pan-cancer.","authors":"Yin He, Xiaosheng Wang","doi":"10.1007/s12094-024-03769-x","DOIUrl":"https://doi.org/10.1007/s12094-024-03769-x","url":null,"abstract":"<p><strong>Background: </strong>Regulated cell death (RCD) pathways play significant roles in tumorigenesis. However, systematic investigation into correlations between RCD and various molecular and clinical features, particularly anti-tumor immunity and immunotherapy response in pan-cancer remains lacking.</p><p><strong>Methods: </strong>Using the single-sample gene set enrichment analysis, we quantified the activities of six RCD pathways (apoptosis, autophagy, ferroptosis, cuproptosis, necroptosis, and pyroptosis) in each cancer specimen. Then, we explored associations of these six RCD pathways with tumor immunity, genomic instability, tumor phenotypes and clinical features, and responses to immunotherapy and targeted therapies in pan-cancer by statistical analyses.</p><p><strong>Results: </strong>Our results showed that the RCD (except autophagy) activities were oncogenic signatures, as evidenced by their hyperactivation in late stage or metastatic cancer patients, positive correlations with tumor proliferation, stemness, genomic instability and intratumor heterogeneity, and correlation with worse survival outcomes in cancer. In contrast, autophagy was a tumor suppressive signature as its associations with molecular and clinical features in cancer shows an opposite pattern compared to the other RCD pathways. Furthermore, heightened RCD (except cuproptosis) activities were correlated with increased sensitivity to immune checkpoint inhibitors. Additionally, elevated activities of pyroptosis, autophagy, cuproptosis and necroptosis were associated with increased drug sensitivity in a broad spectrum of anti-tumor targeted therapies, while the elevated activity of ferroptosis was correlated with decreased sensitivity to numerous targeted therapies.</p><p><strong>Conclusion: </strong>RCD (except autophagy) activities correlate with unfavorable cancer prognosis, while the autophagy activity correlate with favorable clinical outcomes. RCD (except cuproptosis) activities are positive biomarkers for anti-tumor immunity and immunotherapy response.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142565043","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-02DOI: 10.1007/s12094-024-03763-3
Muhammet Göktaş, Derya Karabulut, Ayhan Ünlü, Gkioulsoum Achmet, Nermin Tunçbilek
Purpose: To evaluate the prognostic effects of apparent diffusion coefficient (ADC) values, qualitative MRI findings, and XRCC1 polymorphism in patients with pancreatic ductal adenocarcinoma (PDAC).
Methods: Between January 2019 and December 2021, 41 PDAC patients (23 males; 66.6 ± 8.9 years) diagnosed with MRI and treated with chemotherapy were included in this prospective, unicenter study. Quantitative b:0-800 ADC values were calculated at the workstation using a circular region of interest with a diameter of 2 cm2. Polymerase chain reaction restriction fragment length polymorphism was used to detect XRCC1 genotypes from blood samples. Demographic data, MRI findings, and survival times were recorded. Sensitivity, specificity of ADCmin values, and relationship between XRCC1 genes in predicting survival were calculated and compared.
Results: The median overall survival time was calculated as 9.27 ± 1.4 months. The cut-off value of ADCmin was found to be 0.996 × 10-3 mm2/s for predicting a 4.6-month survival with 77.3% sensitivity and 84.2% specificity. The distribution of XRCC1 codon 399 polymorphism was determined as 26.8% (n = 11) GG, 65.9% (n = 27) AG, and 7.3% (n = 3) AA. There was no statistical correlation between ADCmin values and XRCC1 gene polymorphism (p > 0.05). ADCmin < 0.996 × 10-3 and the XRCC1 codon 399 AG/AA genotype was the subgroup with the worst prognosis (p = 0.035). The mean age at diagnosis was 71.0 ± 9.1 years in cases with GG genotype, while it was 64.6 ± 8.9 years in cases with AG/AA genotype, which was statistically significant (p = 0.038).
Conclusions: ADCmin values are useful for predicting prognosis in patients with PDAC. XRCC1 gene polymorphism may affect the age at diagnosis.
{"title":"MRI-based radiogenomics analysis for predicting prognosis and XRCC1 gene polymorphism in pancreatic ductal adenocarcinoma.","authors":"Muhammet Göktaş, Derya Karabulut, Ayhan Ünlü, Gkioulsoum Achmet, Nermin Tunçbilek","doi":"10.1007/s12094-024-03763-3","DOIUrl":"https://doi.org/10.1007/s12094-024-03763-3","url":null,"abstract":"<p><strong>Purpose: </strong>To evaluate the prognostic effects of apparent diffusion coefficient (ADC) values, qualitative MRI findings, and XRCC1 polymorphism in patients with pancreatic ductal adenocarcinoma (PDAC).</p><p><strong>Methods: </strong>Between January 2019 and December 2021, 41 PDAC patients (23 males; 66.6 ± 8.9 years) diagnosed with MRI and treated with chemotherapy were included in this prospective, unicenter study. Quantitative b:0-800 ADC values were calculated at the workstation using a circular region of interest with a diameter of 2 cm<sup>2</sup>. Polymerase chain reaction restriction fragment length polymorphism was used to detect XRCC1 genotypes from blood samples. Demographic data, MRI findings, and survival times were recorded. Sensitivity, specificity of ADCmin values, and relationship between XRCC1 genes in predicting survival were calculated and compared.</p><p><strong>Results: </strong>The median overall survival time was calculated as 9.27 ± 1.4 months. The cut-off value of ADCmin was found to be 0.996 × 10<sup>-3</sup> mm<sup>2</sup>/s for predicting a 4.6-month survival with 77.3% sensitivity and 84.2% specificity. The distribution of XRCC1 codon 399 polymorphism was determined as 26.8% (n = 11) GG, 65.9% (n = 27) AG, and 7.3% (n = 3) AA. There was no statistical correlation between ADCmin values and XRCC1 gene polymorphism (p > 0.05). ADCmin < 0.996 × 10<sup>-3</sup> and the XRCC1 codon 399 AG/AA genotype was the subgroup with the worst prognosis (p = 0.035). The mean age at diagnosis was 71.0 ± 9.1 years in cases with GG genotype, while it was 64.6 ± 8.9 years in cases with AG/AA genotype, which was statistically significant (p = 0.038).</p><p><strong>Conclusions: </strong>ADCmin values are useful for predicting prognosis in patients with PDAC. XRCC1 gene polymorphism may affect the age at diagnosis.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142565049","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-01Epub Date: 2024-06-24DOI: 10.1007/s12094-024-03559-5
Carles Pericay, Clara Montagut, Juan José Reina, Marcos Melian, Julia Alcaide, Noelia Tarazona, Ana Ruiz-Casado, Encarnación González-Flores, Begoña Graña, Cristina Grávalos
Colorectal cancer (CRC) has a 5-year overall survival rate of over 60%. The decrease in the rate of metastatic disease is due to screening programs and the population's awareness of healthy lifestyle. Similarly, advancements in surgical methods and the use of adjuvant chemotherapy have contributed to a decrease in the recurrence of resected disease. Before evaluating a patient's treatment, it is recommended to be discussed in a multidisciplinary tumor board. In stage II tumors, the pathologic characteristics of poor prognosis must be known (T4, number of lymph nodes analyzed less than 12, lymphovascular or perineural invasion, obstruction or perforation, poor histologic grade, presence of tumor budding) and it is mandatory to determine the MSI/MMR status for avoiding administering fluoropyridimidines in monotherapy to patients with MSI-H/dMMR tumors. In stage III tumors, the standard treatment consists of a combination of fluoropyrimidine (oral or intravenous) with oxaliplatin for 6 months although the administration of CAPOX can be considered for 3 months in low-risk tumors. Neoadjuvant treatment is not consolidated yet although immunotherapy is achieving very good preliminary results in MSI-H patients. The use of ctDNA to define the treatment and monitoring of resected tumors is only recommended within studies. These guidelines are intended to help decision-making to offer the best management of patients with non-metastatic colon cancer.
{"title":"SEOM-GEMCAD-TTD clinical guidelines for the adjuvant treatment of colon cancer (2023).","authors":"Carles Pericay, Clara Montagut, Juan José Reina, Marcos Melian, Julia Alcaide, Noelia Tarazona, Ana Ruiz-Casado, Encarnación González-Flores, Begoña Graña, Cristina Grávalos","doi":"10.1007/s12094-024-03559-5","DOIUrl":"10.1007/s12094-024-03559-5","url":null,"abstract":"<p><p>Colorectal cancer (CRC) has a 5-year overall survival rate of over 60%. The decrease in the rate of metastatic disease is due to screening programs and the population's awareness of healthy lifestyle. Similarly, advancements in surgical methods and the use of adjuvant chemotherapy have contributed to a decrease in the recurrence of resected disease. Before evaluating a patient's treatment, it is recommended to be discussed in a multidisciplinary tumor board. In stage II tumors, the pathologic characteristics of poor prognosis must be known (T4, number of lymph nodes analyzed less than 12, lymphovascular or perineural invasion, obstruction or perforation, poor histologic grade, presence of tumor budding) and it is mandatory to determine the MSI/MMR status for avoiding administering fluoropyridimidines in monotherapy to patients with MSI-H/dMMR tumors. In stage III tumors, the standard treatment consists of a combination of fluoropyrimidine (oral or intravenous) with oxaliplatin for 6 months although the administration of CAPOX can be considered for 3 months in low-risk tumors. Neoadjuvant treatment is not consolidated yet although immunotherapy is achieving very good preliminary results in MSI-H patients. The use of ctDNA to define the treatment and monitoring of resected tumors is only recommended within studies. These guidelines are intended to help decision-making to offer the best management of patients with non-metastatic colon cancer.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11467085/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141447439","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}