Pub Date : 2024-09-19DOI: 10.1007/s00432-024-05952-7
Zhang Zhang, Fangfang Chen, Xiaoxiao Deng
Purpose
This study aims to utilize bioinformatics methods to systematically screen and identify susceptibility genes for cervical cancer, as well as to construct and validate an mitophagy-related genes (MRGs) diagnostic model. The objective is to increase the understanding of the disease’s pathogenesis and improve early diagnosis and treatment.
Method
We initially collected a large amount of genomic data, including gene expression profile and single nucleotide polymorphism (SNP) data, from the control group and Cervical cancer (CC) patients. Through bioinformatics analysis, which employs methods such as differential gene expression analysis and pathway enrichment analysis, we identified a set of candidate susceptibility genes associated with cervical cancer.
Results
MRGs were extracted from single-cell RNA sequencing data, and a network graph was constructed on the basis of intercellular interaction data. Furthermore, using machine learning algorithms, we constructed a clinical prognostic model and validated and optimized it via extensive clinical data. Through bioinformatics analysis, we successfully identified a group of genes whose expression significantly differed during the development of CC and revealed the biological pathways in which these genes are involved. Moreover, our constructed clinical prognostic model demonstrated excellent performance in the validation phase, accurately predicting the clinical prognosis of patients.
Conclusion
This study delves into the susceptibility genes of cervical cancer through bioinformatics approaches and successfully builds a reliable clinical prognostic model. This study not only helps uncover potential pathogenic mechanisms of cervical cancer but also provides new directions for early diagnosis and treatment of the disease.
{"title":"Screening and identification of susceptibility genes for cervical cancer via bioinformatics analysis and the construction of an mitophagy-related genes diagnostic model","authors":"Zhang Zhang, Fangfang Chen, Xiaoxiao Deng","doi":"10.1007/s00432-024-05952-7","DOIUrl":"https://doi.org/10.1007/s00432-024-05952-7","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>This study aims to utilize bioinformatics methods to systematically screen and identify susceptibility genes for cervical cancer, as well as to construct and validate an mitophagy-related genes (MRGs) diagnostic model. The objective is to increase the understanding of the disease’s pathogenesis and improve early diagnosis and treatment.</p><h3 data-test=\"abstract-sub-heading\">Method</h3><p>We initially collected a large amount of genomic data, including gene expression profile and single nucleotide polymorphism (SNP) data, from the control group and Cervical cancer (CC) patients. Through bioinformatics analysis, which employs methods such as differential gene expression analysis and pathway enrichment analysis, we identified a set of candidate susceptibility genes associated with cervical cancer.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>MRGs were extracted from single-cell RNA sequencing data, and a network graph was constructed on the basis of intercellular interaction data. Furthermore, using machine learning algorithms, we constructed a clinical prognostic model and validated and optimized it via extensive clinical data. Through bioinformatics analysis, we successfully identified a group of genes whose expression significantly differed during the development of CC and revealed the biological pathways in which these genes are involved. Moreover, our constructed clinical prognostic model demonstrated excellent performance in the validation phase, accurately predicting the clinical prognosis of patients.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>This study delves into the susceptibility genes of cervical cancer through bioinformatics approaches and successfully builds a reliable clinical prognostic model. This study not only helps uncover potential pathogenic mechanisms of cervical cancer but also provides new directions for early diagnosis and treatment of the disease.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"40 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142260979","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 : 2024-09-19DOI: 10.1007/s00432-024-05956-3
Xibin Xiao, Mengmeng Hu, Huawei Jiang, Panpan Chen, Huyi Lei
Purpose: The retrospective study was to explore the effectiveness and safety of GemOx (gemcitabine, oxaliplatin) plus sintilimab (belongs to the class of drugs known as immune checkpoint inhibitors, particularly targeting the PD-1 receptor) in relapse or refractory nodal PTCLs.
Methods: Patients with nodal PTCL who initiated salvage therapy with sintilimab and GemOx between January 2020 to September 2021 were identified from the database of the hematology department of the Second Affiliated Hospital of Zhejiang University School of Medicine. All patients received 2-4 cycles (3 weeks/cycle) of treatment of sintilimab (200 mg, I.V, D1) in combination with GemOx. Treatment response was assessed every six weeks during the salvage treatment phase. Eligible patients received maintenance therapy according to the investigator's decision. Follow-ups were routinely conducted every three months.
Results: 31 patients with r/r nodal PTCLs were enrolled, including 23 PTCL-NOS, 4 AITL, and 4 ALCL. 21 (67.7%) patients received at least two lines of therapy. 71.0% (95% CI, 53.4%-83.9%) of patients documented objective response of 2-4 cycles of sintilimab plus GemOx therapy, including 9 complete response and 13 partial response. 21 (67.7%) patients received consolidation therapy, including 5 autologous stem-cell transplantation and 12 histone deacetylase inhibitors. After a median 25.6 months follow-up, the median PFS was 22.0 (95% CI,11.8-24.7) months, and the median OS was 26.2 (95% CI, 24.4 -NA) months. 29 (93.5%) patients experienced at least one adverse event, and 26 (83.9% patients only had mild (grade 1-2) AEs.Univariable Cox regression showed the progression risk of AITL is 22.7 (3.9- 131.0, p < 0.01) times of PTCL-NOS, while the HR of ALCL was 1.14 (0.33-3.96,p = 0.833).
Conclusion: Sintilimab plus GemOx showed encouraging activity and manageable toxicity for patients with r/r PTCL.
{"title":"Sintilimab plus GemOx is an effective salvage therapy in patients with refractory/relapsing nodal peripheral T cell lymphomas.","authors":"Xibin Xiao, Mengmeng Hu, Huawei Jiang, Panpan Chen, Huyi Lei","doi":"10.1007/s00432-024-05956-3","DOIUrl":"10.1007/s00432-024-05956-3","url":null,"abstract":"<p><strong>Purpose: </strong>The retrospective study was to explore the effectiveness and safety of GemOx (gemcitabine, oxaliplatin) plus sintilimab (belongs to the class of drugs known as immune checkpoint inhibitors, particularly targeting the PD-1 receptor) in relapse or refractory nodal PTCLs.</p><p><strong>Methods: </strong>Patients with nodal PTCL who initiated salvage therapy with sintilimab and GemOx between January 2020 to September 2021 were identified from the database of the hematology department of the Second Affiliated Hospital of Zhejiang University School of Medicine. All patients received 2-4 cycles (3 weeks/cycle) of treatment of sintilimab (200 mg, I.V, D1) in combination with GemOx. Treatment response was assessed every six weeks during the salvage treatment phase. Eligible patients received maintenance therapy according to the investigator's decision. Follow-ups were routinely conducted every three months.</p><p><strong>Results: </strong>31 patients with r/r nodal PTCLs were enrolled, including 23 PTCL-NOS, 4 AITL, and 4 ALCL. 21 (67.7%) patients received at least two lines of therapy. 71.0% (95% CI, 53.4%-83.9%) of patients documented objective response of 2-4 cycles of sintilimab plus GemOx therapy, including 9 complete response and 13 partial response. 21 (67.7%) patients received consolidation therapy, including 5 autologous stem-cell transplantation and 12 histone deacetylase inhibitors. After a median 25.6 months follow-up, the median PFS was 22.0 (95% CI,11.8-24.7) months, and the median OS was 26.2 (95% CI, 24.4 -NA) months. 29 (93.5%) patients experienced at least one adverse event, and 26 (83.9% patients only had mild (grade 1-2) AEs.Univariable Cox regression showed the progression risk of AITL is 22.7 (3.9- 131.0, p < 0.01) times of PTCL-NOS, while the HR of ALCL was 1.14 (0.33-3.96,p = 0.833).</p><p><strong>Conclusion: </strong>Sintilimab plus GemOx showed encouraging activity and manageable toxicity for patients with r/r PTCL.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"425"},"PeriodicalIF":2.7,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11413062/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142287963","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}
This research aimed to evaluate the expression level of Homeobox A9 (HOXA9) and its role in tumorigenesis of hepatocellular carcinoma (HCC).
Methods
Bioinformatic analysis, qPCR and Western blot analysis of clinical samples were employed to evaluate mRNA and protein levels of HOXA9 in HCC patients and cell lines. In vitro cell proliferation, migration and invasion, cloning formation, xenograft tumor model, wound healing and apoptosis assays, RNA sequencing analysis of RPL38-silenced HCC-LM3 cells and qPCR, Western blot analysis were performed for validation. Analysis of HOXA9-related genes were conducted to identify their relationships between HOXA9.
Results
HOXA9 is dramatically upregulated in HCC. Upregulation of HOXA9 in HCC predicts poor survival of patients. Besides, HOXA9 promotes proliferation, metastasis and prevents apoptosis in HCC in vitro. In addition, HOXA9 controlled by Ribosomal protein RPL38 is upregulated in HCC. Bioinformatic analysis also indicated that HOXA9 is involved in the regulation of DNA methylation and immune infiltration in HCC.
Conclusion
HOXA9 is dramatically upregulated in hepatocellular carcinoma and predicts poor prognosis. Besides, HOXA9 promoted proliferation and metastasis and prevented apoptosis in vitro, which is regulated by Ribosomal protein RPL38 in HCC.
{"title":"HOXA9 promotes proliferation, metastasis and prevents apoptosis in hepatocellular carcinoma","authors":"Guojian Bao, Haowei Wei, Jiawu Yan, Yunzheng Li, Cailin Xue, Rao Fu, Minglu Zhang, Jialu Ding, Hengqian He, Decai Yu, Fei Yang, Beicheng Sun","doi":"10.1007/s00432-024-05950-9","DOIUrl":"https://doi.org/10.1007/s00432-024-05950-9","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>This research aimed to evaluate the expression level of Homeobox A9 (HOXA9) and its role in tumorigenesis of hepatocellular carcinoma (HCC).</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>Bioinformatic analysis, qPCR and Western blot analysis of clinical samples were employed to evaluate mRNA and protein levels of HOXA9 in HCC patients and cell lines. In vitro cell proliferation, migration and invasion, cloning formation, xenograft tumor model, wound healing and apoptosis assays, RNA sequencing analysis of RPL38-silenced HCC-LM3 cells and qPCR, Western blot analysis were performed for validation. Analysis of HOXA9-related genes were conducted to identify their relationships between HOXA9.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>HOXA9 is dramatically upregulated in HCC. Upregulation of HOXA9 in HCC predicts poor survival of patients. Besides, HOXA9 promotes proliferation, metastasis and prevents apoptosis in HCC in vitro. In addition, HOXA9 controlled by Ribosomal protein RPL38 is upregulated in HCC. Bioinformatic analysis also indicated that HOXA9 is involved in the regulation of DNA methylation and immune infiltration in HCC.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>HOXA9 is dramatically upregulated in hepatocellular carcinoma and predicts poor prognosis. Besides, HOXA9 promoted proliferation and metastasis and prevented apoptosis in vitro, which is regulated by Ribosomal protein RPL38 in HCC.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"29 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142261078","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 : 2024-09-17DOI: 10.1007/s00432-024-05943-8
Zohreh Sadat Miripour, Mina Aminifar, Parisa Hoseinpour, Fereshteh Abbasvandi, Koosha Karimi, Alireza Ghahremani, Mohammad Parniani, Mohammadreza Ghaderinia, Faride Makiyan, Parisa Aghaee, Mohammad Esmaeil Akbari, Mohammad Abdolahad
<h3 data-test="abstract-sub-heading">Purpose</h3><p>Cancer-associated fibroblasts (CAFs) are one of the most critical cells in the tumor environment, with crucial roles in cancer progression and metastasis. Due to Field-Effect phenomena (also called field cancerization), the adjacent cavity side area of the margin is histologically normal, but it has been entered into neoplastic transformation due to MCT4 and MCT1 pathways activated by H<sub>2</sub>O<sub>2</sub>/ROS oxidative stress agents secreted by CAF in adjacent tumor bed microenvironment. This paper specifically focused on the role of cancer-associated fibroblast in breast tumor beds and its correlation with the presence of scattered cancer cells or onco-protein-activated cells (may be high risk but not completely transformed cancer cells) in the cavity side margins.</p><h3 data-test="abstract-sub-heading">Methods</h3><p>In this study, the glycolytic behavior of non-tumoral cavity side margins was examined using carbon nanotube-based electrochemical biosensors integrated into a cancer diagnostic probe. This method enabled the detection of CAF accumulation sites in non-cancerous neighboring tissues of tumors, with a correlation to CAF concentration. Subsequently, RT-PCR, fluorescent, histopathological, and invasion assays were conducted on hyperglycolytic lesions to explore any correlation between the abundance of CAFs and the electrochemical responses of the non-cancerous tissues surrounding the tumor, as well as their neoplastic potential.</p><h3 data-test="abstract-sub-heading">Results</h3><p>We observed overexpression of cancer-associated transcriptomes as well as the presence and hyperactivation of CAFs in cavity-side regions in which glycolytic metabolism was recorded, independent of the histopathological state of the lesion. At mean 70.4%, 66.7%, 70.4%, and 44.5% increments were observed in GLUT-1, MMP-2, N-cadherin, and MMP-9 transcriptomes by highly glycolytic but histologically cancer-free expression samples in comparison with negative controls (histologically non-cancer lesions with low glycolytic behavior).</p><h3 data-test="abstract-sub-heading">Conclusion</h3><p>The presence of CAFs is correlated with the presence of high glycolytic metabolism in the cavity margin lesion, high ROS level in the lesion, and finally aggressive cancer-associated proteins (such as MMP2, …) in the margin while these metabolomes, molecules, and proteins are absent in the margins with negatively scored CDP response and low ROS level. So, it seems that when we observe CAFs in glycolytic lesions with high ROS levels, some high-risk epithelial breast cells may exist while no histological trace of cancer cells was observed. Further research on CAFs could provide valuable insights into the local recurrence of malignant breast diseases. Hence, real-time sensors can be used to detect and investigate CAFs in the non-tumoral regions surrounding tumors in cancer patients, potentially aiding in the prevention of
{"title":"The presence of cancer-associated fibroblast in breast cavity side margins is in correlation with the expression of oncoproteins by adjacent epithelial cells: a new era in cancerous potential","authors":"Zohreh Sadat Miripour, Mina Aminifar, Parisa Hoseinpour, Fereshteh Abbasvandi, Koosha Karimi, Alireza Ghahremani, Mohammad Parniani, Mohammadreza Ghaderinia, Faride Makiyan, Parisa Aghaee, Mohammad Esmaeil Akbari, Mohammad Abdolahad","doi":"10.1007/s00432-024-05943-8","DOIUrl":"https://doi.org/10.1007/s00432-024-05943-8","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>Cancer-associated fibroblasts (CAFs) are one of the most critical cells in the tumor environment, with crucial roles in cancer progression and metastasis. Due to Field-Effect phenomena (also called field cancerization), the adjacent cavity side area of the margin is histologically normal, but it has been entered into neoplastic transformation due to MCT4 and MCT1 pathways activated by H<sub>2</sub>O<sub>2</sub>/ROS oxidative stress agents secreted by CAF in adjacent tumor bed microenvironment. This paper specifically focused on the role of cancer-associated fibroblast in breast tumor beds and its correlation with the presence of scattered cancer cells or onco-protein-activated cells (may be high risk but not completely transformed cancer cells) in the cavity side margins.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>In this study, the glycolytic behavior of non-tumoral cavity side margins was examined using carbon nanotube-based electrochemical biosensors integrated into a cancer diagnostic probe. This method enabled the detection of CAF accumulation sites in non-cancerous neighboring tissues of tumors, with a correlation to CAF concentration. Subsequently, RT-PCR, fluorescent, histopathological, and invasion assays were conducted on hyperglycolytic lesions to explore any correlation between the abundance of CAFs and the electrochemical responses of the non-cancerous tissues surrounding the tumor, as well as their neoplastic potential.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>We observed overexpression of cancer-associated transcriptomes as well as the presence and hyperactivation of CAFs in cavity-side regions in which glycolytic metabolism was recorded, independent of the histopathological state of the lesion. At mean 70.4%, 66.7%, 70.4%, and 44.5% increments were observed in GLUT-1, MMP-2, N-cadherin, and MMP-9 transcriptomes by highly glycolytic but histologically cancer-free expression samples in comparison with negative controls (histologically non-cancer lesions with low glycolytic behavior).</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>The presence of CAFs is correlated with the presence of high glycolytic metabolism in the cavity margin lesion, high ROS level in the lesion, and finally aggressive cancer-associated proteins (such as MMP2, …) in the margin while these metabolomes, molecules, and proteins are absent in the margins with negatively scored CDP response and low ROS level. So, it seems that when we observe CAFs in glycolytic lesions with high ROS levels, some high-risk epithelial breast cells may exist while no histological trace of cancer cells was observed. Further research on CAFs could provide valuable insights into the local recurrence of malignant breast diseases. Hence, real-time sensors can be used to detect and investigate CAFs in the non-tumoral regions surrounding tumors in cancer patients, potentially aiding in the prevention of ","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"35 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142261077","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 : 2024-09-16DOI: 10.1007/s00432-024-05937-6
Ying Fang, Jun Wan, Yukai Zeng
Background
This study aims to establish a predictive model for assessing the risk of esophageal cancer lung metastasis using machine learning techniques.
Methods
Data on esophageal cancer patients from 2010 to 2020 were extracted from the surveillance, epidemiology, and end results (SEER) database. Through univariate and multivariate logistic regression analyses, eight indicators related to the risk of lung metastasis were selected. These indicators were incorporated into six machine learning classifiers to develop corresponding predictive models. The performance of these models was evaluated and compared using metrics such as The area under curve (AUC), accuracy, sensitivity, specificity, and F1 score.
Results
A total of 20,249 confirmed cases of esophageal cancer were included in this study. Among them, 14,174 cases (70%) were assigned to the training set while 6075 cases (30%) constituted the internal test set. Primary site location, tumor histology, tumor grade classification system T staging criteria N staging criteria brain metastasis bone metastasis liver metastasis emerged as independent risk factors for esophageal cancer with lung metastasis. Amongst the six constructed models, the GBM algorithm-based machine learning model demonstrated superior performance during internal dataset validation. AUC, accuracy, sensitivity, and specificity values achieved by this model stood at respectively at 0.803, 0.849, 0.604, and 0.867.
Conclusion
We have developed an online calculator based on the GBM model (https://lvgrkyxcgdvo7ugoyxyywe.streamlit.app/)to aid clinical decision-making and treatment planning.
背景本研究旨在利用机器学习技术建立一个评估食管癌肺转移风险的预测模型。方法从监测、流行病学和最终结果(SEER)数据库中提取了2010年至2020年食管癌患者的数据。通过单变量和多变量逻辑回归分析,选出了与肺转移风险相关的八个指标。这些指标被纳入六个机器学习分类器,以建立相应的预测模型。研究使用曲线下面积(AUC)、准确性、灵敏度、特异性和 F1 分数等指标对这些模型的性能进行了评估和比较。其中,14174 个病例(70%)被分配到训练集,6075 个病例(30%)构成内部测试集。原发部位、肿瘤组织学、肿瘤分级分类系统 T 分期标准 N 分期标准 脑转移 骨转移 肝转移成为食管癌肺转移的独立危险因素。在所构建的六个模型中,基于 GBM 算法的机器学习模型在内部数据集验证中表现出卓越的性能。该模型的AUC、准确性、灵敏度和特异性值分别为0.803、0.849、0.604和0.867。结论我们开发了一种基于GBM模型的在线计算器(https://lvgrkyxcgdvo7ugoyxyywe.streamlit.app/),以帮助临床决策和治疗规划。
{"title":"Use machine learning to predict pulmonary metastasis of esophageal cancer: a population-based study","authors":"Ying Fang, Jun Wan, Yukai Zeng","doi":"10.1007/s00432-024-05937-6","DOIUrl":"https://doi.org/10.1007/s00432-024-05937-6","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Background</h3><p>This study aims to establish a predictive model for assessing the risk of esophageal cancer lung metastasis using machine learning techniques.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>Data on esophageal cancer patients from 2010 to 2020 were extracted from the surveillance, epidemiology, and end results (SEER) database. Through univariate and multivariate logistic regression analyses, eight indicators related to the risk of lung metastasis were selected. These indicators were incorporated into six machine learning classifiers to develop corresponding predictive models. The performance of these models was evaluated and compared using metrics such as The area under curve (AUC), accuracy, sensitivity, specificity, and F1 score.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>A total of 20,249 confirmed cases of esophageal cancer were included in this study. Among them, 14,174 cases (70%) were assigned to the training set while 6075 cases (30%) constituted the internal test set. Primary site location, tumor histology, tumor grade classification system T staging criteria N staging criteria brain metastasis bone metastasis liver metastasis emerged as independent risk factors for esophageal cancer with lung metastasis. Amongst the six constructed models, the GBM algorithm-based machine learning model demonstrated superior performance during internal dataset validation. AUC, accuracy, sensitivity, and specificity values achieved by this model stood at respectively at 0.803, 0.849, 0.604, and 0.867.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>We have developed an online calculator based on the GBM model (https://lvgrkyxcgdvo7ugoyxyywe.streamlit.app/)to aid clinical decision-making and treatment planning.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"210 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142261080","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 : 2024-09-12DOI: 10.1007/s00432-024-05933-w
Yuan Li, Aidong Gu, Lili Yang, Qingbo Wang
Background
Hepatocellular carcinoma (LIHC) has severe consequences due to late diagnosis and the lack of effective therapies. Currently, potential biomarkers for the diagnosis and prognosis of LIHC have not been systematically evaluated. Previous studies have reported that RAC1 is associated with the B cell receptor signaling pathway in various tumor microenvironments, but its relationship with LIHC remains unclear. We investigated the relationship between RAC1 and the prognosis and immune infiltration microenvironment of LIHC, exploring its potential as a prognostic biomarker for this type of cancer.
Methods
In this study, we analyzed data from The Cancer Genome Atlas (TCGA) using the Wilcoxon signed-rank test and logistic regression to assess the association between RAC1 expression and clinical characteristics in LIHC patients. Additionally, Kaplan-Meier and Cox regression methods were employed to confirm the impact of RAC1 expression levels on overall survival. Immunohistochemistry was used to validate RAC1 protein expression in LIHC. We constructed RAC1 knockdown LIHC cells and studied the effects of RAC1 protein on cell proliferation and migration at both cellular and animal levels.
Results
RAC1 expression levels were significantly elevated in LIHC tissues compared to normal tissues. High RAC1 expression was strongly associated with advanced pathological stages and was identified as an independent factor negatively affecting overall survival. At both cellular and animal levels, RAC1 knockdown significantly inhibited the proliferation and migration of LIHC cells. Furthermore, RAC1 expression was positively correlated with the infiltration of Th2 cells and macrophages in the tumor microenvironment, suggesting that RAC1 may contribute to the deterioration of the tumor immunosuppressive microenvironment and potentially lead to reduced patient survival.
Conclusion
These findings indicate that RAC1 expression promotes LIHC proliferation and migration and influences the landscape of immune cell infiltration in the tumor microenvironment. Based on these results, RAC1 is proposed as a potential prognostic biomarker for LIHC, associated with both cancer progression and tumor immune cell infiltration.
{"title":"RAC1 serves as a prognostic factor and correlated with immune infiltration in liver hepatocellular carcinoma","authors":"Yuan Li, Aidong Gu, Lili Yang, Qingbo Wang","doi":"10.1007/s00432-024-05933-w","DOIUrl":"https://doi.org/10.1007/s00432-024-05933-w","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Background</h3><p>Hepatocellular carcinoma (LIHC) has severe consequences due to late diagnosis and the lack of effective therapies. Currently, potential biomarkers for the diagnosis and prognosis of LIHC have not been systematically evaluated. Previous studies have reported that RAC1 is associated with the B cell receptor signaling pathway in various tumor microenvironments, but its relationship with LIHC remains unclear. We investigated the relationship between RAC1 and the prognosis and immune infiltration microenvironment of LIHC, exploring its potential as a prognostic biomarker for this type of cancer.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>In this study, we analyzed data from The Cancer Genome Atlas (TCGA) using the Wilcoxon signed-rank test and logistic regression to assess the association between RAC1 expression and clinical characteristics in LIHC patients. Additionally, Kaplan-Meier and Cox regression methods were employed to confirm the impact of RAC1 expression levels on overall survival. Immunohistochemistry was used to validate RAC1 protein expression in LIHC. We constructed RAC1 knockdown LIHC cells and studied the effects of RAC1 protein on cell proliferation and migration at both cellular and animal levels.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>RAC1 expression levels were significantly elevated in LIHC tissues compared to normal tissues. High RAC1 expression was strongly associated with advanced pathological stages and was identified as an independent factor negatively affecting overall survival. At both cellular and animal levels, RAC1 knockdown significantly inhibited the proliferation and migration of LIHC cells. Furthermore, RAC1 expression was positively correlated with the infiltration of Th2 cells and macrophages in the tumor microenvironment, suggesting that RAC1 may contribute to the deterioration of the tumor immunosuppressive microenvironment and potentially lead to reduced patient survival.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>These findings indicate that RAC1 expression promotes LIHC proliferation and migration and influences the landscape of immune cell infiltration in the tumor microenvironment. Based on these results, RAC1 is proposed as a potential prognostic biomarker for LIHC, associated with both cancer progression and tumor immune cell infiltration.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"28 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142198898","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 : 2024-09-12DOI: 10.1007/s00432-024-05917-w
Dou-Dou Li, Teng Zhou, Jing Gao, Guan-Lin Wu, Guang-Rui Yang
Background and Objectives
Circadian rhythms, the endogenous biological clocks that govern physiological processes, have emerged as pivotal regulators in the development and progression of breast cancer. This comprehensive review delves into the intricate interplay between circadian disruption and breast tumorigenesis from multifaceted perspectives, encompassing biological rhythms, circadian gene regulation, tumor microenvironment dynamics, and genetic polymorphisms.
Methods and Results
Epidemiological evidence underscores the profound impact of external factors, such as night shift work, jet lag, dietary patterns, and exercise routines, on breast cancer risk and progression through the perturbation of circadian homeostasis. The review elucidates the distinct roles of key circadian genes, including CLOCK, BMAL1, PER, and CRY, in breast cancer biology, highlighting their therapeutic potential as molecular targets. Additionally, it investigates how circadian rhythm dysregulation shapes the tumor microenvironment, fostering epithelial-mesenchymal transition, chronic inflammation, and immunosuppression, thereby promoting tumor progression and metastasis. Furthermore, the review sheds light on the association between circadian gene polymorphisms and breast cancer susceptibility, paving the way for personalized risk assessment and tailored treatment strategies.
Conclusions
Importantly, it explores innovative therapeutic modalities that harness circadian rhythms, including chronotherapy, melatonin administration, and traditional Chinese medicine interventions. Overall, this comprehensive review emphasizes the critical role of circadian rhythms in the pathogenesis of breast cancer and highlights the promising prospects for the development of circadian rhythm-based interventions to enhance treatment efficacy and improve patient outcomes.
{"title":"Circadian rhythms and breast cancer: from molecular level to therapeutic advancements","authors":"Dou-Dou Li, Teng Zhou, Jing Gao, Guan-Lin Wu, Guang-Rui Yang","doi":"10.1007/s00432-024-05917-w","DOIUrl":"https://doi.org/10.1007/s00432-024-05917-w","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Background and Objectives</h3><p>Circadian rhythms, the endogenous biological clocks that govern physiological processes, have emerged as pivotal regulators in the development and progression of breast cancer. This comprehensive review delves into the intricate interplay between circadian disruption and breast tumorigenesis from multifaceted perspectives, encompassing biological rhythms, circadian gene regulation, tumor microenvironment dynamics, and genetic polymorphisms.</p><h3 data-test=\"abstract-sub-heading\">Methods and Results</h3><p>Epidemiological evidence underscores the profound impact of external factors, such as night shift work, jet lag, dietary patterns, and exercise routines, on breast cancer risk and progression through the perturbation of circadian homeostasis. The review elucidates the distinct roles of key circadian genes, including CLOCK, BMAL1, PER, and CRY, in breast cancer biology, highlighting their therapeutic potential as molecular targets. Additionally, it investigates how circadian rhythm dysregulation shapes the tumor microenvironment, fostering epithelial-mesenchymal transition, chronic inflammation, and immunosuppression, thereby promoting tumor progression and metastasis. Furthermore, the review sheds light on the association between circadian gene polymorphisms and breast cancer susceptibility, paving the way for personalized risk assessment and tailored treatment strategies.</p><h3 data-test=\"abstract-sub-heading\">Conclusions</h3><p>Importantly, it explores innovative therapeutic modalities that harness circadian rhythms, including chronotherapy, melatonin administration, and traditional Chinese medicine interventions. Overall, this comprehensive review emphasizes the critical role of circadian rhythms in the pathogenesis of breast cancer and highlights the promising prospects for the development of circadian rhythm-based interventions to enhance treatment efficacy and improve patient outcomes.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"8 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142225492","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 : 2024-09-11DOI: 10.1007/s00432-024-05936-7
O. Ortmann, S. Schüler-Toprak, K. Kast
Purpose
To support doctors in counselling women with genetic predisposition for breast or gynecologic cancers on endocrine interventions.
Methods
Evidence on the safety of endocrine interventions for fertility treatment, contraception, hormone replacement therapy after risk-reducing salpingo-oophorectomy (RRSO) or treatment of symptoms during peri- and postmenopause was analysed for carriers of probably pathogenic and pathogenic variants in BRCA1 or BRCA2 (BRCA1/2-pV), in other breast and ovarian cancer genes and the Lynch Syndrome. Cancer risks were compared with data on risks for the general population.
Results
Data on risk modulation of endocrine interventions in women with genetic predisposition is limited. Ovarian hyperstimulation for fertility treatment may be performed. Oral contraceptives should not be used to reduce ovarian cancer risk in BRCA1/2-pV carriers. Premenopausal BRCA1/2-pV carriers and carriers of pV in Lynch Syndrome genes should be offered hormone replacement therapy (HRT) after RRSO, to prevent diseases caused by estrogen deficiency.
Conclusion
Effect direction and strength of risk modulation by endocrine interventions is similar to the general population. Participation of individuals at risk in prospective registries is recommended.
{"title":"The risk of endocrine interventions in carriers of a genetic predisposition for breast and gynecologic cancers: recommendations of the German Consortium for Hereditary Breast and Ovarian Cancer","authors":"O. Ortmann, S. Schüler-Toprak, K. Kast","doi":"10.1007/s00432-024-05936-7","DOIUrl":"https://doi.org/10.1007/s00432-024-05936-7","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>To support doctors in counselling women with genetic predisposition for breast or gynecologic cancers on endocrine interventions.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>Evidence on the safety of endocrine interventions for fertility treatment, contraception, hormone replacement therapy after risk-reducing salpingo-oophorectomy (RRSO) or treatment of symptoms during peri- and postmenopause was analysed for carriers of probably pathogenic and pathogenic variants in <i>BRCA1</i> or <i>BRCA2</i> (<i>BRCA1/2</i>-pV), in other breast and ovarian cancer genes and the Lynch Syndrome. Cancer risks were compared with data on risks for the general population.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>Data on risk modulation of endocrine interventions in women with genetic predisposition is limited. Ovarian hyperstimulation for fertility treatment may be performed. Oral contraceptives should not be used to reduce ovarian cancer risk in <i>BRCA1/2</i>-pV carriers. Premenopausal <i>BRCA1/2</i>-pV carriers and carriers of pV in Lynch Syndrome genes should be offered hormone replacement therapy (HRT) after RRSO, to prevent diseases caused by estrogen deficiency.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>Effect direction and strength of risk modulation by endocrine interventions is similar to the general population. Participation of individuals at risk in prospective registries is recommended.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"42 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142225493","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}
Background: Gastric cancer (GC), a prevalent malignant tumor which is a leading cause of death from malignancy around the world. Peritoneal metastasis accounts for the major cause of mortality in patients with GC. Despite hyperthermia intraperitoneal chemotherapy (HIPEC) improves the therapeutic effect of GC, it's equivocal about the mechanism under HIPEC.
Methods: MiR-183-5p expression was sifted from miRNA chip and detected in both GC patients and cell lines by qRT-PCR. Gene interference and rescue experiments were performed to identified biological function in vitro and vivo. Next, we affirmed PPP2CA as targeted of miR-183-5p by dual luciferase reporter assay. Finally, the potential relationship between HIPEC and miR-183-5p was explored.
Results: MiR-183-5p is up-regulated in GC and associated with advanced stage and poor prognosis. MiR-183-5p accelerate GC migration in vitro which is influenced by miR-183-5p/PPP2CA/AKT/GSK3β/β-catenin Axis. HIPEC exerts migration inhibition via attenuating miR-183-5p expression.
Conclusion: MiR-183-5p can be used as a potential HIPEC biomarker in patients with CC.
{"title":"Paclitaxel hyperthermia suppresses gastric cancer migration through MiR-183-5p/PPP2CA/AKT/GSK3β/β-catenin axis.","authors":"Xiansheng Yang, Chang Liu, Zheng Li, Juncai Wen, Jinfu He, Yunxin Lu, Quanxing Liao, Tian Wang, Hongsheng Tang, Xianzi Yang, Lisi Zeng","doi":"10.1007/s00432-024-05923-y","DOIUrl":"10.1007/s00432-024-05923-y","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC), a prevalent malignant tumor which is a leading cause of death from malignancy around the world. Peritoneal metastasis accounts for the major cause of mortality in patients with GC. Despite hyperthermia intraperitoneal chemotherapy (HIPEC) improves the therapeutic effect of GC, it's equivocal about the mechanism under HIPEC.</p><p><strong>Methods: </strong>MiR-183-5p expression was sifted from miRNA chip and detected in both GC patients and cell lines by qRT-PCR. Gene interference and rescue experiments were performed to identified biological function in vitro and vivo. Next, we affirmed PPP2CA as targeted of miR-183-5p by dual luciferase reporter assay. Finally, the potential relationship between HIPEC and miR-183-5p was explored.</p><p><strong>Results: </strong>MiR-183-5p is up-regulated in GC and associated with advanced stage and poor prognosis. MiR-183-5p accelerate GC migration in vitro which is influenced by miR-183-5p/PPP2CA/AKT/GSK3β/β-catenin Axis. HIPEC exerts migration inhibition via attenuating miR-183-5p expression.</p><p><strong>Conclusion: </strong>MiR-183-5p can be used as a potential HIPEC biomarker in patients with CC.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"416"},"PeriodicalIF":2.7,"publicationDate":"2024-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11383839/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154079","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}
Pub Date : 2024-09-09DOI: 10.1007/s00432-024-05928-7
Axel Hegele, Rainer Häußermann, Stefan Schultheis, Lennart Skrobek, Meike Vink, Sebastian Hollwegs, Martin Ludwig, Petra Huwe, Manfred Maywurm, Anke Bartsch-Polle, Jost Weber, Markus Thiemer, Denny Varughese
Purpose: Apalutamide plus androgen-deprivation therapy (ADT) improved outcomes in patients with non-metastatic castration-resistant prostate cancer (nmCRPC). Nevertheless real-world data are limited. The aim of this multicenter study was to generate real-world data from nmCRPC patients treated with ADT plus apalutamide.
Methods: In this observational cohort based investigator initiated trial data of nmCRPC patients receiving apalutamide plus ADT were collected focusing on patient demographic data, prostate-specific antigen (PSA) declines, safety profile including dose modification/discontinuation as well as subsequent therapy and metastasis-free survival (MFS).
Results: Data from a total of 31 nmCRPC patients were documented. Compared to the Phase III study Spartan real-world patients are older, showed a higher ECOG-PS and more aggressive tumors. In the cohort PSA decreased about 98.1%, 74% of patients showed a PSA decrease over 90% and 54.8% reached a PSA-level < 0.2ng/ml. Apalutamide was well tolerated in real world patients: adverse events occurred in 67.7% but were in the majority mild (≥ grade 3: 6.5%). Dose reduction was necessary in 38.7% and 32.2% discontinued apalutamide treatment. MFS was 43 months and majority of patients were subsequently treated with abiraterone.
Conclusion: In real world more comorbid nmCRPC patients with a higher ECOG-PS and more aggressive tumors are treated with apalutamide plus ADT. Nevertheless efficacy results as well as side effects are similar in real-world compared to Spartan trial showing also a rapid, durable and deep PSA response with a median MFS of 43 months.
{"title":"Apalutamide for non-metastatic castration-resistant prostate cancer (nmCRPC): real world data of a multicenter study.","authors":"Axel Hegele, Rainer Häußermann, Stefan Schultheis, Lennart Skrobek, Meike Vink, Sebastian Hollwegs, Martin Ludwig, Petra Huwe, Manfred Maywurm, Anke Bartsch-Polle, Jost Weber, Markus Thiemer, Denny Varughese","doi":"10.1007/s00432-024-05928-7","DOIUrl":"10.1007/s00432-024-05928-7","url":null,"abstract":"<p><strong>Purpose: </strong>Apalutamide plus androgen-deprivation therapy (ADT) improved outcomes in patients with non-metastatic castration-resistant prostate cancer (nmCRPC). Nevertheless real-world data are limited. The aim of this multicenter study was to generate real-world data from nmCRPC patients treated with ADT plus apalutamide.</p><p><strong>Methods: </strong>In this observational cohort based investigator initiated trial data of nmCRPC patients receiving apalutamide plus ADT were collected focusing on patient demographic data, prostate-specific antigen (PSA) declines, safety profile including dose modification/discontinuation as well as subsequent therapy and metastasis-free survival (MFS).</p><p><strong>Results: </strong>Data from a total of 31 nmCRPC patients were documented. Compared to the Phase III study Spartan real-world patients are older, showed a higher ECOG-PS and more aggressive tumors. In the cohort PSA decreased about 98.1%, 74% of patients showed a PSA decrease over 90% and 54.8% reached a PSA-level < 0.2ng/ml. Apalutamide was well tolerated in real world patients: adverse events occurred in 67.7% but were in the majority mild (≥ grade 3: 6.5%). Dose reduction was necessary in 38.7% and 32.2% discontinued apalutamide treatment. MFS was 43 months and majority of patients were subsequently treated with abiraterone.</p><p><strong>Conclusion: </strong>In real world more comorbid nmCRPC patients with a higher ECOG-PS and more aggressive tumors are treated with apalutamide plus ADT. Nevertheless efficacy results as well as side effects are similar in real-world compared to Spartan trial showing also a rapid, durable and deep PSA response with a median MFS of 43 months.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"414"},"PeriodicalIF":2.7,"publicationDate":"2024-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11384626/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154077","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}