Pub Date : 2024-10-24DOI: 10.1186/s12885-024-12841-2
Armando Santoro, Garrido Pilar, Daniel S W Tan, Jon Zugazagoitia, Frances A Shepherd, Alessandra Bearz, Fabrice Barlesi, Tae Min Kim, Tobias R Overbeck, Enriqueta Felip, Can Cai, Eddy Simantini, Tracey McCulloch, Eric S Schaefer
Background: Despite promising outcomes of treatment with anti-programmed cell death (PD)-1/PD-ligand (L)1 agents in combination with platinum-doublet chemotherapy (PDC) in the first-line setting, a significant unmet medical need remains in patients with PD-L1-unselected non-small cell lung cancer (NSCLC).
Methods: This multicenter, open-label, phase 1b study comprising dose-confirmation and dose-expansion parts investigated the combination of spartalizumab and various PDC regimens, with or without canakinumab, in treatment-naïve patients with PD-L1-unselected, metastatic NSCLC. The primary objectives were to determine maximum tolerated dose (MTD) and/or recommended dose for expansion (RDE) of spartalizumab, with or without canakinumab, in combination with PDC in the dose-confirmation part and antitumor activity of spartalizumab in the dose-expansion part.
Results: The MTD/RDE of spartalizumab was 300 mg every 3 weeks (Q3W) when administered with either gemcitabine (1250 mg/m2)/cisplatin (75 mg/m2) (group A; no dose-limiting toxicities [DLTs]), pemetrexed (500 mg/m2)/cisplatin (group B; 2 DLTs: grade 2 posterior reversible encephalopathy syndrome and grade 4 hyponatremia), or paclitaxel (200 mg/m2)/carboplatin area under the curve 6 min*mg/mL (group C; 1 DLT: grade 4 neutropenic colitis). The RDE of canakinumab combined with spartalizumab and pemetrexed/cisplatin (group E; no DLTs) was 200 mg Q3W (no dose-expansion part was initiated). No new safety signals were identified. In groups A, B, C, and E, the overall response rates were 57.6%, 55.3%, 51.5%, and 57.1%, respectively. Group B compared with other groups had the longest median progression-free survival (10.4 months vs. 6.2-7.5 months), overall survival (29.7 months vs. 16.1-21.0 months), and duration of response (30.1 months vs. 6.0-8.2 months).
Conclusions: The combination of spartalizumab and PDC, with or without canakinumab, was well tolerated across treatment groups. The antitumor activity across treatment groups was comparable with that of pembrolizumab and pemetrexed combination. Canakinumab did not appear to improve the antitumor activity when combined with spartalizumab, pemetrexed and cisplatin.
Trial registration: The trial was registered in Clinicaltrials.gov with identifier no. NCT03064854. Date of Registration: 06 February 2017.
背景:尽管抗程序性细胞死亡(PD)-1/PD配体(L)1药物联合铂双联化疗(PDC)在一线治疗中取得了良好的疗效,但PD-L1未被选择的非小细胞肺癌(NSCLC)患者仍有大量医疗需求未得到满足:这项多中心、开放标签、1b期研究包括剂量确认和剂量扩大两部分,研究了在PD-L1未被选择的转移性NSCLC患者中,联合使用斯帕妥珠单抗和各种PDC方案,同时使用或不使用卡那单抗。主要目的是在剂量确认部分确定斯帕妥珠单抗(无论有无卡纳库单抗)联合PDC的最大耐受剂量(MTD)和/或推荐扩大剂量(RDE),在剂量扩大部分确定斯帕妥珠单抗的抗肿瘤活性:当与吉西他滨(1250毫克/平方米)/顺铂(75毫克/平方米)(A组;无剂量限制性毒性[DLTs])、培美曲塞(500毫克/平方米)/顺铂(B组;2个DLTs:或紫杉醇(200 毫克/平方米)/卡铂曲线下面积 6 分钟*毫克/毫升(C 组;1 个 DLT:4 级中性粒细胞减少性结肠炎)。卡那单抗联合斯帕妥珠单抗和培美曲塞/顺铂(E组;无DLT)的RDE为200毫克Q3W(未启动剂量扩展部分)。未发现新的安全信号。A、B、C和E组的总体反应率分别为57.6%、55.3%、51.5%和57.1%。与其他组相比,B组的中位无进展生存期(10.4个月 vs. 6.2-7.5个月)、总生存期(29.7个月 vs. 16.1-21.0个月)和应答持续时间(30.1个月 vs. 6.0-8.2个月)最长:各治疗组患者对斯帕妥珠单抗和PDC联合用药(无论是否使用卡那单抗)的耐受性良好。各治疗组的抗肿瘤活性与彭博利珠单抗和培美曲塞联合疗法相当。卡纳库单抗与斯帕妥珠单抗、培美曲塞和顺铂联合治疗似乎并没有提高抗肿瘤活性:该试验已在Clinicaltrials.gov上注册,识别号为NCT03064854。NCT03064854。注册日期:2017年2月6日:2017年2月6日。
{"title":"Spartalizumab in combination with platinum-doublet chemotherapy with or without canakinumab in patients with PD-L1-unselected, metastatic NSCLC.","authors":"Armando Santoro, Garrido Pilar, Daniel S W Tan, Jon Zugazagoitia, Frances A Shepherd, Alessandra Bearz, Fabrice Barlesi, Tae Min Kim, Tobias R Overbeck, Enriqueta Felip, Can Cai, Eddy Simantini, Tracey McCulloch, Eric S Schaefer","doi":"10.1186/s12885-024-12841-2","DOIUrl":"10.1186/s12885-024-12841-2","url":null,"abstract":"<p><strong>Background: </strong>Despite promising outcomes of treatment with anti-programmed cell death (PD)-1/PD-ligand (L)1 agents in combination with platinum-doublet chemotherapy (PDC) in the first-line setting, a significant unmet medical need remains in patients with PD-L1-unselected non-small cell lung cancer (NSCLC).</p><p><strong>Methods: </strong>This multicenter, open-label, phase 1b study comprising dose-confirmation and dose-expansion parts investigated the combination of spartalizumab and various PDC regimens, with or without canakinumab, in treatment-naïve patients with PD-L1-unselected, metastatic NSCLC. The primary objectives were to determine maximum tolerated dose (MTD) and/or recommended dose for expansion (RDE) of spartalizumab, with or without canakinumab, in combination with PDC in the dose-confirmation part and antitumor activity of spartalizumab in the dose-expansion part.</p><p><strong>Results: </strong>The MTD/RDE of spartalizumab was 300 mg every 3 weeks (Q3W) when administered with either gemcitabine (1250 mg/m<sup>2</sup>)/cisplatin (75 mg/m<sup>2</sup>) (group A; no dose-limiting toxicities [DLTs]), pemetrexed (500 mg/m<sup>2</sup>)/cisplatin (group B; 2 DLTs: grade 2 posterior reversible encephalopathy syndrome and grade 4 hyponatremia), or paclitaxel (200 mg/m<sup>2</sup>)/carboplatin area under the curve 6 min*mg/mL (group C; 1 DLT: grade 4 neutropenic colitis). The RDE of canakinumab combined with spartalizumab and pemetrexed/cisplatin (group E; no DLTs) was 200 mg Q3W (no dose-expansion part was initiated). No new safety signals were identified. In groups A, B, C, and E, the overall response rates were 57.6%, 55.3%, 51.5%, and 57.1%, respectively. Group B compared with other groups had the longest median progression-free survival (10.4 months vs. 6.2-7.5 months), overall survival (29.7 months vs. 16.1-21.0 months), and duration of response (30.1 months vs. 6.0-8.2 months).</p><p><strong>Conclusions: </strong>The combination of spartalizumab and PDC, with or without canakinumab, was well tolerated across treatment groups. The antitumor activity across treatment groups was comparable with that of pembrolizumab and pemetrexed combination. Canakinumab did not appear to improve the antitumor activity when combined with spartalizumab, pemetrexed and cisplatin.</p><p><strong>Trial registration: </strong>The trial was registered in Clinicaltrials.gov with identifier no. NCT03064854. Date of Registration: 06 February 2017.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515544/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-24DOI: 10.1186/s12885-024-13007-w
Grazia Fenu, Carmen Griñán-Lisón, Andrea Pisano, Aitor González-Titos, Cristiano Farace, Giovanni Fiorito, Federica Etzi, Teresa Perra, Angela Sabalic, Belén Toledo, Macarena Perán, Maria Giuliana Solinas, Alberto Porcu, Juan Antonio Marchal, Roberto Madeddu
Background: Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge due to late-stage diagnoses resulting from nonspecific early symptoms and the absence of early diagnostic biomarkers. MicroRNAs (miRNAs) play a crucial role in regulating diverse biological processes, and their abnormal expression is observed in various diseases, including cancer. Cancer stem cells (CSCs) are thought to act as a driving force in PDAC spread and recurrence. In pursuing the goal of unravelling the complexities of PDAC and its underlying molecular mechanisms, our study aimed to identify PDAC-associated miRNAs and relate them to disease progression, focusing on their involvement in various PDAC stages in patients and in reliable in vitro models, including pancreatic CSC (PaCSC) models.
Methods: The miRNA profiling datasets of serum and solid biopsies of PDAC patients deposited in GEO DataSets were analyzed by REML-based meta-analysis. The panel was then investigated by Real Time PCR in serum and solid biopsies of 37 PDAC patients enrolled in the study, as well as on BxPC-3 and AsPC-1 PDAC cell lines. We extended our focus towards a possible role of PDAC-associated miRNAs in the CSC phenotype, by inducing CSC-enriched pancreatospheres from BxPC-3 and AsPC-1 PDAC cell lines and performed differential miRNA expression analysis between PaCSCs and monolayer-grown PDAC cell lines.
Results: Meta-analysis showed differentially expressed miRNAs in blood samples and cancerous tissues of PDAC patients, allowing the identification of a panel of 9 PDAC-associated miRNAs. The results emerging from our patients fully confirmed the meta-analysis for the majority of miRNAs under investigation. In vitro tasks confirmed the aberrant expression of the panel of PDAC-associated miRNAs, with a dramatic dysregulation in PaCSC models. Notably, PaCSCs have shown significant overexpression of miR-4486, miR-216a-5p, and miR-216b-5p compared to PDAC cell lines, suggesting the recruitment of such miRNAs in stemness-related molecular mechanisms. Globally, our results showed a dual behaviour of miR-216a-5p and miR-216b-5p in PDAC while miR-4486, miR-361-3p, miR-125a-5p, miR-320d expression changes during the disease suggest they could promote PDAC initiation and progression.
Conclusions: This study contributed to an enhanced comprehension of the role of miRNAs in the development and progression of PDAC, shedding new light on the miRNA landscape in PDAC and its intricate interplay with CSCs, and providing specific insights useful in the development of miRNA-based diagnostic biomarkers and therapeutic targets.
{"title":"Unveiling the microRNA landscape in pancreatic ductal adenocarcinoma patients and cancer cell models.","authors":"Grazia Fenu, Carmen Griñán-Lisón, Andrea Pisano, Aitor González-Titos, Cristiano Farace, Giovanni Fiorito, Federica Etzi, Teresa Perra, Angela Sabalic, Belén Toledo, Macarena Perán, Maria Giuliana Solinas, Alberto Porcu, Juan Antonio Marchal, Roberto Madeddu","doi":"10.1186/s12885-024-13007-w","DOIUrl":"10.1186/s12885-024-13007-w","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge due to late-stage diagnoses resulting from nonspecific early symptoms and the absence of early diagnostic biomarkers. MicroRNAs (miRNAs) play a crucial role in regulating diverse biological processes, and their abnormal expression is observed in various diseases, including cancer. Cancer stem cells (CSCs) are thought to act as a driving force in PDAC spread and recurrence. In pursuing the goal of unravelling the complexities of PDAC and its underlying molecular mechanisms, our study aimed to identify PDAC-associated miRNAs and relate them to disease progression, focusing on their involvement in various PDAC stages in patients and in reliable in vitro models, including pancreatic CSC (PaCSC) models.</p><p><strong>Methods: </strong>The miRNA profiling datasets of serum and solid biopsies of PDAC patients deposited in GEO DataSets were analyzed by REML-based meta-analysis. The panel was then investigated by Real Time PCR in serum and solid biopsies of 37 PDAC patients enrolled in the study, as well as on BxPC-3 and AsPC-1 PDAC cell lines. We extended our focus towards a possible role of PDAC-associated miRNAs in the CSC phenotype, by inducing CSC-enriched pancreatospheres from BxPC-3 and AsPC-1 PDAC cell lines and performed differential miRNA expression analysis between PaCSCs and monolayer-grown PDAC cell lines.</p><p><strong>Results: </strong>Meta-analysis showed differentially expressed miRNAs in blood samples and cancerous tissues of PDAC patients, allowing the identification of a panel of 9 PDAC-associated miRNAs. The results emerging from our patients fully confirmed the meta-analysis for the majority of miRNAs under investigation. In vitro tasks confirmed the aberrant expression of the panel of PDAC-associated miRNAs, with a dramatic dysregulation in PaCSC models. Notably, PaCSCs have shown significant overexpression of miR-4486, miR-216a-5p, and miR-216b-5p compared to PDAC cell lines, suggesting the recruitment of such miRNAs in stemness-related molecular mechanisms. Globally, our results showed a dual behaviour of miR-216a-5p and miR-216b-5p in PDAC while miR-4486, miR-361-3p, miR-125a-5p, miR-320d expression changes during the disease suggest they could promote PDAC initiation and progression.</p><p><strong>Conclusions: </strong>This study contributed to an enhanced comprehension of the role of miRNAs in the development and progression of PDAC, shedding new light on the miRNA landscape in PDAC and its intricate interplay with CSCs, and providing specific insights useful in the development of miRNA-based diagnostic biomarkers and therapeutic targets.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515555/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495110","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The HER-2 status of breast cancer (BC) has been classified as negative or positive for a long time. Given the efficacy of novel anti-HER2-targeted antibody drug conjugates (ADCs) in HER2-low BC, a distinct subgroup of HER2-low tumors has emerged within BC. The biology and prognostic impact of HER2-low expression are not yet well defined, and inconsistent results were reported. This study aims to evaluate the impact of low HER-2 status on the response to neoadjuvant chemotherapy (NACT) and disease- free survival (DFS) rates.
Methods: We retrospectively analyzed BC patients treated with NACT from 2017 to 2023 in two cancer centers. HER2-negative patients were included. HER-2 low status was defined by IHC + 1 or + 2/ISH non-amplified, and HER2-zero was defined by IHC 0. Pathological complete response (pCR) rates and DFS between HER2-low and HER2-zero populations were compared.
Results: 170 patients were identified. 122 (72%) of these patients were HER2- zero BC, whereas 48 (28%) were HER2-low BC. Overall, pCR was achieved in 35 (20.5%) patients. Of these, pCR was observed in 30 patients (44.6%) from the HER2- zero group, compared to 5 patients (10.4%) from the HER2-low group (p = 0.046), but significance was lost in multivariate analysis. Among the hormone receptor (HR) positive subtype, pCR was achieved 19.8% of HER2-zero tumors and 7.5% of HER2-low tumors (p = 0.08). For HR-negative subtype 34.1% HER2-zero tumors had pCR and 25% of the HER2-low tumors had pCR (p = 0.614). There was no association between DFS and HER2-low status.
Conclusions: Our study indicates that HER2-low status had no impact on pCR or DFS.
{"title":"The impact of HER2-low status on pathological complete response and disease-free survival in early-stage breast cancer.","authors":"Gülin Alkan Şen, Esra Aydın, Murad Guliyev, Nihan Şentürk Öztaş, Ezgi Değerli, Nebi Serkan Demirci, Zeynep Hande Turna, Fuat Hulusi Demirelli","doi":"10.1186/s12885-024-13064-1","DOIUrl":"10.1186/s12885-024-13064-1","url":null,"abstract":"<p><strong>Background: </strong>The HER-2 status of breast cancer (BC) has been classified as negative or positive for a long time. Given the efficacy of novel anti-HER2-targeted antibody drug conjugates (ADCs) in HER2-low BC, a distinct subgroup of HER2-low tumors has emerged within BC. The biology and prognostic impact of HER2-low expression are not yet well defined, and inconsistent results were reported. This study aims to evaluate the impact of low HER-2 status on the response to neoadjuvant chemotherapy (NACT) and disease- free survival (DFS) rates.</p><p><strong>Methods: </strong>We retrospectively analyzed BC patients treated with NACT from 2017 to 2023 in two cancer centers. HER2-negative patients were included. HER-2 low status was defined by IHC + 1 or + 2/ISH non-amplified, and HER2-zero was defined by IHC 0. Pathological complete response (pCR) rates and DFS between HER2-low and HER2-zero populations were compared.</p><p><strong>Results: </strong>170 patients were identified. 122 (72%) of these patients were HER2- zero BC, whereas 48 (28%) were HER2-low BC. Overall, pCR was achieved in 35 (20.5%) patients. Of these, pCR was observed in 30 patients (44.6%) from the HER2- zero group, compared to 5 patients (10.4%) from the HER2-low group (p = 0.046), but significance was lost in multivariate analysis. Among the hormone receptor (HR) positive subtype, pCR was achieved 19.8% of HER2-zero tumors and 7.5% of HER2-low tumors (p = 0.08). For HR-negative subtype 34.1% HER2-zero tumors had pCR and 25% of the HER2-low tumors had pCR (p = 0.614). There was no association between DFS and HER2-low status.</p><p><strong>Conclusions: </strong>Our study indicates that HER2-low status had no impact on pCR or DFS.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515358/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495108","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-24DOI: 10.1186/s12885-024-13059-y
Patrik Karlsson, Malin Nygren-Bonnier, Simon Torikka, Andrea Porserud, Lars Henningsohn, Christina B Olsson, Elisabeth Rydwik, Maria Hagströmer
Background: Physical activity is thought to be a key component in reducing postoperative complications following major abdominal surgery. The available literature on exercise interventions following radical cystectomy in patients with bladder cancer is scarce but suggests that physical activity and exercise might improve physical function and health-related quality of life, thus calling for further investigation. The CanMoRe-trial is a single-blinded randomised controlled trial (Clinicals Trials NCT03998579 25/06/2019), aimed at evaluating the impact of an exercise intervention in primary care following robot-assisted radical cystectomy. This study seeks to explore patients' experiences of the exercise intervention in the CanMoRe-trial to gain a better understanding of facilitating aspects and potential barriers.
Methods: A qualitative study was conducted involving 20 patients from the intervention group of the CanMoRe-trial who were interviewed individually between October 2020 and March 2023 using a semi-structured interview guide. The interviews were recorded and transcribed verbatim and reflexive thematic analysis was used to analyse the data.
Results: Four main themes were identified: Having to adapt to new circumstances, describing the challenges regarding physical activity patients face after discharge. Optimising conditions for rehabilitation, describing how practical conditions affect patients' ability to exercise. Motivated to get back to normal, describing patients´ desire to get back to normal life and factors influencing motivation. Importance of a supportive environment, describing the impact of social support, support from physiotherapists, and how the environment where exercise takes place impacts patients' ability to exercise.
Conclusion: This study found that patients participating in the CanMoRe-trial are positive towards physical exercise in PC following radical RARC. They are motivated to get back to normal life but face major challenges when arriving home following surgery, which affect their ability to perform physical activity and engage in exercise. Conditions need to be optimised to support patients' ability to engage in exercise by providing an accessible PC location to perform exercise in. A supportive environment is also needed, including guidance from healthcare professionals regarding which type of exercise, intensity and amount of exercise that should be performed, enabling patients gradually to develop self-efficacy regarding exercise and focusing on goals related to patients' normal lives before surgery.
{"title":"Patients´ experiences of an exercise intervention in primary care following robot-assisted radical cystectomy due to bladder cancer: a qualitative study.","authors":"Patrik Karlsson, Malin Nygren-Bonnier, Simon Torikka, Andrea Porserud, Lars Henningsohn, Christina B Olsson, Elisabeth Rydwik, Maria Hagströmer","doi":"10.1186/s12885-024-13059-y","DOIUrl":"10.1186/s12885-024-13059-y","url":null,"abstract":"<p><strong>Background: </strong>Physical activity is thought to be a key component in reducing postoperative complications following major abdominal surgery. The available literature on exercise interventions following radical cystectomy in patients with bladder cancer is scarce but suggests that physical activity and exercise might improve physical function and health-related quality of life, thus calling for further investigation. The CanMoRe-trial is a single-blinded randomised controlled trial (Clinicals Trials NCT03998579 25/06/2019), aimed at evaluating the impact of an exercise intervention in primary care following robot-assisted radical cystectomy. This study seeks to explore patients' experiences of the exercise intervention in the CanMoRe-trial to gain a better understanding of facilitating aspects and potential barriers.</p><p><strong>Methods: </strong>A qualitative study was conducted involving 20 patients from the intervention group of the CanMoRe-trial who were interviewed individually between October 2020 and March 2023 using a semi-structured interview guide. The interviews were recorded and transcribed verbatim and reflexive thematic analysis was used to analyse the data.</p><p><strong>Results: </strong>Four main themes were identified: Having to adapt to new circumstances, describing the challenges regarding physical activity patients face after discharge. Optimising conditions for rehabilitation, describing how practical conditions affect patients' ability to exercise. Motivated to get back to normal, describing patients´ desire to get back to normal life and factors influencing motivation. Importance of a supportive environment, describing the impact of social support, support from physiotherapists, and how the environment where exercise takes place impacts patients' ability to exercise.</p><p><strong>Conclusion: </strong>This study found that patients participating in the CanMoRe-trial are positive towards physical exercise in PC following radical RARC. They are motivated to get back to normal life but face major challenges when arriving home following surgery, which affect their ability to perform physical activity and engage in exercise. Conditions need to be optimised to support patients' ability to engage in exercise by providing an accessible PC location to perform exercise in. A supportive environment is also needed, including guidance from healthcare professionals regarding which type of exercise, intensity and amount of exercise that should be performed, enabling patients gradually to develop self-efficacy regarding exercise and focusing on goals related to patients' normal lives before surgery.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515536/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495092","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-23DOI: 10.1186/s12885-024-13071-2
Yalan Tong, Xiaosha Wan, Chang Yin, Ting Lei, Shan Gao, Yinghua Li, Xiaojing Du
Background: There is a growing interest in utilizing a combination of brain radiotherapy (RT) and tyrosine kinase inhibitors (TKIs) for patients diagnosed with brain metastases (BM) in epidermal growth factor receptor (EGFR) mutation-positive lung adenocarcinoma (LAC). The current status of this treatment strategy remains a subject of debate.
Methods: We initiated our study by conducting a comprehensive literature search using the SCI-expanded database of Web of Science Core Collection (WoSCC). We utilized the VOSPviewer software to analyze various aspects of the research, including the year of publication, authorship, keywords, and country.Subsequently, we performed an extensive and systematic literature search on popular online databases. Our primary outcome measures were overall survival (OS) and intracranial progression-free survival (iPFS), both quantified by hazard ratios (HRs). Additionally, for data verification, we included data from patients in non-small cell lung cancer with brain metastasis who underwent therapeutic intervention at the Cancer Prevention and Treatment Center of Sun Yat-sen University and the Radiotherapy Department of Hanzhong Central Hospital between August 2012 and November 2021.
Results: The bibliometric analysis revealed an increasing trend in research focused on the combination of RT and TKIs for the management of lung cancer brain metastases over the previous decade. Then, nine studies consistent with the research direction were included for meta-analysis. The meta-analysis showed that the OS (HR = 0.81, 95% confidence interval: 0.69-0.94; P = 0.007) and iPFS (HR = 0.71, 95% confidence interval: 0.61-0.82; P < 0.001) of the combination therapy were significantly prolonged. Finally, 168 EGFR-mutated BM advanced LAC patients in the real world were verified, and the median iPFS of the combination therapy (n = 88 and EGFR-TKIs alone (n = 80) were 16.0 and 9.0 months, respectively, (P < 0.001). The median OS was 29.0 and 27.0 months, respectively, with no dramatic difference (P = 0.188).
Conclusions: Research on EGFR-mutant LAC brain metastasis has turned towards exploring optimal treatment strategies for this condition. Our meta-analysis and real-world data analysis consistently demonstrate that combination therapy offers a substantial improvement in patient survival compared to EGFR-TKI monotherapy. Notably, among patients undergoing salvage radiotherapy (RT), our subgroup analysis reveals that those initially treated with third-generation TKIs experience more significant benefits than those treated with first- or second-generation TKIs.
{"title":"In-depth exploration of the focus issues of TKI combined with radiotherapy for EGFR-mutant lung adenocarcinoma patients with brain metastasis: a systematic analysis based on literature metrology, meta-analysis, and real-world observational data.","authors":"Yalan Tong, Xiaosha Wan, Chang Yin, Ting Lei, Shan Gao, Yinghua Li, Xiaojing Du","doi":"10.1186/s12885-024-13071-2","DOIUrl":"10.1186/s12885-024-13071-2","url":null,"abstract":"<p><strong>Background: </strong>There is a growing interest in utilizing a combination of brain radiotherapy (RT) and tyrosine kinase inhibitors (TKIs) for patients diagnosed with brain metastases (BM) in epidermal growth factor receptor (EGFR) mutation-positive lung adenocarcinoma (LAC). The current status of this treatment strategy remains a subject of debate.</p><p><strong>Methods: </strong>We initiated our study by conducting a comprehensive literature search using the SCI-expanded database of Web of Science Core Collection (WoSCC). We utilized the VOSPviewer software to analyze various aspects of the research, including the year of publication, authorship, keywords, and country.Subsequently, we performed an extensive and systematic literature search on popular online databases. Our primary outcome measures were overall survival (OS) and intracranial progression-free survival (iPFS), both quantified by hazard ratios (HRs). Additionally, for data verification, we included data from patients in non-small cell lung cancer with brain metastasis who underwent therapeutic intervention at the Cancer Prevention and Treatment Center of Sun Yat-sen University and the Radiotherapy Department of Hanzhong Central Hospital between August 2012 and November 2021.</p><p><strong>Results: </strong>The bibliometric analysis revealed an increasing trend in research focused on the combination of RT and TKIs for the management of lung cancer brain metastases over the previous decade. Then, nine studies consistent with the research direction were included for meta-analysis. The meta-analysis showed that the OS (HR = 0.81, 95% confidence interval: 0.69-0.94; P = 0.007) and iPFS (HR = 0.71, 95% confidence interval: 0.61-0.82; P < 0.001) of the combination therapy were significantly prolonged. Finally, 168 EGFR-mutated BM advanced LAC patients in the real world were verified, and the median iPFS of the combination therapy (n = 88 and EGFR-TKIs alone (n = 80) were 16.0 and 9.0 months, respectively, (P < 0.001). The median OS was 29.0 and 27.0 months, respectively, with no dramatic difference (P = 0.188).</p><p><strong>Conclusions: </strong>Research on EGFR-mutant LAC brain metastasis has turned towards exploring optimal treatment strategies for this condition. Our meta-analysis and real-world data analysis consistently demonstrate that combination therapy offers a substantial improvement in patient survival compared to EGFR-TKI monotherapy. Notably, among patients undergoing salvage radiotherapy (RT), our subgroup analysis reveals that those initially treated with third-generation TKIs experience more significant benefits than those treated with first- or second-generation TKIs.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515526/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background and objectives: Lung cancer is one of the prevailing malignancies worldwide. Surgical interventions hold an important position in the treatment framework for lung cancer. Pleural metastasis is often assumed to be a surgical contraindication, but not all instances of pleural metastasis can be accurately identified before surgery. The question of how to address pleural metastasis detected intraoperatively is still undecided.
Methods: This retrospective study included 187 lung cancer patients who underwent surgery from 2005 to 2017 in whom pleural metastasis was discovered incidentally during the operation. Data on demographic, surgical, pathological, postoperative treatment, and survival information were collected for further analysis.
Results: For patients with intraoperatively detected pleural metastasis, two independent protective prognostic factors were receiving primary tumor resection (compared to only receiving pleural nodule biopsy, HR = 0.079, p = 0.022) and receiving postoperative adjuvant chemotherapy (HR = 0.081, p < 0.001). Simultaneously, performing systematic lymph node dissection during primary tumor resection was found to be detrimental to long-term prognosis (HR = 2.375, p = 0.044). However, the resection of pleural metastatic lesions did not significantly impact patient prognosis.
Conclusion: Our study supports the implementation of major tumor resection in patients with pleural metastasis detected intraoperatively but not lymph node dissection or the resection of pleural metastatic lesions. Postoperative chemotherapy is also necessary.
{"title":"Comprehensive analysis of surgical strategies and prognosis for non-small cell lung cancer with pleural metastasis detected intraoperatively.","authors":"Fanmao Meng, Na Ren, Guochao Zhang, Ruifeng Xu, Mengbai Tian, Xin Sun, Liang Zhao","doi":"10.1186/s12885-024-13029-4","DOIUrl":"https://doi.org/10.1186/s12885-024-13029-4","url":null,"abstract":"<p><strong>Background and objectives: </strong>Lung cancer is one of the prevailing malignancies worldwide. Surgical interventions hold an important position in the treatment framework for lung cancer. Pleural metastasis is often assumed to be a surgical contraindication, but not all instances of pleural metastasis can be accurately identified before surgery. The question of how to address pleural metastasis detected intraoperatively is still undecided.</p><p><strong>Methods: </strong>This retrospective study included 187 lung cancer patients who underwent surgery from 2005 to 2017 in whom pleural metastasis was discovered incidentally during the operation. Data on demographic, surgical, pathological, postoperative treatment, and survival information were collected for further analysis.</p><p><strong>Results: </strong>For patients with intraoperatively detected pleural metastasis, two independent protective prognostic factors were receiving primary tumor resection (compared to only receiving pleural nodule biopsy, HR = 0.079, p = 0.022) and receiving postoperative adjuvant chemotherapy (HR = 0.081, p < 0.001). Simultaneously, performing systematic lymph node dissection during primary tumor resection was found to be detrimental to long-term prognosis (HR = 2.375, p = 0.044). However, the resection of pleural metastatic lesions did not significantly impact patient prognosis.</p><p><strong>Conclusion: </strong>Our study supports the implementation of major tumor resection in patients with pleural metastasis detected intraoperatively but not lymph node dissection or the resection of pleural metastatic lesions. Postoperative chemotherapy is also necessary.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11494768/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495085","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1186/s12885-024-13023-w
Daniele Gennuso, Angela Baldelli, Loredana Gigli, Ilaria Ruotolo, Giovanni Galeoto, Daniela Gaburri, Giovanni Sellitto
Background: Patients with cancer (PwC) who undergo specific treatments reported greater fatigue and reduced functional capacity as predominant outcomes, compromising their QoL during and following the treatment. Prehabilitation intervention, provided after diagnosis and before treatments, is to optimize the physiological reserve and address modifiable risk factors before surgery or chemotherapy to improve post-treatment results. The primary aim of this study is to obtain a common line of efficacy compared with prehabilitation treatment; the secondary endpoint is to assess the methodological quality of the studies eligible in the review.
Methods: The systematic review was conducted from September to February 2024, in accordance with PRISMA guidelines. Databases consulted were MEDLINE, Scopus, Web of science and CINAHL, RCTs related to Prehabilitation intervention on PwC were included. The methodological quality of the included studies was assessed through the RoB2 Cochrane tool and the PEDro scale. Meta-analysis was performed to estimate relative treatment effects among evaluated outcomes.
Results: Forty-two studies were included in the systematic review, 13 were included in the quantitative analysis. The results of the studies reporting reduced postoperative hospital stay, improved endurance, muscle strength, respiratory function, quality of life, and urinary incontinence. Critical analysis of the articles using the PEDro scale revealed 28 RCTs with a good rating, 9 with a fair rating, and 5 with a poor rating; in contrast, the Cochrane RoB2 tool revealed that all articles were at high risk of bias. Meta-analysis showed statistically significant values for 6MWT (38.53, 95%CI 33.03 - 44.04); HADS-depression (-0.71, 95%CI -0.93 -0.49) and HADS-anxiety (-0.49, 95%CI -0.76 -0.23).
Conclusions: Prehabilitation represents a specific intervention that aims to improve postoperative outcomes in fragile patients undergoing surgery, increasing their preoperative physiological reserve in anticipation of the stress they will face and facilitating the postoperative recovery of functional capacity. Prehabilitation is a good intervention to use, especially in terms of functional capacity and mental health, the latter being very impactful in terms of reduced levels of anxiety and depression. These data make it possible to justify supportive intervention by physical therapists aimed at improving and restoring health-related QoL especially in the short term.
背景:接受特定治疗的癌症患者(PwC)报告称,他们在治疗期间和治疗后的主要结果是更加疲劳和功能减退,影响了他们的生活质量。在确诊后和治疗前进行的预康复干预旨在优化生理储备,并在手术或化疗前解决可改变的风险因素,以改善治疗后的效果。本研究的主要目的是获得与康复前治疗相比的共同疗效;次要终点是评估符合综述条件的研究的方法学质量:系统综述于 2024 年 9 月至 2 月进行,符合 PRISMA 指南。查阅的数据库包括 MEDLINE、Scopus、Web of science 和 CINAHL,纳入了与 PwC 前期康复干预相关的 RCT。纳入研究的方法学质量通过 RoB2 Cochrane 工具和 PEDro 量表进行评估。进行了 Meta 分析,以估算评估结果中的相对治疗效果:42项研究被纳入系统综述,13项被纳入定量分析。研究结果表明,术后住院时间缩短,耐力、肌力、呼吸功能、生活质量和尿失禁得到改善。使用 PEDro 量表对文章进行批判性分析后发现,28 篇研究性临床试验获得了良好评价,9 篇获得了一般评价,5 篇获得了较差评价;而 Cochrane RoB2 工具则显示,所有文章都存在较高的偏倚风险。Meta 分析表明,6MWT(38.53,95%CI 33.03 -44.04)、HADS-抑郁(-0.71,95%CI -0.93 -0.49)和 HADS-焦虑(-0.49,95%CI -0.76 -0.23)具有显著的统计学意义:术前康复是一种特殊的干预措施,旨在改善接受手术的脆弱患者的术后效果,增加他们术前的生理储备以应对即将面临的压力,并促进术后功能的恢复。术前康复训练是一种很好的干预措施,特别是在功能能力和心理健康方面,后者对降低焦虑和抑郁水平有很大影响。这些数据证明,物理治疗师有理由采取支持性干预措施,以改善和恢复与健康相关的生活质量,尤其是在短期内。
{"title":"Efficacy of Prehabilitation in cancer patients: an Rcts systematic review with meta-analysis.","authors":"Daniele Gennuso, Angela Baldelli, Loredana Gigli, Ilaria Ruotolo, Giovanni Galeoto, Daniela Gaburri, Giovanni Sellitto","doi":"10.1186/s12885-024-13023-w","DOIUrl":"10.1186/s12885-024-13023-w","url":null,"abstract":"<p><strong>Background: </strong>Patients with cancer (PwC) who undergo specific treatments reported greater fatigue and reduced functional capacity as predominant outcomes, compromising their QoL during and following the treatment. Prehabilitation intervention, provided after diagnosis and before treatments, is to optimize the physiological reserve and address modifiable risk factors before surgery or chemotherapy to improve post-treatment results. The primary aim of this study is to obtain a common line of efficacy compared with prehabilitation treatment; the secondary endpoint is to assess the methodological quality of the studies eligible in the review.</p><p><strong>Methods: </strong>The systematic review was conducted from September to February 2024, in accordance with PRISMA guidelines. Databases consulted were MEDLINE, Scopus, Web of science and CINAHL, RCTs related to Prehabilitation intervention on PwC were included. The methodological quality of the included studies was assessed through the RoB2 Cochrane tool and the PEDro scale. Meta-analysis was performed to estimate relative treatment effects among evaluated outcomes.</p><p><strong>Results: </strong>Forty-two studies were included in the systematic review, 13 were included in the quantitative analysis. The results of the studies reporting reduced postoperative hospital stay, improved endurance, muscle strength, respiratory function, quality of life, and urinary incontinence. Critical analysis of the articles using the PEDro scale revealed 28 RCTs with a good rating, 9 with a fair rating, and 5 with a poor rating; in contrast, the Cochrane RoB2 tool revealed that all articles were at high risk of bias. Meta-analysis showed statistically significant values for 6MWT (38.53, 95%CI 33.03 - 44.04); HADS-depression (-0.71, 95%CI -0.93 -0.49) and HADS-anxiety (-0.49, 95%CI -0.76 -0.23).</p><p><strong>Conclusions: </strong>Prehabilitation represents a specific intervention that aims to improve postoperative outcomes in fragile patients undergoing surgery, increasing their preoperative physiological reserve in anticipation of the stress they will face and facilitating the postoperative recovery of functional capacity. Prehabilitation is a good intervention to use, especially in terms of functional capacity and mental health, the latter being very impactful in terms of reduced levels of anxiety and depression. These data make it possible to justify supportive intervention by physical therapists aimed at improving and restoring health-related QoL especially in the short term.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11494964/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495087","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Cervical cancer is the most common cause of cancer-related mortality among women in Africa. Cervical cancer screening in women is associated with decreased incidence and mortality of cervical cancer. There is a dearth of recent data regarding the prevalence and associated factors of cervical cancer screening in sub-Saharan Africa. Therefore, this study is intended to determine the prevalence and associated factors impacting cervical cancer screening among women in four sub-Saharan African countries.
Methods: Data from the recent demographic and health surveys of four countries in sub-Saharan Africa conducted between 2022 and 2023. Multilevel mixed-effects logistic regression was used to determine the factors associated with the outcome variable. Variables with a p-value < 0.05 were declared statistically significant.
Results: The prevalence of cervical cancer screening among women of childbearing age in four sub-Saharan African countries was 8.90% (95% CI: 8.67%, 9.13%). At the individual level, being older, educated, non-breastfeeding, employed, wealthier, sexually active, using contraceptives, having media exposure, visiting healthcare facilities in the last 12 months, and residing in urban areas were associated with higher odds of cervical cancer screening. At the community level, being from communities with a high level of literacy and media exposure increases the odds of cervical cancer screening among women in sub-Saharan Africa.
Conclusion: The prevalence of cervical cancer screening among childbearing-age women was found to be low. To improve cervical cancer screening practices among women of childbearing age, it is therefore advised to support women's empowerment, mass media campaigns, and regular visits to healthcare facilities.
{"title":"Determinants of cervical cancer screening among women of childbearing age in four sub-Saharan African countries: insights from large population surveys.","authors":"Enyew Getaneh Mekonen, Deresse Abebe Gebrehana, Tadesse Tarik Tamir","doi":"10.1186/s12885-024-13079-8","DOIUrl":"10.1186/s12885-024-13079-8","url":null,"abstract":"<p><strong>Background: </strong>Cervical cancer is the most common cause of cancer-related mortality among women in Africa. Cervical cancer screening in women is associated with decreased incidence and mortality of cervical cancer. There is a dearth of recent data regarding the prevalence and associated factors of cervical cancer screening in sub-Saharan Africa. Therefore, this study is intended to determine the prevalence and associated factors impacting cervical cancer screening among women in four sub-Saharan African countries.</p><p><strong>Methods: </strong>Data from the recent demographic and health surveys of four countries in sub-Saharan Africa conducted between 2022 and 2023. Multilevel mixed-effects logistic regression was used to determine the factors associated with the outcome variable. Variables with a p-value < 0.05 were declared statistically significant.</p><p><strong>Results: </strong>The prevalence of cervical cancer screening among women of childbearing age in four sub-Saharan African countries was 8.90% (95% CI: 8.67%, 9.13%). At the individual level, being older, educated, non-breastfeeding, employed, wealthier, sexually active, using contraceptives, having media exposure, visiting healthcare facilities in the last 12 months, and residing in urban areas were associated with higher odds of cervical cancer screening. At the community level, being from communities with a high level of literacy and media exposure increases the odds of cervical cancer screening among women in sub-Saharan Africa.</p><p><strong>Conclusion: </strong>The prevalence of cervical cancer screening among childbearing-age women was found to be low. To improve cervical cancer screening practices among women of childbearing age, it is therefore advised to support women's empowerment, mass media campaigns, and regular visits to healthcare facilities.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11520148/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-21DOI: 10.1186/s12885-024-13037-4
Sarah Tadhg Ferrier, Mingyang Li, Julia V Burnier
Background: Uveal melanoma (UM) is the most common primary intraocular tumour in adults, and approximately 50% of patients will develop metastasis. Epigenetic changes are a major factor in cancer progression. We aimed to determine whether methylation profiles could be altered using a DNA methyltransferase (DNMT) inhibitor in UM cell lines.
Methods: Four primary and metastatic UM cell lines were treated with azacytidine and analysed for cell proliferation, colony formation, and BAP1 protein expression. Genomic and cell-free (cf)DNA methylation were compared.
Results: In all cell lines, azacytidine treatment resulted in dose-dependent effects on proliferation, colony formation, and radiosensitivity. Methylation profiling revealed differences in methylation between cell lines according to BAP1 expression. Matched primary and metastatic cell lines showed very similar patterns. Alterations were seen in pathways known to be important in UM progression, such as PI3K/Akt and MAPK signaling, and in pathways involved in cancer progression, such as regulation of stemlike potential, cell motility, and invasion. These changes were maintained in genomic and cell-free DNA.
Conclusions: This data suggests that DNMT inhibitors cause changes in UM cells that are maintained in cfDNA. The results suggest that targeting methylation in UM treatment and monitoring response to treatment using cfDNA methylation could be a valuable tool.
背景:葡萄膜黑色素瘤(UM)是成人最常见的原发性眼内肿瘤,约 50%的患者会发生转移。表观遗传变化是癌症进展的一个主要因素。我们旨在确定在 UM 细胞系中使用 DNA 甲基转移酶(DNMT)抑制剂能否改变甲基化图谱:方法:用氮杂胞苷处理四种原发性和转移性 UM 细胞系,分析细胞增殖、集落形成和 BAP1 蛋白表达。比较了基因组和无细胞(cf)DNA甲基化情况:结果:在所有细胞系中,氮杂胞苷处理都会对细胞增殖、集落形成和放射敏感性产生剂量依赖性影响。甲基化分析表明,根据 BAP1 的表达,不同细胞系之间的甲基化存在差异。匹配的原代细胞系和转移细胞系显示出非常相似的模式。在已知对荨麻疹进展很重要的通路(如 PI3K/Akt 和 MAPK 信号转导)和涉及癌症进展的通路(如干细胞潜能、细胞运动和侵袭的调节)中都出现了变化。这些变化在基因组和无细胞DNA中得以维持:这些数据表明,DNMT 抑制剂会导致 UM 细胞发生变化,并在 cfDNA 中保持不变。结果表明,在 UM 治疗中以甲基化为靶点,并利用 cfDNA 甲基化监测治疗反应,可能是一种有价值的工具。
{"title":"Azacytidine treatment affects the methylation pattern of genomic and cell-free DNA in uveal melanoma cell lines.","authors":"Sarah Tadhg Ferrier, Mingyang Li, Julia V Burnier","doi":"10.1186/s12885-024-13037-4","DOIUrl":"10.1186/s12885-024-13037-4","url":null,"abstract":"<p><strong>Background: </strong>Uveal melanoma (UM) is the most common primary intraocular tumour in adults, and approximately 50% of patients will develop metastasis. Epigenetic changes are a major factor in cancer progression. We aimed to determine whether methylation profiles could be altered using a DNA methyltransferase (DNMT) inhibitor in UM cell lines.</p><p><strong>Methods: </strong>Four primary and metastatic UM cell lines were treated with azacytidine and analysed for cell proliferation, colony formation, and BAP1 protein expression. Genomic and cell-free (cf)DNA methylation were compared.</p><p><strong>Results: </strong>In all cell lines, azacytidine treatment resulted in dose-dependent effects on proliferation, colony formation, and radiosensitivity. Methylation profiling revealed differences in methylation between cell lines according to BAP1 expression. Matched primary and metastatic cell lines showed very similar patterns. Alterations were seen in pathways known to be important in UM progression, such as PI3K/Akt and MAPK signaling, and in pathways involved in cancer progression, such as regulation of stemlike potential, cell motility, and invasion. These changes were maintained in genomic and cell-free DNA.</p><p><strong>Conclusions: </strong>This data suggests that DNMT inhibitors cause changes in UM cells that are maintained in cfDNA. The results suggest that targeting methylation in UM treatment and monitoring response to treatment using cfDNA methylation could be a valuable tool.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11495039/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142457962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-21DOI: 10.1186/s12885-024-12927-x
Yao Zhang, Xiaolong Tang, Lin Liu, Dan Cai, Shuang Gou, Siyu Hao, Yan Li, Jing Shen, Yu Chen, Yueshui Zhao, Xu Wu, Mingxing Li, Meijuan Chen, Xiaobing Li, Yuhong Sun, Li Gu, Wanping Li, Fang Wang, Zhuo Zhang, Xiaodong Wang, Shuai Deng, Zhangang Xiao, Lei Yao, Fukuan Du
Background: Hepatocellular carcinoma (HCC) is a malignant tumor characterized by a high mortality rate. The occurrence and progression of HCC are linked to oxidative stress. Glyoxalase-1 (GLO1) plays an important role in regulating oxidative stress, yet the underlying mechanism remains unclear. GLO1 may serve as a prognostic biomarker and therapeutic target for HCC.
Methods: Based on TCGA database hepatocellular carcinoma samples, we conducted a bioinformatics analysis to explore the correlation between GLO1 expression and HCC cell proliferation and viability. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that differentially expressed genes (DEGs) were mainly enriched in the cell cycle pathway. We analyzed the relationships between GLO1 and 24 genes enriched in the cell cycle pathway using a protein-protein interaction (PPI) network. Finally, experimental validation was performed to assess GLO1's impact on the distribution of cells at different cell cycle stages and on the proliferation and migration of HCC cells.
Results: Our study demonstrated that GLO1 was overexpressed in HCC tissues and was associated with a poor prognosis. Data analysis indicated that overexpression of GLO1 activated the cell cycle pathway and positively correlated with expression of the majority of key cell cycle genes. Experimental validation showed that GLO1 expression affects the number of HCC cells in G2 and S phases and regulates HCC cell proliferation and migration.
Conclusions: GLO1 represents a promising therapeutic target for HCC, providing valuable insights into its role in the viability and proliferation of HCC cells.
{"title":"GLO1 regulates hepatocellular carcinoma proliferation and migration through the cell cycle pathway.","authors":"Yao Zhang, Xiaolong Tang, Lin Liu, Dan Cai, Shuang Gou, Siyu Hao, Yan Li, Jing Shen, Yu Chen, Yueshui Zhao, Xu Wu, Mingxing Li, Meijuan Chen, Xiaobing Li, Yuhong Sun, Li Gu, Wanping Li, Fang Wang, Zhuo Zhang, Xiaodong Wang, Shuai Deng, Zhangang Xiao, Lei Yao, Fukuan Du","doi":"10.1186/s12885-024-12927-x","DOIUrl":"10.1186/s12885-024-12927-x","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is a malignant tumor characterized by a high mortality rate. The occurrence and progression of HCC are linked to oxidative stress. Glyoxalase-1 (GLO1) plays an important role in regulating oxidative stress, yet the underlying mechanism remains unclear. GLO1 may serve as a prognostic biomarker and therapeutic target for HCC.</p><p><strong>Methods: </strong>Based on TCGA database hepatocellular carcinoma samples, we conducted a bioinformatics analysis to explore the correlation between GLO1 expression and HCC cell proliferation and viability. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that differentially expressed genes (DEGs) were mainly enriched in the cell cycle pathway. We analyzed the relationships between GLO1 and 24 genes enriched in the cell cycle pathway using a protein-protein interaction (PPI) network. Finally, experimental validation was performed to assess GLO1's impact on the distribution of cells at different cell cycle stages and on the proliferation and migration of HCC cells.</p><p><strong>Results: </strong>Our study demonstrated that GLO1 was overexpressed in HCC tissues and was associated with a poor prognosis. Data analysis indicated that overexpression of GLO1 activated the cell cycle pathway and positively correlated with expression of the majority of key cell cycle genes. Experimental validation showed that GLO1 expression affects the number of HCC cells in G2 and S phases and regulates HCC cell proliferation and migration.</p><p><strong>Conclusions: </strong>GLO1 represents a promising therapeutic target for HCC, providing valuable insights into its role in the viability and proliferation of HCC cells.</p>","PeriodicalId":9131,"journal":{"name":"BMC Cancer","volume":null,"pages":null},"PeriodicalIF":3.4,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11492659/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142457982","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}