Pub Date : 2025-12-12DOI: 10.1007/s00432-025-06393-6
Juanwen Zhang, Jin Liu, Alireza Bayani
Hematologic malignancies remain among the most difficult cancers to treat, challenged by profound heterogeneity, treatment-induced immune dysfunction, and the frequent emergence of drug resistance. Beyond tumor-intrinsic mechanisms, dysbiosis of the gut microbiome is increasingly recognized as a critical determinant of therapeutic outcomes, shaping hematopoiesis, immune responses, and drug metabolism. Bacteriophage (phage) therapy has re-emerged as a precision tool capable of selectively eradicating pathogenic taxa while preserving commensal short-chain fatty acid-producing communities. Preclinical and early human studies demonstrate that phages can recalibrate microbial ecosystems, disrupt antibiotic-tolerant biofilms, and enrich metabolites such as butyrate that support mucosal integrity and immune balance. Mechanistically, phage DNA enriched with CpG motifs engages Toll-like receptor 9, activating dendritic cells and enhancing cytotoxic T lymphocyte responses, suggesting dual benefits in infection control and anti-tumor immunity. Emerging applications extend further, with engineered phages serving as vectors for CRISPR-Cas gene editing, targeted cytokine delivery, and nanocarrier platforms for leukemia therapy. Despite translational promise, major hurdles persist, including immunogenicity, horizontal gene transfer, resistance evolution, and regulatory uncertainty. Addressing these challenges through GMP-compliant manufacturing, metagenomics-guided personalization, and AI-optimized cocktail design could establish phage therapy as a microbiome-informed adjunct to overcome drug resistance in blood cancers. However, direct clinical evidence of phage therapy efficacy in hematologic malignancies remains limited, and current data are largely derived from preclinical and compassionate-use contexts.
{"title":"Phage therapy and the microbiome in hematologic malignancies: opportunities, mechanisms, and early evidence.","authors":"Juanwen Zhang, Jin Liu, Alireza Bayani","doi":"10.1007/s00432-025-06393-6","DOIUrl":"https://doi.org/10.1007/s00432-025-06393-6","url":null,"abstract":"<p><p>Hematologic malignancies remain among the most difficult cancers to treat, challenged by profound heterogeneity, treatment-induced immune dysfunction, and the frequent emergence of drug resistance. Beyond tumor-intrinsic mechanisms, dysbiosis of the gut microbiome is increasingly recognized as a critical determinant of therapeutic outcomes, shaping hematopoiesis, immune responses, and drug metabolism. Bacteriophage (phage) therapy has re-emerged as a precision tool capable of selectively eradicating pathogenic taxa while preserving commensal short-chain fatty acid-producing communities. Preclinical and early human studies demonstrate that phages can recalibrate microbial ecosystems, disrupt antibiotic-tolerant biofilms, and enrich metabolites such as butyrate that support mucosal integrity and immune balance. Mechanistically, phage DNA enriched with CpG motifs engages Toll-like receptor 9, activating dendritic cells and enhancing cytotoxic T lymphocyte responses, suggesting dual benefits in infection control and anti-tumor immunity. Emerging applications extend further, with engineered phages serving as vectors for CRISPR-Cas gene editing, targeted cytokine delivery, and nanocarrier platforms for leukemia therapy. Despite translational promise, major hurdles persist, including immunogenicity, horizontal gene transfer, resistance evolution, and regulatory uncertainty. Addressing these challenges through GMP-compliant manufacturing, metagenomics-guided personalization, and AI-optimized cocktail design could establish phage therapy as a microbiome-informed adjunct to overcome drug resistance in blood cancers. However, direct clinical evidence of phage therapy efficacy in hematologic malignancies remains limited, and current data are largely derived from preclinical and compassionate-use contexts.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"8"},"PeriodicalIF":2.8,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742699","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-08DOI: 10.1007/s00432-025-06378-5
Valerie Catherine Linz, Emma Liebau, Markus Schepers, Katharina Gillen, Marco Johannes Battista, Michael Mohr, Mona Wanda Schmidt, Marcus Schmidt, Annette Hasenburg
{"title":"Impact of preoperative geriatric screening and comorbidity assessment in patients with vulvar and vaginal cancer.","authors":"Valerie Catherine Linz, Emma Liebau, Markus Schepers, Katharina Gillen, Marco Johannes Battista, Michael Mohr, Mona Wanda Schmidt, Marcus Schmidt, Annette Hasenburg","doi":"10.1007/s00432-025-06378-5","DOIUrl":"10.1007/s00432-025-06378-5","url":null,"abstract":"","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"7"},"PeriodicalIF":2.8,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12686312/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145707691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-04DOI: 10.1007/s00432-025-06384-7
Anika Kästner, Christopher Hampf, Pia Naumann, Lizon Fiedler-Lacombe, Anna Kron, Anna Spier, Dusan Simic, Leonie Eilers, Aleksandra Graw, Sebastian Bartholomäus, Andreas Stang, Daniela Reil, Renate Kirschner-Schwabe, Jessica Isabel Selig, Jörg Wulff, Patrik Dröge, Thomas Ruhnke, Christian Günster, Uwe Nußbaum, Ursula Marschall, Juliane Mohnke, Anja Hebbelmann, Uwe Lührig, Anna Rasokat, Vanessa Mildenberger, Stephanie Stock, Florian Kron, Jürgen Wolf, Martin Bialke, Dana Stahl, Neeltje van den Berg, Wolfgang Hoffmann
Objective: While cancer registry and health insurance data are valuable resources for oncological health services research, these are rarely linked at the individual level due to data protection concerns and technical limitations. The prospective, controlled cohort study DigiNet aims to optimize personalized care for patients with stage IV non-small cell lung cancer (NSCLC) in the German study regions Berlin, Saxony and North Rhine-Westphalia. The population-based control group (pCG) was identified through cohort matching within the participating cancer registries. For health economic analyses, case-specific linkage of cancer registry data with claims data without informed consent was required.
Methods: A privacy-preserving record linkage (PPRL) concept was developed, ensuring that no conclusions about individual identities can be drawn. The approach relied on irreversible encryption of the statutory health insurance number (KVNR) within the data-holding institutions, using a study-specific configuration of a publicly available software.
Results: Following cohort matching in the cancer registries, N = 9,597 pCG cases with stage IV NSCLC diagnosis between June 2022 and March 2024 were identified. Of these, n = 1,437 (15.0%) had insurance coverage with one of three participating statutory health insurance funds and were eligible for PPRL. Among those, 94.2% (N = 1,354) were successfully linked with claims data. A trusted third party performed the linkage based on encrypted identifiers, removed the linkage keys, and provided the data to the evaluating parties.
Conclusions: This study demonstrates the feasibility of PPRL of cancer registry and claims data in a real-world oncological research setting. The concept is transferable to other research contexts requiring secure, identifier-based linkage without disclosure of personal identifiers.
{"title":"Concept and feasibility of privacy-preserving record linkage of cancer registry data and claims data in Germany: results from the DigiNet study on stage IV non-small cell lung cancer.","authors":"Anika Kästner, Christopher Hampf, Pia Naumann, Lizon Fiedler-Lacombe, Anna Kron, Anna Spier, Dusan Simic, Leonie Eilers, Aleksandra Graw, Sebastian Bartholomäus, Andreas Stang, Daniela Reil, Renate Kirschner-Schwabe, Jessica Isabel Selig, Jörg Wulff, Patrik Dröge, Thomas Ruhnke, Christian Günster, Uwe Nußbaum, Ursula Marschall, Juliane Mohnke, Anja Hebbelmann, Uwe Lührig, Anna Rasokat, Vanessa Mildenberger, Stephanie Stock, Florian Kron, Jürgen Wolf, Martin Bialke, Dana Stahl, Neeltje van den Berg, Wolfgang Hoffmann","doi":"10.1007/s00432-025-06384-7","DOIUrl":"10.1007/s00432-025-06384-7","url":null,"abstract":"<p><strong>Objective: </strong>While cancer registry and health insurance data are valuable resources for oncological health services research, these are rarely linked at the individual level due to data protection concerns and technical limitations. The prospective, controlled cohort study DigiNet aims to optimize personalized care for patients with stage IV non-small cell lung cancer (NSCLC) in the German study regions Berlin, Saxony and North Rhine-Westphalia. The population-based control group (pCG) was identified through cohort matching within the participating cancer registries. For health economic analyses, case-specific linkage of cancer registry data with claims data without informed consent was required.</p><p><strong>Methods: </strong>A privacy-preserving record linkage (PPRL) concept was developed, ensuring that no conclusions about individual identities can be drawn. The approach relied on irreversible encryption of the statutory health insurance number (KVNR) within the data-holding institutions, using a study-specific configuration of a publicly available software.</p><p><strong>Results: </strong>Following cohort matching in the cancer registries, N = 9,597 pCG cases with stage IV NSCLC diagnosis between June 2022 and March 2024 were identified. Of these, n = 1,437 (15.0%) had insurance coverage with one of three participating statutory health insurance funds and were eligible for PPRL. Among those, 94.2% (N = 1,354) were successfully linked with claims data. A trusted third party performed the linkage based on encrypted identifiers, removed the linkage keys, and provided the data to the evaluating parties.</p><p><strong>Conclusions: </strong>This study demonstrates the feasibility of PPRL of cancer registry and claims data in a real-world oncological research setting. The concept is transferable to other research contexts requiring secure, identifier-based linkage without disclosure of personal identifiers.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"6"},"PeriodicalIF":2.8,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678689/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668224","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-04DOI: 10.1007/s00432-025-06368-7
Liang Xie, Cuiping Song, Jialin Qin, Qianqian Xu, Jianchun Yin, Yuanyuan Ma, Hong Chen, Chao Li, Bing Hong, Ni Chen, Xiaoxi Pang
Gadolinium-neutron capture therapy (Gd-NCT) employs Gadolinium (Gd) isotopes and thermal neutrons to specifically target and kill cancer at cells level. This study investigates the targeting efficacy of 157Gd-DOTA-HK (DHK), a novel agent designed for Gd-NCT and MRI. We synthesized 157Gd-DHK, which combines a Gd-DOTA complex as a neutron capturer and MRI probe with αvβ6 binding peptide (HK) for targeted Pancreas adenocarcinoma (PDAC) therapy. 157Gd-DHK demonstrated a significantly high binding affinity for BxPC-3 cells. scintigraphy revealed that the optimal time window for Gd-NCT was 26 h after injection. The conjugate's imaging capabilities and its potential as an MRI contrast agent were validated. The conjugate effectively triggered nuclear reactions via Gd-NCT, leading to efficient tumor cell destruction. Photon sensitization studies showed that 157Gd-DHK induced photon-mediated phototoxicity in cancer cells while exhibiting minimal toxicity. 157Gd-DHK shows great potential as a theranostic agent for targeted imaging of PDAC and Gd-NCT applications. It presents a novel strategy to enhance the specificity and efficacy of Gd-NCT in cancer treatment.
{"title":"<sup>157</sup>Gd-DHK: enhancing targeted gadolinium neutron capture for pancreatic adenocarcinoma.","authors":"Liang Xie, Cuiping Song, Jialin Qin, Qianqian Xu, Jianchun Yin, Yuanyuan Ma, Hong Chen, Chao Li, Bing Hong, Ni Chen, Xiaoxi Pang","doi":"10.1007/s00432-025-06368-7","DOIUrl":"10.1007/s00432-025-06368-7","url":null,"abstract":"<p><p>Gadolinium-neutron capture therapy (Gd-NCT) employs Gadolinium (Gd) isotopes and thermal neutrons to specifically target and kill cancer at cells level. This study investigates the targeting efficacy of <sup>157</sup>Gd-DOTA-HK (DHK), a novel agent designed for Gd-NCT and MRI. We synthesized <sup>157</sup>Gd-DHK, which combines a Gd-DOTA complex as a neutron capturer and MRI probe with αvβ6 binding peptide (HK) for targeted Pancreas adenocarcinoma (PDAC) therapy. <sup>157</sup>Gd-DHK demonstrated a significantly high binding affinity for BxPC-3 cells. scintigraphy revealed that the optimal time window for Gd-NCT was 26 h after injection. The conjugate's imaging capabilities and its potential as an MRI contrast agent were validated. The conjugate effectively triggered nuclear reactions via Gd-NCT, leading to efficient tumor cell destruction. Photon sensitization studies showed that <sup>157</sup>Gd-DHK induced photon-mediated phototoxicity in cancer cells while exhibiting minimal toxicity. <sup>157</sup>Gd-DHK shows great potential as a theranostic agent for targeted imaging of PDAC and Gd-NCT applications. It presents a novel strategy to enhance the specificity and efficacy of Gd-NCT in cancer treatment.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"5"},"PeriodicalIF":2.8,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678673/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668282","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.1007/s00432-025-06381-w
Po-Ting Lin, Wei Teng, Wei-Ting Chen, Chung-Wei Su, Yi-Chung Hsieh, Chen-Chun Lin, Chun-Yen Lin, Shi-Ming Lin
Background: Combination regimens and monotherapy are both employed in the treatment of hepatocellular carcinoma (HCC). However, the optimal combination regimen for specific scenarios remains unclear. Therefore, our aim is to identify predictors of survival and determine whether combination therapy or monotherapy provides better outcomes for HCC patients.
Methods: From August 2015 to July 2020, a total of 129 unresectable HCC patients receiving immune checkpoint inhibitors (ICI) were recruited. Patients were divided into high tumor burden and low tumor burden groups according to the up-to-7 criteria which is defined as the sum of the size of the largest tumor and the total number of tumors. The combination use of tyrosine kinase inhibitors (TKI) was documented and overall survival was analyzed.
Results: Among the 129 patients receiving ICI, the median age was 63.2 years old, and 76.6% were male. Eighty-five patients (65.9%) were beyond up-to-7 criteria. Patients receiving ICI had median overall survival time of 15 months and had progression-free survival time of 6.2 months. In multivariate Cox regression analysis, the Child-Turcotte-Pugh (CTP) score B (adjusted HR: 3.103 (CI: 1.629-5.912), P = 0.0006), AFP decreased more than 10% from the baseline (adjusted HR: 0.463 (CI: 0.270-0.794), P = 0.0052), and out of up-to-7 criteria (adjusted HR:1.808 (CI:1.007-3.245), P = 0.0472) were the independent predictive factors for mortality. Among patients beyond up-to-7 criteria, 23.5% (n = 20) received combination treatment with TKI and ICI. Moreover, patients using combination treatment showed significantly better survival than those without combination treatment (1st year cumulative survival rate 61% vs 30%, log-rank P = 0.018).
Conclusion: Baseline CTP score, tumor burden, as well as AFP response during ICI treatment were the independent predictive factors for survival. Furthermore, among HCC patients beyond up-to-7 criteria, combination treatment was associated with improved survival than those without combination treatment.
背景:肝细胞癌(HCC)的治疗均采用联合治疗方案和单一治疗方案。然而,针对具体情况的最佳联合方案仍不清楚。因此,我们的目的是确定生存的预测因素,并确定联合治疗或单一治疗是否为HCC患者提供更好的结果。方法:2015年8月至2020年7月,共招募129例接受免疫检查点抑制剂(ICI)治疗的不可切除HCC患者。按照最大肿瘤大小与肿瘤总数之和为7级的标准将患者分为高肿瘤负担组和低肿瘤负担组。记录联合使用酪氨酸激酶抑制剂(TKI)并分析总生存率。结果:129例患者中位年龄为63.2岁,男性占76.6%。85例(65.9%)患者超过7级标准。接受ICI的患者中位总生存期为15个月,无进展生存期为6.2个月。在多因素Cox回归分析中,child - turcote - pugh (CTP)评分B(校正HR: 3.103 (CI: 1.629-5.912), P = 0.0006)、AFP较基线下降10%以上(校正HR: 0.463 (CI: 0.270-0.794), P = 0.0052)和≥7的标准(校正HR:1.808 (CI:1.007-3.245), P = 0.0472)是死亡率的独立预测因素。在超过7级标准的患者中,23.5% (n = 20)接受TKI和ICI联合治疗。此外,联合治疗的患者生存率明显优于未联合治疗的患者(1年累计生存率61% vs 30%, log-rank P = 0.018)。结论:基线CTP评分、肿瘤负荷以及ICI治疗期间AFP反应是生存的独立预测因素。此外,在超过7级标准的HCC患者中,联合治疗比未联合治疗的患者生存率更高。
{"title":"TKI plus ICI versus ICI alone in high tumor burden hepatocellular carcinoma: a retrospective cohort study.","authors":"Po-Ting Lin, Wei Teng, Wei-Ting Chen, Chung-Wei Su, Yi-Chung Hsieh, Chen-Chun Lin, Chun-Yen Lin, Shi-Ming Lin","doi":"10.1007/s00432-025-06381-w","DOIUrl":"10.1007/s00432-025-06381-w","url":null,"abstract":"<p><strong>Background: </strong>Combination regimens and monotherapy are both employed in the treatment of hepatocellular carcinoma (HCC). However, the optimal combination regimen for specific scenarios remains unclear. Therefore, our aim is to identify predictors of survival and determine whether combination therapy or monotherapy provides better outcomes for HCC patients.</p><p><strong>Methods: </strong>From August 2015 to July 2020, a total of 129 unresectable HCC patients receiving immune checkpoint inhibitors (ICI) were recruited. Patients were divided into high tumor burden and low tumor burden groups according to the up-to-7 criteria which is defined as the sum of the size of the largest tumor and the total number of tumors. The combination use of tyrosine kinase inhibitors (TKI) was documented and overall survival was analyzed.</p><p><strong>Results: </strong>Among the 129 patients receiving ICI, the median age was 63.2 years old, and 76.6% were male. Eighty-five patients (65.9%) were beyond up-to-7 criteria. Patients receiving ICI had median overall survival time of 15 months and had progression-free survival time of 6.2 months. In multivariate Cox regression analysis, the Child-Turcotte-Pugh (CTP) score B (adjusted HR: 3.103 (CI: 1.629-5.912), P = 0.0006), AFP decreased more than 10% from the baseline (adjusted HR: 0.463 (CI: 0.270-0.794), P = 0.0052), and out of up-to-7 criteria (adjusted HR:1.808 (CI:1.007-3.245), P = 0.0472) were the independent predictive factors for mortality. Among patients beyond up-to-7 criteria, 23.5% (n = 20) received combination treatment with TKI and ICI. Moreover, patients using combination treatment showed significantly better survival than those without combination treatment (1st year cumulative survival rate 61% vs 30%, log-rank P = 0.018).</p><p><strong>Conclusion: </strong>Baseline CTP score, tumor burden, as well as AFP response during ICI treatment were the independent predictive factors for survival. Furthermore, among HCC patients beyond up-to-7 criteria, combination treatment was associated with improved survival than those without combination treatment.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"4"},"PeriodicalIF":2.8,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12675898/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-02DOI: 10.1007/s00432-025-06357-w
Anne Marie Jelsig, Maria Bejerholm Boelman, Ulf Birkedal, Jane Hübertz, Lana Al-Zehawi, Charlotte Lautrup, John Gásdal Karstensen, Thomas van Overeem Hansen
{"title":"The c.1744G > C, p.(Glu582Gln) missense variant in coding exon 14 of APC increases skipping of a natural occurring isoform and causes Familial Adenomatous Polyposis.","authors":"Anne Marie Jelsig, Maria Bejerholm Boelman, Ulf Birkedal, Jane Hübertz, Lana Al-Zehawi, Charlotte Lautrup, John Gásdal Karstensen, Thomas van Overeem Hansen","doi":"10.1007/s00432-025-06357-w","DOIUrl":"10.1007/s00432-025-06357-w","url":null,"abstract":"","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"3"},"PeriodicalIF":2.8,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12672996/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653443","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-29DOI: 10.1007/s00432-025-06382-9
Peyman Tabnak, Mohammad Ebrahimnezhad
Gastric cancer remains a significant global health challenge due to its aggressive behavior, high mortality rates, and limited treatment success. Growing research underscores the critical role of SUMOylation, a reversible post-translational modification, in regulating key cellular processes such as DNA repair, gene expression, genomic stability, and signaling pathways. Aberrant SUMOylation is closely linked to gastric cancer development, metastasis, and resistance to therapies, positioning it as a potential therapeutic target. This review consolidates current knowledge of the SUMOylation system, which involves activating (E1), conjugating (E2), and ligating (E3) enzymes, alongside SUMO-specific proteases (SENPs) that reverse the modification. We explore how SUMOylation influences cancer-promoting and tumor-suppressing pathways by stabilizing or degrading critical proteins, modulating immune responses, driving epithelial-mesenchymal transition, and contributing to chemoresistance. Furthermore, we discuss promising therapeutic approaches targeting SUMOylation, including small-molecule inhibitors, natural bioactive compounds, and synthetic lethal strategies that exploit SUMO pathway weaknesses. Preclinical studies suggest these approaches could enhance chemotherapy and immunotherapy effectiveness, offering new precision treatment options. Deepening our understanding of SUMOylation in gastric cancer is vital for developing innovative, mechanism-driven therapies to improve patient outcomes.
{"title":"Insights into SUMOylation in gastric cancer: molecular mechanisms and emerging therapeutic opportunities.","authors":"Peyman Tabnak, Mohammad Ebrahimnezhad","doi":"10.1007/s00432-025-06382-9","DOIUrl":"10.1007/s00432-025-06382-9","url":null,"abstract":"<p><p>Gastric cancer remains a significant global health challenge due to its aggressive behavior, high mortality rates, and limited treatment success. Growing research underscores the critical role of SUMOylation, a reversible post-translational modification, in regulating key cellular processes such as DNA repair, gene expression, genomic stability, and signaling pathways. Aberrant SUMOylation is closely linked to gastric cancer development, metastasis, and resistance to therapies, positioning it as a potential therapeutic target. This review consolidates current knowledge of the SUMOylation system, which involves activating (E1), conjugating (E2), and ligating (E3) enzymes, alongside SUMO-specific proteases (SENPs) that reverse the modification. We explore how SUMOylation influences cancer-promoting and tumor-suppressing pathways by stabilizing or degrading critical proteins, modulating immune responses, driving epithelial-mesenchymal transition, and contributing to chemoresistance. Furthermore, we discuss promising therapeutic approaches targeting SUMOylation, including small-molecule inhibitors, natural bioactive compounds, and synthetic lethal strategies that exploit SUMO pathway weaknesses. Preclinical studies suggest these approaches could enhance chemotherapy and immunotherapy effectiveness, offering new precision treatment options. Deepening our understanding of SUMOylation in gastric cancer is vital for developing innovative, mechanism-driven therapies to improve patient outcomes.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"2"},"PeriodicalIF":2.8,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12664871/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145633963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1007/s00432-025-06372-x
Yuexin Liu, Lizhou Jia, Liu Yang, Zhang Ning, Yanmei Li
Aim: The clinical management of gastric cancer (GC) is frequently challenged by the development of drug resistance, leading to poor patient outcomes. This review aims to explore the role of ferroptosis, an iron-dependent form of programmed cell death driven by lipid peroxidation, in overcoming this therapeutic hurdle. Our objective is to provide a theoretical foundation for developing novel strategies to reverse drug resistance in GC by targeting the ferroptosis pathway.
Methods: This review systematically elucidates the core regulatory mechanisms of ferroptosis and analyzes its key role in mediating drug resistance in GC. We synthesize current literature to explore potential therapeutic strategies that leverage ferroptosis induction to sensitize cancer cells. Furthermore, we critically examine the complex interplay between ferroptosis and the tumor microenvironment (TME) and discuss the challenges associated with translating these findings into personalized treatment approaches, integrating insights from emerging technologies.
Results: Our analysis confirms that ferroptosis is governed by a precise regulatory network involving glutathione metabolism, lipid peroxidation, and iron homeostasis, which is frequently dysregulated in GC. We identify that key mechanisms of conventional drug resistance are linked to the evasion of ferroptosis. Consequently, several potential therapeutic strategies, including the use of ferroptosis inducers (FINs) and combination therapies, show promise in resensitizing resistant GC cells. The review also highlights the dual role of the TME, which can either suppress or promote ferroptosis, adding a layer of complexity. Finally, we identify significant challenges in patient stratification and the need for reliable biomarkers to achieve personalized ferroptosis-based therapies.
Conclusion: Targeting ferroptosis presents a promising and innovative research avenue for reversing drug resistance in gastric cancer. Strategies designed to induce ferroptosis effectively overcome common resistance mechanisms and hold significant therapeutic potential. Future research must focus on integrating multi-omics technologies and advanced drug delivery systems to decipher the complex regulatory networks of ferroptosis within the TME and to develop biomarkers for personalized treatment, thereby paving the way for improved clinical outcomes.
{"title":"Research progress on ferroptosis in drug resistance and therapy of gastric cancer.","authors":"Yuexin Liu, Lizhou Jia, Liu Yang, Zhang Ning, Yanmei Li","doi":"10.1007/s00432-025-06372-x","DOIUrl":"10.1007/s00432-025-06372-x","url":null,"abstract":"<p><strong>Aim: </strong>The clinical management of gastric cancer (GC) is frequently challenged by the development of drug resistance, leading to poor patient outcomes. This review aims to explore the role of ferroptosis, an iron-dependent form of programmed cell death driven by lipid peroxidation, in overcoming this therapeutic hurdle. Our objective is to provide a theoretical foundation for developing novel strategies to reverse drug resistance in GC by targeting the ferroptosis pathway.</p><p><strong>Methods: </strong>This review systematically elucidates the core regulatory mechanisms of ferroptosis and analyzes its key role in mediating drug resistance in GC. We synthesize current literature to explore potential therapeutic strategies that leverage ferroptosis induction to sensitize cancer cells. Furthermore, we critically examine the complex interplay between ferroptosis and the tumor microenvironment (TME) and discuss the challenges associated with translating these findings into personalized treatment approaches, integrating insights from emerging technologies.</p><p><strong>Results: </strong>Our analysis confirms that ferroptosis is governed by a precise regulatory network involving glutathione metabolism, lipid peroxidation, and iron homeostasis, which is frequently dysregulated in GC. We identify that key mechanisms of conventional drug resistance are linked to the evasion of ferroptosis. Consequently, several potential therapeutic strategies, including the use of ferroptosis inducers (FINs) and combination therapies, show promise in resensitizing resistant GC cells. The review also highlights the dual role of the TME, which can either suppress or promote ferroptosis, adding a layer of complexity. Finally, we identify significant challenges in patient stratification and the need for reliable biomarkers to achieve personalized ferroptosis-based therapies.</p><p><strong>Conclusion: </strong>Targeting ferroptosis presents a promising and innovative research avenue for reversing drug resistance in gastric cancer. Strategies designed to induce ferroptosis effectively overcome common resistance mechanisms and hold significant therapeutic potential. Future research must focus on integrating multi-omics technologies and advanced drug delivery systems to decipher the complex regulatory networks of ferroptosis within the TME and to develop biomarkers for personalized treatment, thereby paving the way for improved clinical outcomes.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"152 1","pages":"1"},"PeriodicalIF":2.8,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12662952/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145633914","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-25DOI: 10.1007/s00432-025-06380-x
Katharina Kämpfner, Susan Wittig, Bernd Gruhn
Purpose: Complications such as graft-versus-host disease (GVHD), hepatic sinusoidal obstruction syndrome, and infections compromise the success of allogeneic hematopoietic stem cell transplantation (HSCT) as a treatment modality for malignant or genetic diseases. Identification of beneficial non-human leukocyte antigens (HLA), such as cytokines, is one approach to reduce the rate of unintended events. This study investigated the association between single-nucleotide polymorphisms (SNPs) of the gene of the proinflammatory cytokine interleukin-1 (IL-1) and treatment outcomes after allogeneic HSCT in a pediatric population.
Methods: In our single-center study, we retrospectively analyzed a cohort of 270 children and their respective donors. They underwent allogeneic HSCT for the first time, and their conditioning regimen was myeloablative in all cases. We used polymerase chain reaction to genotype the SNPs of IL-1α rs1800587 (C > T), IL-1β rs16944 (C > T), and IL-1β rs1143627 (C > T). The outcome measures included overall survival (OS), event-free survival (EFS), relapse rate (RR), transplant-related mortality (TRM), and the occurrence of acute or chronic GVHD.
Results: The distribution of IL-1α rs1800587 genotype was as follows: we observed the CC genotype in 124 of 256 recipients (48.4%) and 132 of 270 donors (48.9%). We detected the CT genotype in 115 patients (44.9%) and 114 donors (42.2%) and found the homozygous TT genotype in 17 children (6.6%) and 24 of their donors (8.9%). The distribution of the SNP IL-1α rs1800587 is in Hardy-Weinberg equilibrium. The incidence of moderate or severe acute GVHD was significantly decreased in recipients receiving a donor transplant with the TT genotype (4% (TT) versus 25% (CC/CT); p = 0.028). We found no significant SNP IL-1α rs1800587 (C > T) associations for chronic GVHD, RR, TRM, EFS, and OS. For the other genotypes analyzed, IL-1β rs11644 (C > T) and IL-1β rs1143627 (C > T), we also found no significant associations for acute and chronic GVHD, RR, TRM, EFS, and OS, neither in donors nor in recipients. The results of the multivariate analysis revealed a hazard ratio of 0.17 (confidence interval 0.0229-1.27), and a trend that IL-1α rs1800587 could be an independent risk factor for acute GVHD (p = 0.084).
Conclusion: Our study identified the donor IL-1α rs1800587 CC/CT genotype as a possible genetic risk factor for developing moderate to severe acute GVHD (grade II - IV) in pediatric patients who underwent allogeneic HSCT. Once confirmed in further studies, these results may influence the donor selection and prophylaxis to decrease the risk of acute GVHD in children.
{"title":"Association of donor and recipient single-nucleotide polymorphisms of interleukin-1 gene with outcomes after allogeneic hematopoietic stem cell transplantation in childhood.","authors":"Katharina Kämpfner, Susan Wittig, Bernd Gruhn","doi":"10.1007/s00432-025-06380-x","DOIUrl":"10.1007/s00432-025-06380-x","url":null,"abstract":"<p><strong>Purpose: </strong>Complications such as graft-versus-host disease (GVHD), hepatic sinusoidal obstruction syndrome, and infections compromise the success of allogeneic hematopoietic stem cell transplantation (HSCT) as a treatment modality for malignant or genetic diseases. Identification of beneficial non-human leukocyte antigens (HLA), such as cytokines, is one approach to reduce the rate of unintended events. This study investigated the association between single-nucleotide polymorphisms (SNPs) of the gene of the proinflammatory cytokine interleukin-1 (IL-1) and treatment outcomes after allogeneic HSCT in a pediatric population.</p><p><strong>Methods: </strong>In our single-center study, we retrospectively analyzed a cohort of 270 children and their respective donors. They underwent allogeneic HSCT for the first time, and their conditioning regimen was myeloablative in all cases. We used polymerase chain reaction to genotype the SNPs of IL-1α rs1800587 (C > T), IL-1β rs16944 (C > T), and IL-1β rs1143627 (C > T). The outcome measures included overall survival (OS), event-free survival (EFS), relapse rate (RR), transplant-related mortality (TRM), and the occurrence of acute or chronic GVHD.</p><p><strong>Results: </strong>The distribution of IL-1α rs1800587 genotype was as follows: we observed the CC genotype in 124 of 256 recipients (48.4%) and 132 of 270 donors (48.9%). We detected the CT genotype in 115 patients (44.9%) and 114 donors (42.2%) and found the homozygous TT genotype in 17 children (6.6%) and 24 of their donors (8.9%). The distribution of the SNP IL-1α rs1800587 is in Hardy-Weinberg equilibrium. The incidence of moderate or severe acute GVHD was significantly decreased in recipients receiving a donor transplant with the TT genotype (4% (TT) versus 25% (CC/CT); p = 0.028). We found no significant SNP IL-1α rs1800587 (C > T) associations for chronic GVHD, RR, TRM, EFS, and OS. For the other genotypes analyzed, IL-1β rs11644 (C > T) and IL-1β rs1143627 (C > T), we also found no significant associations for acute and chronic GVHD, RR, TRM, EFS, and OS, neither in donors nor in recipients. The results of the multivariate analysis revealed a hazard ratio of 0.17 (confidence interval 0.0229-1.27), and a trend that IL-1α rs1800587 could be an independent risk factor for acute GVHD (p = 0.084).</p><p><strong>Conclusion: </strong>Our study identified the donor IL-1α rs1800587 CC/CT genotype as a possible genetic risk factor for developing moderate to severe acute GVHD (grade II - IV) in pediatric patients who underwent allogeneic HSCT. Once confirmed in further studies, these results may influence the donor selection and prophylaxis to decrease the risk of acute GVHD in children.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"151 12","pages":"331"},"PeriodicalIF":2.8,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12647457/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145604142","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-25DOI: 10.1007/s00432-025-06376-7
Zhen Yu, Xingguo Yang, Jun Wu, Xintao Yu, Jian Cui, Xiang Gao, Bobo Ma, Ji Ke, Baoxun Zhang, Xiaohong Chen, Lei Yu
Objective: The management of pulmonary metastases (PM) from head and neck adenoid cystic carcinoma (ACC) remains controversial, with limited evidence guiding the choice between surgical and non-surgical strategies. This study aimed to compare long-term survival outcomes between patients undergoing surgical resection and those receiving non-surgical management for ACC-derived PM.
Methods: We conducted a retrospective analysis of 204 patients with pulmonary metastases from head and neck ACC treated at our institution between January 2010 and December 2024. Patients were categorized into Surgery (n = 108) and Non-Surgery (n = 96) groups. To address selection bias and improve causal inference in this observational study, propensity score matching (PSM) was performed based on key clinical variables, generating 68 well-matched pairs. Overall survival (OS) and progression-free survival (PFS) were compared using Kaplan-Meier analysis and Cox proportional hazards models.
Results: Before matching, the Surgery group demonstrated significantly higher 5-year OS (97.73% vs. 84.2%, P = 0.0004) and PFS (89.39% vs. 74.66%, P = 0.021). After PSM, the survival advantage persisted, with the Surgery group showing improved 5-year OS (95.75% vs. 82.91%, P = 0.0063) and PFS (86.93% vs. 73.86%, P = 0.0039). Multivariate analysis confirmed surgical resection as an independent prognostic factor for improved OS (HR = 0.08, 95% CI: 0.01-0.42, P = 0.0032), alongside tumor grade, presence of pleural effusion, and International Registry of Lung Metastases (IRLM) stage.
Conclusions: In this propensity-matched analysis, surgical metastasectomy was associated with significantly improved survival in selected patients with ACC lung metastases, supporting its consideration as a valuable therapeutic option for oligometastatic disease within a multidisciplinary framework.
目的:头颈部腺样囊性癌(ACC)肺转移瘤(PM)的治疗仍然存在争议,指导手术和非手术策略选择的证据有限。本研究旨在比较接受手术切除和接受非手术治疗的acc源性PM患者的长期生存结果。方法:我们对2010年1月至2024年12月在我院治疗的204例头颈部ACC肺转移患者进行了回顾性分析。患者分为手术组(n = 108)和非手术组(n = 96)。为了解决本观察性研究中的选择偏倚和改进因果推理,根据关键临床变量进行倾向评分匹配(PSM),产生68对良好匹配的配对。采用Kaplan-Meier分析和Cox比例风险模型比较总生存期(OS)和无进展生存期(PFS)。结果:配对前,手术组5年OS (97.73% vs. 84.2%, P = 0.0004)和PFS (89.39% vs. 74.66%, P = 0.021)明显高于对照组。PSM后,生存优势持续存在,手术组的5年OS (95.75% vs. 82.91%, P = 0.0063)和PFS (86.93% vs. 73.86%, P = 0.0039)均有改善。多因素分析证实手术切除是改善OS的独立预后因素(HR = 0.08, 95% CI: 0.01-0.42, P = 0.0032),以及肿瘤分级、胸腔积液的存在和国际肺转移登记(IRLM)分期。结论:在这个倾向匹配的分析中,手术转移切除术与选定的ACC肺转移患者的生存率显著提高相关,支持其在多学科框架内作为低转移性疾病有价值的治疗选择的考虑。
{"title":"Surgical resection for pulmonary metastases from head and neck adenoid cystic carcinoma: a propensity-matched survival analysis.","authors":"Zhen Yu, Xingguo Yang, Jun Wu, Xintao Yu, Jian Cui, Xiang Gao, Bobo Ma, Ji Ke, Baoxun Zhang, Xiaohong Chen, Lei Yu","doi":"10.1007/s00432-025-06376-7","DOIUrl":"10.1007/s00432-025-06376-7","url":null,"abstract":"<p><strong>Objective: </strong>The management of pulmonary metastases (PM) from head and neck adenoid cystic carcinoma (ACC) remains controversial, with limited evidence guiding the choice between surgical and non-surgical strategies. This study aimed to compare long-term survival outcomes between patients undergoing surgical resection and those receiving non-surgical management for ACC-derived PM.</p><p><strong>Methods: </strong>We conducted a retrospective analysis of 204 patients with pulmonary metastases from head and neck ACC treated at our institution between January 2010 and December 2024. Patients were categorized into Surgery (n = 108) and Non-Surgery (n = 96) groups. To address selection bias and improve causal inference in this observational study, propensity score matching (PSM) was performed based on key clinical variables, generating 68 well-matched pairs. Overall survival (OS) and progression-free survival (PFS) were compared using Kaplan-Meier analysis and Cox proportional hazards models.</p><p><strong>Results: </strong>Before matching, the Surgery group demonstrated significantly higher 5-year OS (97.73% vs. 84.2%, P = 0.0004) and PFS (89.39% vs. 74.66%, P = 0.021). After PSM, the survival advantage persisted, with the Surgery group showing improved 5-year OS (95.75% vs. 82.91%, P = 0.0063) and PFS (86.93% vs. 73.86%, P = 0.0039). Multivariate analysis confirmed surgical resection as an independent prognostic factor for improved OS (HR = 0.08, 95% CI: 0.01-0.42, P = 0.0032), alongside tumor grade, presence of pleural effusion, and International Registry of Lung Metastases (IRLM) stage.</p><p><strong>Conclusions: </strong>In this propensity-matched analysis, surgical metastasectomy was associated with significantly improved survival in selected patients with ACC lung metastases, supporting its consideration as a valuable therapeutic option for oligometastatic disease within a multidisciplinary framework.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"151 12","pages":"330"},"PeriodicalIF":2.8,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12647468/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145604120","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}