Pub Date : 2024-11-07eCollection Date: 2024-01-01DOI: 10.3389/fonc.2024.1483306
Marta Moretti, Antonella Farina, Antonio Angeloni, Emanuela Anastasi
Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive and invasive type of pancreatic cancer and is expected to soon become the second leading cause of cancer-associated death. The high mortality rate is due to the clinical features that allow asymptomatic progression to advanced stages, a period when current therapeutic treatments have limited efficacy. To address these challenges, researchers are focused on identifying new molecular and circulating markers for early PDAC detection and precision medicine. In this mini-review, we report the most well-known and recently identified molecular and circulating biomarkers. This study aimed to emphasize the need for continued innovative research to develop diagnostic algorithms and therapies to improve the management of patients with PDAC.
{"title":"Emerging horizons on molecular and circulating biomarkers in pancreatic adenocarcinoma.","authors":"Marta Moretti, Antonella Farina, Antonio Angeloni, Emanuela Anastasi","doi":"10.3389/fonc.2024.1483306","DOIUrl":"10.3389/fonc.2024.1483306","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive and invasive type of pancreatic cancer and is expected to soon become the second leading cause of cancer-associated death. The high mortality rate is due to the clinical features that allow asymptomatic progression to advanced stages, a period when current therapeutic treatments have limited efficacy. To address these challenges, researchers are focused on identifying new molecular and circulating markers for early PDAC detection and precision medicine. In this mini-review, we report the most well-known and recently identified molecular and circulating biomarkers. This study aimed to emphasize the need for continued innovative research to develop diagnostic algorithms and therapies to improve the management of patients with PDAC.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1483306"},"PeriodicalIF":3.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11578827/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686371","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}
In the management of advanced non-squamous non-small cell lung cancer (NSCLC) without driver gene mutations, the current therapeutic strategies encompass chemotherapy, chemotherapy combined with anti-angiogenic therapy, and chemotherapy combined with immunotherapy. For patients with high programmed death-ligand 1(PD-L1) expression, monotherapy with immune checkpoint inhibitors is a viable option. Recognizing that some patients cannot tolerate or decline chemotherapy, clinical practice has introduced non-chemotherapeutic treatment regimens, which have shown promising results. This article presents a clinical case of advanced NSCLC with low PD-L1 expression and negative driver gene mutations. The patient was treated with a chemotherapy-free regimen combining envafolimab with endostar. After 17 months of follow-up, both the primary tumor and metastatic lesions exhibited significant reduction, and no notable adverse reactions were observed. This case demonstrates the efficacy of envafolimab combined with endostar in the treatment of advanced NSCLC. This regimen enhances treatment safety and patient compliance, potentially offering a novel therapeutic option for patients with advanced NSCLC characterized by low PD-L1 expression and negative driver gene mutations.
{"title":"Case report: Combination therapy of envafolimab with endostar for advanced non-small cell lung cancer with low PD-L1 expression.","authors":"Shuo Wu, Changhong Dong, Chenxi Hu, Kaiyuan Hui, Xiaodong Jiang","doi":"10.3389/fonc.2024.1437260","DOIUrl":"10.3389/fonc.2024.1437260","url":null,"abstract":"<p><p>In the management of advanced non-squamous non-small cell lung cancer (NSCLC) without driver gene mutations, the current therapeutic strategies encompass chemotherapy, chemotherapy combined with anti-angiogenic therapy, and chemotherapy combined with immunotherapy. For patients with high programmed death-ligand 1(PD-L1) expression, monotherapy with immune checkpoint inhibitors is a viable option. Recognizing that some patients cannot tolerate or decline chemotherapy, clinical practice has introduced non-chemotherapeutic treatment regimens, which have shown promising results. This article presents a clinical case of advanced NSCLC with low PD-L1 expression and negative driver gene mutations. The patient was treated with a chemotherapy-free regimen combining envafolimab with endostar. After 17 months of follow-up, both the primary tumor and metastatic lesions exhibited significant reduction, and no notable adverse reactions were observed. This case demonstrates the efficacy of envafolimab combined with endostar in the treatment of advanced NSCLC. This regimen enhances treatment safety and patient compliance, potentially offering a novel therapeutic option for patients with advanced NSCLC characterized by low PD-L1 expression and negative driver gene mutations.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1437260"},"PeriodicalIF":3.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11578815/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686659","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 : 2024-11-07eCollection Date: 2024-01-01DOI: 10.3389/fonc.2024.1406759
Guoxing Zhang, Ni Sun, Xiaohua Li
Background: To explore new modulatory intervention targets for radiation-induced lung injury, bioinformatics analysis technology was used to search for the core driving genes in the pathogenesis of radiation pneumonitis, and the results were verified by a radiation-induced murine lung injury model to find possible new targets for the treatment of radiation lung injury.
Method: Gene Expression Omnibus Database was used to identify differentially expressed genes in radiation pneumonitis. DAVID database was used for gene ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) enrichment analysis. Gene Set Enrichment Analysis was used to analyze abnormal expressions. Protein-protein interaction networks were constructed using STRING and Cytoscape. Discovery Studio 4.5 software was used to find the preferred inhibitor of the specific gene. A radiation-induced lung injury model was induced in female C57BL/6N mice. The specific inhibitors were administered by intraperitoneal injection 24 h before and for 7 consecutive days after radiation. Lungs were harvested for further analysis 14 days and 10 weeks post-irradiation.
Results: We screened Syk as one of the most important driver genes of radiation pneumonitis by bioinformatics analysis and screened the preferred Syk inhibitor fostamatinib from the drug database. Syk was highly expressed in irradiated lung tissue, and fostamatinib inhibited the level of Syk expression. Syk inhibitor significantly alleviated the radiation-induced lung injury and downregulated the increased expression of p38 MAPK, p53, IL-1β, and IL-6 in lung tissue at 2 weeks after radiation. The levels of TGF-β, COL1A1, and α-SMA and degree of pulmonary fibrosis at 10 weeks after radiation were also decreased by Syk inhibitor.
Conclusion: Syk inhibitor may have a potential to be used as a targeted drug to mitigate radiation pneumonitis and inhibit radiation-induced pulmonary fibrosis.
背景:为探索放射性肺损伤的新调控干预靶点,利用生物信息学分析技术寻找放射性肺炎发病机制中的核心驱动基因,并通过放射性诱导的小鼠肺损伤模型对结果进行验证,以寻找治疗放射性肺损伤的可能新靶点:方法:利用基因表达总库(Gene Expression Omnibus Database)鉴定辐射性肺炎的差异表达基因。使用 DAVID 数据库进行基因本体(GO)和京都基因和基因组百科全书(KEGG)富集分析。基因组富集分析用于分析异常表达。使用 STRING 和 Cytoscape 构建了蛋白质-蛋白质相互作用网络。使用 Discovery Studio 4.5 软件查找特定基因的首选抑制剂。在雌性 C57BL/6N 小鼠中诱导辐射诱导的肺损伤模型。在辐射前 24 小时和辐射后连续 7 天腹腔注射特异性抑制剂。分别在辐射后 14 天和 10 周收获肺脏进行进一步分析:结果:我们通过生物信息学分析筛选出Syk是放射性肺炎最重要的驱动基因之一,并从药物数据库中筛选出首选的Syk抑制剂福斯塔替尼。Syk在辐照肺组织中高表达,福斯塔替尼抑制了Syk的表达水平。Syk抑制剂明显减轻了辐射诱导的肺损伤,并下调了辐射2周后肺组织中p38 MAPK、p53、IL-1β和IL-6的表达。Syk抑制剂还能降低辐射10周后肺组织中TGF-β、COL1A1和α-SMA的水平以及肺纤维化程度:结论:Syk抑制剂可作为一种靶向药物用于缓解放射性肺炎和抑制辐射诱导的肺纤维化。
{"title":"Spleen tyrosine kinase inhibition mitigates radiation-induced lung injury through anti-inflammatory effects and downregulation of p38 MAPK and p53.","authors":"Guoxing Zhang, Ni Sun, Xiaohua Li","doi":"10.3389/fonc.2024.1406759","DOIUrl":"10.3389/fonc.2024.1406759","url":null,"abstract":"<p><strong>Background: </strong>To explore new modulatory intervention targets for radiation-induced lung injury, bioinformatics analysis technology was used to search for the core driving genes in the pathogenesis of radiation pneumonitis, and the results were verified by a radiation-induced murine lung injury model to find possible new targets for the treatment of radiation lung injury.</p><p><strong>Method: </strong>Gene Expression Omnibus Database was used to identify differentially expressed genes in radiation pneumonitis. DAVID database was used for gene ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) enrichment analysis. Gene Set Enrichment Analysis was used to analyze abnormal expressions. Protein-protein interaction networks were constructed using STRING and Cytoscape. Discovery Studio 4.5 software was used to find the preferred inhibitor of the specific gene. A radiation-induced lung injury model was induced in female C57BL/6N mice. The specific inhibitors were administered by intraperitoneal injection 24 h before and for 7 consecutive days after radiation. Lungs were harvested for further analysis 14 days and 10 weeks post-irradiation.</p><p><strong>Results: </strong>We screened Syk as one of the most important driver genes of radiation pneumonitis by bioinformatics analysis and screened the preferred Syk inhibitor fostamatinib from the drug database. Syk was highly expressed in irradiated lung tissue, and fostamatinib inhibited the level of Syk expression. Syk inhibitor significantly alleviated the radiation-induced lung injury and downregulated the increased expression of p38 MAPK, p53, IL-1β, and IL-6 in lung tissue at 2 weeks after radiation. The levels of TGF-β, COL1A1, and α-SMA and degree of pulmonary fibrosis at 10 weeks after radiation were also decreased by Syk inhibitor.</p><p><strong>Conclusion: </strong>Syk inhibitor may have a potential to be used as a targeted drug to mitigate radiation pneumonitis and inhibit radiation-induced pulmonary fibrosis.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1406759"},"PeriodicalIF":3.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11578954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686313","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 : 2024-11-07eCollection Date: 2024-01-01DOI: 10.3389/fonc.2024.1361871
Can Qi, Ziwei Zhao, Lin Chen, Le Wang, Yun Zhou, Guochen Duan
<p><strong>Background: </strong>Cancer cells induce neutrophil extracellular traps (NETs) to promote tumor progression and metastasis. However, only a few studies have focused on the role of NETs in Neuroblastoma (NB).</p><p><strong>Methods: </strong>First, based on the expression of NET-related genes, consensus clustering analysis was conducted to cluster NB samples into different subtypes. Differential analysis was performed to identify DEGs between subtypes. Functional items and related pathways of DEGs were identified using enrichment analysis. Univariate Cox analysis and the LASSO algorithm were used to identify biomarkers for prognosis. Furthermore, independent prognostic analysis was performed. Immune infiltration analysis was performed to identify differential immune cells. Finally, the verification of prognostic model genes were taken by the immunohistochemical staining and quantitative real-time PCR.</p><p><strong>Results: </strong>Consensus clustering analysis demonstrated that NB samples were clustered into two subtypes. There were 125 DEGs between the two subtypes of NB. Moreover, the enrichment analysis results showed that the DEGs were mainly associated with 'external side of plasma membrane,' 'immune receptor activity' 'regulation of leukocyte migration' GO items. There were also several GO items related to neutrophils, such as regulation of neutrophil migration and differentiation. KEGG pathways revealed that the DEGs were correlated with in immunity-related activities, including 'Complement and coagulation cascades,' 'Neutrophil extracellular trap formation, 'T cell receptor signaling pathway,' 'PD-L1 expression and PD-1 checkpoint pathway in cancer' and so on. A total of five biomarkers,[Selenoprotein P1 (SEPP1), Fibrinogen-like protein 2 (FGL2), NK cell lectin-like receptor K1 (KLRK1), ATP-binding cassette transporters 6(ABCA6) and Galectins(GAL)], were screened, and a risk model based on the biomarkers was created. Furthermore, a nomogram for forecasting the survival rates of patients with NB was established based on the risk score, age at diagnosis, and MYCN status. Eight differential immune cells (CD8 + T cells, resting mast cells, etc.) were acquired between the two risk subgroups. The expression levels of five prognostic model genes at the protein and mRNA were verified and all results were consistent with the results of our bioinformatics analysis.</p><p><strong>Conclusion: </strong>We initially found that five NET-related genes were significantly differentially expressed in NETs-associated molecular isoforms and two Netrg molecular isoforms were found to be associated with poorer prognosis. This stratification might provide insight into the prediction of prognosis and ideal immunotherapy strategies for patients with NB. However, we also noted that the formation of NETs is a complex biological process involving the regulation of multiple cytokines and cellular interactions. Therefore, the exact roles of these genes and t
{"title":"Exploring the role of neutrophil extracellular traps in neuroblastoma: identification of molecular subtypes and prognostic implications.","authors":"Can Qi, Ziwei Zhao, Lin Chen, Le Wang, Yun Zhou, Guochen Duan","doi":"10.3389/fonc.2024.1361871","DOIUrl":"10.3389/fonc.2024.1361871","url":null,"abstract":"<p><strong>Background: </strong>Cancer cells induce neutrophil extracellular traps (NETs) to promote tumor progression and metastasis. However, only a few studies have focused on the role of NETs in Neuroblastoma (NB).</p><p><strong>Methods: </strong>First, based on the expression of NET-related genes, consensus clustering analysis was conducted to cluster NB samples into different subtypes. Differential analysis was performed to identify DEGs between subtypes. Functional items and related pathways of DEGs were identified using enrichment analysis. Univariate Cox analysis and the LASSO algorithm were used to identify biomarkers for prognosis. Furthermore, independent prognostic analysis was performed. Immune infiltration analysis was performed to identify differential immune cells. Finally, the verification of prognostic model genes were taken by the immunohistochemical staining and quantitative real-time PCR.</p><p><strong>Results: </strong>Consensus clustering analysis demonstrated that NB samples were clustered into two subtypes. There were 125 DEGs between the two subtypes of NB. Moreover, the enrichment analysis results showed that the DEGs were mainly associated with 'external side of plasma membrane,' 'immune receptor activity' 'regulation of leukocyte migration' GO items. There were also several GO items related to neutrophils, such as regulation of neutrophil migration and differentiation. KEGG pathways revealed that the DEGs were correlated with in immunity-related activities, including 'Complement and coagulation cascades,' 'Neutrophil extracellular trap formation, 'T cell receptor signaling pathway,' 'PD-L1 expression and PD-1 checkpoint pathway in cancer' and so on. A total of five biomarkers,[Selenoprotein P1 (SEPP1), Fibrinogen-like protein 2 (FGL2), NK cell lectin-like receptor K1 (KLRK1), ATP-binding cassette transporters 6(ABCA6) and Galectins(GAL)], were screened, and a risk model based on the biomarkers was created. Furthermore, a nomogram for forecasting the survival rates of patients with NB was established based on the risk score, age at diagnosis, and MYCN status. Eight differential immune cells (CD8 + T cells, resting mast cells, etc.) were acquired between the two risk subgroups. The expression levels of five prognostic model genes at the protein and mRNA were verified and all results were consistent with the results of our bioinformatics analysis.</p><p><strong>Conclusion: </strong>We initially found that five NET-related genes were significantly differentially expressed in NETs-associated molecular isoforms and two Netrg molecular isoforms were found to be associated with poorer prognosis. This stratification might provide insight into the prediction of prognosis and ideal immunotherapy strategies for patients with NB. However, we also noted that the formation of NETs is a complex biological process involving the regulation of multiple cytokines and cellular interactions. Therefore, the exact roles of these genes and t","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1361871"},"PeriodicalIF":3.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11578966/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686372","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}
Introduction: When elderly patients have underlying diseases combined with oromaxillofacial diseases requiring surgical treatment, the application of conventional general anesthesia (GA) for oromaxillofacial surgical diseases has become a risk due to underlying disease reasons. The objective of this study was to evaluate the efficacy and safety of ultrasound-guided superficial cervical plexus block (SCPB) anesthesia combined with local infiltration anesthesia (LIA) for partial oral and maxillofacial surgery (OMFS) in patients who with risk for GA due to underlying disease.
Methods: The clinical data of 7 high risk patients with OMFS treated with SCPB anesthesia combined with LIA were retrospectively analyzed. All seven surgeries were performed on one side of the neck. All patients were given ultrasound-guided SCPB anesthesia by the same anesthesiologist, LIA by the same surgeon, and surgery was performed under continuous Electrocardiogram (ECG) monitoring.
Results: Seven patients had stable vital signs and no significant postoperative complications. The results of this study indicated that SCPB anesthesia combined with LIA is a safe and effective anesthesia technique with a high success rate and patient tolerance.
Discussion: For patients with OMFS who have a risk for GA due to underlying diseases, ultrasound-guided cervical superficial plexus block anesthesia combined with LIA is a safe and effective alternative to conventional GA.
导言:当老年患者患有基础疾病并合并口腔颌面部疾病需要手术治疗时,由于基础疾病的原因,应用常规全身麻醉(GA)治疗口腔颌面部手术疾病具有一定的风险。本研究旨在评估超声引导下颈浅神经丛阻滞(SCPB)麻醉联合局部浸润麻醉(LIA)用于口腔颌面外科部分手术(OMFS)的有效性和安全性:方法:回顾性分析了7例采用SCPB麻醉联合LIA治疗的高风险口腔颌面外科手术患者的临床数据。所有七例手术均在一侧颈部进行。所有患者均由同一位麻醉师在超声引导下进行 SCPB 麻醉,由同一位外科医生进行 LIA,手术在持续心电图(ECG)监测下进行:结果:七名患者生命体征平稳,术后无明显并发症。本研究结果表明,SCPB 麻醉联合 LIA 是一种安全有效的麻醉技术,成功率高,患者耐受性好:讨论:对于因基础疾病而有GA风险的OMFS患者,超声引导下颈浅丛阻滞麻醉联合LIA是传统GA的一种安全有效的替代方法。
{"title":"Combination of ultrasound guided superficial cervical plexus block and local infiltration for oromaxillofacial surgeries: a case series.","authors":"Hao-Ran Zhao, Jian-Shuai Hao, Ling-Fa Xue, Jin-Ze Zhao, Yi-Chen Wang, Wen-Lin Xiao","doi":"10.3389/fonc.2024.1412062","DOIUrl":"10.3389/fonc.2024.1412062","url":null,"abstract":"<p><strong>Introduction: </strong>When elderly patients have underlying diseases combined with oromaxillofacial diseases requiring surgical treatment, the application of conventional general anesthesia (GA) for oromaxillofacial surgical diseases has become a risk due to underlying disease reasons. The objective of this study was to evaluate the efficacy and safety of ultrasound-guided superficial cervical plexus block (SCPB) anesthesia combined with local infiltration anesthesia (LIA) for partial oral and maxillofacial surgery (OMFS) in patients who with risk for GA due to underlying disease.</p><p><strong>Methods: </strong>The clinical data of 7 high risk patients with OMFS treated with SCPB anesthesia combined with LIA were retrospectively analyzed. All seven surgeries were performed on one side of the neck. All patients were given ultrasound-guided SCPB anesthesia by the same anesthesiologist, LIA by the same surgeon, and surgery was performed under continuous Electrocardiogram (ECG) monitoring.</p><p><strong>Results: </strong>Seven patients had stable vital signs and no significant postoperative complications. The results of this study indicated that SCPB anesthesia combined with LIA is a safe and effective anesthesia technique with a high success rate and patient tolerance.</p><p><strong>Discussion: </strong>For patients with OMFS who have a risk for GA due to underlying diseases, ultrasound-guided cervical superficial plexus block anesthesia combined with LIA is a safe and effective alternative to conventional GA.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1412062"},"PeriodicalIF":3.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11578926/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686288","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}
Tumor-associated macrophages (TAMs) play a pivotal role in sculpting the tumor microenvironment and influencing cancer progression, particularly through their interactions with various forms of regulated cell death (RCD), including apoptosis, pyroptosis, ferroptosis, and necroptosis. This review examines the interplay between TAMs and these RCD pathways, exploring the mechanisms through which they interact to promote tumor growth and advancement. We examine the underlying mechanisms of these intricate interactions, emphasizing their importance in cancer progression and treatment. Moreover, we present potential therapeutic strategies for targeting TAMs and manipulating RCD to enhance anti-tumor responses. These strategies encompass reprogramming TAMs, inhibiting their recruitment, and selectively eliminating them to enhance anti-tumor functions, alongside modulating RCD pathways to amplify immune responses. These insights offer a novel perspective on tumor biology and provide a foundation for the development of more efficacious cancer therapies.
{"title":"Interactions between tumor-associated macrophages and regulated cell death: therapeutic implications in immuno-oncology.","authors":"Yifei Ge, Lixue Jiang, Chengru Yang, Qingfu Dong, Chengwu Tang, Yi Xu, Xiangyu Zhong","doi":"10.3389/fonc.2024.1449696","DOIUrl":"10.3389/fonc.2024.1449696","url":null,"abstract":"<p><p>Tumor-associated macrophages (TAMs) play a pivotal role in sculpting the tumor microenvironment and influencing cancer progression, particularly through their interactions with various forms of regulated cell death (RCD), including apoptosis, pyroptosis, ferroptosis, and necroptosis. This review examines the interplay between TAMs and these RCD pathways, exploring the mechanisms through which they interact to promote tumor growth and advancement. We examine the underlying mechanisms of these intricate interactions, emphasizing their importance in cancer progression and treatment. Moreover, we present potential therapeutic strategies for targeting TAMs and manipulating RCD to enhance anti-tumor responses. These strategies encompass reprogramming TAMs, inhibiting their recruitment, and selectively eliminating them to enhance anti-tumor functions, alongside modulating RCD pathways to amplify immune responses. These insights offer a novel perspective on tumor biology and provide a foundation for the development of more efficacious cancer therapies.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1449696"},"PeriodicalIF":3.5,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11578871/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686374","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 : 2024-11-06eCollection Date: 2024-01-01DOI: 10.3389/fonc.2024.1441025
Meiling Zhang, Li Wang, Qian Wang, Jiu Yang, Wei Peng, Xiaoyou Li, Meiqi Shi, Kaihua Lu
Background: Non-small cell lung cancer (NSCLC) with human epidermal growth factor receptor 2 (HER2) alterations poses a substantial treatment challenge. Current HER2-targeted therapies offer limited efficacy. Antibody-drug conjugates (ADCs) targeting HER2 have emerged as a promising therapeutic strategy. This study aimed to evaluate the clinical response to a novel ADC drug Disitamab vedotin (RC48) in advanced NSCLC with HER2 alterations.
Methods: This study conducted a retrospective review of patients harboring HER2 alterations treated with RC48 in the real world. Clinical outcomes were evaluated in terms of objective response rate (ORR), disease control rate (DCR), and progression-free survival (PFS).
Results: Out of 22 patients, 21 (95.5%) received RC48 combination therapy, while one received RC48 monotherapy. The ORR of all patients reached 45.5%, and the DCR stood at 90.9%. The median PFS (mPFS) was 7.5 months. Among patients receiving RC48 combination therapy, the ORR was 47.7%, and the mPFS of 8.1 months. The combination of RC48 with platinum+/- bevacizumab resulted in the highest ORR of 71.4% (5 out of 7 patients), with HER2 TKI following at a 50.0% ORR (4 out of 8 patients). First-line (1L) treatment with RC48 showed an ORR of 62.5% (5 out of 8 patients), second-line (2L) treatments had a 57.1% ORR (4 out of 7 patients), and beyond second-line (>2L) treatments exhibited a 14.3% ORR (1 out of 7 patients). Patients with 1L, 2L, or >2L treatment had a mPFS of 8.1 months, 7.2 months, and 7.4 months, respectively. Patients with HER2 mutations or amplifications, and those with concurrent mutations and amplifications at baseline, showed mPFS of 8.1 months, 9.4 months, and 7.4 months, respectively. The mPFS was significantly longer in patients with HER2 amplification. The most common adverse events included hand-foot syndrome (54.5%), asthenia (50.0%), decreased white blood cell count (45.5%), and liver impairment (45.5%). Grade 3 adverse events occurred in one (4.5%) patient.
Conclusion: RC48, particularly in combination regimens, demonstrates promising efficacy in advanced NSCLC with HER2 alterations. These findings underscore the need for further research to validate RC48's application in clinical practice.
{"title":"Efficacy of disitamab vedotin in non-small cell lung cancer with <i>HER2</i> alterations: a multicenter, retrospective real-world study.","authors":"Meiling Zhang, Li Wang, Qian Wang, Jiu Yang, Wei Peng, Xiaoyou Li, Meiqi Shi, Kaihua Lu","doi":"10.3389/fonc.2024.1441025","DOIUrl":"10.3389/fonc.2024.1441025","url":null,"abstract":"<p><strong>Background: </strong>Non-small cell lung cancer (NSCLC) with human epidermal growth factor receptor 2 (<i>HER2</i>) alterations poses a substantial treatment challenge. Current <i>HER2</i>-targeted therapies offer limited efficacy. Antibody-drug conjugates (ADCs) targeting <i>HER2</i> have emerged as a promising therapeutic strategy. This study aimed to evaluate the clinical response to a novel ADC drug Disitamab vedotin (RC48) in advanced NSCLC with <i>HER2</i> alterations.</p><p><strong>Methods: </strong>This study conducted a retrospective review of patients harboring <i>HER2</i> alterations treated with RC48 in the real world. Clinical outcomes were evaluated in terms of objective response rate (ORR), disease control rate (DCR), and progression-free survival (PFS).</p><p><strong>Results: </strong>Out of 22 patients, 21 (95.5%) received RC48 combination therapy, while one received RC48 monotherapy. The ORR of all patients reached 45.5%, and the DCR stood at 90.9%. The median PFS (mPFS) was 7.5 months. Among patients receiving RC48 combination therapy, the ORR was 47.7%, and the mPFS of 8.1 months. The combination of RC48 with platinum+/- bevacizumab resulted in the highest ORR of 71.4% (5 out of 7 patients), with <i>HER2</i> TKI following at a 50.0% ORR (4 out of 8 patients). First-line (1L) treatment with RC48 showed an ORR of 62.5% (5 out of 8 patients), second-line (2L) treatments had a 57.1% ORR (4 out of 7 patients), and beyond second-line (>2L) treatments exhibited a 14.3% ORR (1 out of 7 patients). Patients with 1L, 2L, or >2L treatment had a mPFS of 8.1 months, 7.2 months, and 7.4 months, respectively. Patients with <i>HER2</i> mutations or amplifications, and those with concurrent mutations and amplifications at baseline, showed mPFS of 8.1 months, 9.4 months, and 7.4 months, respectively. The mPFS was significantly longer in patients with <i>HER2</i> amplification. The most common adverse events included hand-foot syndrome (54.5%), asthenia (50.0%), decreased white blood cell count (45.5%), and liver impairment (45.5%). Grade 3 adverse events occurred in one (4.5%) patient.</p><p><strong>Conclusion: </strong>RC48, particularly in combination regimens, demonstrates promising efficacy in advanced NSCLC with <i>HER2</i> alterations. These findings underscore the need for further research to validate RC48's application in clinical practice.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1441025"},"PeriodicalIF":3.5,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576286/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142681104","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 : 2024-11-06eCollection Date: 2024-01-01DOI: 10.3389/fonc.2024.1442168
Hao Wu, Dan Jia, Xianyu Dai, Hongliang Cao, Fulin Wang, Tong Yang, Lei Wang, Tao Xu, Baoshan Gao
Purpose: We sought to develop and validate a nomogram for predicting extra-urinary recurrence (EUR) following radical nephroureterectomy (RNU) in patients with upper urinary tract urothelial carcinoma (UTUC).
Methods: Data from 556 UTUC patients post-RNU at the First Hospital of Jilin University were retrospectively analyzed. These patients were categorized into a training group (n=389) and a validation group (n=167). Variables significantly associated with prognosis were identified using univariate Cox regression and most minor absolute shrinkage and selection operator (LASSO) analysis. These independent predictors were incorporated into the nomogram to estimate extra-urinary recurrence-free survival (EURFS). Validation of the nomogram involved ROC curves, calibration plots, and decision curve analysis (DCA). Patients were stratified into two risk categories based on their nomogram scores to compare EURFS using the Kaplan-Meier method.
Results: Eight predictors were identified: T-stage, N-stage, tumor grade, local and nerve invasion, preoperative hemoglobin level, neutrophil-to-lymphocyte ratio (NLR), and creatinine, all proving to be independent predictors of EUR. A nomogram was created based on these eight factors, and using the ROC, calibration curves, and DCA, good prediction results were shown in both the training and validation groups. The training and validation groups also showed reliable predictive performance. In particular, there was a significant difference in survival between the high-risk and low-risk groups (P<0.0001). We have also built a network calculator that calculates patient survival time. The URL is https://haowu24.shinyapps.io/dynnomapp.
Conclusion: A nomogram for predicting distant metastases in UTUC patients was successfully developed, and its accuracy, reliability, and clinical value were demonstrated. This new tool helps to improve the clinical management of UTUC cases.
{"title":"A new nomogram for predicting extraurothelial recurrence in patients with upper urinary tract urothelial carcinoma following radical nephroureterectomy.","authors":"Hao Wu, Dan Jia, Xianyu Dai, Hongliang Cao, Fulin Wang, Tong Yang, Lei Wang, Tao Xu, Baoshan Gao","doi":"10.3389/fonc.2024.1442168","DOIUrl":"10.3389/fonc.2024.1442168","url":null,"abstract":"<p><strong>Purpose: </strong>We sought to develop and validate a nomogram for predicting extra-urinary recurrence (EUR) following radical nephroureterectomy (RNU) in patients with upper urinary tract urothelial carcinoma (UTUC).</p><p><strong>Methods: </strong>Data from 556 UTUC patients post-RNU at the First Hospital of Jilin University were retrospectively analyzed. These patients were categorized into a training group (n=389) and a validation group (n=167). Variables significantly associated with prognosis were identified using univariate Cox regression and most minor absolute shrinkage and selection operator (LASSO) analysis. These independent predictors were incorporated into the nomogram to estimate extra-urinary recurrence-free survival (EURFS). Validation of the nomogram involved ROC curves, calibration plots, and decision curve analysis (DCA). Patients were stratified into two risk categories based on their nomogram scores to compare EURFS using the Kaplan-Meier method.</p><p><strong>Results: </strong>Eight predictors were identified: T-stage, N-stage, tumor grade, local and nerve invasion, preoperative hemoglobin level, neutrophil-to-lymphocyte ratio (NLR), and creatinine, all proving to be independent predictors of EUR. A nomogram was created based on these eight factors, and using the ROC, calibration curves, and DCA, good prediction results were shown in both the training and validation groups. The training and validation groups also showed reliable predictive performance. In particular, there was a significant difference in survival between the high-risk and low-risk groups (P<0.0001). We have also built a network calculator that calculates patient survival time. The URL is https://haowu24.shinyapps.io/dynnomapp.</p><p><strong>Conclusion: </strong>A nomogram for predicting distant metastases in UTUC patients was successfully developed, and its accuracy, reliability, and clinical value were demonstrated. This new tool helps to improve the clinical management of UTUC cases.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1442168"},"PeriodicalIF":3.5,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576284/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142681445","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 : 2024-11-06eCollection Date: 2024-01-01DOI: 10.3389/fonc.2024.1472725
Anthony Crymes, Mark G Evans, Tolulope Adeyelu, Jack Reid, Ifegwu O Ibe, Matthew J Oberley, Jill H Tseng
The discovery of gene fusions involving Neuregulin-1 (NRG1) within solid tumors has important therapeutic implications, as they are being actively explored as targets for emerging ERBB/ERBB2/ERBB3-directed anti-cancer agents. NRG1 fusions are very uncommon across all tumor types and are infrequently documented in the medical literature. We report a female patient presenting with widespread peritoneal carcinomatosis diagnosed as high grade serous fallopian tube carcinoma, which harbored a previously undescribed MYH10::NRG1 fusion. Moreover, we queried the whole transcriptome sequencing results of neoplasms analyzed by a commercial laboratory (Caris Life Sciences) to determine the overall incidence of NRG1 fusions in carcinomas of the ovary, fallopian tube, and peritoneum (0.18%). Twenty-five additional tumors were found to demonstrate NRG1 fusions, including 20 new genes partners that had not been previously identified in gynecologic carcinomas. Overall, NRG1 fusion events are rare in ovarian, fallopian tube, and primary peritoneal carcinomas, but they may carry diagnostic significance in the context of borderline/low grade serous tumors, which demonstrated exclusively CLU::NRG1 fusions, and could have important predictive implications for response to ERBB/ERBB2/ERBB3-directed therapies.
{"title":"Case report: High grade serous fallopian tube carcinoma with rare <i>NRG1</i> gene fusion presenting as widespread peritoneal carcinomatosis.","authors":"Anthony Crymes, Mark G Evans, Tolulope Adeyelu, Jack Reid, Ifegwu O Ibe, Matthew J Oberley, Jill H Tseng","doi":"10.3389/fonc.2024.1472725","DOIUrl":"10.3389/fonc.2024.1472725","url":null,"abstract":"<p><p>The discovery of gene fusions involving Neuregulin-1 (<i>NRG1</i>) within solid tumors has important therapeutic implications, as they are being actively explored as targets for emerging ERBB/ERBB2/ERBB3-directed anti-cancer agents. <i>NRG1</i> fusions are very uncommon across all tumor types and are infrequently documented in the medical literature. We report a female patient presenting with widespread peritoneal carcinomatosis diagnosed as high grade serous fallopian tube carcinoma, which harbored a previously undescribed <i>MYH10</i>::<i>NRG1</i> fusion. Moreover, we queried the whole transcriptome sequencing results of neoplasms analyzed by a commercial laboratory (Caris Life Sciences) to determine the overall incidence of <i>NRG1</i> fusions in carcinomas of the ovary, fallopian tube, and peritoneum (0.18%). Twenty-five additional tumors were found to demonstrate <i>NRG1</i> fusions, including 20 new genes partners that had not been previously identified in gynecologic carcinomas. Overall, <i>NRG1</i> fusion events are rare in ovarian, fallopian tube, and primary peritoneal carcinomas, but they may carry diagnostic significance in the context of borderline/low grade serous tumors, which demonstrated exclusively <i>CLU::NRG1</i> fusions, and could have important predictive implications for response to ERBB/ERBB2/ERBB3-directed therapies.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1472725"},"PeriodicalIF":3.5,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11580015/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686660","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 : 2024-11-06eCollection Date: 2024-01-01DOI: 10.3389/fonc.2024.1442712
Xiaojing Yang, Hanru Ren, Zhen Li, Jie Fu
Cervical cancer is a common malignant tumor of female reproductive system. Radiation therapy is one of the main methods of cervical cancer treatment, of which brachytherapy is an essential and important part of radiation therapy for locally advanced cervical cancer. With the rapid development of imaging technologies such as computed tomography (CT) and magnetic resonance imaging (MRI), brachytherapy for cervical cancer has gradually developed from traditional two-dimensional image-guided technology to three-dimensional image-guided technology. And there are more and more treatment methods, including intracavitary brachytherapy, interstitial brachytherapy, and intracavitary combined interstitial implantation brachytherapy. We performed a PubMed search for introduce the application progress of intracavity, implantation, intracavity combined implantation brachytherapy and radioactive seed implantation, and discuss the dosimetric feasibility of internal and external fusion irradiation.
{"title":"Brachytherapy for cervical cancer: from intracavitary to interstitial technique.","authors":"Xiaojing Yang, Hanru Ren, Zhen Li, Jie Fu","doi":"10.3389/fonc.2024.1442712","DOIUrl":"10.3389/fonc.2024.1442712","url":null,"abstract":"<p><p>Cervical cancer is a common malignant tumor of female reproductive system. Radiation therapy is one of the main methods of cervical cancer treatment, of which brachytherapy is an essential and important part of radiation therapy for locally advanced cervical cancer. With the rapid development of imaging technologies such as computed tomography (CT) and magnetic resonance imaging (MRI), brachytherapy for cervical cancer has gradually developed from traditional two-dimensional image-guided technology to three-dimensional image-guided technology. And there are more and more treatment methods, including intracavitary brachytherapy, interstitial brachytherapy, and intracavitary combined interstitial implantation brachytherapy. We performed a PubMed search for introduce the application progress of intracavity, implantation, intracavity combined implantation brachytherapy and radioactive seed implantation, and discuss the dosimetric feasibility of internal and external fusion irradiation.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"14 ","pages":"1442712"},"PeriodicalIF":3.5,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576414/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142680884","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}