Pub Date : 2025-12-19DOI: 10.1158/2767-9764.CRC-25-0563
Long-Sheng Chang, Janet L Oblinger, Lai Man Natalie Wu, Cecelia Miller, Sarah S Burns
We previously generated an orthotopic, NF2-deficient meningioma model using the luciferase-expressing Ben-Men-1 cell line established from a sporadic tumor and identified the multi-kinase inhibitor brigatinib and the mTOR kinase inhibitor INK128 to potently impede tumor growth. Here, we describe generation of the telomerase-immortalized AG-NF2-Men cell line from a grade-1 meningioma of an NF2-related schwannomatosis (NF2-SWN) patient. We showed that like Ben-Men-1 cells, AG-NF2-Men cells were NF2-null, expressed several NF2-regulated receptor tyrosine kinases, and responded to their cognate ligands. We also found that brigatinib and INK128 alone inhibited AG-NF2-Men cell proliferation at IC50 values similar to those in Ben-Men-1 cells. Combining brigatinib with INK128 exhibited growth-inhibitory synergy. Mechanistically, the combination not only completely abrogated p-AKT(S473) and its downstream signaling compared to either drug alone but also prevented INK128-mediated re-phosphorylation of AKT on T308. Also, the combination more-effectively blocked ligand-mediated phosphorylation of EGFR, ErbB3, and IGF-1R and elicited major changes in the expression of genes including the upstream regulators of several signaling networks important for meningioma growth. Further, we generated luciferase-expressing AG-NF2-Men cells that readily grew as intracranial xenografts. Importantly, combining brigatinib with INK128 enhanced tumor regression in both the orthotopic AG-NF2-Men and Ben-Men-1 xenograft models. As the first NF2-SWN-related meningioma cell line, AG-NF2-Men is a unique reagent for investigating meningioma biology and therapeutics. A clinical trial to evaluate the combination of brigatinib with an mTOR inhibitor in NF2-deficient meningiomas is warranted.
{"title":"Combining brigatinib with mTOR inhibition to effectively treat NF2-SWN-associated and sporadic NF2-deficient meningiomas.","authors":"Long-Sheng Chang, Janet L Oblinger, Lai Man Natalie Wu, Cecelia Miller, Sarah S Burns","doi":"10.1158/2767-9764.CRC-25-0563","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0563","url":null,"abstract":"<p><p>We previously generated an orthotopic, NF2-deficient meningioma model using the luciferase-expressing Ben-Men-1 cell line established from a sporadic tumor and identified the multi-kinase inhibitor brigatinib and the mTOR kinase inhibitor INK128 to potently impede tumor growth. Here, we describe generation of the telomerase-immortalized AG-NF2-Men cell line from a grade-1 meningioma of an NF2-related schwannomatosis (NF2-SWN) patient. We showed that like Ben-Men-1 cells, AG-NF2-Men cells were NF2-null, expressed several NF2-regulated receptor tyrosine kinases, and responded to their cognate ligands. We also found that brigatinib and INK128 alone inhibited AG-NF2-Men cell proliferation at IC50 values similar to those in Ben-Men-1 cells. Combining brigatinib with INK128 exhibited growth-inhibitory synergy. Mechanistically, the combination not only completely abrogated p-AKT(S473) and its downstream signaling compared to either drug alone but also prevented INK128-mediated re-phosphorylation of AKT on T308. Also, the combination more-effectively blocked ligand-mediated phosphorylation of EGFR, ErbB3, and IGF-1R and elicited major changes in the expression of genes including the upstream regulators of several signaling networks important for meningioma growth. Further, we generated luciferase-expressing AG-NF2-Men cells that readily grew as intracranial xenografts. Importantly, combining brigatinib with INK128 enhanced tumor regression in both the orthotopic AG-NF2-Men and Ben-Men-1 xenograft models. As the first NF2-SWN-related meningioma cell line, AG-NF2-Men is a unique reagent for investigating meningioma biology and therapeutics. A clinical trial to evaluate the combination of brigatinib with an mTOR inhibitor in NF2-deficient meningiomas is warranted.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795426","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 : 2025-12-18DOI: 10.1158/2767-9764.CRC-25-0627
Alexander Zheleznyak, Dhanusha Duraiyan, Prasanth Thunuguntla, Jessica Camacho, Shuang Wu, Jingxia Liu, Jason D Weber, Daniel L J Thorek, Benjamin S Strnad, Ryan C Fields, Matthew A Ciorba, Christine Yoon, Valerie Blanc, Nicholas Davidson, Jessica Silva-Fisher, Christopher D Malone
Molecular signatures predict prognosis in hepatocellular carcinoma (HCC), but their relevance to transarterial radioembolization (TARE) with yttrium-90 (90Y) is unknown. We aimed to identify baseline and treatment-induced pathways associated with response and nominate biomarkers. Ten transcriptomically diverse human liver cancer cell lines were exposed to escalating activities of glass 90Y microspheres for ten days. Normalized area-under-curve (nAUC) values quantified sensitivity. Whole-transcriptome RNA sequencing (RNAseq) at baseline and after treatment was analyzed with elastic-net (EN) regression and gene set enrichment. Findings were corroborated by quantitative reverse transcription polymerase chain reaction (RT-qPCR) and exploratory analysis of pretreatment tumor samples from patients undergoing TARE. Liver cancer cell line responses to 90Y were heterogenous, with resistance aligning to Hoshida S1 and cholangiocarcinoma-like subtypes. Epithelial-mesenchymal transition (EMT) and adhesion pathways were enriched in resistant lines, with CD44 and ITGA3/α3β1 emerging as candidate markers, corroborated by RNA and protein expression. After 90Y exposure, resistant lines upregulated interferon (γ/α), TNFα/inflammatory, and antigen-presentation-related pathways, whereas sensitive lines downregulated these pathways along with DNA-repair and oxidative-phosphorylation. In an exploratory patient cohort, higher tumor CD44 expression trended with early progression. In conclusion, cancer cell lines display marked biological heterogeneity in response to 90Y. Baseline EMT/adhesion signatures and stress-response pathways nominate CD44 and ITGA3/α3β1 as candidate biomarkers of resistance. These findings delineate molecular programs of β-emitter radioresistance and identify candidate pathways for future targeting.
{"title":"Epithelial-Mesenchymal Transition and Stress Adaptations Underlie Yttrium-90 Resistance in Liver Cancer Cell Lines.","authors":"Alexander Zheleznyak, Dhanusha Duraiyan, Prasanth Thunuguntla, Jessica Camacho, Shuang Wu, Jingxia Liu, Jason D Weber, Daniel L J Thorek, Benjamin S Strnad, Ryan C Fields, Matthew A Ciorba, Christine Yoon, Valerie Blanc, Nicholas Davidson, Jessica Silva-Fisher, Christopher D Malone","doi":"10.1158/2767-9764.CRC-25-0627","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0627","url":null,"abstract":"<p><p>Molecular signatures predict prognosis in hepatocellular carcinoma (HCC), but their relevance to transarterial radioembolization (TARE) with yttrium-90 (90Y) is unknown. We aimed to identify baseline and treatment-induced pathways associated with response and nominate biomarkers. Ten transcriptomically diverse human liver cancer cell lines were exposed to escalating activities of glass 90Y microspheres for ten days. Normalized area-under-curve (nAUC) values quantified sensitivity. Whole-transcriptome RNA sequencing (RNAseq) at baseline and after treatment was analyzed with elastic-net (EN) regression and gene set enrichment. Findings were corroborated by quantitative reverse transcription polymerase chain reaction (RT-qPCR) and exploratory analysis of pretreatment tumor samples from patients undergoing TARE. Liver cancer cell line responses to 90Y were heterogenous, with resistance aligning to Hoshida S1 and cholangiocarcinoma-like subtypes. Epithelial-mesenchymal transition (EMT) and adhesion pathways were enriched in resistant lines, with CD44 and ITGA3/α3β1 emerging as candidate markers, corroborated by RNA and protein expression. After 90Y exposure, resistant lines upregulated interferon (γ/α), TNFα/inflammatory, and antigen-presentation-related pathways, whereas sensitive lines downregulated these pathways along with DNA-repair and oxidative-phosphorylation. In an exploratory patient cohort, higher tumor CD44 expression trended with early progression. In conclusion, cancer cell lines display marked biological heterogeneity in response to 90Y. Baseline EMT/adhesion signatures and stress-response pathways nominate CD44 and ITGA3/α3β1 as candidate biomarkers of resistance. These findings delineate molecular programs of β-emitter radioresistance and identify candidate pathways for future targeting.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145783829","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 : 2025-12-18DOI: 10.1158/2767-9764.CRC-25-0150
James Boyle, Jan Zaucha, Felicia Ng, Andreas Spitzmüller, Miljenka Vuko, Felix J Segerer, Monica Azqueta-Gavaldon, Marco Testori, Amelia Raymond, Barrett R B Nuttall, Andrea Ennio Storti, Sakshi Gulati, Shaan Gill, Michael Surace, Paola Marco-Casanova, Emma V Jones, Ben S Sidders, Jorge S Reis-Filho, Maurizio Scaltriti, Anita Semertzidou, Helen K Angell
Ovarian cancers with similar histopathologic but diverse immunogenomic profiles may require different treatment regimens to achieve the best clinical outcomes. Development of optimal treatment regimens will thus require an understanding of the main ways in which ovarian cancers differ with respect to their immunogenomic profiles. We used a multi-modal latent variable model to enable an integrated analysis of somatic mutation, mRNA expression, and multiplex immunofluorescence data, to uncover the principal drivers of inter-tumour heterogeneity across 197 ovarian cancer patients. We found that the majority of the immunogenomic inter-tumour heterogeneity was driven by gene expression and immune infiltration in the tumour core and invasive margin. Moreover, much of this heterogeneity could not be explained by histological subtype; somatic mutation patterns explained much of the difference between high-grade serous and other subtypes. Clustering of samples according to their positions in the latent space revealed a distinct subgroup of Endometrioid Ovarian cancer tumours, characterised by increased CST2 expression and improved prognosis, among other immunogenomic features. We identified a group of collagen-related genes, whose expression in all subtypes was inversely correlated with the density of proliferating tumour cells in the tumour core and associated with increased levels of inflammatory fibroblasts, independent of tumour purity. In summary, our findings advocate for a paradigm shift in how we classify and approach the treatment of ovarian cancer. By moving beyond the constraints of histological subtypes and delving into the molecular intricacies of patient-specific tumour microenvironments, we unveil a new opportunity for targeted immuno-oncological treatment of a subset of ovarian cancer patients.
{"title":"Characterisation of the immunogenomic landscape of ovarian cancer uncovers a distinct subset of endometroid tumours associated with high CST2 expression and a favourable prognosis.","authors":"James Boyle, Jan Zaucha, Felicia Ng, Andreas Spitzmüller, Miljenka Vuko, Felix J Segerer, Monica Azqueta-Gavaldon, Marco Testori, Amelia Raymond, Barrett R B Nuttall, Andrea Ennio Storti, Sakshi Gulati, Shaan Gill, Michael Surace, Paola Marco-Casanova, Emma V Jones, Ben S Sidders, Jorge S Reis-Filho, Maurizio Scaltriti, Anita Semertzidou, Helen K Angell","doi":"10.1158/2767-9764.CRC-25-0150","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0150","url":null,"abstract":"<p><p>Ovarian cancers with similar histopathologic but diverse immunogenomic profiles may require different treatment regimens to achieve the best clinical outcomes. Development of optimal treatment regimens will thus require an understanding of the main ways in which ovarian cancers differ with respect to their immunogenomic profiles. We used a multi-modal latent variable model to enable an integrated analysis of somatic mutation, mRNA expression, and multiplex immunofluorescence data, to uncover the principal drivers of inter-tumour heterogeneity across 197 ovarian cancer patients. We found that the majority of the immunogenomic inter-tumour heterogeneity was driven by gene expression and immune infiltration in the tumour core and invasive margin. Moreover, much of this heterogeneity could not be explained by histological subtype; somatic mutation patterns explained much of the difference between high-grade serous and other subtypes. Clustering of samples according to their positions in the latent space revealed a distinct subgroup of Endometrioid Ovarian cancer tumours, characterised by increased CST2 expression and improved prognosis, among other immunogenomic features. We identified a group of collagen-related genes, whose expression in all subtypes was inversely correlated with the density of proliferating tumour cells in the tumour core and associated with increased levels of inflammatory fibroblasts, independent of tumour purity. In summary, our findings advocate for a paradigm shift in how we classify and approach the treatment of ovarian cancer. By moving beyond the constraints of histological subtypes and delving into the molecular intricacies of patient-specific tumour microenvironments, we unveil a new opportunity for targeted immuno-oncological treatment of a subset of ovarian cancer patients.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145783759","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 : 2025-12-12DOI: 10.1158/2767-9764.CRC-25-0580
Jorge Baquero, Xiao-Han Tang, Daniel Galke, Theresa Scognamiglio, Tuo Zhang, Lorraine J Gudas
Although five-year relative survival rates for oral squamous cell carcinoma (OSCC) have moderately increased in the last 30 years, most patients are diagnosed during the later stages of the disease. B cell-specific Moloney murine leukemia virus integration site 1 (BMI1) is a biomarker of OSCC that is increased in epithelial basal stem cells (SCs) of premalignant oral lesions. However, the molecular functions of BMI1 in early-stage OSCC have not been fully elucidated. Here we used a transgenic mouse line (KrTB) that overexpresses BMI1 in the tongue epithelial SCs to delineate BMI1 actions during these early stages. We observed more oncogenic changes in mice with ectopic BMI1 expression after only a short, 4-week treatment with the carcinogen 4-nitroquinoline 1-oxide (4-NQO). For example, we detected increased proliferation, oxidative stress, and expression of multiple transcripts and proteins linked to human OSCCs in murine tongue epithelia with high, ectopic BMI1 expression. Furthermore, increases in mRNAs encoding multiple metabolic targets, such as SLC16A3, PKM, and GPI1, were greater upon BMI1 overexpression with 4 weeks of 4-NQO treatment. In a human OSCC model (SCC-25 cell line) in which we deleted the BMI1 gene we observed decreases in proliferation, oxidative stress, and expression of the glycolysis-associated protein GLUT1. Thus, BMI1 expression leads to increases in key features of early-stage, carcinogen-induced tumorigenesis, including metabolic reprogramming. Consequently, limiting BMI1 could be a potential target for cancer prevention approaches that merits further consideration and additional functional studies.
{"title":"Key early changes in oral squamous cell carcinogenesis are accelerated by ectopic BMI1 expression.","authors":"Jorge Baquero, Xiao-Han Tang, Daniel Galke, Theresa Scognamiglio, Tuo Zhang, Lorraine J Gudas","doi":"10.1158/2767-9764.CRC-25-0580","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0580","url":null,"abstract":"<p><p>Although five-year relative survival rates for oral squamous cell carcinoma (OSCC) have moderately increased in the last 30 years, most patients are diagnosed during the later stages of the disease. B cell-specific Moloney murine leukemia virus integration site 1 (BMI1) is a biomarker of OSCC that is increased in epithelial basal stem cells (SCs) of premalignant oral lesions. However, the molecular functions of BMI1 in early-stage OSCC have not been fully elucidated. Here we used a transgenic mouse line (KrTB) that overexpresses BMI1 in the tongue epithelial SCs to delineate BMI1 actions during these early stages. We observed more oncogenic changes in mice with ectopic BMI1 expression after only a short, 4-week treatment with the carcinogen 4-nitroquinoline 1-oxide (4-NQO). For example, we detected increased proliferation, oxidative stress, and expression of multiple transcripts and proteins linked to human OSCCs in murine tongue epithelia with high, ectopic BMI1 expression. Furthermore, increases in mRNAs encoding multiple metabolic targets, such as SLC16A3, PKM, and GPI1, were greater upon BMI1 overexpression with 4 weeks of 4-NQO treatment. In a human OSCC model (SCC-25 cell line) in which we deleted the BMI1 gene we observed decreases in proliferation, oxidative stress, and expression of the glycolysis-associated protein GLUT1. Thus, BMI1 expression leads to increases in key features of early-stage, carcinogen-induced tumorigenesis, including metabolic reprogramming. Consequently, limiting BMI1 could be a potential target for cancer prevention approaches that merits further consideration and additional functional studies.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145745866","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 : 2025-12-11DOI: 10.1158/2767-9764.CRC-25-0419
Ivan Brukner, Vincent Gagné, Alex Richard-St-Hilaire, Pascal Tremblay-Dauphinais, Claire Fuchs, Henrique Bittencourt, Teodor Veres, Daniel Sinnett, Maja Krajinovic
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Despite improved therapies, refractory and relapsed ALL remain the leading cause of cancer-related mortality in children. There is a need for accessible biomarkers for frequent, minimally invasive disease monitoring, and prompt intervention. MicroDNA is a novel extrachromosomal DNA that preferentially originates from the gene segments with high transcriptional activity and/or increased chromatin accessibility. We investigated whether microDNA-producing genes repertoire changes in a disease-dependent manner. We characterized microDNAs in 52 paired bone marrow (BM) and plasma samples from pediatric ALL patients at diagnosis, relapse, and remission. No difference in length or number of microDNA was noted across stages, but comparative analysis of microDNA profiles led to the identification of microDNA gene panel associated with active disease. The relative distribution of these genes was significantly different from that expected by chance (P <0.0001). Analyses of BM samples identified a signature comprising 289 distinct microDNA-producing genes present in multiple patients at diagnosis and relapse but absent in remission. The best biomarker candidates were 11 microDNA producing genes identified also in plasma samples at diagnosis and overrepresented in patients who relapsed (p=0.006). MicroDNA from the same genes was confirmed in relapse plasma samples. All signature genes are known to be involved in cancer proliferation or drug response. MicroDNA appears to be a candidate for a novel class of biomarkers for ALL, with the potential to improve precision diagnostics, particularly through their identification in plasma samples. Further validation in an independent cohort of patients is warranted.
{"title":"Extrachromosomal microDNA Signature as Candidate Biomarker in Pediatric Acute Lymphoblastic Leukemia.","authors":"Ivan Brukner, Vincent Gagné, Alex Richard-St-Hilaire, Pascal Tremblay-Dauphinais, Claire Fuchs, Henrique Bittencourt, Teodor Veres, Daniel Sinnett, Maja Krajinovic","doi":"10.1158/2767-9764.CRC-25-0419","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0419","url":null,"abstract":"<p><p>Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Despite improved therapies, refractory and relapsed ALL remain the leading cause of cancer-related mortality in children. There is a need for accessible biomarkers for frequent, minimally invasive disease monitoring, and prompt intervention. MicroDNA is a novel extrachromosomal DNA that preferentially originates from the gene segments with high transcriptional activity and/or increased chromatin accessibility. We investigated whether microDNA-producing genes repertoire changes in a disease-dependent manner. We characterized microDNAs in 52 paired bone marrow (BM) and plasma samples from pediatric ALL patients at diagnosis, relapse, and remission. No difference in length or number of microDNA was noted across stages, but comparative analysis of microDNA profiles led to the identification of microDNA gene panel associated with active disease. The relative distribution of these genes was significantly different from that expected by chance (P <0.0001). Analyses of BM samples identified a signature comprising 289 distinct microDNA-producing genes present in multiple patients at diagnosis and relapse but absent in remission. The best biomarker candidates were 11 microDNA producing genes identified also in plasma samples at diagnosis and overrepresented in patients who relapsed (p=0.006). MicroDNA from the same genes was confirmed in relapse plasma samples. All signature genes are known to be involved in cancer proliferation or drug response. MicroDNA appears to be a candidate for a novel class of biomarkers for ALL, with the potential to improve precision diagnostics, particularly through their identification in plasma samples. Further validation in an independent cohort of patients is warranted.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145745947","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 : 2025-12-10DOI: 10.1158/2767-9764.CRC-25-0381
Pınar Çakılkaya, Ida Marie Egeland Larsen, Qun Jiang, Kirstine Sandal Nørregaard, Henrik Gårdsvoll, Jingli Zhang, Henrik Jessen Jürgensen, Michaela Hansen Blomquist, Alba Martinez Perlado, Oliver Krigslund, Eric Santoni-Rugiu, Lars Henning Engelholm, Raffit Hassan, Niels Behrendt
uPARAP is a collagen-internalizing receptor with emerging relevance as a therapeutic target. The involvement of uPARAP in extracellular matrix turnover and stromal remodeling within the tumor microenvironment (TME) makes it an especially promising target in tumors with a high stromal component. Mesothelioma, characterized by its unique complex TME, is a highly refractory cancer type for which no effective targeted therapy exists. uPARAP has been shown to be expressed across all subtypes of mesothelioma, especially in the sarcomatoid and biphasic subtypes, suggesting that a uPARAP-targeted therapy may benefit a specific patient population. Here, we utilized an antibody-drug conjugate (ADC) comprising an anti-uPARAP monoclonal antibody conjugated to monomethyl auristatin E (MMAE) via a protease-sensitive linker for preclinical studies on mesothelioma treatment. Employing mouse xenograft models with a human mesothelioma cell line or a patient-derived mesothelioma cell isolate, we demonstrated a strong anti-cancer effect following treatment with the uPARAP-targeting ADC. Immunohistochemical studies revealed this to be the case, even in tumors exhibiting a heterogeneous expression of uPARAP, suggesting that uPARAP-expressing cancer-associated fibroblasts also play a role in the anti-cancer effect through a bystander mechanism. Furthermore, we show that combination-treatment with cisplatin, commonly used in first-line mesothelioma therapy, leads to a strongly enhanced anti-cancer effect relative to treatment with either drug alone. Our study demonstrates the potential utility of a uPARAP-targeted ADC as a therapeutic option for mesothelioma, alone or in combination with chemotherapeutics. This perspective is particularly emphasized by a recently initiated clinical trial with a humanized anti-uPARAP-ADC for the treatment of other malignancies.
uPARAP是一种胶原内化受体,作为一种新的治疗靶点。uPARAP参与肿瘤微环境(TME)内的细胞外基质转换和基质重塑,这使得它在具有高基质成分的肿瘤中成为一个特别有希望的靶点。间皮瘤是一种高度难治的癌症类型,其特点是其独特的复杂TME,目前尚无有效的靶向治疗方法。uPARAP已被证明在所有间皮瘤亚型中表达,特别是在肉瘤样亚型和双相亚型中,这表明uPARAP靶向治疗可能使特定患者群体受益。在这里,我们利用了一种抗体-药物偶联物(ADC),该偶联物包括一种抗uparap单克隆抗体,通过蛋白酶敏感的连接物偶联到单甲基aurisatin E (MMAE),用于间皮瘤治疗的临床前研究。利用人类间皮瘤细胞系或患者来源的间皮瘤细胞分离物的小鼠异种移植模型,我们证明了用靶向uparap的ADC治疗后具有很强的抗癌效果。免疫组织化学研究表明,即使在uPARAP异质性表达的肿瘤中也是如此,这表明表达uPARAP的癌症相关成纤维细胞也通过旁观者机制在抗癌作用中发挥作用。此外,我们表明,与一线间皮瘤治疗中常用的顺铂联合治疗,相对于单独使用任何一种药物治疗,可显著增强抗癌效果。我们的研究证明了uparap靶向ADC作为间皮瘤治疗选择的潜在效用,单独或与化疗药物联合使用。最近启动的一项用于治疗其他恶性肿瘤的人源化抗uparap - adc的临床试验特别强调了这一观点。
{"title":"Targeting uPARAP with an antibody-drug conjugate exhibits efficacy against mesothelioma and synergizes with cisplatin.","authors":"Pınar Çakılkaya, Ida Marie Egeland Larsen, Qun Jiang, Kirstine Sandal Nørregaard, Henrik Gårdsvoll, Jingli Zhang, Henrik Jessen Jürgensen, Michaela Hansen Blomquist, Alba Martinez Perlado, Oliver Krigslund, Eric Santoni-Rugiu, Lars Henning Engelholm, Raffit Hassan, Niels Behrendt","doi":"10.1158/2767-9764.CRC-25-0381","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0381","url":null,"abstract":"<p><p>uPARAP is a collagen-internalizing receptor with emerging relevance as a therapeutic target. The involvement of uPARAP in extracellular matrix turnover and stromal remodeling within the tumor microenvironment (TME) makes it an especially promising target in tumors with a high stromal component. Mesothelioma, characterized by its unique complex TME, is a highly refractory cancer type for which no effective targeted therapy exists. uPARAP has been shown to be expressed across all subtypes of mesothelioma, especially in the sarcomatoid and biphasic subtypes, suggesting that a uPARAP-targeted therapy may benefit a specific patient population. Here, we utilized an antibody-drug conjugate (ADC) comprising an anti-uPARAP monoclonal antibody conjugated to monomethyl auristatin E (MMAE) via a protease-sensitive linker for preclinical studies on mesothelioma treatment. Employing mouse xenograft models with a human mesothelioma cell line or a patient-derived mesothelioma cell isolate, we demonstrated a strong anti-cancer effect following treatment with the uPARAP-targeting ADC. Immunohistochemical studies revealed this to be the case, even in tumors exhibiting a heterogeneous expression of uPARAP, suggesting that uPARAP-expressing cancer-associated fibroblasts also play a role in the anti-cancer effect through a bystander mechanism. Furthermore, we show that combination-treatment with cisplatin, commonly used in first-line mesothelioma therapy, leads to a strongly enhanced anti-cancer effect relative to treatment with either drug alone. Our study demonstrates the potential utility of a uPARAP-targeted ADC as a therapeutic option for mesothelioma, alone or in combination with chemotherapeutics. This perspective is particularly emphasized by a recently initiated clinical trial with a humanized anti-uPARAP-ADC for the treatment of other malignancies.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145716591","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 : 2025-12-10DOI: 10.1158/2767-9764.CRC-25-0411
Corey M Perkins, Jinmai Jiang, Kalyanee Shirlekar, Zachary Greenberg, Md Abu Talha Siddique, Jason Brant, Kiley Graim, Mei He, Sarah Kim, Diana J Wilkie, Bo Han, Jamel Ali, Pascal Belleau, Astrid Deschênes, Alexander Krasnitz, Mazhar Kanak, Thomas D Schmittgen
Molecular subtypes exist in pancreatic ductal adenocarcinoma (PDAC), however comparable subtypes are not known to exist in normal pancreas. A large, racially diverse cohort of normal, human pancreatic acinar cells were obtained from islet transplantation centers. RNA isolated from uncultured acini and those acini that had undergone acinar ductal metaplasia (ADM) was sequenced. Data analysis from 69 uncultured, normal acini showed two clusters of samples, one that aligns with exocrine resembling tissue (ERT) signatures and another with the classical and basal (C/B) subtypes. Gene expression associated with the ERT signature decreased following 6 days of ADM transdifferentiation while those assigned to the C/B subtype increased. Subtype classification findings were confirmed in two independent cohorts of transcriptomic data of normal pancreas. A direct correlation exists between ancestral admixture and the molecular index of ADM; those specimens with higher African ancestral admixture showed a greater propensity for ADM compared to those with a higher Ameridigenous admixture. The extent of morphological ADM decreased in the order of Black>White>Hispanic race. Analysis of associated metadata revealed that potential confounders such as diabetes, obesity, age, gender or specimen handling and processing variables did not influence the distribution of samples across subtype classification. We report that normal human pancreatic acinar tissues group into molecular subtypes like those used to classify PDAC and that subtype assignment and ADM are influenced by the donor's race.
{"title":"Transcriptomic analyses of normal human pancreata reveal the presence of cancer subtypes that correlate with acinar ductal metaplasia and donor ancestry.","authors":"Corey M Perkins, Jinmai Jiang, Kalyanee Shirlekar, Zachary Greenberg, Md Abu Talha Siddique, Jason Brant, Kiley Graim, Mei He, Sarah Kim, Diana J Wilkie, Bo Han, Jamel Ali, Pascal Belleau, Astrid Deschênes, Alexander Krasnitz, Mazhar Kanak, Thomas D Schmittgen","doi":"10.1158/2767-9764.CRC-25-0411","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0411","url":null,"abstract":"<p><p>Molecular subtypes exist in pancreatic ductal adenocarcinoma (PDAC), however comparable subtypes are not known to exist in normal pancreas. A large, racially diverse cohort of normal, human pancreatic acinar cells were obtained from islet transplantation centers. RNA isolated from uncultured acini and those acini that had undergone acinar ductal metaplasia (ADM) was sequenced. Data analysis from 69 uncultured, normal acini showed two clusters of samples, one that aligns with exocrine resembling tissue (ERT) signatures and another with the classical and basal (C/B) subtypes. Gene expression associated with the ERT signature decreased following 6 days of ADM transdifferentiation while those assigned to the C/B subtype increased. Subtype classification findings were confirmed in two independent cohorts of transcriptomic data of normal pancreas. A direct correlation exists between ancestral admixture and the molecular index of ADM; those specimens with higher African ancestral admixture showed a greater propensity for ADM compared to those with a higher Ameridigenous admixture. The extent of morphological ADM decreased in the order of Black>White>Hispanic race. Analysis of associated metadata revealed that potential confounders such as diabetes, obesity, age, gender or specimen handling and processing variables did not influence the distribution of samples across subtype classification. We report that normal human pancreatic acinar tissues group into molecular subtypes like those used to classify PDAC and that subtype assignment and ADM are influenced by the donor's race.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145727416","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 : 2025-12-09DOI: 10.1158/2767-9764.CRC-25-0696
Ana Carolina Monteiro, Diego D Garcia, Ana Paula A Fontão, Bárbara Du Rocher, Maria E Globa Masset, Isabella N Alves, Lucas Gonçalves Carvalho, Cecília Vianna de Andrade, Gabriel Fidalgo, Marcos Vinicius Colaço, Liebert Parreiras Nogueira, Maria de Fatima Dias Gaui, Adriana Bonomo
Bone metastases represent a common and devastating complication of breast cancer (BC); however, the immune determinants of skeletal colonization, particularly the role of adaptive lymphocyte subsets, have only recently been elucidated. Here, we identified a non-classical regulatory role of CD19⁺ B cells in the modulation of bone remodeling and BC metastatic progression. Using murine models of metastatic 4T1 and non-metastatic 67NR mammary tumors, we show that 67NR-bearing mice exhibit increased trabecular bone mass, associated with osteoprotegerin (OPG) secretion by CD19⁺IgD⁺IgM⁺CD138⁻ B cells. Functional assays demonstrated that 67NR-primed B cells suppressed bone resorption in vitro and counteracted 4T1-specific T cell-mediated osteolytic activity in vivo. Adoptive transfer of 67NR-primed CD19⁺ B cells into 4T1-bearing immunocompetent or immunodeficient hosts preserved bone architecture, reduced RANKL production, inhibited metastases, and limited tumor growth. These effects are T cell-independent at the effector phase but require T cell licensing for the acquisition of the OPG-producing phenotype. Silencing OPG abrogated this protective function. Mechanistically, B cells must be transferred during the early stages of tumor progression to retain their therapeutic potential. Molecular analyses supported the enrichment of RANKL⁺CD4⁺ T cells in tumors and lymph nodes, whereas OPG⁺ B cells were restricted to tumors. Transcriptomic data from The Cancer Genome Atlas (TCGA) further support the prognostic relevance of these immune phenotypes. In a retrospective human cohort, high RANKL⁺ lymphocyte infiltration in primary tumors correlated with bone metastases, whereas OPG⁺ infiltration predominated in bone metastasis-free cases. These findings uncover a B cell-driven axis that restrains osteolysis and tumor progression.
{"title":"OPG-producing B Cells and RANKL-expressing T cells define immune signatures predictive of bone metastases in breast cancer.","authors":"Ana Carolina Monteiro, Diego D Garcia, Ana Paula A Fontão, Bárbara Du Rocher, Maria E Globa Masset, Isabella N Alves, Lucas Gonçalves Carvalho, Cecília Vianna de Andrade, Gabriel Fidalgo, Marcos Vinicius Colaço, Liebert Parreiras Nogueira, Maria de Fatima Dias Gaui, Adriana Bonomo","doi":"10.1158/2767-9764.CRC-25-0696","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0696","url":null,"abstract":"<p><p>Bone metastases represent a common and devastating complication of breast cancer (BC); however, the immune determinants of skeletal colonization, particularly the role of adaptive lymphocyte subsets, have only recently been elucidated. Here, we identified a non-classical regulatory role of CD19⁺ B cells in the modulation of bone remodeling and BC metastatic progression. Using murine models of metastatic 4T1 and non-metastatic 67NR mammary tumors, we show that 67NR-bearing mice exhibit increased trabecular bone mass, associated with osteoprotegerin (OPG) secretion by CD19⁺IgD⁺IgM⁺CD138⁻ B cells. Functional assays demonstrated that 67NR-primed B cells suppressed bone resorption in vitro and counteracted 4T1-specific T cell-mediated osteolytic activity in vivo. Adoptive transfer of 67NR-primed CD19⁺ B cells into 4T1-bearing immunocompetent or immunodeficient hosts preserved bone architecture, reduced RANKL production, inhibited metastases, and limited tumor growth. These effects are T cell-independent at the effector phase but require T cell licensing for the acquisition of the OPG-producing phenotype. Silencing OPG abrogated this protective function. Mechanistically, B cells must be transferred during the early stages of tumor progression to retain their therapeutic potential. Molecular analyses supported the enrichment of RANKL⁺CD4⁺ T cells in tumors and lymph nodes, whereas OPG⁺ B cells were restricted to tumors. Transcriptomic data from The Cancer Genome Atlas (TCGA) further support the prognostic relevance of these immune phenotypes. In a retrospective human cohort, high RANKL⁺ lymphocyte infiltration in primary tumors correlated with bone metastases, whereas OPG⁺ infiltration predominated in bone metastasis-free cases. These findings uncover a B cell-driven axis that restrains osteolysis and tumor progression.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145709530","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 : 2025-12-08DOI: 10.1158/2767-9764.CRC-25-0249
Todor M Popov, Gergana S Stancheva, Veronika Petkova, Silva Kyurkchiyan, Stoyan Dimitrov, Lyuben Dimitrov, Boryana Ilcheva, Albena D Fakirova, Sylvia Skelina, Tzvetomir Marinov, Radka P Kaneva
The aim of this study was to test whether intratumoral heterogeneity explains inconsistent reports on VEGF-A as a prognostic marker in laryngeal squamous cell carcinoma (LSCC) and to identify expression phenotypes (mRNA/miRNA) associated with recurrence. In a prospective cohort of 60 T3/T4 LSCC patients undergoing primary laryngectomy, we sampled four regions per case (tumor surface, tumor depth, peritumoral mucosa ≤1 cm, paired distant normal mucosa). mRNA levels of HIF-1α/2α/3α, VEGF-A, VEGFR1/2, and ETS-1 were quantified by RT-qPCR; cohort-matched miRNA profiling (microarray with RT-qPCR validation) was integrated. Elevated VEGF-A at tumor depth predicted recurrence (log-rank p = 0.0001), while surface VEGF-A had no prognostic value (p = 0.170). Pairwise testing confirmed higher VEGF-A at depth versus surface (Wilcoxon p = 0.026). A subgroup with depth VEGF-A RQ > 2 and HIF-1α RQ < 2 showed a 64% recurrence rate. An independent subgroup defined by high miR-93-5p/miR-144-3p/miR-210-3p expression also had significantly worse outcomes. The two subgroups were non-overlapping in most patients and accounted for 76% of recurrences. The prognostic relevance of VEGF-A in LSCC is region-dependent, with clinically meaningful value confined to tumor depth. Together with a high-risk three-miRNA signature, these findings delineate two molecular subgroups capturing most recurrences and may inform sampling strategies and risk stratification.
{"title":"Two molecular subgroups predict most recurrences in advanced laryngeal squamous cell carcinoma.","authors":"Todor M Popov, Gergana S Stancheva, Veronika Petkova, Silva Kyurkchiyan, Stoyan Dimitrov, Lyuben Dimitrov, Boryana Ilcheva, Albena D Fakirova, Sylvia Skelina, Tzvetomir Marinov, Radka P Kaneva","doi":"10.1158/2767-9764.CRC-25-0249","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0249","url":null,"abstract":"<p><p>The aim of this study was to test whether intratumoral heterogeneity explains inconsistent reports on VEGF-A as a prognostic marker in laryngeal squamous cell carcinoma (LSCC) and to identify expression phenotypes (mRNA/miRNA) associated with recurrence. In a prospective cohort of 60 T3/T4 LSCC patients undergoing primary laryngectomy, we sampled four regions per case (tumor surface, tumor depth, peritumoral mucosa ≤1 cm, paired distant normal mucosa). mRNA levels of HIF-1α/2α/3α, VEGF-A, VEGFR1/2, and ETS-1 were quantified by RT-qPCR; cohort-matched miRNA profiling (microarray with RT-qPCR validation) was integrated. Elevated VEGF-A at tumor depth predicted recurrence (log-rank p = 0.0001), while surface VEGF-A had no prognostic value (p = 0.170). Pairwise testing confirmed higher VEGF-A at depth versus surface (Wilcoxon p = 0.026). A subgroup with depth VEGF-A RQ > 2 and HIF-1α RQ < 2 showed a 64% recurrence rate. An independent subgroup defined by high miR-93-5p/miR-144-3p/miR-210-3p expression also had significantly worse outcomes. The two subgroups were non-overlapping in most patients and accounted for 76% of recurrences. The prognostic relevance of VEGF-A in LSCC is region-dependent, with clinically meaningful value confined to tumor depth. Together with a high-risk three-miRNA signature, these findings delineate two molecular subgroups capturing most recurrences and may inform sampling strategies and risk stratification.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145709477","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 : 2025-12-08DOI: 10.1158/2767-9764.CRC-25-0445
Simon Cheah, Jared Burke, Fiona J Bruinsma, Melanie Evans, Helen Tsimiklis, Allison M Hodge, Brigid M Lynch, Graham G Giles, Rashmi Sinha, Melissa C Southey, Roger L Milne
Large prospective analyses of human gut microbiome profiles are needed to elucidate the role of microbiome variation in the development of disease. We conducted a pilot study to assess the feasibility of home faecal sample collection within a cohort study. A subset of cohort study participants was randomly selected and randomised into four groups defined by faecal sample collection method and questionnaire components. Of 1,093 invited participants, 610 (56%) opted in and, of those, 88% returned a sample. Of those asked to provide a faecal sample via faecal occult blood test card (FOBT) and complete a short "day of sample" questionnaire (dosQ), 49% returned a sample. Sample return was comparable for participants additionally asked to provide a sample via ethanol tube (51%), complete a food frequency questionnaire (48%), or complete both additional activities (49%). Whole genome sequencing and metagenomic analysis on paired FOBT and ethanol samples showed that both collection methods provided sufficient quality and quantity of DNA for downstream metagenomic analyses and displayed highly concordant microbiome profiles. Home faecal sample collection for microbiome analysis is feasible in a large prospective cohort. Including additional components did not reduce the likelihood of participants completing all requested items.
{"title":"Faecal sample collection for gut microbiome research in a prospective cohort: a pilot study within the Australian Breakthrough Cancer Study.","authors":"Simon Cheah, Jared Burke, Fiona J Bruinsma, Melanie Evans, Helen Tsimiklis, Allison M Hodge, Brigid M Lynch, Graham G Giles, Rashmi Sinha, Melissa C Southey, Roger L Milne","doi":"10.1158/2767-9764.CRC-25-0445","DOIUrl":"https://doi.org/10.1158/2767-9764.CRC-25-0445","url":null,"abstract":"<p><p>Large prospective analyses of human gut microbiome profiles are needed to elucidate the role of microbiome variation in the development of disease. We conducted a pilot study to assess the feasibility of home faecal sample collection within a cohort study. A subset of cohort study participants was randomly selected and randomised into four groups defined by faecal sample collection method and questionnaire components. Of 1,093 invited participants, 610 (56%) opted in and, of those, 88% returned a sample. Of those asked to provide a faecal sample via faecal occult blood test card (FOBT) and complete a short \"day of sample\" questionnaire (dosQ), 49% returned a sample. Sample return was comparable for participants additionally asked to provide a sample via ethanol tube (51%), complete a food frequency questionnaire (48%), or complete both additional activities (49%). Whole genome sequencing and metagenomic analysis on paired FOBT and ethanol samples showed that both collection methods provided sufficient quality and quantity of DNA for downstream metagenomic analyses and displayed highly concordant microbiome profiles. Home faecal sample collection for microbiome analysis is feasible in a large prospective cohort. Including additional components did not reduce the likelihood of participants completing all requested items.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145703184","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}