Pub Date : 2024-11-30Epub Date: 2024-11-27DOI: 10.21037/tcr-24-1619
Jiangang Xu, Liyin Zhang, Menghui Feng, Weijun Hong, Xinming Ye
Background: Research interest into regulation of gene expression by physical activity and its effect on cancer prognosis has intensified. This study investigated the role of an exercise-related gene, NUP155, in the progression of non-small cell lung cancer (NSCLC) and its potential as therapy target.
Methods: Using the GSE41914 dataset, which includes data related to exercise, and the Cancer Genome Atlas (TCGA)-NSCLC dataset, we identified differentially expressed genes (DEGs) and selected NUP155 as a hub gene for further analysis. NUP155 expression levels were measured in NSCLC cell lines and normal lung cells using in vitro assays. The functional roles of NUP155 were investigated through small interfering RNA (siRNA) knockdown experiments, assessing effects on migration, cell proliferation, invasion, and apoptosis. The involvement of the PTEN/AKT signaling pathway was examined using the PTEN inhibitor SF1670.
Results: NUP155 was downregulated in postexercise samples and upregulated in NSCLC samples, indicating its association with poor prognosis in NSCLC. Knockdown of NUP155 in NSCLC cell lines resulted in reduced cell viability, migration, and invasion, alongside increased apoptosis. Western blotting revealed that NUP155 knockdown upregulated PTEN levels and downregulated phosphorylated AKT (p-AKT), without altering total AKT levels. The addition of SF1670 partially reversed the effects of NUP155 knockdown, indicating the involvement of the signaling pathway PTEN/AKT in NUP155-mediated tumorigenesis.
Conclusions: NUP155 is upregulated in NSCLC, which promotes cell invasion and migration via the PTEN/AKT signaling pathway. Targeting NUP155, potentially influenced by exercise, could be a promising therapy. Combining exercise with targeted treatments may enhance patient outcomes.
{"title":"Postexercise downregulation of <i>NUP155</i> in regulating non-small cell lung cancer progression via the PTEN/AKT signaling pathway.","authors":"Jiangang Xu, Liyin Zhang, Menghui Feng, Weijun Hong, Xinming Ye","doi":"10.21037/tcr-24-1619","DOIUrl":"10.21037/tcr-24-1619","url":null,"abstract":"<p><strong>Background: </strong>Research interest into regulation of gene expression by physical activity and its effect on cancer prognosis has intensified. This study investigated the role of an exercise-related gene, <i>NUP155</i>, in the progression of non-small cell lung cancer (NSCLC) and its potential as therapy target.</p><p><strong>Methods: </strong>Using the GSE41914 dataset, which includes data related to exercise, and the Cancer Genome Atlas (TCGA)-NSCLC dataset, we identified differentially expressed genes (DEGs) and selected <i>NUP155</i> as a hub gene for further analysis. <i>NUP155</i> expression levels were measured in NSCLC cell lines and normal lung cells using <i>in vitro</i> assays. The functional roles of <i>NUP155</i> were investigated through small interfering RNA (siRNA) knockdown experiments, assessing effects on migration, cell proliferation, invasion, and apoptosis. The involvement of the PTEN/AKT signaling pathway was examined using the PTEN inhibitor SF1670.</p><p><strong>Results: </strong><i>NUP155</i> was downregulated in postexercise samples and upregulated in NSCLC samples, indicating its association with poor prognosis in NSCLC. Knockdown of <i>NUP155</i> in NSCLC cell lines resulted in reduced cell viability, migration, and invasion, alongside increased apoptosis. Western blotting revealed that <i>NUP155</i> knockdown upregulated PTEN levels and downregulated phosphorylated AKT (p-AKT), without altering total AKT levels. The addition of SF1670 partially reversed the effects of <i>NUP155</i> knockdown, indicating the involvement of the signaling pathway PTEN/AKT in <i>NUP155</i>-mediated tumorigenesis.</p><p><strong>Conclusions: </strong><i>NUP155</i> is upregulated in NSCLC, which promotes cell invasion and migration via the PTEN/AKT signaling pathway. Targeting <i>NUP155</i>, potentially influenced by exercise, could be a promising therapy. Combining exercise with targeted treatments may enhance patient outcomes.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"6323-6335"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651763/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-30Epub Date: 2024-05-21DOI: 10.21037/tcr-23-2234
J Patrick Mershon, Tasha Posid, Keyan Salari, Richard S Matulewicz, Eric A Singer, Shawn Dason
Background: OpenAI's ChatGPT is a large language model-based artificial intelligence (AI) chatbot that can be used to answer unique, user-generated questions without direct training on specific content. Large language models have significant potential in urologic education. We reviewed the primary data surrounding the use of large language models in urology. We also reported findings of our primary study assessing the performance of ChatGPT in renal cell carcinoma (RCC) education.
Methods: For our primary study, we utilized three professional society guidelines addressing RCC to generate fifteen content questions. These questions were inputted into ChatGPT 3.5. ChatGPT responses along with pre- and post-content assessment questions regarding ChatGPT were then presented to evaluators. Evaluators consisted of four urologic oncologists and four non-clinical staff members. Medline was reviewed for additional studies pertaining to the use of ChatGPT in urologic education.
Results: We found that all assessors rated ChatGPT highly on the accuracy and usefulness of information provided with overall mean scores of 3.64 [±0.62 standard deviation (SD)] and 3.58 (±0.75) out of 5, respectively. Clinicians and non-clinicians did not differ in their scoring of responses (P=0.37). Completing content assessment improved confidence in the accuracy of ChatGPT's information (P=0.01) and increased agreement that it should be used for medical education (P=0.007). Attitudes towards use for patient education did not change (P=0.30). We also review the current state of the literature regarding ChatGPT use for patient and trainee education and discuss future steps towards optimization.
Conclusions: ChatGPT has significant potential utility in medical education if it can continue to provide accurate and useful information. We have found it to be a useful adjunct to expert human guidance both for medical trainee and, less so, for patient education. Further work is needed to validate ChatGPT before widespread adoption.
{"title":"Integrating artificial intelligence in renal cell carcinoma: evaluating ChatGPT's performance in educating patients and trainees.","authors":"J Patrick Mershon, Tasha Posid, Keyan Salari, Richard S Matulewicz, Eric A Singer, Shawn Dason","doi":"10.21037/tcr-23-2234","DOIUrl":"10.21037/tcr-23-2234","url":null,"abstract":"<p><strong>Background: </strong>OpenAI's ChatGPT is a large language model-based artificial intelligence (AI) chatbot that can be used to answer unique, user-generated questions without direct training on specific content. Large language models have significant potential in urologic education. We reviewed the primary data surrounding the use of large language models in urology. We also reported findings of our primary study assessing the performance of ChatGPT in renal cell carcinoma (RCC) education.</p><p><strong>Methods: </strong>For our primary study, we utilized three professional society guidelines addressing RCC to generate fifteen content questions. These questions were inputted into ChatGPT 3.5. ChatGPT responses along with pre- and post-content assessment questions regarding ChatGPT were then presented to evaluators. Evaluators consisted of four urologic oncologists and four non-clinical staff members. Medline was reviewed for additional studies pertaining to the use of ChatGPT in urologic education.</p><p><strong>Results: </strong>We found that all assessors rated ChatGPT highly on the accuracy and usefulness of information provided with overall mean scores of 3.64 [±0.62 standard deviation (SD)] and 3.58 (±0.75) out of 5, respectively. Clinicians and non-clinicians did not differ in their scoring of responses (P=0.37). Completing content assessment improved confidence in the accuracy of ChatGPT's information (P=0.01) and increased agreement that it should be used for medical education (P=0.007). Attitudes towards use for patient education did not change (P=0.30). We also review the current state of the literature regarding ChatGPT use for patient and trainee education and discuss future steps towards optimization.</p><p><strong>Conclusions: </strong>ChatGPT has significant potential utility in medical education if it can continue to provide accurate and useful information. We have found it to be a useful adjunct to expert human guidance both for medical trainee and, less so, for patient education. Further work is needed to validate ChatGPT before widespread adoption.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"6246-6254"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651803/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855295","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is a prevalent type of cancer with high incidence and mortality rates. It is the third most common cause of cancer-related deaths. CD8<sup>+</sup> T cell exhaustion (TEX) is a progressive decline in T cell function due to sustained T cell receptor stimulation from continuous antigen exposure. Studies have shown that CD8<sup>+</sup> TEX plays an important role in the anti-tumor immune process and is significantly correlated with patient prognosis. The aim of the research is to establish a reliable CD8<sup>+</sup> TEX-based signature using single-cell RNA sequencing (scRNA-seq) and high-throughput RNA sequencing (RNA-seq), providing a new approach to evaluate HCC patient prognosis and immune microenvironment.</p><p><strong>Methods: </strong>The RNA-seq data of HCC patients were download from three different databases: The Cancer Genome Atlas (TCGA), the Gene Expression Omnibus (GEO), and the International Cancer Genome Consortium (ICGC). HCC's 10× scRNA data were acquired from GSE149614. Based on single-cell sequencing data, CD8<sup>+</sup> TEX-related genes were identified using uniform manifold approximation and projection (UMAP) algorithm, singleR, and marker gene methods. Afterwards, we proceeded to construct CD8<sup>+</sup> TEX signature using differential gene analysis, univariate Cox regression analysis, least absolute shrinkage and selection operator (LASSO) regression, and multivariate Cox regression analysis. We also validated the CD8<sup>+</sup> TEX signature in GEO and ICGC external cohorts and investigated clinical characteristics, chemotherapy sensitivity, mutation landscape, functional analysis, and immune cell infiltration in different risk groups.</p><p><strong>Results: </strong>The CD8<sup>+</sup> TEX signature, consisting of 13 genes (<i>HSPD1</i>, <i>UBB</i>, <i>DNAJB4,</i> <i>CALM1</i>, <i>LGALS3</i>, <i>BATF</i>, <i>COMMD3</i>, <i>IL7R</i>, <i>FDPS</i>, <i>DRAP1</i>, <i>RPS27L</i>, <i>PAPOLA</i>, <i>GPR171</i>), was found to have a strong predictive effect on the prognosis of HCC. The Kaplan-Meier (KM) analysis showed that the overall survival (OS) rate of patients in the low-risk group was higher than that of patients in the high-risk group across different datasets and specific populations. The research findings suggested that the risk score was an independent predictor of HCC prognosis. The model based on clinical features and risk score has a strong predictive effect. We observed significant differences among various risk groups in terms of clinical characteristics, functional analysis, mutation landscape, chemotherapy sensitivity, and immune cell infiltration.</p><p><strong>Conclusions: </strong>We constructed a CD8<sup>+</sup> TEX signature to predict the survival probability of patients with HCC. We also found that the model could predict the sensitivity of targeted drugs and immune cell infiltration, and the risk score was negatively correlated with CD8<sup
{"title":"Identification of the CD8<sup>+</sup> T-cell exhaustion signature of hepatocellular carcinoma for the prediction of prognosis and immune microenvironment by integrated analysis of bulk- and single-cell RNA sequencing data.","authors":"Jianhui Fan, Qinghua Zhang, Tiancong Huang, Haitao Li, Guoxu Fang","doi":"10.21037/tcr-24-650","DOIUrl":"10.21037/tcr-24-650","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is a prevalent type of cancer with high incidence and mortality rates. It is the third most common cause of cancer-related deaths. CD8<sup>+</sup> T cell exhaustion (TEX) is a progressive decline in T cell function due to sustained T cell receptor stimulation from continuous antigen exposure. Studies have shown that CD8<sup>+</sup> TEX plays an important role in the anti-tumor immune process and is significantly correlated with patient prognosis. The aim of the research is to establish a reliable CD8<sup>+</sup> TEX-based signature using single-cell RNA sequencing (scRNA-seq) and high-throughput RNA sequencing (RNA-seq), providing a new approach to evaluate HCC patient prognosis and immune microenvironment.</p><p><strong>Methods: </strong>The RNA-seq data of HCC patients were download from three different databases: The Cancer Genome Atlas (TCGA), the Gene Expression Omnibus (GEO), and the International Cancer Genome Consortium (ICGC). HCC's 10× scRNA data were acquired from GSE149614. Based on single-cell sequencing data, CD8<sup>+</sup> TEX-related genes were identified using uniform manifold approximation and projection (UMAP) algorithm, singleR, and marker gene methods. Afterwards, we proceeded to construct CD8<sup>+</sup> TEX signature using differential gene analysis, univariate Cox regression analysis, least absolute shrinkage and selection operator (LASSO) regression, and multivariate Cox regression analysis. We also validated the CD8<sup>+</sup> TEX signature in GEO and ICGC external cohorts and investigated clinical characteristics, chemotherapy sensitivity, mutation landscape, functional analysis, and immune cell infiltration in different risk groups.</p><p><strong>Results: </strong>The CD8<sup>+</sup> TEX signature, consisting of 13 genes (<i>HSPD1</i>, <i>UBB</i>, <i>DNAJB4,</i> <i>CALM1</i>, <i>LGALS3</i>, <i>BATF</i>, <i>COMMD3</i>, <i>IL7R</i>, <i>FDPS</i>, <i>DRAP1</i>, <i>RPS27L</i>, <i>PAPOLA</i>, <i>GPR171</i>), was found to have a strong predictive effect on the prognosis of HCC. The Kaplan-Meier (KM) analysis showed that the overall survival (OS) rate of patients in the low-risk group was higher than that of patients in the high-risk group across different datasets and specific populations. The research findings suggested that the risk score was an independent predictor of HCC prognosis. The model based on clinical features and risk score has a strong predictive effect. We observed significant differences among various risk groups in terms of clinical characteristics, functional analysis, mutation landscape, chemotherapy sensitivity, and immune cell infiltration.</p><p><strong>Conclusions: </strong>We constructed a CD8<sup>+</sup> TEX signature to predict the survival probability of patients with HCC. We also found that the model could predict the sensitivity of targeted drugs and immune cell infiltration, and the risk score was negatively correlated with CD8<sup","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"5856-5872"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651828/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855262","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-30Epub Date: 2024-11-12DOI: 10.21037/tcr-24-1131
Yoon Jung Jang, Heyjin Kim, Sang-Young Ryu, Moon-Hong Kim, Beob-Jong Kim, Hee Jung Jung, Jisik Kang, Sung Hyun Yang, Im Il Na, Hyo-Rak Lee, Hye Jin Kang
Background: Prior prospective studies have demonstrated the efficacy of poly(adenosine diphosphate-ribose) polymerase inhibitors (PARPis) in various cancers with mutations in the breast cancer gene (BRCA), such as ovarian and breast cancers. However, PARPi have also been associated with an increased incidence of therapy-related myeloid neoplasms (t-MNs). This study aimed to investigate the incidence of t-MNs following PARPi therapy in patients with ovarian or primary peritoneal cancer in Korea and to identify related risk factors.
Methods: We retrospectively analyzed data of patients with ovarian or primary peritoneal cancer who received PARPi therapy between January 2015 and June 2023.
Results: Among 52 patients treated with PARPi, four were diagnosed with t-MNs. All four patients had BRCA mutations, and two of them had breast cancer with no evidence of disease (NED) status following treatment. All patients received radiotherapy and at least one granulocyte-colony stimulating factor (G-CSF) application. The median duration of PARPi therapy was 16.3 (range, 6.2-48.8) months. At the time of analysis, three patients had metastatic ovarian cancer and one maintained the NED status. Next-generation sequencing (NGS) performed in four patients revealed TP53 mutations and complex karyotypes in all tested patients. Among the four patients, three received only supportive care, and one was actively undergoing t-MN treatment.
Conclusions: The incidence of t-MNs after PARPi therapy in the current study was higher than that of overall t-MNs, which is consistent with the results of previous studies on t-MNs after PARPi therapy. Further international studies are needed to elucidate the mechanism and clinical characteristics of t-MNs associated with PARPi therapy.
{"title":"Therapy-related myeloid neoplasms in Korean patients with ovarian or primary peritoneal cancer treated with poly(ADP-ribose) polymerase inhibitors.","authors":"Yoon Jung Jang, Heyjin Kim, Sang-Young Ryu, Moon-Hong Kim, Beob-Jong Kim, Hee Jung Jung, Jisik Kang, Sung Hyun Yang, Im Il Na, Hyo-Rak Lee, Hye Jin Kang","doi":"10.21037/tcr-24-1131","DOIUrl":"10.21037/tcr-24-1131","url":null,"abstract":"<p><strong>Background: </strong>Prior prospective studies have demonstrated the efficacy of poly(adenosine diphosphate-ribose) polymerase inhibitors (PARPis) in various cancers with mutations in the breast cancer gene (<i>BRCA</i>), such as ovarian and breast cancers. However, PARPi have also been associated with an increased incidence of therapy-related myeloid neoplasms (t-MNs). This study aimed to investigate the incidence of t-MNs following PARPi therapy in patients with ovarian or primary peritoneal cancer in Korea and to identify related risk factors.</p><p><strong>Methods: </strong>We retrospectively analyzed data of patients with ovarian or primary peritoneal cancer who received PARPi therapy between January 2015 and June 2023.</p><p><strong>Results: </strong>Among 52 patients treated with PARPi, four were diagnosed with t-MNs. All four patients had <i>BRCA</i> mutations, and two of them had breast cancer with no evidence of disease (NED) status following treatment. All patients received radiotherapy and at least one granulocyte-colony stimulating factor (G-CSF) application. The median duration of PARPi therapy was 16.3 (range, 6.2-48.8) months. At the time of analysis, three patients had metastatic ovarian cancer and one maintained the NED status. Next-generation sequencing (NGS) performed in four patients revealed <i>TP</i>53 mutations and complex karyotypes in all tested patients. Among the four patients, three received only supportive care, and one was actively undergoing t-MN treatment.</p><p><strong>Conclusions: </strong>The incidence of t-MNs after PARPi therapy in the current study was higher than that of overall t-MNs, which is consistent with the results of previous studies on t-MNs after PARPi therapy. Further international studies are needed to elucidate the mechanism and clinical characteristics of t-MNs associated with PARPi therapy.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"6018-6027"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651792/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-30Epub Date: 2024-11-27DOI: 10.21037/tcr-24-533
Long Guo, Na Chen, Mei Qiu, Juliang Yang, Min Zhou, Fei Liu
Background: Immunogenic cell death (ICD) has been verified as a modality of regulated cell death (RCD). Bladder cancer (BC) is a common malignant tumor and ranks tenth in the incidence of global tumor epidemiology. We conducted this study to understand the relationship between ICD and BC and benefit clinical practice.
Methods: Transcriptome and clinical profiling, mutational data of patients were downloaded from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database. BC patients were divided into ICD-high and -low risk subgroups via consensus clusters. Functional enrichment, somatic mutation analysis, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to explore the potential mechanism. An ICD-related risk signature was constructed via least absolute shrinkage and selection operator (LASSO) regression analysis. Immune infiltration was investigated and multiplexed immunofluorescence staining was used to validate the BC microenvironment. Immune landscape was summarized to show the potential of immunotherapy.
Results: A total of 18 differentially expressed ICD-related genes in BC were distinguished from normal tissue. We identified two clusters and BC patients were divided into ICD-high and -low subgroups in the TCGA BC cohort. The ICD-high subgroup exhibited worse clinical outcomes, different mutation profiles, different functional enrichment, higher immune infiltration, and better immunotherapy response. An ICD-related risk signature made of seven ICD-related genes was established and shown to have outstanding predictive power of prognosis via LASSO Cox regression.
Conclusions: An ICD-related risk signature was established that provides a promising classification system to predict the prognosis in BC patients accurately. The signature provides a novel strategy for immunotherapy of BC.
背景:免疫原性细胞死亡(ICD)已被证实是一种调节性细胞死亡(RCD)。膀胱癌(BC)是一种常见的恶性肿瘤,在全球肿瘤流行病学发病率中排名第十。我们进行这项研究是为了了解ICD和BC之间的关系,并有利于临床实践。方法:从The Cancer Genome Atlas (TCGA)和Gene Expression Omnibus (GEO)数据库下载患者的转录组和临床分析、突变数据。通过共识聚类将BC患者分为icd高危亚组和低危亚组。利用功能富集、体细胞突变分析、基因本体(GO)和京都基因与基因组百科全书(KEGG)来探索潜在的机制。通过最小绝对收缩和选择算子(LASSO)回归分析构建了icd相关的风险特征。采用免疫浸润法和多重免疫荧光染色法对BC微环境进行验证。综述了免疫景观,以显示免疫治疗的潜力。结果:BC与正常组织中icd相关基因共有18个差异表达。在TCGA BC队列中,我们确定了两个簇,并将BC患者分为icd高亚组和低亚组。icd高亚组临床预后较差,突变谱不同,功能富集程度不同,免疫浸润程度较高,免疫治疗反应较好。建立了由7个icd相关基因组成的icd相关风险标记,并通过LASSO Cox回归显示出对预后的出色预测能力。结论:建立了与icd相关的风险特征,为准确预测BC患者的预后提供了一个有希望的分类系统。该特征为BC的免疫治疗提供了一种新的策略。
{"title":"Immunogenic cell death-related signature predicts prognosis and immunotherapy efficacy in bladder cancer.","authors":"Long Guo, Na Chen, Mei Qiu, Juliang Yang, Min Zhou, Fei Liu","doi":"10.21037/tcr-24-533","DOIUrl":"10.21037/tcr-24-533","url":null,"abstract":"<p><strong>Background: </strong>Immunogenic cell death (ICD) has been verified as a modality of regulated cell death (RCD). Bladder cancer (BC) is a common malignant tumor and ranks tenth in the incidence of global tumor epidemiology. We conducted this study to understand the relationship between ICD and BC and benefit clinical practice.</p><p><strong>Methods: </strong>Transcriptome and clinical profiling, mutational data of patients were downloaded from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database. BC patients were divided into ICD-high and -low risk subgroups via consensus clusters. Functional enrichment, somatic mutation analysis, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to explore the potential mechanism. An ICD-related risk signature was constructed via least absolute shrinkage and selection operator (LASSO) regression analysis. Immune infiltration was investigated and multiplexed immunofluorescence staining was used to validate the BC microenvironment. Immune landscape was summarized to show the potential of immunotherapy.</p><p><strong>Results: </strong>A total of 18 differentially expressed ICD-related genes in BC were distinguished from normal tissue. We identified two clusters and BC patients were divided into ICD-high and -low subgroups in the TCGA BC cohort. The ICD-high subgroup exhibited worse clinical outcomes, different mutation profiles, different functional enrichment, higher immune infiltration, and better immunotherapy response. An ICD-related risk signature made of seven ICD-related genes was established and shown to have outstanding predictive power of prognosis via LASSO Cox regression.</p><p><strong>Conclusions: </strong>An ICD-related risk signature was established that provides a promising classification system to predict the prognosis in BC patients accurately. The signature provides a novel strategy for immunotherapy of BC.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"5801-5814"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651764/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855269","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p><strong>Background: </strong>Glioblastoma multiforme (GBM), the most prevalent and aggressive primary brain tumor, poses substantial challenges in both treatment and prognosis. Post-translational modifications, like palmitoylation, are known to have critical roles in the development and progression of glioma. Yet, the molecular mechanisms involved in palmitoylation and its prognostic significance in GBM are still not fully understood. This study aimed to explore prognostic biomarkers for GBM based on palmitoylation-related genes and to construct a prognostic risk model.</p><p><strong>Methods: </strong>The messenger ribonucleic acid (mRNA) expressions data and the clinical information were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) to explore palmitoylation-related mechanisms in GBM. The Cox regression analysis was performed to identify prognostic palmitoylation-related genes and the consensus clustering was used for molecular classification. The package "limma" was used for differential gene expression analysis and the least absolute shrinkage and selection operator (LASSO) regression was applied to construct a risk signature. A nomogram model was established using the risk score and clinical variables. Receiver operating characteristic (ROC), calibration curve, and decision curve analysis (DCA) were used to assess the predicted accuracy and clinical benefit of the model. The difference in immune cell infiltration was compared between different risk groups. The drug susceptibility analysis and immunotherapy response prediction were conducted to access the ability of the risk signature in predicting the therapeutic effect.</p><p><strong>Results: </strong>Based on datasets from TCGA, five palmitoylation-related genes were identified as prognostic markers, allowing for the categorization of GBM patients into two subtypes with differing survival rates. Through differential expression analysis, 570 specific genes linked to GBM advancement were uncovered. A total of seven signature genes (<i>COL22A1</i>, <i>IGFBP6</i>, <i>SOD3</i>, <i>UPP1</i>, <i>CA14</i>, <i>TIMP4</i> and <i>FERMT1</i>) were applied to establish a prognostic risk model, which was demonstrated to be an independent prognostic indicator for patients with GBM. Kaplan-Meier analysis indicted that the GBM patients in low-risk group exhibited a better survival outcome compared the patients in high-risk group. The ROC curve analyses demonstrated that the risk score model was reliable. The nomograms showed excellent predictive ability. Two external cohort of patients from the GSE74187 and GSE83300 in the GEO database confirmed the model's strong predictive performance. The immune infiltration, drug sensitivity and immunotherapy responses were significantly different between the low- and high-risk groups.</p><p><strong>Conclusions: </strong>Our study offers insights into the molecular classification and prognostic assessment of GBM, focusing on palmitoy
背景:多形性胶质母细胞瘤(GBM)是最常见和侵袭性最强的原发性脑肿瘤,在治疗和预后方面都面临着巨大的挑战。翻译后修饰,如棕榈酰化,在胶质瘤的发生和发展中起着至关重要的作用。然而,棕榈酰化的分子机制及其在GBM中的预后意义尚不完全清楚。本研究旨在探索基于棕榈酰化相关基因的GBM预后生物标志物,并构建预后风险模型。方法:从Cancer Genome Atlas (TCGA)和Gene Expression Omnibus (GEO)下载信使核糖核酸(mRNA)表达数据和临床资料,探讨棕榈酰化在GBM中的相关机制。采用Cox回归分析确定预后棕榈酰化相关基因,并采用共识聚类进行分子分类。采用“limma”包进行差异基因表达分析,采用最小绝对收缩和选择算子(LASSO)回归构建风险特征。采用风险评分和临床变量建立nomogram模型。采用受试者工作特征(ROC)、校准曲线和决策曲线分析(DCA)评估模型的预测准确性和临床获益。比较不同危险组间免疫细胞浸润的差异。通过药物敏感性分析和免疫治疗反应预测,了解风险特征对疗效的预测能力。结果:基于TCGA的数据集,五个棕榈酰化相关基因被确定为预后标志物,允许将GBM患者分为两种生存率不同的亚型。通过差异表达分析,发现了570个与GBM进展相关的特异性基因。共应用COL22A1、IGFBP6、SOD3、UPP1、CA14、TIMP4、FERMT1 7个特征基因建立预后风险模型,证明该模型是GBM患者独立的预后指标。Kaplan-Meier分析表明,低危组GBM患者的生存结局优于高危组。ROC曲线分析表明,风险评分模型是可靠的。图显示了良好的预测能力。GEO数据库中来自GSE74187和GSE83300的两个外部队列患者证实了该模型的强大预测性能。免疫浸润、药物敏感性和免疫治疗反应在低危组和高危组之间存在显著差异。结论:我们的研究为GBM的分子分类和预后评估提供了见解,重点是棕榈酰化相关机制。我们构建的预后模型为GBM患者定制个性化治疗策略提供了有价值的指导。
{"title":"Construction and validation of a novel prognostic model with palmitoylation-related genes for glioblastoma.","authors":"Guowen Qin, Gang Pang, Shuaishuai Wu, Shuiqing Bi, Shengyong Lan, Xiuwen Tang, Beiquan Hu, Junlin Zhou, Fengning Shi, Chengjian Qin","doi":"10.21037/tcr-24-787","DOIUrl":"10.21037/tcr-24-787","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma multiforme (GBM), the most prevalent and aggressive primary brain tumor, poses substantial challenges in both treatment and prognosis. Post-translational modifications, like palmitoylation, are known to have critical roles in the development and progression of glioma. Yet, the molecular mechanisms involved in palmitoylation and its prognostic significance in GBM are still not fully understood. This study aimed to explore prognostic biomarkers for GBM based on palmitoylation-related genes and to construct a prognostic risk model.</p><p><strong>Methods: </strong>The messenger ribonucleic acid (mRNA) expressions data and the clinical information were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) to explore palmitoylation-related mechanisms in GBM. The Cox regression analysis was performed to identify prognostic palmitoylation-related genes and the consensus clustering was used for molecular classification. The package \"limma\" was used for differential gene expression analysis and the least absolute shrinkage and selection operator (LASSO) regression was applied to construct a risk signature. A nomogram model was established using the risk score and clinical variables. Receiver operating characteristic (ROC), calibration curve, and decision curve analysis (DCA) were used to assess the predicted accuracy and clinical benefit of the model. The difference in immune cell infiltration was compared between different risk groups. The drug susceptibility analysis and immunotherapy response prediction were conducted to access the ability of the risk signature in predicting the therapeutic effect.</p><p><strong>Results: </strong>Based on datasets from TCGA, five palmitoylation-related genes were identified as prognostic markers, allowing for the categorization of GBM patients into two subtypes with differing survival rates. Through differential expression analysis, 570 specific genes linked to GBM advancement were uncovered. A total of seven signature genes (<i>COL22A1</i>, <i>IGFBP6</i>, <i>SOD3</i>, <i>UPP1</i>, <i>CA14</i>, <i>TIMP4</i> and <i>FERMT1</i>) were applied to establish a prognostic risk model, which was demonstrated to be an independent prognostic indicator for patients with GBM. Kaplan-Meier analysis indicted that the GBM patients in low-risk group exhibited a better survival outcome compared the patients in high-risk group. The ROC curve analyses demonstrated that the risk score model was reliable. The nomograms showed excellent predictive ability. Two external cohort of patients from the GSE74187 and GSE83300 in the GEO database confirmed the model's strong predictive performance. The immune infiltration, drug sensitivity and immunotherapy responses were significantly different between the low- and high-risk groups.</p><p><strong>Conclusions: </strong>Our study offers insights into the molecular classification and prognostic assessment of GBM, focusing on palmitoy","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"6117-6135"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651772/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855433","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-30Epub Date: 2024-11-20DOI: 10.21037/tcr-24-611
Rui Luo, Shu Huang, Xiaomin Shi, Huan Xu, Jieyu Peng, Wenjie Lei, Shiqi Li, Wei Zhang, Lei Shi, Yan Peng, Xiaowei Tang
Background: Characterized by its high mortality and easy recurrence, hepatocellular carcinoma (HCC) poses significant clinical challenges. The association between copper metabolism and development of cancer has been identified. However, the underlying mechanisms of copper metabolism-related long non-coding RNAs (CMRLs) in HCC remain elusive. To address the gap, our study analyzed the prognostic and immuno-therapeutic value of CMRLs in HCC.
Methods: This research utilized The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) data (n=424) for analysis, applying the "limma" package in R software for differential gene analysis and construction of a prognostic signature. We validated the signature using training and validation groups stochastically divided at a ratio of 1:1 and assessed prognostic value via Kaplan-Meier, C-index, and receiver operating characteristic (ROC) curves. By multivariate Cox regression, independent prognostic indicators were identified, and a nomogram was formulated for survival forecasting. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses elucidated biological pathways, and the immune landscape was examined through multiple algorithms. Finally, drug sensitivity was determined from Genomics of Drug Sensitivity in Cancer (GDSC), with mutation analysis conducted via maftools.
Results: In this study, a predictive model based on four pivotal CMRLs (PRRT3-AS1, AC108752.1, AC092115.3, AL031985.3) significantly associated with HCC progression and prognosis was constructed and validated with the overall survival (OS) prediction area under the curve (AUC) values for 1, 3, and 5 years of 0.718, 0.688, and 0.669, respectively. The calibration curves and C-index values showed a solid prognostic ability of the nomogram. The high-risk group was notably higher than the low-risk group both in OS and tumor mutational burdens (TMBs). Moreover, functional annotation enrichment analysis of CMRLs revealed that the signature was mainly associated with mitotic function, chromosome, kinetochore, cell cycle, and oocyte meiosis. Furthermore, therapeutic drugs, including fluorouracil, afatinib, alpelisib, cedranib, crizotinib, erlotinib, gefitinib, and ipatasertib, were found to induce higher sensitivity in high-risk group.
Conclusions: The prognostic signature consisting of four CMRLs displays an outstanding predictive performance and improves the precision of immuno-oncology.
背景:肝细胞癌(HCC)具有高死亡率和易复发的特点,给临床带来了重大挑战。铜代谢与癌症发展之间的关系已被确定。然而,铜代谢相关的长链非编码rna (CMRLs)在HCC中的潜在机制仍然是未知的。为了弥补这一空白,我们的研究分析了cmls在HCC中的预后和免疫治疗价值。方法:本研究利用The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC)数据(n=424)进行分析,应用R软件中的“limma”软件包进行差异基因分析并构建预后特征。我们使用训练组和验证组(按1:1的比例随机分组)验证签名,并通过Kaplan-Meier、c -指数和受试者工作特征(ROC)曲线评估预后价值。通过多变量Cox回归,确定了独立的预后指标,并制定了生存预测的nomogram。基因本体(GO)和京都基因与基因组百科全书(KEGG)分析阐明了生物途径,并通过多种算法检查了免疫景观。最后,通过癌症药物敏感性基因组学(GDSC)测定药物敏感性,并通过maftools进行突变分析。结果:本研究构建了基于4个与HCC进展和预后显著相关的关键性CMRLs (PRRT3-AS1、AC108752.1、AC092115.3、AL031985.3)的预测模型,1年、3年和5年的总生存期(OS)预测曲线下面积(AUC)值分别为0.718、0.688和0.669。校准曲线和c指数值显示了nomogram可靠的预测能力。在OS和肿瘤突变负荷(TMBs)方面,高危组明显高于低危组。此外,CMRLs的功能注释富集分析显示,该特征主要与有丝分裂功能、染色体、着丝点、细胞周期和卵母细胞减数分裂有关。此外,治疗药物氟脲嘧啶、阿法替尼、阿非利西、西德拉尼、克里唑替尼、厄洛替尼、吉非替尼和伊帕他塞替在高危人群中具有更高的敏感性。结论:由4个CMRLs组成的预后特征具有较好的预测效果,提高了免疫肿瘤学的准确性。
{"title":"Copper metabolism-related lncRNAs predict prognosis and immune landscape in liver cancer patients.","authors":"Rui Luo, Shu Huang, Xiaomin Shi, Huan Xu, Jieyu Peng, Wenjie Lei, Shiqi Li, Wei Zhang, Lei Shi, Yan Peng, Xiaowei Tang","doi":"10.21037/tcr-24-611","DOIUrl":"10.21037/tcr-24-611","url":null,"abstract":"<p><strong>Background: </strong>Characterized by its high mortality and easy recurrence, hepatocellular carcinoma (HCC) poses significant clinical challenges. The association between copper metabolism and development of cancer has been identified. However, the underlying mechanisms of copper metabolism-related long non-coding RNAs (CMRLs) in HCC remain elusive. To address the gap, our study analyzed the prognostic and immuno-therapeutic value of CMRLs in HCC.</p><p><strong>Methods: </strong>This research utilized The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) data (n=424) for analysis, applying the \"limma\" package in R software for differential gene analysis and construction of a prognostic signature. We validated the signature using training and validation groups stochastically divided at a ratio of 1:1 and assessed prognostic value via Kaplan-Meier, C-index, and receiver operating characteristic (ROC) curves. By multivariate Cox regression, independent prognostic indicators were identified, and a nomogram was formulated for survival forecasting. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses elucidated biological pathways, and the immune landscape was examined through multiple algorithms. Finally, drug sensitivity was determined from Genomics of Drug Sensitivity in Cancer (GDSC), with mutation analysis conducted via maftools.</p><p><strong>Results: </strong>In this study, a predictive model based on four pivotal CMRLs (<i>PRRT3-AS1</i>, <i>AC108752.1</i>, <i>AC092115.3</i>, <i>AL031985.3</i>) significantly associated with HCC progression and prognosis was constructed and validated with the overall survival (OS) prediction area under the curve (AUC) values for 1, 3, and 5 years of 0.718, 0.688, and 0.669, respectively. The calibration curves and C-index values showed a solid prognostic ability of the nomogram. The high-risk group was notably higher than the low-risk group both in OS and tumor mutational burdens (TMBs). Moreover, functional annotation enrichment analysis of CMRLs revealed that the signature was mainly associated with mitotic function, chromosome, kinetochore, cell cycle, and oocyte meiosis. Furthermore, therapeutic drugs, including fluorouracil, afatinib, alpelisib, cedranib, crizotinib, erlotinib, gefitinib, and ipatasertib, were found to induce higher sensitivity in high-risk group.</p><p><strong>Conclusions: </strong>The prognostic signature consisting of four CMRLs displays an outstanding predictive performance and improves the precision of immuno-oncology.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"5784-5800"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651766/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855484","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Ovarian cancer (OC) is a globally prevalent malignancy with significant morbidity and mortality, yet its heterogeneity poses challenges in treatment and prognosis. Recognizing the crucial role of the tumor microenvironment (TME) in OC progression, this study leverages integrative multi-omics and machine learning to uncover TME-associated prognostic biomarkers, paving the way for more personalized therapeutic interventions.
Methods: Employing a rigorous multi-omics approach, this study analyzed single-cell RNA sequencing (scRNA-seq) data from OC and normal tissue samples, including high-grade serous OC (HGSOC) from the Gene Expression Omnibus (GEO: GSE184880) and The Cancer Genome Atlas (TCGA) OC cohort, utilizing the Seurat package to annotate 700 TME-related genes. A prognostic model was developed using the least absolute shrinkage and selection operator (LASSO) regression and independently validated against similarly composed HGSOC datasets. Comprehensive gene expression and immune cell infiltration analyses were conducted, employing advanced algorithms like xCell to delineate the immune landscape of HGSOC.
Results: Our investigation unveiled distinctive immune cell infiltration patterns and gene expression profiles within the TME of HGSOC. Notably, the prevalence of exhausted CD8+ T cells in high-risk patient samples emerged as a critical finding, underscoring the dualistic nature of the immune response in OC. The developed prognostic model, incorporating immune cell markers, exhibited robust predictive accuracy for patient outcomes, showing significant correlations with immunotherapy responses and drug sensitivities.
Conclusions: This study presents a groundbreaking exploration of the OC TME, offering vital insights into its molecular intricacies. By systematically deciphering the TME-associated gene signatures, the research illuminates the potential of these biomarkers in refining patient prognosis and guiding treatment strategies. Our findings underscore the necessity for personalized medicine in OC treatment, potentially enhancing patient survival rates and quality of life. This study marks a significant stride in understanding and combatting the complexities of OC.
{"title":"Integrative multi-omics and machine learning approach reveals tumor microenvironment-associated prognostic biomarkers in ovarian cancer.","authors":"Wenzhi Jiao, Shasha Yang, Yue Li, Yu Li, Shanshan Liu, Jianwei Shi, Minmin Yu","doi":"10.21037/tcr-24-539","DOIUrl":"10.21037/tcr-24-539","url":null,"abstract":"<p><strong>Background: </strong>Ovarian cancer (OC) is a globally prevalent malignancy with significant morbidity and mortality, yet its heterogeneity poses challenges in treatment and prognosis. Recognizing the crucial role of the tumor microenvironment (TME) in OC progression, this study leverages integrative multi-omics and machine learning to uncover TME-associated prognostic biomarkers, paving the way for more personalized therapeutic interventions.</p><p><strong>Methods: </strong>Employing a rigorous multi-omics approach, this study analyzed single-cell RNA sequencing (scRNA-seq) data from OC and normal tissue samples, including high-grade serous OC (HGSOC) from the Gene Expression Omnibus (GEO: GSE184880) and The Cancer Genome Atlas (TCGA) OC cohort, utilizing the Seurat package to annotate 700 TME-related genes. A prognostic model was developed using the least absolute shrinkage and selection operator (LASSO) regression and independently validated against similarly composed HGSOC datasets. Comprehensive gene expression and immune cell infiltration analyses were conducted, employing advanced algorithms like xCell to delineate the immune landscape of HGSOC.</p><p><strong>Results: </strong>Our investigation unveiled distinctive immune cell infiltration patterns and gene expression profiles within the TME of HGSOC. Notably, the prevalence of exhausted CD8<sup>+</sup> T cells in high-risk patient samples emerged as a critical finding, underscoring the dualistic nature of the immune response in OC. The developed prognostic model, incorporating immune cell markers, exhibited robust predictive accuracy for patient outcomes, showing significant correlations with immunotherapy responses and drug sensitivities.</p><p><strong>Conclusions: </strong>This study presents a groundbreaking exploration of the OC TME, offering vital insights into its molecular intricacies. By systematically deciphering the TME-associated gene signatures, the research illuminates the potential of these biomarkers in refining patient prognosis and guiding treatment strategies. Our findings underscore the necessity for personalized medicine in OC treatment, potentially enhancing patient survival rates and quality of life. This study marks a significant stride in understanding and combatting the complexities of OC.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"6182-6200"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651752/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855375","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-30Epub Date: 2024-11-21DOI: 10.21037/tcr-23-2077
Dongdong Luo, Aiping Luo, Su Hu, Hailin Zhao, Xuefeng Yao, Dan Li, Biao Peng
Background: Glioblastoma (GBM) is a frequent malignant tumor in neurosurgery characterized by a high degree of heterogeneity and genetic instability. DNA double-strand breaks generated by homologous recombination deficiency (HRD) are a well-known contributor to genomic instability, which can encourage tumor development. It is unknown, however, whether the molecular characteristics linked with HRD have a predictive role in GBM. The study aims to assess the extent of genomic instability in GBM using HRD score and investigate the prognostic significance of HRD-related molecular features in GBM.
Methods: The discovery cohort comprised 567 GBM patients from The Cancer Genome Atlas (TCGA) database. We established HRD scores using the single nucleotide polymorphism (SNP) array data and analyzed transcriptomic data from patients with different HRD scores to identify biomarkers associated with HRD. A prognostic model was built by using HRD-related differentially expressed genes (DEGs) and validated in a distinct cohort from the Chinese Glioma Genome Atlas (CGGA) database.
Results: Based on the SNP array data, the gene expression profile data, and the clinical characteristics of GBM patients, we found that patients with a high HRD score had a better prognosis than those with a low HRD score. The DNA damage repair (DDR) signaling pathways were notably enriched in the HRD-positive subgroup. The prognostic model was developed by including HRD-related DEGs that could evaluate the clinical prognosis of patients more efficiently than the HRD score. In addition, patients with a low-risk score had a considerably augmented signature of γδT cells. Finally, through univariate and multivariate Cox regression analyses, it was demonstrated that the prognostic model was superior to other prognostic markers.
Conclusions: In conclusion, our research has not only demonstrated that a high HRD score is a valid prognostic biomarker in GBM patients but also built a stable prognosis model [odds ratio (OR) 0.18, 95% confidence interval (CI): 0.11-0.23, P<0.001] that is more accurate than conventional prognostic markers such as O6-methylguanine-DNA methyltransferase (MGMT) methylation (OR 0.55, 95% CI: 0.33-0.91, P=0.02).
背景:胶质母细胞瘤(GBM)是神经外科中一种常见的恶性肿瘤,具有高度的异质性和遗传不稳定性。同源重组缺陷(HRD)引起的DNA双链断裂是众所周知的基因组不稳定因素,它可以促进肿瘤的发展。然而,与HRD相关的分子特征是否在GBM中具有预测作用尚不清楚。本研究旨在利用HRD评分评估GBM基因组不稳定性的程度,并探讨HRD相关分子特征在GBM中的预后意义。方法:发现队列包括来自癌症基因组图谱(TCGA)数据库的567例GBM患者。我们使用单核苷酸多态性(SNP)阵列数据建立了HRD评分,并分析了不同HRD评分患者的转录组数据,以确定与HRD相关的生物标志物。利用hdd相关差异表达基因(DEGs)建立预后模型,并在中国胶质瘤基因组图谱(CGGA)数据库中的不同队列中进行验证。结果:基于SNP阵列数据、基因表达谱数据以及GBM患者的临床特点,我们发现HRD评分高的患者预后优于HRD评分低的患者。DNA损伤修复(DDR)信号通路在hrd阳性亚组中显著富集。通过纳入与HRD相关的deg来建立预后模型,该模型可以比HRD评分更有效地评估患者的临床预后。此外,低风险评分的患者具有显著增强的γδT细胞特征。最后,通过单因素和多因素Cox回归分析,证明该预后模型优于其他预后指标。结论:总之,我们的研究不仅证明了高HRD评分是GBM患者有效的预后生物标志物,而且建立了稳定的预后模型[优势比(OR) 0.18, 95%可信区间(CI): 0.11-0.23, P
{"title":"Prognostic signature detects homologous recombination deficient in glioblastoma.","authors":"Dongdong Luo, Aiping Luo, Su Hu, Hailin Zhao, Xuefeng Yao, Dan Li, Biao Peng","doi":"10.21037/tcr-23-2077","DOIUrl":"10.21037/tcr-23-2077","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GBM) is a frequent malignant tumor in neurosurgery characterized by a high degree of heterogeneity and genetic instability. DNA double-strand breaks generated by homologous recombination deficiency (HRD) are a well-known contributor to genomic instability, which can encourage tumor development. It is unknown, however, whether the molecular characteristics linked with HRD have a predictive role in GBM. The study aims to assess the extent of genomic instability in GBM using HRD score and investigate the prognostic significance of HRD-related molecular features in GBM.</p><p><strong>Methods: </strong>The discovery cohort comprised 567 GBM patients from The Cancer Genome Atlas (TCGA) database. We established HRD scores using the single nucleotide polymorphism (SNP) array data and analyzed transcriptomic data from patients with different HRD scores to identify biomarkers associated with HRD. A prognostic model was built by using HRD-related differentially expressed genes (DEGs) and validated in a distinct cohort from the Chinese Glioma Genome Atlas (CGGA) database.</p><p><strong>Results: </strong>Based on the SNP array data, the gene expression profile data, and the clinical characteristics of GBM patients, we found that patients with a high HRD score had a better prognosis than those with a low HRD score. The DNA damage repair (DDR) signaling pathways were notably enriched in the HRD-positive subgroup. The prognostic model was developed by including HRD-related DEGs that could evaluate the clinical prognosis of patients more efficiently than the HRD score. In addition, patients with a low-risk score had a considerably augmented signature of γδT cells. Finally, through univariate and multivariate Cox regression analyses, it was demonstrated that the prognostic model was superior to other prognostic markers.</p><p><strong>Conclusions: </strong>In conclusion, our research has not only demonstrated that a high HRD score is a valid prognostic biomarker in GBM patients but also built a stable prognosis model [odds ratio (OR) 0.18, 95% confidence interval (CI): 0.11-0.23, P<0.001] that is more accurate than conventional prognostic markers such as O6-methylguanine-DNA methyltransferase (MGMT) methylation (OR 0.55, 95% CI: 0.33-0.91, P=0.02).</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"5883-5897"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651736/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-30Epub Date: 2024-10-14DOI: 10.21037/tcr-24-123
Theodore Wang, Jongmyung Kim, Ritesh Kumar, Rebecca A Deek, Ryan Stephenson, Tina Mayer, Biren Saraiya, Saum Ghodoussipour, Thomas Jang, David Golombos, Vignesh Packiam, Ronald Ennis, Lara Hathout, Salma K Jabbour, Ozan Guler, Cem Onal, Matthew P Deek
Background: Tumor suppressors are well known drivers of cancer invasion and metastasis in metastatic castration sensitive prostate cancer (mCSPC). However, oncogenes are also known to be altered in this state, however the frequency and prognosis of these alterations are unclear. Thus, we aimed to study the spectrum of oncogene mutations in mCSPC and study the significance of these alteration on outcomes.
Methods: Four hundred and seventy-seven patients with mCSPC were included who underwent next generation sequencing. Oncogene alterations were defined as mutations in ALK, AKT1-3, BRAF, CCND1-3, CTNNB1, EGFR, ERBB2, FGFR1, FGFR2, HRAS, KRAS, MDM2, MET, MITF, MYC, NOTCH1-3, NRAS, PIK3CA, PI3KCB, PIK3R1, RET. Endpoints of interests were radiographic progression-free survival (rPFS), time to development of CRPC (tdCRPC), and overall survival (OS). Kaplan Meier analysis was performed and Cox regression hazard ratios (HR) calculated.
Results: A total of 477 patients were included with baseline characteristics with 117 patients (24.5%) harbored a mutation within an oncogene. A total of 172 oncogene mutations were found within the population with the most common being MYC (n=29; 16.9%), PIK3CA (n=24; 14%), CTNNB1 (n=22, 12.8%), BRAF (n=10, 5.8%), and CCND1 (n=10, 5.8%). Oncogene mutations were associated with inferior rPFS (19.2 vs. 32.2 months, P=0.03), tdCRPC (15.7 vs. 32.4 months, P<0.001), and OS (5-year OS 75.3% vs. 55.4%, P=0.01). On multivariable analysis oncogene mutations were strongly associated with tdCRPC (HR 1.42, P=0.03).
Conclusions: Oncogenes are frequency mutated in mCSPC and associated with aggressive features and inferior outcomes. Future work will need to validate these results to better assess its significance in allowing for personalization of care.
{"title":"Landscape and prognostic significance of oncogene drivers in metastatic castration sensitive prostate cancer.","authors":"Theodore Wang, Jongmyung Kim, Ritesh Kumar, Rebecca A Deek, Ryan Stephenson, Tina Mayer, Biren Saraiya, Saum Ghodoussipour, Thomas Jang, David Golombos, Vignesh Packiam, Ronald Ennis, Lara Hathout, Salma K Jabbour, Ozan Guler, Cem Onal, Matthew P Deek","doi":"10.21037/tcr-24-123","DOIUrl":"10.21037/tcr-24-123","url":null,"abstract":"<p><strong>Background: </strong>Tumor suppressors are well known drivers of cancer invasion and metastasis in metastatic castration sensitive prostate cancer (mCSPC). However, oncogenes are also known to be altered in this state, however the frequency and prognosis of these alterations are unclear. Thus, we aimed to study the spectrum of oncogene mutations in mCSPC and study the significance of these alteration on outcomes.</p><p><strong>Methods: </strong>Four hundred and seventy-seven patients with mCSPC were included who underwent next generation sequencing. Oncogene alterations were defined as mutations in <i>ALK, AKT1-3, BRAF, CCND1-3, CTNNB1, EGFR, ERBB2, FGFR1, FGFR2, HRAS, KRAS, MDM2, MET, MITF, MYC, NOTCH1-3, NRAS, PIK3CA, PI3KCB, PIK3R1, RET.</i> Endpoints of interests were radiographic progression-free survival (rPFS), time to development of CRPC (tdCRPC), and overall survival (OS). Kaplan Meier analysis was performed and Cox regression hazard ratios (HR) calculated.</p><p><strong>Results: </strong>A total of 477 patients were included with baseline characteristics with 117 patients (24.5%) harbored a mutation within an oncogene. A total of 172 oncogene mutations were found within the population with the most common being <i>MYC</i> (n=29; 16.9%), <i>PIK3CA</i> (n=24; 14%), <i>CTNNB1</i> (n=22, 12.8%), <i>BRAF</i> (n=10, 5.8%), and <i>CCND1</i> (n=10, 5.8%). Oncogene mutations were associated with inferior rPFS (19.2 <i>vs.</i> 32.2 months, P=0.03), tdCRPC (15.7 <i>vs.</i> 32.4 months, P<0.001), and OS (5-year OS 75.3% <i>vs.</i> 55.4%, P=0.01). On multivariable analysis oncogene mutations were strongly associated with tdCRPC (HR 1.42, P=0.03).</p><p><strong>Conclusions: </strong>Oncogenes are frequency mutated in mCSPC and associated with aggressive features and inferior outcomes. Future work will need to validate these results to better assess its significance in allowing for personalization of care.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 11","pages":"6235-6245"},"PeriodicalIF":1.5,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11651787/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855308","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}