首页 > 最新文献

Cancer Cell International最新文献

英文 中文
Circulating IFNγ-associated protein signatures predict response to neoadjuvant immunotherapy in patients with stage III melanoma. 循环ifn γ相关蛋白特征预测III期黑色素瘤患者对新辅助免疫治疗的反应。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-25 DOI: 10.1186/s12935-025-04067-4
Fei Yang, Su Yin Lim, Ines Pires da Silva, Lijia Yu, Jordan W Conway, Alexander M Menzies, Georgina V Long, Jean Yh Yang, Helen Rizos

Background: Immune checkpoint inhibitors (ICIs), such as anti-programmed cell death protein 1 (PD-1) and anti-cytotoxic T-lymphocyte associated protein 4 (CTLA-4), have transformed the management of stage III melanoma in the neoadjuvant setting. However, a substantial proportion of patients do not derive benefit from ICI therapy. To improve clinical outcomes, there remains a critical unmet need to identify early biomarkers of response to neoadjuvant immunotherapy in stage III melanoma.

Methods: In this study, we performed longitudinal serum proteomic profiling in 39 patients undergoing neoadjuvant combination anti-PD-1 and anti-CTLA-4 therapy. Using a multiplex proximity extension assay, we measured 702 proteins at three timepoints: baseline, early on-treatment (3-4 weeks after treatment initiation), and pre-surgery (4-8 weeks post-treatment).

Results: The most pronounced differences between major pathological responders (MPR) and non-MPR patients were detected at baseline and were linked to interferon gamma (IFNγ) signalling, but these differences diminish at later timepoints. A 10-protein IFNγ-associated signature derived from baseline serum profile achieved an AUC of 0.68 for predicting pathological response, comparable to a previously reported tumour-based IFNγ gene signature (AUC = 0.67).

Conclusions: These findings support the use of circulating protein signatures as minimally invasive, scalable biomarkers to inform early treatment decisions in the neoadjuvant setting.

背景:免疫检查点抑制剂(ICIs),如抗程序性细胞死亡蛋白1 (PD-1)和抗细胞毒性t淋巴细胞相关蛋白4 (CTLA-4),已经在新辅助治疗中改变了III期黑色素瘤的治疗。然而,相当比例的患者并没有从ICI治疗中获益。为了改善临床结果,确定III期黑色素瘤对新辅助免疫治疗反应的早期生物标志物仍然是一个关键的未满足需求。方法:在本研究中,我们对39例接受新辅助联合抗pd -1和抗ctla -4治疗的患者进行了纵向血清蛋白质组学分析。使用多重接近扩展试验,我们在三个时间点测量了702种蛋白质:基线、治疗早期(治疗开始后3-4周)和手术前(治疗后4-8周)。结果:主要病理应答者(MPR)和非MPR患者之间最显著的差异在基线时被检测到,并且与干扰素γ (IFNγ)信号传导有关,但这些差异在较晚的时间点减弱。来自基线血清谱的10蛋白IFNγ相关标记在预测病理反应方面的AUC为0.68,与先前报道的基于肿瘤的IFNγ基因标记(AUC = 0.67)相当。结论:这些发现支持将循环蛋白特征作为微创、可扩展的生物标志物,为新辅助治疗的早期治疗决策提供信息。
{"title":"Circulating IFNγ-associated protein signatures predict response to neoadjuvant immunotherapy in patients with stage III melanoma.","authors":"Fei Yang, Su Yin Lim, Ines Pires da Silva, Lijia Yu, Jordan W Conway, Alexander M Menzies, Georgina V Long, Jean Yh Yang, Helen Rizos","doi":"10.1186/s12935-025-04067-4","DOIUrl":"10.1186/s12935-025-04067-4","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs), such as anti-programmed cell death protein 1 (PD-1) and anti-cytotoxic T-lymphocyte associated protein 4 (CTLA-4), have transformed the management of stage III melanoma in the neoadjuvant setting. However, a substantial proportion of patients do not derive benefit from ICI therapy. To improve clinical outcomes, there remains a critical unmet need to identify early biomarkers of response to neoadjuvant immunotherapy in stage III melanoma.</p><p><strong>Methods: </strong>In this study, we performed longitudinal serum proteomic profiling in 39 patients undergoing neoadjuvant combination anti-PD-1 and anti-CTLA-4 therapy. Using a multiplex proximity extension assay, we measured 702 proteins at three timepoints: baseline, early on-treatment (3-4 weeks after treatment initiation), and pre-surgery (4-8 weeks post-treatment).</p><p><strong>Results: </strong>The most pronounced differences between major pathological responders (MPR) and non-MPR patients were detected at baseline and were linked to interferon gamma (IFNγ) signalling, but these differences diminish at later timepoints. A 10-protein IFNγ-associated signature derived from baseline serum profile achieved an AUC of 0.68 for predicting pathological response, comparable to a previously reported tumour-based IFNγ gene signature (AUC = 0.67).</p><p><strong>Conclusions: </strong>These findings support the use of circulating protein signatures as minimally invasive, scalable biomarkers to inform early treatment decisions in the neoadjuvant setting.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"424"},"PeriodicalIF":6.0,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12648901/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145602357","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
HAUS6 as a potential prognostic and immunological biomarker: validation from pan-cancer analysis to hepatocellular carcinoma. HAUS6作为一种潜在的预后和免疫生物标志物:从泛癌分析到肝细胞癌的验证
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-24 DOI: 10.1186/s12935-025-04081-6
Haixiang Xie, Zuyin Wan, Xianwei Lu, Kejian Yang, Chongjiu Qin, Yu Chen, Chenlu Lan, Kai Peng, Wei Qin, Shutian Mo, Xin Zhou, Chengkun Yang, Xiwen Liao, Tao Peng
{"title":"HAUS6 as a potential prognostic and immunological biomarker: validation from pan-cancer analysis to hepatocellular carcinoma.","authors":"Haixiang Xie, Zuyin Wan, Xianwei Lu, Kejian Yang, Chongjiu Qin, Yu Chen, Chenlu Lan, Kai Peng, Wei Qin, Shutian Mo, Xin Zhou, Chengkun Yang, Xiwen Liao, Tao Peng","doi":"10.1186/s12935-025-04081-6","DOIUrl":"https://doi.org/10.1186/s12935-025-04081-6","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145595499","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrated scRNA-seq and transcriptome analyses uncover the effects of UBE2H on the immune microenvironment regulation in pancreatic cancer. 综合scRNA-seq和转录组分析揭示了UBE2H在胰腺癌免疫微环境调节中的作用。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-24 DOI: 10.1186/s12935-025-04059-4
Fuxin Huang, Zhongyan Zhang, Jike Fang, Yue Chen, Jinhui Wei, Quanzhang Li, Chuanzhao Zhang, Shanzhou Huang, Baohua Hou

Background: Pancreatic cancer, recognized as a refractory tumor, has an overall survival rate of less than 10%, and its mortality rate continues to rise. Due to the low immune activity induced by its unique tumor microenvironment, pancreatic cancer is classified as a "cold" tumor and is insensitive to current immunotherapies. However, little is still known about the identification and functional mechanisms of key regulatory molecules in the formation of "cold" tumors.

Methods: In this study, we identified the pancreatic ductal cell niche through single-cell sequencing. By calculating CNV scores using inferCNV to distinguish malignant from non-malignant cells and analyzing differences in transcriptional levels, we constructed a 15-gene model. The inhibitory effect of UBE2H on the immune microenvironment was studied through single-cell sequencing analysis, including the inhibition of T cell function, impairment of antigen presentation in macrophages and neutrophils, and activation of neutrophil extracellular traps (NETs).

Results: Our results revealed the presence of a UBE2H+ cell population within the pancreatic ductal cell niche, primarily observed in pancreatic cancer samples. Notably, single-cell sequencing analysis of UBE2H overexpression revealed an enhanced T cell PD-L1 interaction axis and a downregulation of antigen presentation pathways in macrophages and neutrophils (antigen processing and presentation, macrophage activation, and neutrophil activation). Further in vivo experiments confirmed that high UBE2H expression promotes tumor progression, leading to increased T cell exhaustion (PD1) and decreased activation (CD69, GZMB, IFNR) in the immune microenvironment, impaired antigen presentation of macrophages and neutrophils (H2Kb, I-Ab, I-A/E), and increased neutrophil extracellular trap formation (MPO, NE, CITH3).

Conclusions: This study revealed the presence of a UBE2H+ cell population within the pancreatic ductal cell niche and analyzed the inhibition of UBE2H on the immune microenvironment of pancreatic cancer through single-cell sequencing and in vivo experiments, providing important clues for the formation of "cold" tumors in pancreatic cancer and opening new directions for exploring new treatment strategies.

背景:胰腺癌是公认的难治性肿瘤,总生存率不足10%,且死亡率持续上升。由于其独特的肿瘤微环境诱导的低免疫活性,胰腺癌被归类为“冷”肿瘤,对目前的免疫疗法不敏感。然而,对于“冷”肿瘤形成的关键调控分子的鉴定和功能机制仍然知之甚少。方法:本研究通过单细胞测序鉴定胰腺导管细胞生态位。通过使用intercnv计算CNV评分来区分恶性和非恶性细胞,并分析转录水平的差异,我们构建了一个15个基因的模型。通过单细胞测序分析,研究UBE2H对免疫微环境的抑制作用,包括抑制T细胞功能、破坏巨噬细胞和中性粒细胞抗原呈递、激活中性粒细胞胞外陷阱(NETs)等。结果:我们的研究结果揭示了UBE2H+细胞群存在于胰腺导管细胞生态位中,主要在胰腺癌样本中观察到。值得注意的是,UBE2H过表达的单细胞测序分析显示,T细胞PD-L1相互作用轴增强,巨噬细胞和中性粒细胞抗原递呈途径下调(抗原加工和递呈、巨噬细胞活化和中性粒细胞活化)。进一步的体内实验证实,UBE2H高表达促进肿瘤进展,导致免疫微环境中T细胞衰竭(PD1)增加,活化(CD69、GZMB、IFNR)降低,巨噬细胞和中性粒细胞(H2Kb、I-Ab、I-A/E)抗原呈递受损,中性粒细胞胞外陷阱(MPO、NE、CITH3)形成增加。结论:本研究通过单细胞测序和体内实验,揭示了胰腺导管细胞生态位内存在UBE2H+细胞群,并分析了UBE2H对胰腺癌免疫微环境的抑制作用,为胰腺癌“冷”肿瘤的形成提供了重要线索,为探索新的治疗策略开辟了新的方向。
{"title":"Integrated scRNA-seq and transcriptome analyses uncover the effects of UBE2H on the immune microenvironment regulation in pancreatic cancer.","authors":"Fuxin Huang, Zhongyan Zhang, Jike Fang, Yue Chen, Jinhui Wei, Quanzhang Li, Chuanzhao Zhang, Shanzhou Huang, Baohua Hou","doi":"10.1186/s12935-025-04059-4","DOIUrl":"10.1186/s12935-025-04059-4","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic cancer, recognized as a refractory tumor, has an overall survival rate of less than 10%, and its mortality rate continues to rise. Due to the low immune activity induced by its unique tumor microenvironment, pancreatic cancer is classified as a \"cold\" tumor and is insensitive to current immunotherapies. However, little is still known about the identification and functional mechanisms of key regulatory molecules in the formation of \"cold\" tumors.</p><p><strong>Methods: </strong>In this study, we identified the pancreatic ductal cell niche through single-cell sequencing. By calculating CNV scores using inferCNV to distinguish malignant from non-malignant cells and analyzing differences in transcriptional levels, we constructed a 15-gene model. The inhibitory effect of UBE2H on the immune microenvironment was studied through single-cell sequencing analysis, including the inhibition of T cell function, impairment of antigen presentation in macrophages and neutrophils, and activation of neutrophil extracellular traps (NETs).</p><p><strong>Results: </strong>Our results revealed the presence of a UBE2H+ cell population within the pancreatic ductal cell niche, primarily observed in pancreatic cancer samples. Notably, single-cell sequencing analysis of UBE2H overexpression revealed an enhanced T cell PD-L1 interaction axis and a downregulation of antigen presentation pathways in macrophages and neutrophils (antigen processing and presentation, macrophage activation, and neutrophil activation). Further in vivo experiments confirmed that high UBE2H expression promotes tumor progression, leading to increased T cell exhaustion (PD1) and decreased activation (CD69, GZMB, IFNR) in the immune microenvironment, impaired antigen presentation of macrophages and neutrophils (H2Kb, I-Ab, I-A/E), and increased neutrophil extracellular trap formation (MPO, NE, CITH3).</p><p><strong>Conclusions: </strong>This study revealed the presence of a UBE2H+ cell population within the pancreatic ductal cell niche and analyzed the inhibition of UBE2H on the immune microenvironment of pancreatic cancer through single-cell sequencing and in vivo experiments, providing important clues for the formation of \"cold\" tumors in pancreatic cancer and opening new directions for exploring new treatment strategies.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"422"},"PeriodicalIF":6.0,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12645782/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145595428","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The lncrnas: innovative multifunctional players of drug resistance in colorectal cancer. lncrnas:结直肠癌耐药的创新多功能参与者。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-23 DOI: 10.1186/s12935-025-03951-3
Hossein Maghsoudi, Motahareh Rasoulzadeh, Maryam Abbastabar, Ahmad Fazilat, Farhad Sheikhnia, Bita Azizzadeh, Mohamad Valilo, Maryam Majidinia
{"title":"The lncrnas: innovative multifunctional players of drug resistance in colorectal cancer.","authors":"Hossein Maghsoudi, Motahareh Rasoulzadeh, Maryam Abbastabar, Ahmad Fazilat, Farhad Sheikhnia, Bita Azizzadeh, Mohamad Valilo, Maryam Majidinia","doi":"10.1186/s12935-025-03951-3","DOIUrl":"https://doi.org/10.1186/s12935-025-03951-3","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-11-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585783","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unraveling the role of the solute carrier family in hepatocellular carcinoma: mechanisms and therapeutic prospects. 溶质载体家族在肝细胞癌中的作用:机制和治疗前景。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-22 DOI: 10.1186/s12935-025-03960-2
Yi Xue, Xuanran Yang, Chi Zhang, Huan Li, Chuanfei Zeng, Mingkai Chen

The solute carrier (SLC) family is one of the largest families of membrane transport proteins, essential for cellular metabolism and communication. Studies have shown that the SLC family plays a critical role in the metabolic processes and intrinsic biological behaviors of hepatocellular carcinoma (HCC). SLC38s contribute to the progression of HCC through signaling pathways such as PI3K/AKT/mTOR and Wnt/β-catenin/MYC. The regulation of upstream factors like miR-10b-5p, the LINC01559-miR-511-SLC38A1 axis, miRNA-432, and other genes are also implicated in HCC development. Additionally, SLC7A11 influences the proliferation of HCC cells via pathways such as SMYD3/SLC7A11 and JAK/STAT/SLC7A11. Other members of the SLC family also play significant roles in HCC through various mechanisms. Given the importance of SLCs in HCC and the lack of a comprehensive understanding of their molecular mechanisms, this study explores metabolic alterations in liver cancer cells and their surrounding immune cells. It also examines how the metal ions in liver cancer impact HCC growth and the efficacy of anticancer immunotherapy. Furthermore, we discuss the vital role of SLCs as key transporters for the hepatic uptake of anionic anticancer drugs, highlighting novel therapeutic opportunities.

溶质载体(SLC)家族是最大的膜转运蛋白家族之一,对细胞代谢和通讯至关重要。研究表明,SLC家族在肝细胞癌(HCC)的代谢过程和内在生物学行为中起着至关重要的作用。slc38通过PI3K/AKT/mTOR和Wnt/β-catenin/MYC等信号通路促进HCC的进展。上游因子如miR-10b-5p、LINC01559-miR-511-SLC38A1轴、miRNA-432等基因的调控也与HCC的发展有关。此外,SLC7A11通过SMYD3/SLC7A11和JAK/STAT/SLC7A11等途径影响HCC细胞的增殖。SLC家族的其他成员也通过各种机制在HCC中发挥重要作用。鉴于SLCs在HCC中的重要性以及对其分子机制缺乏全面了解,本研究探讨了肝癌细胞及其周围免疫细胞的代谢改变。它还研究了肝癌中的金属离子如何影响HCC的生长和抗癌免疫治疗的疗效。此外,我们讨论了SLCs作为阴离子抗癌药物肝脏摄取的关键转运体的重要作用,强调了新的治疗机会。
{"title":"Unraveling the role of the solute carrier family in hepatocellular carcinoma: mechanisms and therapeutic prospects.","authors":"Yi Xue, Xuanran Yang, Chi Zhang, Huan Li, Chuanfei Zeng, Mingkai Chen","doi":"10.1186/s12935-025-03960-2","DOIUrl":"https://doi.org/10.1186/s12935-025-03960-2","url":null,"abstract":"<p><p>The solute carrier (SLC) family is one of the largest families of membrane transport proteins, essential for cellular metabolism and communication. Studies have shown that the SLC family plays a critical role in the metabolic processes and intrinsic biological behaviors of hepatocellular carcinoma (HCC). SLC38s contribute to the progression of HCC through signaling pathways such as PI3K/AKT/mTOR and Wnt/β-catenin/MYC. The regulation of upstream factors like miR-10b-5p, the LINC01559-miR-511-SLC38A1 axis, miRNA-432, and other genes are also implicated in HCC development. Additionally, SLC7A11 influences the proliferation of HCC cells via pathways such as SMYD3/SLC7A11 and JAK/STAT/SLC7A11. Other members of the SLC family also play significant roles in HCC through various mechanisms. Given the importance of SLCs in HCC and the lack of a comprehensive understanding of their molecular mechanisms, this study explores metabolic alterations in liver cancer cells and their surrounding immune cells. It also examines how the metal ions in liver cancer impact HCC growth and the efficacy of anticancer immunotherapy. Furthermore, we discuss the vital role of SLCs as key transporters for the hepatic uptake of anionic anticancer drugs, highlighting novel therapeutic opportunities.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":" ","pages":""},"PeriodicalIF":6.0,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581615","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GPR34 inhibition reprograms tumor-associated macrophages and enhances the sensitivity of anti-PD-1 therapy in hepatocellular carcinoma. GPR34抑制可重编程肿瘤相关巨噬细胞,增强抗pd -1治疗在肝癌中的敏感性。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-21 DOI: 10.1186/s12935-025-04030-3
Wenfeng Liu, Qiang Yu, Xinyi Liu, Feng Zhang, Qiuyu Jiang, Wenqing Tang, Ling Dong

Background: Macrophage play a dual role in tumor progression. However, the mechanisms underlying tumor-associated macrophage (TAM) polarization in the hepatocellular carcinoma (HCC) microenvironment remain elusive.

Methods: We analyzed the proportions of tumor-infiltrating immune cells in The Cancer Genome Atlas Liver Hepatocellular Carcinoma cohort. Subsequently, a risk prediction model was constructed for patients with HCC. The expression patterns and clinical relevance of G protein-coupled receptor 34 (GPR34) were evaluated. The potential functions and mechanisms of GPR34 in macrophages were also investigated. Additionally, HCC mouse models were used to assess the therapeutic potential of a GPR34 inhibitor in enhancing immune checkpoint blockade (ICB) therapy.

Results: A novel prognostic prediction model based on macrophage-related genes was established to predict the outcomes of patients with HCC. In this model, GPR34 was identified as significantly upregulated and associated with a poor clinical prognosis in HCC. Mechanistically, GPR34 facilitated M2 macrophage polarization by activating the PI3K/AKT signaling axis. The pharmacological inhibition of GPR34 effectively suppressed tumor growth. Moreover, the combination of a GPR34 inhibitor with a PD-1 inhibitor demonstrated synergistic antitumor effects.

Conclusion: GPR34 is pivotal in driving macrophage M2 polarization, and inhibitor targeting GPR34 represents promising antitumor agent capable of augmenting the efficacy of anti-PD-1 therapy.

背景:巨噬细胞在肿瘤进展中起双重作用。然而,肝细胞癌(HCC)微环境中肿瘤相关巨噬细胞(TAM)极化的机制尚不清楚。方法:我们分析了肝癌基因组图谱中肿瘤浸润免疫细胞的比例。随后,构建HCC患者的风险预测模型。评价G蛋白偶联受体34 (GPR34)的表达模式及临床意义。研究了GPR34在巨噬细胞中的潜在功能和机制。此外,HCC小鼠模型被用来评估GPR34抑制剂在增强免疫检查点阻断(ICB)治疗中的治疗潜力。结果:建立了一种基于巨噬细胞相关基因的肝癌预后预测模型。在该模型中,GPR34在HCC中被鉴定为显著上调并与不良临床预后相关。在机制上,GPR34通过激活PI3K/AKT信号轴促进M2巨噬细胞极化。药理抑制GPR34可有效抑制肿瘤生长。此外,GPR34抑制剂与PD-1抑制剂联合使用显示出协同抗肿瘤作用。结论:GPR34在驱动巨噬细胞M2极化中起关键作用,靶向GPR34的抑制剂是一种有前景的抗肿瘤药物,可以增强抗pd -1治疗的疗效。
{"title":"GPR34 inhibition reprograms tumor-associated macrophages and enhances the sensitivity of anti-PD-1 therapy in hepatocellular carcinoma.","authors":"Wenfeng Liu, Qiang Yu, Xinyi Liu, Feng Zhang, Qiuyu Jiang, Wenqing Tang, Ling Dong","doi":"10.1186/s12935-025-04030-3","DOIUrl":"10.1186/s12935-025-04030-3","url":null,"abstract":"<p><strong>Background: </strong>Macrophage play a dual role in tumor progression. However, the mechanisms underlying tumor-associated macrophage (TAM) polarization in the hepatocellular carcinoma (HCC) microenvironment remain elusive.</p><p><strong>Methods: </strong>We analyzed the proportions of tumor-infiltrating immune cells in The Cancer Genome Atlas Liver Hepatocellular Carcinoma cohort. Subsequently, a risk prediction model was constructed for patients with HCC. The expression patterns and clinical relevance of G protein-coupled receptor 34 (GPR34) were evaluated. The potential functions and mechanisms of GPR34 in macrophages were also investigated. Additionally, HCC mouse models were used to assess the therapeutic potential of a GPR34 inhibitor in enhancing immune checkpoint blockade (ICB) therapy.</p><p><strong>Results: </strong>A novel prognostic prediction model based on macrophage-related genes was established to predict the outcomes of patients with HCC. In this model, GPR34 was identified as significantly upregulated and associated with a poor clinical prognosis in HCC. Mechanistically, GPR34 facilitated M2 macrophage polarization by activating the PI3K/AKT signaling axis. The pharmacological inhibition of GPR34 effectively suppressed tumor growth. Moreover, the combination of a GPR34 inhibitor with a PD-1 inhibitor demonstrated synergistic antitumor effects.</p><p><strong>Conclusion: </strong>GPR34 is pivotal in driving macrophage M2 polarization, and inhibitor targeting GPR34 represents promising antitumor agent capable of augmenting the efficacy of anti-PD-1 therapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"419"},"PeriodicalIF":6.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12639727/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573104","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ammonia in cancer: dual roles and therapeutic strategies. 氨在癌症中的双重作用和治疗策略。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-21 DOI: 10.1186/s12935-025-04065-6
Jinhui Wei, Yue Chen, Quanzhang Li, Fuxin Huang, Chuanzhao Zhang, Baohua Hou, Shanzhou Huang

Ammonia, a toxic nitrogenous metabolic byproduct, has garnered increasing attention for its pivotal role in tumor biology. The human body has developed intricate detoxification mechanisms to regulate ammonia homeostasis and maintain acid‒base equilibrium. Through their adaptation mechanisms, cancer cells can exploit ammonia to facilitate their growth and modulate the tumor immune microenvironment. As ammonia is a toxic substance, it can have a toxic effect on tumor, thereby inhibiting tumor growth. This article critically examines the sources and destinations of ammonia within the tumor microenvironment (TME), offering an innovative synthesis of its dual roles in both promoting and inhibiting tumor progression. Additionally, it explores therapeutic strategies targeting ammonia metabolism and anticipates future research trajectories, thereby providing valuable insights and a theoretical framework for ammonia-based therapies in oncology.

氨是一种有毒的含氮代谢副产物,因其在肿瘤生物学中的关键作用而受到越来越多的关注。人体已经发展出复杂的解毒机制来调节氨稳态和维持酸碱平衡。癌细胞通过自身的适应机制,利用氨促进自身生长,调节肿瘤免疫微环境。氨是一种有毒物质,可以对肿瘤产生毒性作用,从而抑制肿瘤生长。本文批判性地研究了肿瘤微环境(TME)中氨的来源和目的地,提供了其在促进和抑制肿瘤进展中的双重作用的创新合成。此外,它还探索了针对氨代谢的治疗策略,并预测了未来的研究轨迹,从而为肿瘤氨基治疗提供了有价值的见解和理论框架。
{"title":"Ammonia in cancer: dual roles and therapeutic strategies.","authors":"Jinhui Wei, Yue Chen, Quanzhang Li, Fuxin Huang, Chuanzhao Zhang, Baohua Hou, Shanzhou Huang","doi":"10.1186/s12935-025-04065-6","DOIUrl":"10.1186/s12935-025-04065-6","url":null,"abstract":"<p><p>Ammonia, a toxic nitrogenous metabolic byproduct, has garnered increasing attention for its pivotal role in tumor biology. The human body has developed intricate detoxification mechanisms to regulate ammonia homeostasis and maintain acid‒base equilibrium. Through their adaptation mechanisms, cancer cells can exploit ammonia to facilitate their growth and modulate the tumor immune microenvironment. As ammonia is a toxic substance, it can have a toxic effect on tumor, thereby inhibiting tumor growth. This article critically examines the sources and destinations of ammonia within the tumor microenvironment (TME), offering an innovative synthesis of its dual roles in both promoting and inhibiting tumor progression. Additionally, it explores therapeutic strategies targeting ammonia metabolism and anticipates future research trajectories, thereby providing valuable insights and a theoretical framework for ammonia-based therapies in oncology.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"420"},"PeriodicalIF":6.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12639695/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573070","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
S100A9 regulates eosinophil extracellular trap and activates NF-κB signaling in endometrial cancer: a machine learning-based biomarker discovery. S100A9在子宫内膜癌中调节嗜酸性粒细胞胞外陷阱和激活NF-κB信号:一个基于机器学习的生物标志物发现。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-21 DOI: 10.1186/s12935-025-04058-5
Juanjuan Ni, Yanan Mao, Qiaofeng Ma, Ziyao Wu, Min Su

Endometrial cancer (EC) is a leading cause of gynecological malignancy with poor prognosis in advanced stages. This study aimed to identify key eosinophil extracellular trap (EET) regulators involved in EC progression and explore their prognostic value using machine learning-based models. Through differential expression analysis, we identified 108 EET regulators whose expression was significantly altered in tumor tissues compared to normal tissues. Survival analysis further demonstrated that S100A9 and other EET-related genes, such as CCL26 and CD40, were significantly associated with poor patient outcomes. Unsupervised clustering analysis revealed two distinct molecular subtypes of EC, with Cluster A showing upregulation of most EET regulators and worse clinical outcomes. We assessed immune infiltration profiles and found elevated eosinophil infiltration in Cluster A. Machine learning models incorporating S100A9 achieved superior predictive performance, with the Lasso + RSF model demonstrating robust accuracy (C-index = 0.864) for predicting patient survival. Experimental validation of S100A9 function in endometrial cancer cell lines demonstrated that S100A9 knockdown effectively reduced its expression, leading to the disruption of the NF-κB pathway, as confirmed by Western blot analysis. Further, S100A9 overexpression in Ishikawa and KLE cells resulted in increased levels of apoptotic markers, indicating its role in apoptosis regulation. These findings suggest S100A9 as a potential prognostic biomarker for EC, influencing immune infiltration, NF-κB signaling, and tumor progression, with implications for new therapeutic strategies targeting EET-related pathways.

子宫内膜癌(EC)是妇科恶性肿瘤的主要病因,晚期预后较差。本研究旨在确定参与EC进展的关键嗜酸性粒细胞胞外陷阱(EET)调节因子,并利用基于机器学习的模型探索其预后价值。通过差异表达分析,我们鉴定出108个EET调节因子在肿瘤组织中的表达与正常组织相比有显著改变。生存分析进一步表明,S100A9和其他eet相关基因,如CCL26和CD40,与患者预后不良显著相关。无监督聚类分析显示EC有两种不同的分子亚型,A类显示大多数EET调节因子上调,临床结果较差。我们评估了免疫浸润谱,发现集群a中嗜酸性粒细胞浸润升高。纳入S100A9的机器学习模型具有优越的预测性能,Lasso + RSF模型在预测患者生存方面显示出强大的准确性(c -指数= 0.864)。通过实验验证S100A9在子宫内膜癌细胞系中的功能,经Western blot分析证实,敲低S100A9可有效降低其表达,导致NF-κB通路中断。此外,S100A9在Ishikawa细胞和KLE细胞中的过表达导致凋亡标志物水平升高,表明其在细胞凋亡调节中起作用。这些研究结果表明,S100A9是EC的潜在预后生物标志物,影响免疫浸润、NF-κB信号传导和肿瘤进展,对针对eet相关通路的新治疗策略具有重要意义。
{"title":"S100A9 regulates eosinophil extracellular trap and activates NF-κB signaling in endometrial cancer: a machine learning-based biomarker discovery.","authors":"Juanjuan Ni, Yanan Mao, Qiaofeng Ma, Ziyao Wu, Min Su","doi":"10.1186/s12935-025-04058-5","DOIUrl":"10.1186/s12935-025-04058-5","url":null,"abstract":"<p><p>Endometrial cancer (EC) is a leading cause of gynecological malignancy with poor prognosis in advanced stages. This study aimed to identify key eosinophil extracellular trap (EET) regulators involved in EC progression and explore their prognostic value using machine learning-based models. Through differential expression analysis, we identified 108 EET regulators whose expression was significantly altered in tumor tissues compared to normal tissues. Survival analysis further demonstrated that S100A9 and other EET-related genes, such as CCL26 and CD40, were significantly associated with poor patient outcomes. Unsupervised clustering analysis revealed two distinct molecular subtypes of EC, with Cluster A showing upregulation of most EET regulators and worse clinical outcomes. We assessed immune infiltration profiles and found elevated eosinophil infiltration in Cluster A. Machine learning models incorporating S100A9 achieved superior predictive performance, with the Lasso + RSF model demonstrating robust accuracy (C-index = 0.864) for predicting patient survival. Experimental validation of S100A9 function in endometrial cancer cell lines demonstrated that S100A9 knockdown effectively reduced its expression, leading to the disruption of the NF-κB pathway, as confirmed by Western blot analysis. Further, S100A9 overexpression in Ishikawa and KLE cells resulted in increased levels of apoptotic markers, indicating its role in apoptosis regulation. These findings suggest S100A9 as a potential prognostic biomarker for EC, influencing immune infiltration, NF-κB signaling, and tumor progression, with implications for new therapeutic strategies targeting EET-related pathways.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"421"},"PeriodicalIF":6.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12639692/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573144","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A vascularized 3D bioengineered lung tumor model for anticancer drug screening. 用于抗癌药物筛选的血管化三维生物工程肺肿瘤模型。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-20 DOI: 10.1186/s12935-025-04064-7
Mingqi Wang, Yunming Ye, Ruqiang Yuan, Xu Zheng, Xuelu Li, Yinhe Han, Lina Guo, Ting Lei, Xiuli Wang

The limited clinical translation of preclinical anticancer drug efficacy underscores the urgent need for advanced models that faithfully replicate tumor pathophysiology. While three-dimensional (3D) tumor cultures improve the fidelity of microenvironmental modeling, most existing systems lack vascularization, which is a critical element influencing tumor progression and treatment resistance. In this study, a vascularized 3D lung cancer model was established by co-seeding decellularized lung scaffold with human embryonic stem cell-derived endothelial cells, pericytes, and A549 adenocarcinoma cells. This tri-culture system successfully formed a hierarchical vascular network and recapitulated key features of the tumor microenvironment, including hypoxia-driven lysyl oxidase (LOX) overexpression, and integrin-mediated fibronectin-rich desmoplastic niches accumulation. Compared to traditional cultures, this 3D bioengineered platform demonstrated excellent cell compatibility and architectural complexity, supporting enhanced cell migration and MUC5AC hypersecretion. Importantly, cancer cells cultured in this 3D vascularized system exhibited reduced chemosensitivity relative to monolayer cultures. Moreover, patient-derived lung cancer organoids were integrated into the pre-vascularized 3D compartment for individualized drug response testing. Mechanically, hypoxia-activated HIF-1α/LOX signaling promoted ITGA5/FN1-dependent extracellular matrix remodeling and contributed to a chemoprotective niche. This vascularized 3D lung cancer model offers a physiologically relevant and translationally valuable platform for investigating non-small cell lung cancer progression and optimizing patient-specific drug screening.

临床前抗癌药物疗效的有限临床转化强调了对忠实地复制肿瘤病理生理的先进模型的迫切需要。虽然三维(3D)肿瘤培养提高了微环境建模的保真度,但大多数现有系统缺乏血管化,而血管化是影响肿瘤进展和治疗耐药性的关键因素。本研究通过将去细胞化的肺支架与人胚胎干细胞来源的内皮细胞、周细胞和A549腺癌细胞共植,建立了血管化的三维肺癌模型。这个三培养系统成功地形成了一个分层的血管网络,并概括了肿瘤微环境的关键特征,包括缺氧驱动的赖氨酸氧化酶(LOX)过表达和整合素介导的富含纤维连接蛋白的结缔组织生态位积累。与传统培养物相比,这种3D生物工程平台具有良好的细胞相容性和结构复杂性,支持增强的细胞迁移和MUC5AC的高分泌。重要的是,与单层培养相比,在这种三维血管化系统中培养的癌细胞表现出较低的化学敏感性。此外,患者来源的肺癌类器官被整合到预血管化的3D室中,用于个体化药物反应测试。机械上,缺氧激活的HIF-1α/LOX信号促进ITGA5/ fn1依赖的细胞外基质重塑,并有助于化学保护生态位。这种血管化的三维肺癌模型为研究非小细胞肺癌的进展和优化患者特异性药物筛选提供了一个生理学上相关的和有翻译价值的平台。
{"title":"A vascularized 3D bioengineered lung tumor model for anticancer drug screening.","authors":"Mingqi Wang, Yunming Ye, Ruqiang Yuan, Xu Zheng, Xuelu Li, Yinhe Han, Lina Guo, Ting Lei, Xiuli Wang","doi":"10.1186/s12935-025-04064-7","DOIUrl":"10.1186/s12935-025-04064-7","url":null,"abstract":"<p><p>The limited clinical translation of preclinical anticancer drug efficacy underscores the urgent need for advanced models that faithfully replicate tumor pathophysiology. While three-dimensional (3D) tumor cultures improve the fidelity of microenvironmental modeling, most existing systems lack vascularization, which is a critical element influencing tumor progression and treatment resistance. In this study, a vascularized 3D lung cancer model was established by co-seeding decellularized lung scaffold with human embryonic stem cell-derived endothelial cells, pericytes, and A549 adenocarcinoma cells. This tri-culture system successfully formed a hierarchical vascular network and recapitulated key features of the tumor microenvironment, including hypoxia-driven lysyl oxidase (LOX) overexpression, and integrin-mediated fibronectin-rich desmoplastic niches accumulation. Compared to traditional cultures, this 3D bioengineered platform demonstrated excellent cell compatibility and architectural complexity, supporting enhanced cell migration and MUC5AC hypersecretion. Importantly, cancer cells cultured in this 3D vascularized system exhibited reduced chemosensitivity relative to monolayer cultures. Moreover, patient-derived lung cancer organoids were integrated into the pre-vascularized 3D compartment for individualized drug response testing. Mechanically, hypoxia-activated HIF-1α/LOX signaling promoted ITGA5/FN1-dependent extracellular matrix remodeling and contributed to a chemoprotective niche. This vascularized 3D lung cancer model offers a physiologically relevant and translationally valuable platform for investigating non-small cell lung cancer progression and optimizing patient-specific drug screening.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"418"},"PeriodicalIF":6.0,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12632099/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145562800","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multi-omics analysis unveils the role of cancer-associated fibroblasts in cutaneous squamous cell carcinoma. 多组学分析揭示了癌症相关成纤维细胞在皮肤鳞状细胞癌中的作用。
IF 6 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-19 DOI: 10.1186/s12935-025-04061-w
Xiaochuan Wang, Tingrui Li, Yichao Jin, Jingjing Chen, XinYang, Zhen Guan, Mei Jin, Jingxian Zhang, Liangheng Xu, Sizhen Tao, Chunguang Li, Chunping Ao

Background: Human papillomavirus (HPV) infection is associated with an increased risk of cutaneous squamous cell carcinoma (CSCC). A comprehensive understanding of the cellular heterogeneity of HPV-positive and -negative CSCC is crucial for improving diagnosis and preventing tumor progression.

Methods: We conducted an integrated analysis of single-cell RNA and spatial transcriptomic data from different skin tissue sources to map the cellular landscape of the tumor microenvironment (TME) in both HPV positive and negative CSCC. Results were validated through multiplex immunohistochemistry (mIHC) and in vitro experiments.

Results: We identified 10 major cell types in CSCC and normal skin samples, including epithelial cells, myeloid cells, T cells, fibroblasts, endothelial cells, B cells, smooth muscle cells, mast cells, melanocytes, and hair follicle cells. Notably, fibroblasts were found to be associated with tumor progression in CSCC with or without HPV infected. We further identified eight major CAF subtypes in CSCC, with iCAFs-CXCL2 promoting tumor progression, while iCAFs-PLA2G2A acted to suppress tumor growth. The MDK-ITGA6 pair was found to mediate interactions between fibroblasts and epithelial cells in CSCC. mIHC analysis confirmed elevated expression of MDK and ITGA6 in CSCC samples. Additionally, cell co-culture experiments confirmed that MDK-mediated CAFs were shown to enhance tumor cell migration and invasion in CSCC.

Conclusion: Our findings provide a comprehensive cellular atlas of CSCC, highlighting the association of CAFs in HPV infection and tumor progression of CSCC. These results also offer potential diagnostic and prognostic biomarkers for CSCC patients.

背景:人乳头瘤病毒(HPV)感染与皮肤鳞状细胞癌(CSCC)的风险增加有关。全面了解hpv阳性和阴性CSCC的细胞异质性对于提高诊断和预防肿瘤进展至关重要。方法:我们对来自不同皮肤组织来源的单细胞RNA和空间转录组数据进行了综合分析,以绘制HPV阳性和阴性CSCC中肿瘤微环境(TME)的细胞景观。多重免疫组化(mIHC)和体外实验验证了结果。结果:我们在CSCC和正常皮肤样本中鉴定出10种主要细胞类型,包括上皮细胞、髓样细胞、T细胞、成纤维细胞、内皮细胞、B细胞、平滑肌细胞、肥大细胞、黑素细胞和毛囊细胞。值得注意的是,在感染或未感染HPV的CSCC中,发现成纤维细胞与肿瘤进展有关。我们进一步确定了CSCC中的8种主要CAF亚型,其中iCAFs-CXCL2促进肿瘤进展,而iCAFs-PLA2G2A抑制肿瘤生长。MDK-ITGA6对被发现介导CSCC成纤维细胞和上皮细胞之间的相互作用。mIHC分析证实MDK和ITGA6在CSCC样品中表达升高。此外,细胞共培养实验证实mdk介导的CAFs可增强CSCC中肿瘤细胞的迁移和侵袭。结论:我们的研究结果提供了CSCC的全面细胞图谱,突出了cas与HPV感染和CSCC肿瘤进展的关系。这些结果也为CSCC患者提供了潜在的诊断和预后生物标志物。
{"title":"Multi-omics analysis unveils the role of cancer-associated fibroblasts in cutaneous squamous cell carcinoma.","authors":"Xiaochuan Wang, Tingrui Li, Yichao Jin, Jingjing Chen, XinYang, Zhen Guan, Mei Jin, Jingxian Zhang, Liangheng Xu, Sizhen Tao, Chunguang Li, Chunping Ao","doi":"10.1186/s12935-025-04061-w","DOIUrl":"10.1186/s12935-025-04061-w","url":null,"abstract":"<p><strong>Background: </strong>Human papillomavirus (HPV) infection is associated with an increased risk of cutaneous squamous cell carcinoma (CSCC). A comprehensive understanding of the cellular heterogeneity of HPV-positive and -negative CSCC is crucial for improving diagnosis and preventing tumor progression.</p><p><strong>Methods: </strong>We conducted an integrated analysis of single-cell RNA and spatial transcriptomic data from different skin tissue sources to map the cellular landscape of the tumor microenvironment (TME) in both HPV positive and negative CSCC. Results were validated through multiplex immunohistochemistry (mIHC) and in vitro experiments.</p><p><strong>Results: </strong>We identified 10 major cell types in CSCC and normal skin samples, including epithelial cells, myeloid cells, T cells, fibroblasts, endothelial cells, B cells, smooth muscle cells, mast cells, melanocytes, and hair follicle cells. Notably, fibroblasts were found to be associated with tumor progression in CSCC with or without HPV infected. We further identified eight major CAF subtypes in CSCC, with iCAFs-CXCL2 promoting tumor progression, while iCAFs-PLA2G2A acted to suppress tumor growth. The MDK-ITGA6 pair was found to mediate interactions between fibroblasts and epithelial cells in CSCC. mIHC analysis confirmed elevated expression of MDK and ITGA6 in CSCC samples. Additionally, cell co-culture experiments confirmed that MDK-mediated CAFs were shown to enhance tumor cell migration and invasion in CSCC.</p><p><strong>Conclusion: </strong>Our findings provide a comprehensive cellular atlas of CSCC, highlighting the association of CAFs in HPV infection and tumor progression of CSCC. These results also offer potential diagnostic and prognostic biomarkers for CSCC patients.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"417"},"PeriodicalIF":6.0,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12628552/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145548468","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer Cell International
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1