首页 > 最新文献

Journal of Cancer最新文献

英文 中文
Molecular Signature of Cisplatin Resistance in Ovarian Cancer Identifies Therapeutic Opportunities for Re-sensitization. 卵巢癌顺铂耐药的分子特征确定了再敏化的治疗机会。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.124252
Mahmoud Mansour, Sabrina van Ginkel, Maaz Alata, Ibrahim Bani, Isra Elhussin

Cisplatin remains a standard first-line therapy for epithelial ovarian cancer; however, chemoresistance leads to poor prognosis and high recurrence. Analysis of The Cancer Genome Atlas confirmed improved overall survival in cisplatin-sensitive tumors, underscoring the need for strategies to overcome resistance in clinical settings. Integrative bioinformatics of cisplatin-treated ovarian cancer datasets from the Gene Expression Omnibus (n=255) identified six molecular drivers of resistance: Kaiso (ZBTB33), pregnane X receptor (PXR), NF-κB, HER2 (ERBB2), P-glycoprotein (P-gp/ABCB1), and HIF1A. These targets were validated in ovarian tumor specimens via immunohistochemistry, confirming elevated expression in chemo-resistant disease. Additionally, the quantitative real-time PCR analysis confirms the transcriptional upregulation of the six resistance-associated genes in cisplatin-resistant SKOV3 and OVCAR-5 ovarian cancer cells, consistent with the immunohistochemistry findings. The average fold change in mRNA transcripts ranged from 2.4 for P-glycoprotein to 5 for both NF-kB and Kaiso. Although less well studied in ovarian cancer, Kaiso is known to regulate EMT and tumor invasion in other solid tumors. Functional studies using SKOV3 and OVCAR-5 cell lines demonstrated that knockdown of Kaiso via RNA interference significantly increased cisplatin-induced cell death, indicating a direct role in therapeutic resistance. Furthermore, we investigated the synergistic effects of combining stearidonic acid (SDA), a plant-based omega-3 fatty acid known to inhibit NF-κB, with cisplatin on cell death in SKOV3 and OVCAR-5 cell lines, and compared the results with those of each compound used individually. Interestingly, co-treatment with stearidonic acid (SDA) synergistically enhanced the cytotoxicity of cisplatin at a lower dose in both cell models. These findings reveal a clinically relevant resistance signature and highlight the therapeutic potential of combinatorial strategies that target both transcriptional regulators (e.g., Kaiso) and inflammatory signaling (e.g., NF-κB). Dual targeting of these pathways may resensitize tumors to cisplatin and improve outcomes for patients with advanced ovarian cancer.

顺铂仍然是上皮性卵巢癌的标准一线治疗;然而,化疗耐药导致预后差和高复发率。癌症基因组图谱的分析证实了顺铂敏感肿瘤的总生存率提高,强调了在临床环境中克服耐药策略的必要性。来自基因表达Omnibus (n=255)的顺铂治疗卵巢癌数据集的综合生物信息学鉴定出6个耐药分子驱动因素:Kaiso (ZBTB33)、妊娠X受体(PXR)、NF-κB、HER2 (ERBB2)、p -糖蛋白(P-gp/ABCB1)和HIF1A。这些靶点通过免疫组化在卵巢肿瘤标本中得到验证,证实在化疗耐药疾病中表达升高。此外,实时荧光定量PCR分析证实了顺铂耐药的SKOV3和OVCAR-5卵巢癌细胞中6个耐药相关基因的转录上调,与免疫组化结果一致。p -糖蛋白mRNA转录本的平均折叠变化从2.4倍到NF-kB和Kaiso的5倍不等。虽然对卵巢癌的研究较少,但已知Kaiso在其他实体肿瘤中调节EMT和肿瘤侵袭。对SKOV3和OVCAR-5细胞系的功能研究表明,通过RNA干扰敲低Kaiso可显著增加顺铂诱导的细胞死亡,表明其在治疗耐药中起直接作用。此外,我们还研究了硬脂酸(SDA)(一种已知可抑制NF-κB的植物基omega-3脂肪酸)与顺铂联合使用对SKOV3和OVCAR-5细胞系细胞死亡的协同作用,并将结果与每种化合物单独使用的结果进行了比较。有趣的是,在两种细胞模型中,与硬脂酸(SDA)共同治疗在较低剂量下协同增强顺铂的细胞毒性。这些发现揭示了临床相关的耐药特征,并强调了针对转录调节因子(如Kaiso)和炎症信号(如NF-κB)的组合策略的治疗潜力。这些途径的双重靶向可能会使肿瘤对顺铂重新敏感,并改善晚期卵巢癌患者的预后。
{"title":"Molecular Signature of Cisplatin Resistance in Ovarian Cancer Identifies Therapeutic Opportunities for Re-sensitization.","authors":"Mahmoud Mansour, Sabrina van Ginkel, Maaz Alata, Ibrahim Bani, Isra Elhussin","doi":"10.7150/jca.124252","DOIUrl":"10.7150/jca.124252","url":null,"abstract":"<p><p>Cisplatin remains a standard first-line therapy for epithelial ovarian cancer; however, chemoresistance leads to poor prognosis and high recurrence. Analysis of The Cancer Genome Atlas confirmed improved overall survival in cisplatin-sensitive tumors, underscoring the need for strategies to overcome resistance in clinical settings. Integrative bioinformatics of cisplatin-treated ovarian cancer datasets from the Gene Expression Omnibus (n=255) identified six molecular drivers of resistance: Kaiso (ZBTB33), pregnane X receptor (PXR), NF-κB, HER2 (ERBB2), P-glycoprotein (P-gp/ABCB1), and HIF1A. These targets were validated in ovarian tumor specimens via immunohistochemistry, confirming elevated expression in chemo-resistant disease. Additionally, the quantitative real-time PCR analysis confirms the transcriptional upregulation of the six resistance-associated genes in cisplatin-resistant SKOV3 and OVCAR-5 ovarian cancer cells, consistent with the immunohistochemistry findings. The average fold change in mRNA transcripts ranged from 2.4 for P-glycoprotein to 5 for both NF-kB and Kaiso. Although less well studied in ovarian cancer, Kaiso is known to regulate EMT and tumor invasion in other solid tumors. Functional studies using SKOV3 and OVCAR-5 cell lines demonstrated that knockdown of Kaiso via RNA interference significantly increased cisplatin-induced cell death, indicating a direct role in therapeutic resistance. Furthermore, we investigated the synergistic effects of combining stearidonic acid (SDA), a plant-based omega-3 fatty acid known to inhibit NF-κB, with cisplatin on cell death in SKOV3 and OVCAR-5 cell lines, and compared the results with those of each compound used individually. Interestingly, co-treatment with stearidonic acid (SDA) synergistically enhanced the cytotoxicity of cisplatin at a lower dose in both cell models. These findings reveal a clinically relevant resistance signature and highlight the therapeutic potential of combinatorial strategies that target both transcriptional regulators (e.g., Kaiso) and inflammatory signaling (e.g., NF-κB). Dual targeting of these pathways may resensitize tumors to cisplatin and improve outcomes for patients with advanced ovarian cancer.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"49-58"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719565/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819259","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}
引用次数: 0
Loss of CDKN2A/B is a Hallmark of RTK II Glioblastomas. CDKN2A/B缺失是RTK II型胶质母细胞瘤的标志。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.122609
Celina K Langwieder, Dorothee Hölzl, Georg Hutarew, Hans U Schlicker, Beate Alinger-Scharinger, Christoph Schwartz, Karl Sotlar, Theo F J Kraus

Glioblastomas represent the most prevalent primary brain tumors in adults. Due to their highly malignant biological behavior, they are classified as grade 4 according to the World Health Organization (WHO) classification of brain tumors. Despite the progress in understanding the molecular pathogenesis of these tumors, no curative therapy has been developed for patients with glioblastoma. In this study, an integrated comparative analysis of cyclin-dependent kinase inhibitor (CDKN) 2A/B chromosomal deletion was performed on 45 glioblastomas, representing the most frequent molecular subtypes of glioblastomas, receptor tyrosine kinase (RTK) I (n=13), RTK II (n=15), and the mesenchymal subtype (MES) (n=17). The analysis of copy number variation (CNV) profiles was conducted on CDKN2A/B losses. Subsequent statistical analysis was then applied to correlate the collected data with molecular glioblastoma epigenotypes. Loss of CDKN2A/B was found 44% (20/45) of all glioblastomas, thereby, in 46% (6/13) of RTK I, 67% (10/15) RTK II, and 24% (4/17) of MES. Statistical analysis showed that loss of CDKN2A/B is significant (p < 0.01) in RTK II compared with MES. Even though CDKN2A/B does not per se function as a molecular target, there is great potential for enhancing treatment outcomes through the restoration of the tumor-suppressing capabilities of CDKN2A/B. This strategy can be employed in therapeutic interventions and is a promising avenue for research. This efficacy of this approach demonstrates high potential, as evidenced by its efficacy in other tumors, including melanoma.

胶质母细胞瘤是成人中最常见的原发性脑肿瘤。由于其高度恶性的生物学行为,根据世界卫生组织(WHO)的脑肿瘤分类,它们被列为第4级。尽管在了解这些肿瘤的分子发病机制方面取得了进展,但尚无针对胶质母细胞瘤患者的有效治疗方法。在本研究中,对45个胶质母细胞瘤进行了细胞周期蛋白依赖性激酶抑制剂(CDKN) 2A/B染色体缺失的综合比较分析,代表了胶质母细胞瘤最常见的分子亚型,受体酪氨酸激酶(RTK) I (n=13), RTK II (n=15)和间充质亚型(MES) (n=17)。对CDKN2A/B基因缺失的拷贝数变异(CNV)谱进行分析。随后进行统计分析,将收集到的数据与分子胶质母细胞瘤表观基因型相关联。CDKN2A/B缺失在所有胶质母细胞瘤中占44%(20/45),因此在RTK I中占46% (6/13),RTK II中占67% (10/15),MES中占24%(4/17)。统计分析显示,与MES相比,RTK II中CDKN2A/B的缺失显著(p < 0.01)。尽管CDKN2A/B本身不作为分子靶点起作用,但通过恢复CDKN2A/B的肿瘤抑制能力,有很大的潜力来提高治疗效果。该策略可用于治疗干预,是一种有前途的研究途径。这种方法的疗效显示出很高的潜力,正如它在其他肿瘤(包括黑色素瘤)中的疗效所证明的那样。
{"title":"Loss of <i>CDKN2A/B</i> is a Hallmark of RTK II Glioblastomas.","authors":"Celina K Langwieder, Dorothee Hölzl, Georg Hutarew, Hans U Schlicker, Beate Alinger-Scharinger, Christoph Schwartz, Karl Sotlar, Theo F J Kraus","doi":"10.7150/jca.122609","DOIUrl":"10.7150/jca.122609","url":null,"abstract":"<p><p>Glioblastomas represent the most prevalent primary brain tumors in adults. Due to their highly malignant biological behavior, they are classified as grade 4 according to the World Health Organization (WHO) classification of brain tumors. Despite the progress in understanding the molecular pathogenesis of these tumors, no curative therapy has been developed for patients with glioblastoma. In this study, an integrated comparative analysis of <i>cyclin-dependent kinase inhibitor (CDKN) 2A/B</i> chromosomal deletion was performed on 45 glioblastomas, representing the most frequent molecular subtypes of glioblastomas, receptor tyrosine kinase (RTK) I (n=13), RTK II (n=15), and the mesenchymal subtype (MES) (n=17). The analysis of copy number variation (CNV) profiles was conducted on <i>CDKN2A/B</i> losses. Subsequent statistical analysis was then applied to correlate the collected data with molecular glioblastoma epigenotypes. Loss of <i>CDKN2A/B</i> was found 44% (20/45) of all glioblastomas, thereby, in 46% (6/13) of RTK I, 67% (10/15) RTK II, and 24% (4/17) of MES. Statistical analysis showed that loss of <i>CDKN2A/B</i> is significant (p < 0.01) in RTK II compared with MES. Even though <i>CDKN2A/B</i> does not per se function as a molecular target, there is great potential for enhancing treatment outcomes through the restoration of the tumor-suppressing capabilities of <i>CDKN2A/B</i>. This strategy can be employed in therapeutic interventions and is a promising avenue for research. This efficacy of this approach demonstrates high potential, as evidenced by its efficacy in other tumors, including melanoma.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"1-9"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719383/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819263","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}
引用次数: 0
Interleukin-17 receptor A drives cancer stem-like properties in colorectal cancer through STAT3 activation. 白细胞介素-17受体A通过STAT3激活驱动结直肠癌的癌干样特性。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.121654
Jeng-Kai Jiang, Chi-Hung Lin, Chun-Chi Lin, Liang-Chuan Lo, Po-Yen Sung, Zhen-Yu Wen, Chien-Ping Lin, Ting-An Chang, Chih-Yung Yang

Cancer stem cells (CSCs) play pivotal roles in tumor relapse, metastasis, and therapy resistance. Interleukin 17 receptor A (IL-17RA) is a key mediator in colorectal cancer (CRC) pathogenesis and progression. Our recent study demonstrated that reduced IL-17RA expression correlates with favorable prognosis in CRC patients and suppresses tumor growth in murine models. This study aimed to investigate the role of IL-17RA in promoting cancer stem-like properties and its impact on colorectal cancer prognosis and chemoresistance. Expression levels of IL-17RA and CSC markers in CRC cells were evaluated using quantitative real-time polymerase chain reaction and Western blotting. Kaplan-Meier analysis of 68 CRC patients revealed that high IL-17RA expression is associated with poor clinical outcomes. To investigate IL-17RA's functional role, CRC cells with stable IL-17RA overexpression were analyzed for changes in CSC marker expression, sphere formation, and 5-fluorouracil (5-FU) resistance. IL-17RA overexpression significantly increased CSC marker expression, including cluster of differentiation 133 (CD133), leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), sex determining region Y-box 2 (SOX2), and enhanced tumor sphere formation and 5-FU resistance in SW620 cells. Specific inhibitors of IL-17RA signaling, such as the STAT3 inhibitor Stattic, reduced the expression of CD133, LGR5, ALDHA1, SOX2 and c-MYC, as well as tumor sphere formation in SW620 cells. These findings elucidate a novel IL-17RA-STAT3 axis that regulates CSC properties in CRC and highlight IL-17RA as a promising prognostic biomarker and therapeutic target for CRC treatment.

肿瘤干细胞(CSCs)在肿瘤复发、转移和治疗抵抗中起着关键作用。白细胞介素17受体A (IL-17RA)是结直肠癌(CRC)发病和进展的关键调节因子。我们最近的研究表明,在小鼠模型中,IL-17RA表达的降低与CRC患者的良好预后相关,并抑制肿瘤生长。本研究旨在探讨IL-17RA在促进肿瘤干细胞样特性中的作用及其对结直肠癌预后和化疗耐药的影响。采用实时定量聚合酶链反应和Western blotting检测CRC细胞中IL-17RA和CSC标志物的表达水平。对68例结直肠癌患者的Kaplan-Meier分析显示,高IL-17RA表达与较差的临床预后相关。为了研究IL-17RA的功能作用,我们分析了IL-17RA稳定过表达的CRC细胞在CSC标志物表达、球体形成和5-氟尿嘧啶(5-FU)耐药性方面的变化。IL-17RA过表达显著增加CSC标志物的表达,包括分化簇133 (CD133)、富含亮氨酸的重复- G蛋白偶联受体5 (LGR5)、性别决定区Y-box 2 (SOX2),并增强SW620细胞的肿瘤球形成和5- fu耐药性。IL-17RA信号的特异性抑制剂,如STAT3抑制剂Stattic,可降低SW620细胞中CD133、LGR5、ALDHA1、SOX2和c-MYC的表达以及肿瘤球的形成。这些发现阐明了一种新的IL-17RA- stat3轴调节CRC中CSC的特性,并突出了IL-17RA作为CRC治疗的有希望的预后生物标志物和治疗靶点。
{"title":"Interleukin-17 receptor A drives cancer stem-like properties in colorectal cancer through STAT3 activation.","authors":"Jeng-Kai Jiang, Chi-Hung Lin, Chun-Chi Lin, Liang-Chuan Lo, Po-Yen Sung, Zhen-Yu Wen, Chien-Ping Lin, Ting-An Chang, Chih-Yung Yang","doi":"10.7150/jca.121654","DOIUrl":"10.7150/jca.121654","url":null,"abstract":"<p><p>Cancer stem cells (CSCs) play pivotal roles in tumor relapse, metastasis, and therapy resistance. Interleukin 17 receptor A (IL-17RA) is a key mediator in colorectal cancer (CRC) pathogenesis and progression. Our recent study demonstrated that reduced IL-17RA expression correlates with favorable prognosis in CRC patients and suppresses tumor growth in murine models. This study aimed to investigate the role of IL-17RA in promoting cancer stem-like properties and its impact on colorectal cancer prognosis and chemoresistance. Expression levels of IL-17RA and CSC markers in CRC cells were evaluated using quantitative real-time polymerase chain reaction and Western blotting. Kaplan-Meier analysis of 68 CRC patients revealed that high IL-17RA expression is associated with poor clinical outcomes. To investigate IL-17RA's functional role, CRC cells with stable IL-17RA overexpression were analyzed for changes in CSC marker expression, sphere formation, and 5-fluorouracil (5-FU) resistance. IL-17RA overexpression significantly increased CSC marker expression, including cluster of differentiation 133 (CD133), leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), sex determining region Y-box 2 (SOX2), and enhanced tumor sphere formation and 5-FU resistance in SW620 cells. Specific inhibitors of IL-17RA signaling, such as the STAT3 inhibitor Stattic, reduced the expression of CD133, LGR5, ALDHA1, SOX2 and c-MYC, as well as tumor sphere formation in SW620 cells. These findings elucidate a novel IL-17RA-STAT3 axis that regulates CSC properties in CRC and highlight IL-17RA as a promising prognostic biomarker and therapeutic target for CRC treatment.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"74-85"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719591/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819267","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}
引用次数: 0
The Role of Inflammatory Biomarkers in PIPAC: Predicting Survival and Treatment Completion in Patients with Peritoneal Metastasis. 炎症生物标志物在PIPAC中的作用:预测腹膜转移患者的生存和治疗完成。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.123687
Signe Roensholdt, Martin Graversen, Sönke Detlefsen, Claus Fristrup, Per Pfeiffer, Michael Bau Mortensen

Introduction: Appropriate patient selection is essential for optimising outcomes in individuals with peritoneal metastasis (PM) undergoing treatment with Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC). This study investigated the prognostic value of pretreatment inflammatory biomarkers and explored their ability to predict the possibility of completion of three or more PIPAC treatments. Method: This observational study analysed prospectively collected data from patients with PM of gastrointestinal or ovarian origin enrolled in the PIPAC OPC-1 or OPC-2 studies between March 2015 and January 2022. Six biomarkers were examined: Neutrophil-to-Lymphocyte Ratio (NLR), Platelet-to-Lymphocyte Ratio, Systemic Immune-Inflammation Index (SII), C-reactive protein, modified Glasgow Prognostic Score, and Prognostic Nutritional Index. Biomarkers were obtained pretreatment, and treated as continuous variables. Survival was assessed using Kaplan-Meier and Cox regression analyses, adjusting for covariates available prior to the first PIPAC. ROC analysis was used to evaluate the predictive performance. A p-value less than 0.05 was considered statistically significant. Results: The cohort consisted of 130 patients, with a median overall survival (OS) of 8.7 months. Sixty percent of the patients received three or more PIPAC treatments. Elevated levels of all six biomarkers were significantly associated with reduced OS. In the multivariate analysis, five biomarkers remained independently associated with survival. NLR and SII demonstrated moderate discriminatory power (AUC > 0.70) for predicting the completion of three or more treatments. Conclusion: Pretreatment inflammatory biomarkers are objective, readily accessible and clinically applicable tools that may support the selection of appropriate candidates for PIPAC. The findings of this study encourage the integration of biomarker assessments into future PIPAC research protocols.

简介:适当的患者选择对于腹膜转移(PM)患者接受加压腹膜内气溶胶化疗(PIPAC)治疗的结果优化至关重要。本研究探讨了预处理炎症生物标志物的预后价值,并探讨了它们预测三次或更多PIPAC治疗完成可能性的能力。方法:本观察性研究对2015年3月至2022年1月期间参加PIPAC OPC-1或OPC-2研究的胃肠道或卵巢源性PM患者的前瞻性数据进行了分析。检测六项生物标志物:中性粒细胞与淋巴细胞比率(NLR)、血小板与淋巴细胞比率、全身免疫炎症指数(SII)、c反应蛋白、改良格拉斯哥预后评分和预后营养指数。预处理获得生物标志物,并作为连续变量处理。使用Kaplan-Meier和Cox回归分析评估生存率,调整第一次PIPAC之前可用的协变量。采用ROC分析评估预测效果。p值小于0.05被认为具有统计学意义。结果:该队列包括130例患者,中位总生存期(OS)为8.7个月。60%的患者接受了三次或更多的PIPAC治疗。所有六种生物标志物水平升高与OS降低显著相关。在多变量分析中,五个生物标志物仍然独立地与生存相关。NLR和SII在预测三种或三种以上治疗的完成度方面表现出中等的区分力(AUC > 0.70)。结论:预处理炎症生物标志物是客观的、容易获得的和临床适用的工具,可以支持选择合适的PIPAC候选药物。这项研究的发现鼓励将生物标志物评估纳入未来的PIPAC研究方案。
{"title":"The Role of Inflammatory Biomarkers in PIPAC: Predicting Survival and Treatment Completion in Patients with Peritoneal Metastasis.","authors":"Signe Roensholdt, Martin Graversen, Sönke Detlefsen, Claus Fristrup, Per Pfeiffer, Michael Bau Mortensen","doi":"10.7150/jca.123687","DOIUrl":"10.7150/jca.123687","url":null,"abstract":"<p><p><b>Introduction:</b> Appropriate patient selection is essential for optimising outcomes in individuals with peritoneal metastasis (PM) undergoing treatment with Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC). This study investigated the prognostic value of pretreatment inflammatory biomarkers and explored their ability to predict the possibility of completion of three or more PIPAC treatments. <b>Method:</b> This observational study analysed prospectively collected data from patients with PM of gastrointestinal or ovarian origin enrolled in the PIPAC OPC-1 or OPC-2 studies between March 2015 and January 2022. Six biomarkers were examined: Neutrophil-to-Lymphocyte Ratio (NLR), Platelet-to-Lymphocyte Ratio, Systemic Immune-Inflammation Index (SII), C-reactive protein, modified Glasgow Prognostic Score, and Prognostic Nutritional Index. Biomarkers were obtained pretreatment, and treated as continuous variables. Survival was assessed using Kaplan-Meier and Cox regression analyses, adjusting for covariates available prior to the first PIPAC. ROC analysis was used to evaluate the predictive performance. A p-value less than 0.05 was considered statistically significant. <b>Results:</b> The cohort consisted of 130 patients, with a median overall survival (OS) of 8.7 months. Sixty percent of the patients received three or more PIPAC treatments. Elevated levels of all six biomarkers were significantly associated with reduced OS. In the multivariate analysis, five biomarkers remained independently associated with survival. NLR and SII demonstrated moderate discriminatory power (AUC > 0.70) for predicting the completion of three or more treatments. <b>Conclusion:</b> Pretreatment inflammatory biomarkers are objective, readily accessible and clinically applicable tools that may support the selection of appropriate candidates for PIPAC. The findings of this study encourage the integration of biomarker assessments into future PIPAC research protocols.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"10-20"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719556/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819211","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}
引用次数: 0
Multi-Omics and Single-Cell Dissection of Exostosin Glycosyltransferases (EXT1/EXT2) Reveals Divergent Oncogenic Roles and Therapeutic Vulnerabilities in Gliomas. 外泌素糖基转移酶(EXT1/EXT2)的多组学和单细胞解剖揭示了胶质瘤中不同的致癌作用和治疗脆弱性。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.123965
Yi-Chun Chiang, Chih-Yang Wang, Neethu Palekkode, Shun-Fa Yang, Kai-Fu Chang, Ching-Chung Ko, Chih-Hsuan Chang, Hui-Ru Lin, Chi-Jen Wu, Yu-Cheng Ho, Chih-Chun Lin, Chien-Han Yuan, Sachin Kumar, Dahlak Daniel Solomon, Juan Lorell Ngadio, Fitria Sari Wulandari, Do Thi Minh Xuan, Chung-Bao Hsieh, Meng-Chi Yen, I-Jeng Yeh, Pi-Chan Ko, Chia-Lung Shih, Hoi-Bor Chan, Yung-Kuo Lee, Ngoc Uyen Nhi Nguyen

Exostosin glycosyltransferase 1 (EXT1) and exostosin glycosyltransferase 2 (EXT2) catalyze heparan sulfate chain elongation and are increasingly implicated in cancer biology, but their roles in gliomas remain incompletely defined. Here, we performed an integrative multi-omics analysis to dissect the transcriptional, epigenetic, and microenvironmental landscape of EXT1 and EXT2 across gliomas. Bulk transcriptomic data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) revealed that both EXT1 and EXT2 are upregulated in high-grade gliomas and associate with adverse survival, with EXT1 showing the strongest and most consistent prognostic impact. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) indicated that EXT1-high tumors are enriched for DNA damage and replication stress programs, cell cycle progression, inflammatory response, and stromal activation pathways, whereas EXT2 expression is preferentially linked to extracellular matrix remodeling, cytoskeletal organization and angiogenesis-related signaling. Single-cell RNA sequencing and Immune deconvolution using Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) showed that EXT1 correlates with increased stromal and immune scores, and reduced cytotoxic T cell signatures, consistent with an immunosuppressive tumor microenvironment. EXT2 expression is enriched in gliomas with pronounced vascular and mesenchymal features, supporting a complementary role in invasive growth and tissue remodeling. Immunohistochemistry on a glioma tissue microarray validated the upregulation of EXT1 protein in high-grade tumors. The study findings identified EXT1 as a central glycosylation-linked regulator of replication stress tolerance and immune remodeling in gliomas, and suggest that EXT2 contributes to extracellular matrix and cytoskeletal reprogramming. The exostosin axis represents a promising source of prognostic biomarkers and potential therapeutic targets in glioma.

Exostosin糖基转移酶1 (EXT1)和Exostosin糖基转移酶2 (EXT2)催化硫酸肝素链延伸,并越来越多地参与癌症生物学,但它们在胶质瘤中的作用仍不完全明确。在这里,我们进行了一项综合多组学分析,以解剖神经胶质瘤中EXT1和EXT2的转录、表观遗传和微环境景观。来自癌症基因组图谱(TCGA)和中国胶质瘤基因组图谱(CGGA)的大量转录组学数据显示,EXT1和EXT2在高级别胶质瘤中均上调,并与不良生存相关,其中EXT1表现出最强且最一致的预后影响。基因集富集分析(GSEA)和基因集变异分析(GSVA)表明,ext1高的肿瘤富含DNA损伤和复制应激程序、细胞周期进程、炎症反应和基质激活途径,而EXT2的表达优先与细胞外基质重塑、细胞骨架组织和血管生成相关的信号传导有关。单细胞RNA测序和免疫反褶积使用细胞类型鉴定通过估计RNA转录物的相对亚群(CIBERSORT)和估计恶性肿瘤组织中的基质和免疫细胞使用表达数据(ESTIMATE)表明,EXT1与增加基质和免疫评分相关,并减少细胞毒性T细胞特征,与免疫抑制肿瘤微环境一致。EXT2在具有明显血管和间质特征的胶质瘤中表达丰富,在侵袭性生长和组织重塑中支持互补作用。神经胶质瘤组织芯片上的免疫组织化学证实了EXT1蛋白在高级别肿瘤中的上调。研究结果表明,EXT1是神经胶质瘤中复制应激耐受性和免疫重塑的中心糖基化调控因子,并表明EXT2有助于细胞外基质和细胞骨架重编程。外泌素轴是胶质瘤预后生物标志物和潜在治疗靶点的一个有希望的来源。
{"title":"Multi-Omics and Single-Cell Dissection of Exostosin Glycosyltransferases (EXT1/EXT2) Reveals Divergent Oncogenic Roles and Therapeutic Vulnerabilities in Gliomas.","authors":"Yi-Chun Chiang, Chih-Yang Wang, Neethu Palekkode, Shun-Fa Yang, Kai-Fu Chang, Ching-Chung Ko, Chih-Hsuan Chang, Hui-Ru Lin, Chi-Jen Wu, Yu-Cheng Ho, Chih-Chun Lin, Chien-Han Yuan, Sachin Kumar, Dahlak Daniel Solomon, Juan Lorell Ngadio, Fitria Sari Wulandari, Do Thi Minh Xuan, Chung-Bao Hsieh, Meng-Chi Yen, I-Jeng Yeh, Pi-Chan Ko, Chia-Lung Shih, Hoi-Bor Chan, Yung-Kuo Lee, Ngoc Uyen Nhi Nguyen","doi":"10.7150/jca.123965","DOIUrl":"10.7150/jca.123965","url":null,"abstract":"<p><p>Exostosin glycosyltransferase 1 (<i>EXT1</i>) and exostosin glycosyltransferase 2 (<i>EXT2</i>) catalyze heparan sulfate chain elongation and are increasingly implicated in cancer biology, but their roles in gliomas remain incompletely defined. Here, we performed an integrative multi-omics analysis to dissect the transcriptional, epigenetic, and microenvironmental landscape of <i>EXT1</i> and <i>EXT2</i> across gliomas. Bulk transcriptomic data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) revealed that both EXT1 and EXT2 are upregulated in high-grade gliomas and associate with adverse survival, with EXT1 showing the strongest and most consistent prognostic impact. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) indicated that EXT1-high tumors are enriched for DNA damage and replication stress programs, cell cycle progression, inflammatory response, and stromal activation pathways, whereas EXT2 expression is preferentially linked to extracellular matrix remodeling, cytoskeletal organization and angiogenesis-related signaling. Single-cell RNA sequencing and Immune deconvolution using Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) showed that <i>EXT1</i> correlates with increased stromal and immune scores, and reduced cytotoxic T cell signatures, consistent with an immunosuppressive tumor microenvironment. <i>EXT2</i> expression is enriched in gliomas with pronounced vascular and mesenchymal features, supporting a complementary role in invasive growth and tissue remodeling. Immunohistochemistry on a glioma tissue microarray validated the upregulation of <i>EXT1</i> protein in high-grade tumors. The study findings identified <i>EXT1</i> as a central glycosylation-linked regulator of replication stress tolerance and immune remodeling in gliomas, and suggest that <i>EXT2</i> contributes to extracellular matrix and cytoskeletal reprogramming. The exostosin axis represents a promising source of prognostic biomarkers and potential therapeutic targets in glioma.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"177-196"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719590/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819214","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}
引用次数: 0
Cancer Cell Dormancy and Chemotherapy Resistance. 癌细胞休眠和化疗耐药性。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.123742
Jingxian Hu, Wei Zhou, Ying Zhao, Peiran Li, Zhenying Zhao, Wei Wang, Wenhong Wang, Yijia Wang

Cancer cell dormancy is associated with tumor recurrence and metastasis. Chemotherapy usually induces dormancy as external pressure on the tumor. Dormant cells have considerable resistance to antitumor drugs, although they are not harmful to the host if they do not wake up. Chemotherapy induces a dormant phenotype through remodeling of the tumor microenvironment and alteration of intracellular signaling networks. Multiple adaptive mechanisms that confer drug resistance have been identified in these dormant cells, including the unfolded protein response to endoplasmic reticulum stress, metabolic reprogramming favoring oxidative phosphorylation to avoid damage from oxidative stress, and autophagy to realize the circular utilization of energy. However, dormancy is reversible. The conversion between dormancy and awakening of the tumor during chemotherapy and the recovery period after treatment is modulated by several factors, including the dose and cycle of treatment, and individual differences among patients. The direct elimination of cancer cells or permanent dormancy by chemotherapy predicts favorable outcomes. According to this theory, understanding the mechanisms of cancer dormancy and awakening under chemotherapy and improving prognosis using suitable treatment strategies requires further investigation. This review analyzed studies on cancer cell dormancy and response to chemotherapy to identify potential novel interests for future studies and probable strategies to optimize chemotherapy in clinical trials.

肿瘤细胞休眠与肿瘤的复发和转移有关。化疗通常以外部压迫肿瘤的方式诱导休眠。休眠细胞对抗肿瘤药物有相当大的抵抗力,尽管如果它们不醒来对宿主是无害的。化疗通过重塑肿瘤微环境和改变细胞内信号网络诱导休眠表型。在这些休眠细胞中已经发现了多种赋予耐药性的适应性机制,包括未折叠蛋白对内质网应激的反应,有利于氧化磷酸化的代谢重编程以避免氧化应激的损伤,以及实现能量循环利用的自噬。然而,休眠是可逆的。肿瘤在化疗期间的休眠和苏醒与治疗后恢复期之间的转换受多种因素的调节,包括治疗剂量和周期以及患者的个体差异。通过化疗直接消除癌细胞或使其永久休眠预示着有利的结果。根据这一理论,了解化疗下肿瘤休眠和觉醒的机制,并采用合适的治疗策略改善预后,需要进一步研究。这篇综述分析了关于癌细胞休眠和化疗反应的研究,以确定未来研究的潜在新兴趣和优化临床试验化疗的可能策略。
{"title":"Cancer Cell Dormancy and Chemotherapy Resistance.","authors":"Jingxian Hu, Wei Zhou, Ying Zhao, Peiran Li, Zhenying Zhao, Wei Wang, Wenhong Wang, Yijia Wang","doi":"10.7150/jca.123742","DOIUrl":"10.7150/jca.123742","url":null,"abstract":"<p><p>Cancer cell dormancy is associated with tumor recurrence and metastasis. Chemotherapy usually induces dormancy as external pressure on the tumor. Dormant cells have considerable resistance to antitumor drugs, although they are not harmful to the host if they do not wake up. Chemotherapy induces a dormant phenotype through remodeling of the tumor microenvironment and alteration of intracellular signaling networks. Multiple adaptive mechanisms that confer drug resistance have been identified in these dormant cells, including the unfolded protein response to endoplasmic reticulum stress, metabolic reprogramming favoring oxidative phosphorylation to avoid damage from oxidative stress, and autophagy to realize the circular utilization of energy. However, dormancy is reversible. The conversion between dormancy and awakening of the tumor during chemotherapy and the recovery period after treatment is modulated by several factors, including the dose and cycle of treatment, and individual differences among patients. The direct elimination of cancer cells or permanent dormancy by chemotherapy predicts favorable outcomes. According to this theory, understanding the mechanisms of cancer dormancy and awakening under chemotherapy and improving prognosis using suitable treatment strategies requires further investigation. This review analyzed studies on cancer cell dormancy and response to chemotherapy to identify potential novel interests for future studies and probable strategies to optimize chemotherapy in clinical trials.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"59-73"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719572/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819315","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}
引用次数: 0
CES3 promotes NSCLC progression via lipid metabolic reprogramming regulated by TFAP2A. CES3通过TFAP2A调控的脂质代谢重编程促进NSCLC进展。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.118395
Pengfei Luo, Zirui Huang, Sijuan Ding, Zhangwen Tang, Yanhong Wei, Shaohui Jiang, Ruoting Tang, Fang Li, Han Yang, Lujun Zhao

Metabolic reprogramming is an important feature in non-small cell lung cancer (NSCLC) that can result in therapeutic resistance. Exploring dysregulated lipid metabolism in NSCLC will accelerate the development of potential lipid biomarkers to target and control the malignant progression of NSCLC. In this study, RNA next-generation sequencing of 25 paired NSCLC specimens and adjacent normal tissues was used to find that carboxylesterase 3 (CES3) was upregulated in NSCLC. Knockdown of CES3 significantly inhibited NSCLC cell proliferation and invasion. Additionally, CES3 inhibition promoted lipid accumulation in NSCLC cells. Furthermore, we found transcription factor AP-2α (TFAP2A) could regulate CES3 levels in NSCLC. TFAP2A was found upregulated in NSCLC and correlated with poorer outcome. Inhibiting TFAP2A resulted in suppressed cell proliferation as well as invasion while increasing the lipid accumulation in NSCLC. CES3 overexpression could reverse the impact of TFAP2A inhibition on NSCLC progression. In summary, TFAP2A dysregulation resulted in CES3 overexpression and the following NSCLC tumorigenesis. Targeting the TFAP2A/CES3 axis may represent a promising therapeutic strategy for NSCLC in the future.

代谢重编程是非小细胞肺癌(NSCLC)的一个重要特征,可导致治疗耐药。探索非小细胞肺癌中脂质代谢失调将加速开发潜在的脂质生物标志物,以靶向和控制非小细胞肺癌的恶性进展。本研究通过对25个配对的NSCLC标本及邻近正常组织进行RNA新一代测序,发现羧酸酯酶3 (CES3)在NSCLC中表达上调。低表达CES3可显著抑制NSCLC细胞的增殖和侵袭。此外,CES3抑制促进了非小细胞肺癌细胞的脂质积累。此外,我们发现转录因子AP-2α (TFAP2A)可以调节NSCLC中CES3的水平。TFAP2A在NSCLC中表达上调,并与预后较差相关。抑制TFAP2A可抑制非小细胞肺癌的细胞增殖和侵袭,同时增加脂质积累。CES3过表达可以逆转TFAP2A抑制对NSCLC进展的影响。综上所述,TFAP2A失调导致CES3过表达和随后的NSCLC肿瘤发生。靶向TFAP2A/CES3轴可能是未来治疗非小细胞肺癌的一种有希望的治疗策略。
{"title":"CES3 promotes NSCLC progression via lipid metabolic reprogramming regulated by TFAP2A.","authors":"Pengfei Luo, Zirui Huang, Sijuan Ding, Zhangwen Tang, Yanhong Wei, Shaohui Jiang, Ruoting Tang, Fang Li, Han Yang, Lujun Zhao","doi":"10.7150/jca.118395","DOIUrl":"10.7150/jca.118395","url":null,"abstract":"<p><p>Metabolic reprogramming is an important feature in non-small cell lung cancer (NSCLC) that can result in therapeutic resistance. Exploring dysregulated lipid metabolism in NSCLC will accelerate the development of potential lipid biomarkers to target and control the malignant progression of NSCLC. In this study, RNA next-generation sequencing of 25 paired NSCLC specimens and adjacent normal tissues was used to find that carboxylesterase 3 (CES3) was upregulated in NSCLC. Knockdown of CES3 significantly inhibited NSCLC cell proliferation and invasion. Additionally, CES3 inhibition promoted lipid accumulation in NSCLC cells. Furthermore, we found transcription factor AP-2α (TFAP2A) could regulate CES3 levels in NSCLC. TFAP2A was found upregulated in NSCLC and correlated with poorer outcome. Inhibiting TFAP2A resulted in suppressed cell proliferation as well as invasion while increasing the lipid accumulation in NSCLC. CES3 overexpression could reverse the impact of TFAP2A inhibition on NSCLC progression. In summary, TFAP2A dysregulation resulted in CES3 overexpression and the following NSCLC tumorigenesis. Targeting the TFAP2A/CES3 axis may represent a promising therapeutic strategy for NSCLC in the future.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"99-108"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719574/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819379","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}
引用次数: 0
Macrophages in Colorectal Cancer: from Normal Mucosa to Distant Metastasis: Beyond the M1/M2 Paradigm. 巨噬细胞在结直肠癌中的作用:从正常粘膜到远处转移:超越M1/M2范式。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.126772
Sergii Pavlov, Esraa Ali, Filip Ambrozkiewicz, Wenjing Ye, Marie Rajtmajerová, Václav Liška, Kari Hemminki, Andriy Trailin

Colorectal cancer (CRC) is the third most common malignancy and leading cause of mortality worldwide. Tumor microenvironment (TME) strongly influences CRC growth, immune evasion, and metastasis. Among various immune cells, tumor-associated macrophages (TAMs) act as key regulators of cancer progression. Although traditionally classified as M1 (pro-inflammatory, anti-tumor) or M2 (anti-inflammatory, pro-tumor), single-cell RNA sequencing and spatial transcriptomics have revealed that macrophage phenotypes exist along a continuum, challenging the classic dichotomy. This review investigates macrophages throughout CRC development, from normal mucosa to adenoma, primary tumor, and liver metastasis. Early adenomas feature M1-like macrophages that drive local inflammation, whereas advanced adenomas and invasive CRC comprise M2-like macrophages promoting angiogenesis, extracellular matrix remodeling, and immunosuppression. TAMs are crucial in CRC metastasis, particularly to the liver. M2-polarized Kupffer cells express CD206 and CD163, secrete hepatocyte growth factor, and activate PI3K/AKT signaling, thus aiding extravasation, survival, and proliferation of metastatic cells. They also foster lymphangiogenesis and immunosuppression through release of IL-10 and TGF-β. CRC's consensus molecular subtype (CMS) impacts the profile of TAMs: CMS1 (microsatellite instability-high) tumors typically harbor an anti-tumor M1 macrophages, while CMS4 (mesenchymal) tumors are enriched in M2-like TAMs, which facilitate stromal remodeling and angiogenesis, ultimately contributing to a poor prognosis. Spatial distribution also matters. Abundant M1 macrophages at the invasive margin correlate with better outcomes, whereas M2 macrophages in tumor centers and metastatic sites drive disease progression. Some CD206+ macrophages, however, support vascular normalization, which can limit metastasis. These findings underscore the complexity of TAMs in CRC and highlight the necessity of multi-marker phenotyping. Given the limitations of the M1/M2 paradigm, advanced techniques such as spatial transcriptomics and single-cell RNA sequencing offer novel insights into TAM heterogeneity. Future therapeutic strategies targeting TAMs, including metabolic reprogramming, epigenetic modulators, and immune checkpoint inhibitors, hold promise for improving CRC patient outcomes by shifting the balance toward an anti-tumor immune response.

结直肠癌(CRC)是世界上第三大最常见的恶性肿瘤,也是导致死亡的主要原因。肿瘤微环境(Tumor microenvironment, TME)强烈影响结直肠癌的生长、免疫逃避和转移。在各种免疫细胞中,肿瘤相关巨噬细胞(tam)是癌症进展的关键调节因子。虽然传统上被分类为M1(促炎、抗肿瘤)或M2(抗炎、促肿瘤),但单细胞RNA测序和空间转录组学显示,巨噬细胞表型是连续存在的,挑战了传统的两分法。本文综述了巨噬细胞在结直肠癌发展过程中的作用,从正常粘膜到腺瘤、原发肿瘤和肝转移。早期腺瘤以驱动局部炎症的m1样巨噬细胞为特征,而晚期腺瘤和侵袭性结直肠癌则包括促进血管生成、细胞外基质重塑和免疫抑制的m2样巨噬细胞。tam在结直肠癌转移中起关键作用,特别是转移到肝脏。m2极化Kupffer细胞表达CD206和CD163,分泌肝细胞生长因子,激活PI3K/AKT信号,从而帮助转移细胞外溢、存活和增殖。它们还通过释放IL-10和TGF-β促进淋巴管生成和免疫抑制。CRC的共识分子亚型(CMS)影响tam的分布:CMS1(微卫星不稳定性高)肿瘤通常含有抗肿瘤的M1巨噬细胞,而CMS4(间质)肿瘤富含m2样tam,促进基质重塑和血管生成,最终导致预后不良。空间分布也很重要。侵袭边缘丰富的M1巨噬细胞与更好的预后相关,而肿瘤中心和转移部位的M2巨噬细胞驱动疾病进展。然而,一些CD206+巨噬细胞支持血管正常化,可以限制转移。这些发现强调了CRC中tam的复杂性,并强调了多标记表型的必要性。考虑到M1/M2模式的局限性,空间转录组学和单细胞RNA测序等先进技术为TAM异质性提供了新的见解。未来针对tam的治疗策略,包括代谢重编程、表观遗传调节剂和免疫检查点抑制剂,有望通过将平衡转向抗肿瘤免疫反应来改善结直肠癌患者的预后。
{"title":"Macrophages in Colorectal Cancer: from Normal Mucosa to Distant Metastasis: Beyond the M1/M2 Paradigm.","authors":"Sergii Pavlov, Esraa Ali, Filip Ambrozkiewicz, Wenjing Ye, Marie Rajtmajerová, Václav Liška, Kari Hemminki, Andriy Trailin","doi":"10.7150/jca.126772","DOIUrl":"10.7150/jca.126772","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is the third most common malignancy and leading cause of mortality worldwide. Tumor microenvironment (TME) strongly influences CRC growth, immune evasion, and metastasis. Among various immune cells, tumor-associated macrophages (TAMs) act as key regulators of cancer progression. Although traditionally classified as M1 (pro-inflammatory, anti-tumor) or M2 (anti-inflammatory, pro-tumor), single-cell RNA sequencing and spatial transcriptomics have revealed that macrophage phenotypes exist along a continuum, challenging the classic dichotomy. This review investigates macrophages throughout CRC development, from normal mucosa to adenoma, primary tumor, and liver metastasis. Early adenomas feature M1-like macrophages that drive local inflammation, whereas advanced adenomas and invasive CRC comprise M2-like macrophages promoting angiogenesis, extracellular matrix remodeling, and immunosuppression. TAMs are crucial in CRC metastasis, particularly to the liver. M2-polarized Kupffer cells express CD206 and CD163, secrete hepatocyte growth factor, and activate PI3K/AKT signaling, thus aiding extravasation, survival, and proliferation of metastatic cells. They also foster lymphangiogenesis and immunosuppression through release of IL-10 and TGF-β. CRC's consensus molecular subtype (CMS) impacts the profile of TAMs: CMS1 (microsatellite instability-high) tumors typically harbor an anti-tumor M1 macrophages, while CMS4 (mesenchymal) tumors are enriched in M2-like TAMs, which facilitate stromal remodeling and angiogenesis, ultimately contributing to a poor prognosis. Spatial distribution also matters. Abundant M1 macrophages at the invasive margin correlate with better outcomes, whereas M2 macrophages in tumor centers and metastatic sites drive disease progression. Some CD206+ macrophages, however, support vascular normalization, which can limit metastasis. These findings underscore the complexity of TAMs in CRC and highlight the necessity of multi-marker phenotyping. Given the limitations of the M1/M2 paradigm, advanced techniques such as spatial transcriptomics and single-cell RNA sequencing offer novel insights into TAM heterogeneity. Future therapeutic strategies targeting TAMs, including metabolic reprogramming, epigenetic modulators, and immune checkpoint inhibitors, hold promise for improving CRC patient outcomes by shifting the balance toward an anti-tumor immune response.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"157-176"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719567/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819285","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}
引用次数: 0
Comprehensive characterization of AP-1 adaptor complex genes in lung cancer reveals AP1AR as a novel prognostic and therapeutic biomarker. AP-1接头复合物基因在肺癌中的综合表征揭示了AP1AR作为一种新的预后和治疗生物标志物。
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.125763
Dahlak Daniel Solomon, I-Jeng Yeh, Hsin-Liang Liu, Che-Yu Su, Yung-Kuo Lee, Ching-Chung Ko, Hui-Ru Lin, Sachin Kumar, Do Thi Minh Xuan, Neethu Palekkode, Ayman Fathima, Hung-Yun Lin, Chih-Yang Wang, Meng-Chi Yen

Lung cancer remains the leading cause of cancer mortality. The AP-1 adaptor complex, including AP1AR, AP1S1, AP1S2, AP1S3, AP1M1, AP1M2, AP1B1, and AP1G1, functions as a conserved hub of vesicular trafficking, selecting cargo and coordinating clathrin-mediated transport. By shaping receptor recycling, membrane composition, and signal duration, AP-1 influences core cancer phenotypes such as proliferation, migration, and therapy response. However, the family-level role of AP-1 adaptors in lung cancer is incompletely defined. We systematically profiled all eight AP-1 adaptor genes using multi-omics datasets, survival resources, pharmacogenomic panels, Human Protein Atlas data, pathway enrichment, and single-cell RNA sequencing with cell-cell communication modeling. AP1AR was consistently upregulated in lung adenocarcinoma and independently associated with poorer overall survival. It was linked to cell-cycle progression, DNA replication checkpoints, hypoxia, and epithelial-to-mesenchymal transition (EMT). At single cell resolution, AP1AR also regulate malignant epithelial and fibroblast cell types. Pseudotime analyses revealed progressive activation along proliferative and EMT axes, and CellChat modeling indicated enhanced stromal and epithelial signaling. AP1S3 and AP1S1 showed complementary roles, associated with oncogenic/inflammatory signaling and immune-metabolic programs, respectively. These findings identify AP1AR as a clinically relevant biomarker and highlight AP-1 adaptor biology as an underexplored contributor to lung adenocarcinoma progression and therapeutic stratification.

肺癌仍然是癌症死亡的主要原因。AP-1接头复合体,包括AP1AR、AP1S1、AP1S2、AP1S3、AP1M1、AP1M2、AP1B1和AP1G1,作为囊泡运输的保守枢纽,选择货物和协调网格蛋白介导的运输。通过塑造受体循环、膜组成和信号持续时间,AP-1影响核心癌症表型,如增殖、迁移和治疗反应。然而,AP-1接头在肺癌中的家族作用尚未完全确定。我们使用多组学数据集、生存资源、药物基因组学面板、人类蛋白质图谱数据、途径富集和单细胞RNA测序以及细胞-细胞通讯模型系统地分析了所有八个AP-1接头基因。AP1AR在肺腺癌中持续上调,并与较差的总生存率独立相关。它与细胞周期进程、DNA复制检查点、缺氧和上皮到间质转化(EMT)有关。在单细胞分辨率下,AP1AR还调节恶性上皮细胞和成纤维细胞类型。伪时间分析显示沿增殖轴和EMT轴渐进式激活,CellChat模型显示间质和上皮信号传导增强。AP1S3和AP1S1显示互补作用,分别与致癌/炎症信号和免疫代谢程序相关。这些发现确定了AP1AR是一种临床相关的生物标志物,并强调了AP-1受体生物学在肺腺癌进展和治疗分层中的作用。
{"title":"Comprehensive characterization of AP-1 adaptor complex genes in lung cancer reveals AP1AR as a novel prognostic and therapeutic biomarker.","authors":"Dahlak Daniel Solomon, I-Jeng Yeh, Hsin-Liang Liu, Che-Yu Su, Yung-Kuo Lee, Ching-Chung Ko, Hui-Ru Lin, Sachin Kumar, Do Thi Minh Xuan, Neethu Palekkode, Ayman Fathima, Hung-Yun Lin, Chih-Yang Wang, Meng-Chi Yen","doi":"10.7150/jca.125763","DOIUrl":"10.7150/jca.125763","url":null,"abstract":"<p><p>Lung cancer remains the leading cause of cancer mortality. The AP-1 adaptor complex, including AP1AR, AP1S1, AP1S2, AP1S3, AP1M1, AP1M2, AP1B1, and AP1G1, functions as a conserved hub of vesicular trafficking, selecting cargo and coordinating clathrin-mediated transport. By shaping receptor recycling, membrane composition, and signal duration, AP-1 influences core cancer phenotypes such as proliferation, migration, and therapy response. However, the family-level role of AP-1 adaptors in lung cancer is incompletely defined. We systematically profiled all eight AP-1 adaptor genes using multi-omics datasets, survival resources, pharmacogenomic panels, Human Protein Atlas data, pathway enrichment, and single-cell RNA sequencing with cell-cell communication modeling. <i>AP1AR</i> was consistently upregulated in lung adenocarcinoma and independently associated with poorer overall survival. It was linked to cell-cycle progression, DNA replication checkpoints, hypoxia, and epithelial-to-mesenchymal transition (EMT). At single cell resolution, <i>AP1AR</i> also regulate malignant epithelial and fibroblast cell types. Pseudotime analyses revealed progressive activation along proliferative and EMT axes, and CellChat modeling indicated enhanced stromal and epithelial signaling. <i>AP1S3</i> and <i>AP1S1</i> showed complementary roles, associated with oncogenic/inflammatory signaling and immune-metabolic programs, respectively. These findings identify <i>AP1AR</i> as a clinically relevant biomarker and highlight AP-1 adaptor biology as an underexplored contributor to lung adenocarcinoma progression and therapeutic stratification.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"142-156"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719575/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819437","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}
引用次数: 0
Ganoderma microsporum Immunomodulatory Protein (GMI) Enhances Phagocytosis by Suppressing STAT3/CD47 Signaling in EGFR-Mutant NSCLC Resistant to Gefitinib and Osimertinib. 在对吉非替尼和奥西替尼耐药的egfr突变型NSCLC中,小孢子灵芝免疫调节蛋白(GMI)通过抑制STAT3/CD47信号增强吞噬作用
IF 3.2 3区 医学 Q2 ONCOLOGY Pub Date : 2026-01-01 DOI: 10.7150/jca.124363
Ya-Chu Hsieh, I-Lun Hsin, Ling-Yen Chiu, Yu-Chien Hung, Yu-Ting Kang, Hui-Yi Chang, Ching-Hsiung Lin, Jiunn-Liang Ko, Yu-Fan Liu

Target therapy is effective for epidermal growth factor receptor (EGFR) mutation in non-small cell lung cancer (NSCLC). However, resistance often occurs after treatment for several months. Macrophages have difficulty in devouring resistant cells. Ganoderma immunomodulatory protein (GMI) exhibits anti-tumour and immunomodulatory activities. This study aimed to investigate whether GMI overcomes Osimertinib (Tagrisso) and Gefitinib (Iressa) resistance via enhancing macrophage polarization. GMI attenuated signal transducer and activator of transcription 3 (STAT3) phosphorylation and downstream CD47 expression in parental and resistant cells via Western blot and RT-qPCR. Overexpressed STAT3 restored GMI-induced apoptosis and GMI-reduced transcription of CD47 in HCC827 and H1975 lung cancer cells. Phospho-STAT3 inhibitor (W1131) also reduced the expression of CD47 in NSCLC cells. The interaction between GMI and W1131 was effective in reducing phosphorylated STAT3 and CD47. ImageXpress Pico analysis revealed that GMI enhanced phagocytotic activity of macrophages toward tumour cells with Red CMTPX and Green CMFDA dyes. The results showed that GMI enhanced macrophage phagocytosis of lung cancer cells by inhibiting STAT3 and reducing CD47 expression. In addition, GMI enhanced M1 inhibition of M2 polarization but had no effect on M1 differentiation. This is the first study to demonstrate that GMI enhances macrophage phagocytosis and modulates the STAT3-CD47 axis to overcome EGFR-TKI resistance in NSCLC, highlighting its potential as a novel adjunct immunotherapeutic agent.

靶向治疗对表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)有效。然而,耐药性通常在治疗几个月后出现。巨噬细胞很难吞噬耐药细胞。灵芝免疫调节蛋白(GMI)具有抗肿瘤和免疫调节活性。本研究旨在探讨GMI是否通过增强巨噬细胞极化来克服奥西替尼(Tagrisso)和吉非替尼(Iressa)耐药。通过Western blot和RT-qPCR, GMI减弱了亲本和耐药细胞中转录3 (STAT3)磷酸化和下游CD47的表达。STAT3过表达恢复了gmi诱导的HCC827和H1975肺癌细胞的凋亡,gmi降低了CD47的转录。Phospho-STAT3 inhibitor (W1131)也能降低NSCLC细胞中CD47的表达。GMI和W1131之间的相互作用可有效降低磷酸化的STAT3和CD47。ImageXpress Pico分析显示,GMI增强了巨噬细胞对红色CMTPX和绿色CMFDA染色的肿瘤细胞的吞噬活性。结果表明,GMI通过抑制STAT3和降低CD47的表达,增强了肺癌细胞的巨噬细胞吞噬能力。此外,GMI增强了M1对M2极化的抑制作用,但对M1分化没有影响。这是第一个证明GMI增强巨噬细胞吞噬并调节STAT3-CD47轴以克服NSCLC中EGFR-TKI耐药的研究,突出了其作为一种新型辅助免疫治疗剂的潜力。
{"title":"<i>Ganoderma microsporum</i> Immunomodulatory Protein (GMI) Enhances Phagocytosis by Suppressing STAT3/CD47 Signaling in EGFR-Mutant NSCLC Resistant to Gefitinib and Osimertinib.","authors":"Ya-Chu Hsieh, I-Lun Hsin, Ling-Yen Chiu, Yu-Chien Hung, Yu-Ting Kang, Hui-Yi Chang, Ching-Hsiung Lin, Jiunn-Liang Ko, Yu-Fan Liu","doi":"10.7150/jca.124363","DOIUrl":"10.7150/jca.124363","url":null,"abstract":"<p><p>Target therapy is effective for epidermal growth factor receptor (EGFR) mutation in non-small cell lung cancer (NSCLC). However, resistance often occurs after treatment for several months. Macrophages have difficulty in devouring resistant cells. <i>Ganoderma</i> immunomodulatory protein (GMI) exhibits anti-tumour and immunomodulatory activities. This study aimed to investigate whether GMI overcomes Osimertinib (Tagrisso) and Gefitinib (Iressa) resistance via enhancing macrophage polarization. GMI attenuated signal transducer and activator of transcription 3 (STAT3) phosphorylation and downstream CD47 expression in parental and resistant cells via Western blot and RT-qPCR. Overexpressed STAT3 restored GMI-induced apoptosis and GMI-reduced transcription of CD47 in HCC827 and H1975 lung cancer cells. Phospho-STAT3 inhibitor (W1131) also reduced the expression of CD47 in NSCLC cells. The interaction between GMI and W1131 was effective in reducing phosphorylated STAT3 and CD47. ImageXpress Pico analysis revealed that GMI enhanced phagocytotic activity of macrophages toward tumour cells with Red CMTPX and Green CMFDA dyes. The results showed that GMI enhanced macrophage phagocytosis of lung cancer cells by inhibiting STAT3 and reducing CD47 expression. In addition, GMI enhanced M1 inhibition of M2 polarization but had no effect on M1 differentiation. This is the first study to demonstrate that GMI enhances macrophage phagocytosis and modulates the STAT3-CD47 axis to overcome EGFR-TKI resistance in NSCLC, highlighting its potential as a novel adjunct immunotherapeutic agent.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"17 1","pages":"86-98"},"PeriodicalIF":3.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719595/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145819350","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}
引用次数: 0
期刊
Journal of Cancer
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