Pub Date : 2026-01-01Epub Date: 2026-01-02DOI: 10.1177/14230380251410478
Kateryna Maslova, Magdalena Chechlinska, Irmina Maria Michalek, Lukasz Taraszkiewicz, Paulina Kober, Inga Trulson, Karolina Worf, Sophie Gabriel, Laura Knoblauch, Fiona Campbell, Ian A Cree, Stefan Holdenrieder, Magdalena Kowalewska
BackgroundTumour biomarkers have become increasingly important in oncology, shaping cancer diagnostics, classification, and patient management. Despite their potential, the use of cancer biomarkers in clinical settings remains limited.ObjectiveThis paper aims to outline biomarker development, from classical, serum protein markers to emerging tumour biomarkers, including meta-biomarkers, to show their diversity and point out the challenges in their development, reporting, and implementation in clinical practice as well as their relevance in evidence-based pathology and cancer classification.MethodsA literature-based analysis, incorporating insights from our ongoing research, is presented.ResultsAlthough numerous potential biomarkers, biomarker signatures, and meta-biomarkers, are being discovered, existing innovations are often not supported by sufficiently rigorous research methodologies and standardised reporting practices to enable their translation into clinical practice.ConclusionsTo ensure that biomarker discoveries are both scientifically sound and clinically useful, improved research and validation methods, along with adherence to established reporting standards, are essential. We propose the use of the Hierarchy of Evidence for Tumour Pathology as a framework to evaluate and map existing evidence and identify knowledge gaps and research priorities.
{"title":"Tumour markers and evidence-based pathology.","authors":"Kateryna Maslova, Magdalena Chechlinska, Irmina Maria Michalek, Lukasz Taraszkiewicz, Paulina Kober, Inga Trulson, Karolina Worf, Sophie Gabriel, Laura Knoblauch, Fiona Campbell, Ian A Cree, Stefan Holdenrieder, Magdalena Kowalewska","doi":"10.1177/14230380251410478","DOIUrl":"https://doi.org/10.1177/14230380251410478","url":null,"abstract":"<p><p>BackgroundTumour biomarkers have become increasingly important in oncology, shaping cancer diagnostics, classification, and patient management. Despite their potential, the use of cancer biomarkers in clinical settings remains limited.ObjectiveThis paper aims to outline biomarker development, from classical, serum protein markers to emerging tumour biomarkers, including meta-biomarkers, to show their diversity and point out the challenges in their development, reporting, and implementation in clinical practice as well as their relevance in evidence-based pathology and cancer classification.MethodsA literature-based analysis, incorporating insights from our ongoing research, is presented.ResultsAlthough numerous potential biomarkers, biomarker signatures, and meta-biomarkers, are being discovered, existing innovations are often not supported by sufficiently rigorous research methodologies and standardised reporting practices to enable their translation into clinical practice.ConclusionsTo ensure that biomarker discoveries are both scientifically sound and clinically useful, improved research and validation methods, along with adherence to established reporting standards, are essential. We propose the use of the Hierarchy of Evidence for Tumour Pathology as a framework to evaluate and map existing evidence and identify knowledge gaps and research priorities.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"48 ","pages":"14230380251410478"},"PeriodicalIF":0.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145893293","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-03-17DOI: 10.1177/14230380251316788
Milou M F Schuurbiers, Freek A van Delft, Hendrik Koffijberg, Maarten J IJzerman, Kim Monkhorst, Marjolijn J L Ligtenberg, Daan van den Broek, Huub H van Rossum, Michel M van den Heuvel
BackgroundImmune checkpoint inhibitors (ICIs) provide a significant survival benefit in non-small cell lung cancer (NSCLC) patients; however, accurately predicting which patients will benefit remains a challenge. As previously shown, the STOP model, a machine learning model based on serum tumor markers, is capable of identifying non-responders after 6 weeks of ICIs.ObjectiveThis study aims to externally validate this model and to assess the predictive value in combination with radiological response assessment using RECIST criteria.MethodsIn a cohort of 242 metastatic NSCLC patients, CYFRA, CEA, and NSE were measured before start and after 6 weeks of ICI treatment. The ability of the STOP model to predict no durable benefit (NDB; progressive disease, death within 6 months or disease control of less than 6 months) was assessed using specificity and positive predictive value (PPV). Moreover, a combination of the STOP model with RECIST after 6-8 weeks of ICIs was investigated.ResultsThe STOP model achieved a specificity of 96% (95% CI 95%-97%) and a PPV of predicting NDB of 88.1% (95% CI 85.9%-90.3%). Combining the STOP model with RECIST improved specificity and PPV to 100% and predicted NDB on average 11.6 weeks (IQR 1.8-18.0 weeks) prior to developing radiologically defined progression.ConclusionsAfter 6 weeks of ICIs, the blood-based STOP model was capable of accurately predicting NDB in metastatic NSCLC patients, earlier than conventional radiological assessment. The combined serological and radiological response assessment creates an early opportunity to safely stop ICI treatment in patients who will not benefit, although the clinical utility of the assay is limited since the high specificity comes at the cost of a lower sensitivity.
免疫检查点抑制剂(ICIs)为非小细胞肺癌(NSCLC)患者提供了显著的生存益处;然而,准确预测哪些患者将受益仍然是一个挑战。如前所述,STOP模型是一种基于血清肿瘤标志物的机器学习模型,能够在6周的ICIs后识别无反应。目的对该模型进行外部验证,并结合RECIST标准评估放射反应的预测价值。方法在242例转移性NSCLC患者中,在ICI治疗开始前和6周后测量CYFRA、CEA和NSE。STOP模型预测无持久效益(NDB;使用特异性和阳性预测值(PPV)评估进展性疾病、6个月内死亡或疾病控制少于6个月的患者。此外,在6-8周的ICIs后,研究了STOP模型与RECIST的结合。结果STOP模型的特异性为96% (95% CI 95%-97%),预测NDB的PPV为88.1% (95% CI 85.9%-90.3%)。将STOP模型与RECIST相结合可将特异性和PPV提高到100%,并在放射学定义的进展发生前平均11.6周(IQR 1.8-18.0周)预测NDB。结论经过6周的ICIs后,基于血液的STOP模型能够准确预测转移性NSCLC患者的NDB,比传统的放射评估更早。血清学和放射学反应联合评估为不能获益的患者安全停止ICI治疗创造了早期机会,尽管该检测的临床效用有限,因为高特异性是以较低敏感性为代价的。
{"title":"External validation of a serum tumor marker algorithm for early prediction of no durable benefit to immunotherapy in metastastic non-small cell lung carcinoma.","authors":"Milou M F Schuurbiers, Freek A van Delft, Hendrik Koffijberg, Maarten J IJzerman, Kim Monkhorst, Marjolijn J L Ligtenberg, Daan van den Broek, Huub H van Rossum, Michel M van den Heuvel","doi":"10.1177/14230380251316788","DOIUrl":"10.1177/14230380251316788","url":null,"abstract":"<p><p>BackgroundImmune checkpoint inhibitors (ICIs) provide a significant survival benefit in non-small cell lung cancer (NSCLC) patients; however, accurately predicting which patients will benefit remains a challenge. As previously shown, the STOP model, a machine learning model based on serum tumor markers, is capable of identifying non-responders after 6 weeks of ICIs.ObjectiveThis study aims to externally validate this model and to assess the predictive value in combination with radiological response assessment using RECIST criteria.MethodsIn a cohort of 242 metastatic NSCLC patients, CYFRA, CEA, and NSE were measured before start and after 6 weeks of ICI treatment. The ability of the STOP model to predict no durable benefit (NDB; progressive disease, death within 6 months or disease control of less than 6 months) was assessed using specificity and positive predictive value (PPV). Moreover, a combination of the STOP model with RECIST after 6-8 weeks of ICIs was investigated.ResultsThe STOP model achieved a specificity of 96% (95% CI 95%-97%) and a PPV of predicting NDB of 88.1% (95% CI 85.9%-90.3%). Combining the STOP model with RECIST improved specificity and PPV to 100% and predicted NDB on average 11.6 weeks (IQR 1.8-18.0 weeks) prior to developing radiologically defined progression.ConclusionsAfter 6 weeks of ICIs, the blood-based STOP model was capable of accurately predicting NDB in metastatic NSCLC patients, earlier than conventional radiological assessment. The combined serological and radiological response assessment creates an early opportunity to safely stop ICI treatment in patients who will not benefit, although the clinical utility of the assay is limited since the high specificity comes at the cost of a lower sensitivity.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"47 ","pages":"14230380251316788"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143650843","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-12-24DOI: 10.1177/14230380251410470
Md Shahedur Rahman, Rizone Al Hasib, Md Rezanur Rahman, Polash Kumar Biswas, Abu Reza, Munzura Khatun, Mohammad Abu Hena Mostofa Jamal
BackgroundIn recent years, the significance of sirtuins in cancer biology has become increasingly evident, but their molecular mechanisms and prognostic impacts remain elusive.ObjectiveThe present study aimed to investigate the differential expression of the sirtuin gene family across cancers and to evaluate their prognostic value.MethodsWe used various bioinformatics databases and methodologies, including Oncomine, GEPIA, OncoDB, cBioPortal, R2 Kaplan-Meier Scanner, STRING, etc., to determine the expression pattern of the sirtuin family genes, along with their mutations and prognostic values in human cancers.ResultsIn the current study, SIRT1, SIRT2, SIRT4, and SIRT5 were downregulated in lymphoma, whereas SIRT6 and SIRT7 were overexpressed. In breast cancer, SIRT3, SIRT5, and SIRT7 were overexpressed, and in terms of kidney cancer, higher expression of SIRT2, SIRT3, and SIRT5 was observed. In contrast, for leukemia, bladder, and brain cancers, most sirtuin family members showed reduced expression. We found that most mutations occurred in uterine cancer, chRCC (chromophobe renal cell carcinoma), DLBCL (diffuse large B-cell lymphoma), melanoma, pRCC (papillary renal cell carcinoma), and esophageal cancer. Moreover, we identified the relevant functional proteins through protein-protein interaction analysis to evaluate copy number alterations (CNAs) in sirtuins. The most frequent alterations were amplifications and deep deletions. Survival analysis demonstrated that SIRT1 and SIRT2 overexpression correlated with improved overall survival in low-grade glioma but predicted poorer outcomes in ovarian cancer. Downregulation of SIRT1, SIRT3, and SIRT5 was associated with better prognosis in DLBCL, while SIRT3 and SIRT4 upregulation predicted favorable survival in testicular germ cell tumors. SIRT6 overexpression was linked to favorable prognosis in esophageal carcinoma and sarcoma, while unfavorable outcomes were observed in hepatocellular carcinoma and cholangiocarcinoma. SIRT7 upregulation was significantly associated with reduced survival in esophageal, liver, and uterine cancers, but surprisingly correlated with improved outcomes in urothelial carcinoma and cervical squamous cell carcinoma.ConclusionsTogether, this multi-omics analysis reveals the correlation and prognostic values of sirtuins across multiple types of human cancers and suggests that sirtuins may serve as promising biomarkers for different cancers.
{"title":"An integrated bioinformatics and multi-omics investigation of the sirtuin family to identify their prognostic importance in human cancers.","authors":"Md Shahedur Rahman, Rizone Al Hasib, Md Rezanur Rahman, Polash Kumar Biswas, Abu Reza, Munzura Khatun, Mohammad Abu Hena Mostofa Jamal","doi":"10.1177/14230380251410470","DOIUrl":"https://doi.org/10.1177/14230380251410470","url":null,"abstract":"<p><p>BackgroundIn recent years, the significance of sirtuins in cancer biology has become increasingly evident, but their molecular mechanisms and prognostic impacts remain elusive.ObjectiveThe present study aimed to investigate the differential expression of the sirtuin gene family across cancers and to evaluate their prognostic value.MethodsWe used various bioinformatics databases and methodologies, including Oncomine, GEPIA, OncoDB, cBioPortal, R2 Kaplan-Meier Scanner, STRING, etc., to determine the expression pattern of the sirtuin family genes, along with their mutations and prognostic values in human cancers.ResultsIn the current study, <i>SIRT1</i>, <i>SIRT2</i>, <i>SIRT4</i>, and <i>SIRT5</i> were downregulated in lymphoma, whereas <i>SIRT6</i> and <i>SIRT7</i> were overexpressed. In breast cancer, <i>SIRT3</i>, <i>SIRT5</i>, and <i>SIRT7</i> were overexpressed, and in terms of kidney cancer, higher expression of <i>SIRT2</i>, <i>SIRT3</i>, and <i>SIRT5</i> was observed. In contrast, for leukemia, bladder, and brain cancers, most sirtuin family members showed reduced expression. We found that most mutations occurred in uterine cancer, chRCC (chromophobe renal cell carcinoma), DLBCL (diffuse large B-cell lymphoma), melanoma, pRCC (papillary renal cell carcinoma), and esophageal cancer. Moreover, we identified the relevant functional proteins through protein-protein interaction analysis to evaluate copy number alterations (CNAs) in sirtuins. The most frequent alterations were amplifications and deep deletions. Survival analysis demonstrated that <i>SIRT1</i> and <i>SIRT2</i> overexpression correlated with improved overall survival in low-grade glioma but predicted poorer outcomes in ovarian cancer. Downregulation of <i>SIRT1</i>, <i>SIRT3</i>, and <i>SIRT5</i> was associated with better prognosis in DLBCL, while <i>SIRT3</i> and <i>SIRT4</i> upregulation predicted favorable survival in testicular germ cell tumors. <i>SIRT6</i> overexpression was linked to favorable prognosis in esophageal carcinoma and sarcoma, while unfavorable outcomes were observed in hepatocellular carcinoma and cholangiocarcinoma. <i>SIRT7</i> upregulation was significantly associated with reduced survival in esophageal, liver, and uterine cancers, but surprisingly correlated with improved outcomes in urothelial carcinoma and cervical squamous cell carcinoma.ConclusionsTogether, this multi-omics analysis reveals the correlation and prognostic values of sirtuins across multiple types of human cancers and suggests that sirtuins may serve as promising biomarkers for different cancers.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"47 ","pages":"14230380251410470"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145821102","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-09-23DOI: 10.1177/14230380251375818
Mariana Concentino Menezes Brum, Annie Cristhine Moraes Sousa Squiavinato, Luciana da Torre Carneiro, Luciana Bueno Ferreira, Alessandra Serain, Mariana Boroni, G Nestal de Moraes, Erp Gimba
BackgroundOsteopontin is a glycophosphoprotein aberrantly expressed in several tumor types, which exhibits several isoforms generated by post-translational and post-transcriptional mechanisms, including alternative splicing. Among total osteopontin (tOPN), the osteopontin-c (OPN-c) splice variant has been the most explored with an oncogenic role described for a range of tumor types. Especially in ovarian cancer (OC) cells, OPN-c is found overexpressed, presenting both diagnostic and prognostic implications.ObjectiveIn this review article, we aim to outline OPN-c roles in cancer, particularly in OC, in which it has been reported as a diagnostic biomarker.MethodsWe used PubMed search, and experimental procedures were summarized at the Figure legends.ResultsWe identified cytoplasmic, perinuclear, and nuclear OPN-c in OC cells that overexpress this OPN splice variant. Moreover, we report that OPN-c splicing isoform is found highly expressed in endometrioid OC patients' samples, compared to non-neoplastic ovarian tissues. Also, OPN-c expression levels have been associated with worse overall survival and worse progression-free survival in patients with both endometrioid and serous OC. Furthermore, OPN-c may be involved in a wide range of tumor features evoked by signaling pathways, such as AKT, ERK, and FAK.ConclusionsTherefore, a better comprehension of OPN-c roles in OC can further contribute to its application as a biomarker as well as a target for putative treatment strategies, especially those aiming to sensitize tumor cells to chemotherapeutic agents currently used in the OC treatment.
{"title":"Osteopontin-c gene expression and subcellular localization in ovarian cancer cells: Implications for prognosis and therapeutic responses.","authors":"Mariana Concentino Menezes Brum, Annie Cristhine Moraes Sousa Squiavinato, Luciana da Torre Carneiro, Luciana Bueno Ferreira, Alessandra Serain, Mariana Boroni, G Nestal de Moraes, Erp Gimba","doi":"10.1177/14230380251375818","DOIUrl":"https://doi.org/10.1177/14230380251375818","url":null,"abstract":"<p><p>BackgroundOsteopontin is a glycophosphoprotein aberrantly expressed in several tumor types, which exhibits several isoforms generated by post-translational and post-transcriptional mechanisms, including alternative splicing. Among total osteopontin (tOPN), the osteopontin-c (OPN-c) splice variant has been the most explored with an oncogenic role described for a range of tumor types. Especially in ovarian cancer (OC) cells, OPN-c is found overexpressed, presenting both diagnostic and prognostic implications.ObjectiveIn this review article, we aim to outline OPN-c roles in cancer, particularly in OC, in which it has been reported as a diagnostic biomarker.MethodsWe used PubMed search, and experimental procedures were summarized at the Figure legends.ResultsWe identified cytoplasmic, perinuclear, and nuclear OPN-c in OC cells that overexpress this OPN splice variant. Moreover, we report that OPN-c splicing isoform is found highly expressed in endometrioid OC patients' samples, compared to non-neoplastic ovarian tissues. Also, OPN-c expression levels have been associated with worse overall survival and worse progression-free survival in patients with both endometrioid and serous OC. Furthermore, OPN-c may be involved in a wide range of tumor features evoked by signaling pathways, such as AKT, ERK, and FAK.ConclusionsTherefore, a better comprehension of OPN-c roles in OC can further contribute to its application as a biomarker as well as a target for putative treatment strategies, especially those aiming to sensitize tumor cells to chemotherapeutic agents currently used in the OC treatment.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"47 ","pages":"14230380251375818"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145132009","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-12-24DOI: 10.1177/14230380251411266
{"title":"Expression of concern: \"Prognostic value of preoperative peripheral monocyte count in patients with hepatocellular carcinoma after liver transplantation\".","authors":"","doi":"10.1177/14230380251411266","DOIUrl":"https://doi.org/10.1177/14230380251411266","url":null,"abstract":"","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"47 ","pages":"14230380251411266"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145820680","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-03-18DOI: 10.1177/10104283241313441
Kimberly Hernandez, Caitlin H Nguyen, Girdhari Rijal
BackgroundMigrating strategies of the triple-negative breast cancer (TNBC) together with its role in the establishment of tumor microenvironment (TME), supporting metastasis, have been extensively studied. Extracellular matrix (ECM) is a major player for the TME, establishing the 3D spatial networks with interconnected pores necessary for the mechano-physiological function of the cells. Certain collagen aligners and cross-linkers which are necessary for the formation and the stabilization of ECM networks, however, have not been studied either in normal or in abnormal tissues. Complexities in cell-cell and cell-matrix interactions, and different in types and ratios of ECM proteins in a TME challenge to reveal the precise function of a particular protein that is exhibited by special cells and if specifically present in insignificant amount. Cancer-associated fibroblasts (CAFs) predominantly occupy the major stroma of a solid tumor where they deposit extracellular proteins in the excessive amount compared to other tumor-associated cells. For example, the TNBC tumor itself is positive for asporin (ASPN) since CAFs are major ASPN exhibitors. However, the TNBC cells express it insignificantly.ObjectiveThe increase in ECM and its networks suppresses the metastasis.MethodsHere, we studied the expression of collagen type I and ASPN in CAFS and MDA-MB-231 (MM231), and evaluated the role of ASPN in collagen alignment and crosslinking.ResultsTNBC cells have an insignificant expression of ASPN and scanty collagen fibers, some of which aggregate to form the stiff deranged fibers, forming large-size pores in ECM of cancer-cell-dominant outer core of TNBC that support cancer cell invasion and metastasis. Exogenous ASPN and fibroblast-ASPN supported for the collagen alignment and crosslinking that established the small-size pores in the ECM, inhibiting the cancer cell invasion.ConclusionsThe collagen aligner and the cross-linker, ASPN increases the ECM networks and decreases the migration, and this preliminary study provides the hope that ASPN might be used as an anti-metastatic drug after its confirmation through extensive studies in animal, and positive outcomes through preclinical trials.
{"title":"Asporin increases the extracellular matrix cross-links and inhibits the cancer cell migration.","authors":"Kimberly Hernandez, Caitlin H Nguyen, Girdhari Rijal","doi":"10.1177/10104283241313441","DOIUrl":"10.1177/10104283241313441","url":null,"abstract":"<p><p>BackgroundMigrating strategies of the triple-negative breast cancer (TNBC) together with its role in the establishment of tumor microenvironment (TME), supporting metastasis, have been extensively studied. Extracellular matrix (ECM) is a major player for the TME, establishing the 3D spatial networks with interconnected pores necessary for the mechano-physiological function of the cells. Certain collagen aligners and cross-linkers which are necessary for the formation and the stabilization of ECM networks, however, have not been studied either in normal or in abnormal tissues. Complexities in cell-cell and cell-matrix interactions, and different in types and ratios of ECM proteins in a TME challenge to reveal the precise function of a particular protein that is exhibited by special cells and if specifically present in insignificant amount. Cancer-associated fibroblasts (CAFs) predominantly occupy the major stroma of a solid tumor where they deposit extracellular proteins in the excessive amount compared to other tumor-associated cells. For example, the TNBC tumor itself is positive for asporin (ASPN) since CAFs are major ASPN exhibitors. However, the TNBC cells express it insignificantly.ObjectiveThe increase in ECM and its networks suppresses the metastasis.MethodsHere, we studied the expression of collagen type I and ASPN in CAFS and MDA-MB-231 (MM231), and evaluated the role of ASPN in collagen alignment and crosslinking.ResultsTNBC cells have an insignificant expression of ASPN and scanty collagen fibers, some of which aggregate to form the stiff deranged fibers, forming large-size pores in ECM of cancer-cell-dominant outer core of TNBC that support cancer cell invasion and metastasis. Exogenous ASPN and fibroblast-ASPN supported for the collagen alignment and crosslinking that established the small-size pores in the ECM, inhibiting the cancer cell invasion.ConclusionsThe collagen aligner and the cross-linker, ASPN increases the ECM networks and decreases the migration, and this preliminary study provides the hope that ASPN might be used as an anti-metastatic drug after its confirmation through extensive studies in animal, and positive outcomes through preclinical trials.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"47 ","pages":"10104283241313441"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143658739","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2025-12-24DOI: 10.1177/14230380251411267
{"title":"Expression of concern: \"Association between donor and recipient smoothened gene polymorphisms and the risk of hepatocellular carcinoma recurrence following orthotopic liver transplantation in a Han Chinese population\".","authors":"","doi":"10.1177/14230380251411267","DOIUrl":"https://doi.org/10.1177/14230380251411267","url":null,"abstract":"","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"47 ","pages":"14230380251411267"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145820664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Alexander Rupp, Sophie Bahlmann, Nicolai Trimpop, Joachim von Pawel, Stefan Holdenrieder
Background: Lung cancer is a major burden to global health and is still among the most frequent and most lethal malignant diseases. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in a variety of processes including tumorigenesis, formation of a tumor microenvironment and metastasis. It is therefore a potential prognostic biomarker in malignant diseases.
Objective: In this study, we investigated the applicability of MIF in serum samples as a biomarker in lung cancer.
Methods: In a retrospective approach, we analyzed the sera of 79 patients with non-small-cell lung cancer (NSCLC) and 14 patients with small-cell lung cancer (SCLC) before the start of chemotherapy, as well as before the second and third chemotherapy cycle, respectively. Serum MIF levels were measured using a sandwich immunoassay with a sulfo-tag-labelled detection antibody, while pro-gastrin releasing peptide (proGRP) levels were determined with an enzyme-linked immunosorbent assay.
Results: No difference in serum MIF levels between responders and non-responders to chemotherapy was observed at all time points, while proGRP levels were significantly lower in responders before the second chemotherapy cycle (p = 0.012). No differences in biomarker levels depending on the histopathological classification of NSCLC patients was found. Moreover, in ROC curve analyses MIF was not able to distinguish between responders and non-responders to therapy. proGRP could differentiate between responders and non-responders before the second chemotherapy cycle (p = 0.015) with sensitivities of 43% at 90% and 95% specificity, respectively. Likewise, proGRP yielded significantly longer survival times of patients with low proGRP concentrations before the second chemotherapy cycle (p = 0.015) in Kaplan-Meier analyses, yet MIF showed no significant differences in survival times at all time points. Comparison with the biomarkers CEA and CYFRA 21-1 in the same cohort showed that these established biomarkers clearly performed superior to MIF and proGRP.
Conclusions: From the present results, there is no indication that serum MIF may serve as a biomarker in prognosis and monitoring of response to therapy in lung cancer. Limitations of this study include its retrospective design, the inclusion of a larger NSCLC and a smaller SCLC subgroup, the classical chemotherapeutic treatment, the use of a non-diagnostic immunoassay (RUO-test) for MIF measurement and the lack of a validation cohort. Strengths of the study are its highly standardized procedures concerning sample collection, preanalytic treatment, measurements and quality control of the laboratory assays.
{"title":"Lack of clinical utility of serum macrophage migration inhibitory factor (MIF) for monitoring therapy response and estimating prognosis in advanced lung cancer.","authors":"Alexander Rupp, Sophie Bahlmann, Nicolai Trimpop, Joachim von Pawel, Stefan Holdenrieder","doi":"10.3233/TUB-230006","DOIUrl":"10.3233/TUB-230006","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is a major burden to global health and is still among the most frequent and most lethal malignant diseases. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in a variety of processes including tumorigenesis, formation of a tumor microenvironment and metastasis. It is therefore a potential prognostic biomarker in malignant diseases.</p><p><strong>Objective: </strong>In this study, we investigated the applicability of MIF in serum samples as a biomarker in lung cancer.</p><p><strong>Methods: </strong>In a retrospective approach, we analyzed the sera of 79 patients with non-small-cell lung cancer (NSCLC) and 14 patients with small-cell lung cancer (SCLC) before the start of chemotherapy, as well as before the second and third chemotherapy cycle, respectively. Serum MIF levels were measured using a sandwich immunoassay with a sulfo-tag-labelled detection antibody, while pro-gastrin releasing peptide (proGRP) levels were determined with an enzyme-linked immunosorbent assay.</p><p><strong>Results: </strong>No difference in serum MIF levels between responders and non-responders to chemotherapy was observed at all time points, while proGRP levels were significantly lower in responders before the second chemotherapy cycle (p = 0.012). No differences in biomarker levels depending on the histopathological classification of NSCLC patients was found. Moreover, in ROC curve analyses MIF was not able to distinguish between responders and non-responders to therapy. proGRP could differentiate between responders and non-responders before the second chemotherapy cycle (p = 0.015) with sensitivities of 43% at 90% and 95% specificity, respectively. Likewise, proGRP yielded significantly longer survival times of patients with low proGRP concentrations before the second chemotherapy cycle (p = 0.015) in Kaplan-Meier analyses, yet MIF showed no significant differences in survival times at all time points. Comparison with the biomarkers CEA and CYFRA 21-1 in the same cohort showed that these established biomarkers clearly performed superior to MIF and proGRP.</p><p><strong>Conclusions: </strong>From the present results, there is no indication that serum MIF may serve as a biomarker in prognosis and monitoring of response to therapy in lung cancer. Limitations of this study include its retrospective design, the inclusion of a larger NSCLC and a smaller SCLC subgroup, the classical chemotherapeutic treatment, the use of a non-diagnostic immunoassay (RUO-test) for MIF measurement and the lack of a validation cohort. Strengths of the study are its highly standardized procedures concerning sample collection, preanalytic treatment, measurements and quality control of the laboratory assays.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":" ","pages":"S341-S353"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9946783","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Thomas Muley, Mark A Schneider, Michael Meister, Michael Thomas, Claus Peter Heußel, Mark Kriegsmann, Stefan Holdenrieder, Birgit Wehnl, Vinzent Rolny, Anika Mang, Rebecca Gerber, Felix Herth
Background: Serum tumor markers (STM) may complement imaging and provide additional clinical information for patients with non-small cell lung cancer (NSCLC).
Objective: To determine whether STMs can predict outcomes in patients with stable disease (SD) after initial treatment.
Methods: This single-center, prospective, observational trial enrolled 395 patients with stage III/IV treatment-naïve NSCLC; of which 263 patients were included in this analysis. Computed Tomography (CT) scans were performed and STMs measured before and after initial treatment (two cycles of chemotherapy and/or an immune checkpoint inhibitor or tyrosine kinase inhibitor); analyses were based on CT and STM measurements obtained at first CT performed after cycle 2 only PFS and OS were analyzed by Kaplan-Meier curves and Cox-proportional hazard models.
Results: When patients with SD (n = 100) were split into high- and low-risk groups based on CYFRA 21-1, CEA and CA 125 measurements using an optimized cut-off, a 4-fold increase risk of progression or death was estimated for high- vs low-risk SD patients (PFS, HR 4.17; OS, 3.99; both p < 0.0001). Outcomes were similar between patients with high-risk SD or progressive disease (n = 35) (OS, HR 1.17) and between patients with low-risk SD or partial response (n = 128) (PFS, HR 0.98; OS, 1.14).
Conclusions: STMs can provide further guidance in patients with indeterminate CT responses by separating them into high- and low-risk groups for future PFS and OS events.
{"title":"CYFRA 21-1, CA 125 and CEA provide additional prognostic value in NSCLC patients with stable disease at first CT scan.","authors":"Thomas Muley, Mark A Schneider, Michael Meister, Michael Thomas, Claus Peter Heußel, Mark Kriegsmann, Stefan Holdenrieder, Birgit Wehnl, Vinzent Rolny, Anika Mang, Rebecca Gerber, Felix Herth","doi":"10.3233/TUB-220042","DOIUrl":"10.3233/TUB-220042","url":null,"abstract":"<p><strong>Background: </strong>Serum tumor markers (STM) may complement imaging and provide additional clinical information for patients with non-small cell lung cancer (NSCLC).</p><p><strong>Objective: </strong>To determine whether STMs can predict outcomes in patients with stable disease (SD) after initial treatment.</p><p><strong>Methods: </strong>This single-center, prospective, observational trial enrolled 395 patients with stage III/IV treatment-naïve NSCLC; of which 263 patients were included in this analysis. Computed Tomography (CT) scans were performed and STMs measured before and after initial treatment (two cycles of chemotherapy and/or an immune checkpoint inhibitor or tyrosine kinase inhibitor); analyses were based on CT and STM measurements obtained at first CT performed after cycle 2 only PFS and OS were analyzed by Kaplan-Meier curves and Cox-proportional hazard models.</p><p><strong>Results: </strong>When patients with SD (n = 100) were split into high- and low-risk groups based on CYFRA 21-1, CEA and CA 125 measurements using an optimized cut-off, a 4-fold increase risk of progression or death was estimated for high- vs low-risk SD patients (PFS, HR 4.17; OS, 3.99; both p < 0.0001). Outcomes were similar between patients with high-risk SD or progressive disease (n = 35) (OS, HR 1.17) and between patients with low-risk SD or partial response (n = 128) (PFS, HR 0.98; OS, 1.14).</p><p><strong>Conclusions: </strong>STMs can provide further guidance in patients with indeterminate CT responses by separating them into high- and low-risk groups for future PFS and OS events.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":" ","pages":"S163-S175"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41238982","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Stefan Holdenrieder, Huub H van Rossum, Michel van den Heuvel
Blood-based diagnostics for lung cancer support the diagnosis, estimation of prognosis, prediction, and monitoring of therapy response in lung cancer patients. The clinical utility of serum tumor markers has considerably increased due to developments in serum protein tumor markers analytics and clinical biomarker studies, the exploration of preanalytical and influencing conditions, the interpretation of biomarker combinations and individual biomarker kinetics, as well as the implementation of biostatistical models. In addition, circulating tumor DNA (ctDNA) and other liquid biopsy markers are playing an increasingly prominent role in the molecular tumor characterization and the monitoring of tumor evolution over time. Thus, modern lung cancer biomarkers may considerably contribute to an individualized companion diagnostics and provide a sensitive guidance for patients throughout the course of their disease. In this special edition on Tumor Markers in Lung Cancer, experts summarize recent developments in clinical laboratory diagnostics of lung cancer and give an outlook on future challenges and opportunities.
{"title":"Lung cancer biomarkers: Raising the clinical value of the classical and the new ones.","authors":"Stefan Holdenrieder, Huub H van Rossum, Michel van den Heuvel","doi":"10.3233/TUB-240004","DOIUrl":"10.3233/TUB-240004","url":null,"abstract":"<p><p>Blood-based diagnostics for lung cancer support the diagnosis, estimation of prognosis, prediction, and monitoring of therapy response in lung cancer patients. The clinical utility of serum tumor markers has considerably increased due to developments in serum protein tumor markers analytics and clinical biomarker studies, the exploration of preanalytical and influencing conditions, the interpretation of biomarker combinations and individual biomarker kinetics, as well as the implementation of biostatistical models. In addition, circulating tumor DNA (ctDNA) and other liquid biopsy markers are playing an increasingly prominent role in the molecular tumor characterization and the monitoring of tumor evolution over time. Thus, modern lung cancer biomarkers may considerably contribute to an individualized companion diagnostics and provide a sensitive guidance for patients throughout the course of their disease. In this special edition on Tumor Markers in Lung Cancer, experts summarize recent developments in clinical laboratory diagnostics of lung cancer and give an outlook on future challenges and opportunities.</p>","PeriodicalId":23364,"journal":{"name":"Tumor Biology","volume":"46 s1","pages":"S1-S7"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140190182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}