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.
{"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}
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}
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.
{"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}
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.
{"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}
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.
{"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}
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.
{"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}
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.
{"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}
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.
{"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}
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.
{"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}
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.
{"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}