Pub Date : 2025-12-01Epub Date: 2025-06-04DOI: 10.1002/1878-0261.70063
Jenifer Brea-Iglesias, Ana Oitabén, Sonia Zumalave, Bernardo Rodriguez-Martin, María Gallardo-Gómez, Martín Santamarina, Ana Pequeño-Valtierra, Laura Juaneda-Magdalena, Ramón García-Escudero, José Luis López-Cedrún, Máximo Fraga, José M C Tubio, Mónica Martínez-Fernández
The relevant role of LINE-1 (L1) retrotransposition in cancer has been recurrently demonstrated in recent years. However, the repetitive nature of retrotransposons makes their identification and detection inaccessible for clinical practice. Also, its clinical relevance for cancer patients is still limited. Here, we developed RetroTest, a new efficient method to quantify L1 activation based on targeted sequencing and a sophisticated bioinformatic pipeline, allowing its application in tumor biopsies. Firstly, we performed RetroTest benchmarking to confirm its high specificity and reliability. Then, we unraveled L1 activation in head-and-neck squamous cell carcinoma (HNSCC) according to an extensive patient cohort including all tumor stages. L1 retrotransposition estimation revealed a surprisingly early activation in HNSCC progression, contrary to its classical association with advanced stages. In addition, L1 activation together with genomic mutational profiling in normal adjacent tissues supported a field cancerization process, a phenomenon where a tissue develops multiple patches of cells with genetic and/or epigenetic alterations, increasing the risk of cancer development in that area. Overall, our results underline an early L1 activation in HNSCC and field characterization, raising L1 as a promising early diagnostic biomarker and supporting the importance of estimating L1 retrotransposition in clinical practice toward a more efficient diagnosis in HNSCC.
{"title":"Unraveling LINE-1 retrotransposition in head and neck squamous cell carcinoma.","authors":"Jenifer Brea-Iglesias, Ana Oitabén, Sonia Zumalave, Bernardo Rodriguez-Martin, María Gallardo-Gómez, Martín Santamarina, Ana Pequeño-Valtierra, Laura Juaneda-Magdalena, Ramón García-Escudero, José Luis López-Cedrún, Máximo Fraga, José M C Tubio, Mónica Martínez-Fernández","doi":"10.1002/1878-0261.70063","DOIUrl":"10.1002/1878-0261.70063","url":null,"abstract":"<p><p>The relevant role of LINE-1 (L1) retrotransposition in cancer has been recurrently demonstrated in recent years. However, the repetitive nature of retrotransposons makes their identification and detection inaccessible for clinical practice. Also, its clinical relevance for cancer patients is still limited. Here, we developed RetroTest, a new efficient method to quantify L1 activation based on targeted sequencing and a sophisticated bioinformatic pipeline, allowing its application in tumor biopsies. Firstly, we performed RetroTest benchmarking to confirm its high specificity and reliability. Then, we unraveled L1 activation in head-and-neck squamous cell carcinoma (HNSCC) according to an extensive patient cohort including all tumor stages. L1 retrotransposition estimation revealed a surprisingly early activation in HNSCC progression, contrary to its classical association with advanced stages. In addition, L1 activation together with genomic mutational profiling in normal adjacent tissues supported a field cancerization process, a phenomenon where a tissue develops multiple patches of cells with genetic and/or epigenetic alterations, increasing the risk of cancer development in that area. Overall, our results underline an early L1 activation in HNSCC and field characterization, raising L1 as a promising early diagnostic biomarker and supporting the importance of estimating L1 retrotransposition in clinical practice toward a more efficient diagnosis in HNSCC.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3769-3783"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688165/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144225976","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-14DOI: 10.1002/1878-0261.70162
{"title":"Correction to 'Inhibition of autophagy as a novel treatment for neurofibromatosis type 1 tumors'.","authors":"","doi":"10.1002/1878-0261.70162","DOIUrl":"10.1002/1878-0261.70162","url":null,"abstract":"","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3821"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688160/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145523964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Eleonora Piumatti, Giovanni Germano, Pietro Paolo Vitiello, Alberto Bardelli
Mismatch repair-proficient (pMMR) colorectal cancers (CRC) have long been considered nonresponsive to immune checkpoint blockade (ICB), in contrast to their mismatch repair-deficient (dMMR) counterparts. Recent evidence indicates that neoadjuvant immunotherapy can be used to treat pMMR CRC before surgery, potentially reducing postoperative relapse. Tan et al. report results from the NICHE-2 trial, which achieved a 26% response rate in early-stage pMMR colon cancer (CC) patients. Molecular studies show that despite low tumor mutational burden (TMB), responders exhibit higher chromosomal instability (CIN), TP53 mutations, and enrichment of proliferative and cell-cycle signatures, associated with higher density of Ki-67+ tumor and CD8+ T cells. In contrast, nonresponders display metabolic and stromal reprogramming, enhanced TGF-β signaling, and immune exclusion. Circulating tumor DNA (ctDNA) clearance correlated with pathological response and long-term disease-free survival postsurgery. While the biological and molecular determinants underlying the response rates observed in the NICHE-2 trial remain to be fully elucidated, the work by Tan et al. suggests that biomarker-guided neoadjuvant immunotherapy could represent a valuable strategy to achieve pathological responses in early-stage pMMR CC, despite its clinical relevance requiring further evaluation.
{"title":"A subset of MMR-proficient colon cancers responds to neoadjuvant immunotherapy.","authors":"Eleonora Piumatti, Giovanni Germano, Pietro Paolo Vitiello, Alberto Bardelli","doi":"10.1002/1878-0261.70178","DOIUrl":"https://doi.org/10.1002/1878-0261.70178","url":null,"abstract":"<p><p>Mismatch repair-proficient (pMMR) colorectal cancers (CRC) have long been considered nonresponsive to immune checkpoint blockade (ICB), in contrast to their mismatch repair-deficient (dMMR) counterparts. Recent evidence indicates that neoadjuvant immunotherapy can be used to treat pMMR CRC before surgery, potentially reducing postoperative relapse. Tan et al. report results from the NICHE-2 trial, which achieved a 26% response rate in early-stage pMMR colon cancer (CC) patients. Molecular studies show that despite low tumor mutational burden (TMB), responders exhibit higher chromosomal instability (CIN), TP53 mutations, and enrichment of proliferative and cell-cycle signatures, associated with higher density of Ki-67<sup>+</sup> tumor and CD8<sup>+</sup> T cells. In contrast, nonresponders display metabolic and stromal reprogramming, enhanced TGF-β signaling, and immune exclusion. Circulating tumor DNA (ctDNA) clearance correlated with pathological response and long-term disease-free survival postsurgery. While the biological and molecular determinants underlying the response rates observed in the NICHE-2 trial remain to be fully elucidated, the work by Tan et al. suggests that biomarker-guided neoadjuvant immunotherapy could represent a valuable strategy to achieve pathological responses in early-stage pMMR CC, despite its clinical relevance requiring further evaluation.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145649074","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lucia Cottone, James Dunford, Eleanor Calcutt, Vicki Gamble, Filiz Senbabaoglu Aksu, Lorena Ligammari, Georgina Wherry, Giorgia Gaeta, John C Christianson, Adrienne M Flanagan, Udo Oppermann, Adam P Cribbs
Chordoma, a rare primary bone malignancy, currently lacks effective targeted therapies. Despite surgical resection and adjuvant radiotherapy, prognosis remains poor. Recent preclinical studies have highlighted potential therapeutic targets, including the transcription factor T-box transcription factor T (TBXT). However, clinical outcomes associated with therapies targeting TBXT remain underexplored or have been modest, warranting further investigation. In this study, we investigated the therapeutic potential of transfer RNA (tRNA) synthetase inhibitors in chordoma treatment. Focused compound screening identified distinct chemotypes targeting human glutamyl-prolyl-tRNA synthetase (EPRS) as being effective in reducing cell viability in chordoma cell lines through a cyclic AMP-dependent transcription factor (ATF4)-mediated stress response rather than through TBXT regulation. Mechanistically significant upregulation of ATF4 and associated stress response genes was identified with consecutive pro-apoptotic DNA damage-inducible transcript 3 protein (DDIT3)-mediated cell death. The prototypic EPRS inhibitor halofuginone demonstrated significant tumour growth inhibition in an in vivo patient-derived xenograft model. These results suggest that targeting metabolic stress pathways via ATF4 activation presents a novel therapeutic approach for chordoma, warranting further clinical investigation.
{"title":"ATF4-mediated stress response as a therapeutic vulnerability in chordoma.","authors":"Lucia Cottone, James Dunford, Eleanor Calcutt, Vicki Gamble, Filiz Senbabaoglu Aksu, Lorena Ligammari, Georgina Wherry, Giorgia Gaeta, John C Christianson, Adrienne M Flanagan, Udo Oppermann, Adam P Cribbs","doi":"10.1002/1878-0261.70176","DOIUrl":"https://doi.org/10.1002/1878-0261.70176","url":null,"abstract":"<p><p>Chordoma, a rare primary bone malignancy, currently lacks effective targeted therapies. Despite surgical resection and adjuvant radiotherapy, prognosis remains poor. Recent preclinical studies have highlighted potential therapeutic targets, including the transcription factor T-box transcription factor T (TBXT). However, clinical outcomes associated with therapies targeting TBXT remain underexplored or have been modest, warranting further investigation. In this study, we investigated the therapeutic potential of transfer RNA (tRNA) synthetase inhibitors in chordoma treatment. Focused compound screening identified distinct chemotypes targeting human glutamyl-prolyl-tRNA synthetase (EPRS) as being effective in reducing cell viability in chordoma cell lines through a cyclic AMP-dependent transcription factor (ATF4)-mediated stress response rather than through TBXT regulation. Mechanistically significant upregulation of ATF4 and associated stress response genes was identified with consecutive pro-apoptotic DNA damage-inducible transcript 3 protein (DDIT3)-mediated cell death. The prototypic EPRS inhibitor halofuginone demonstrated significant tumour growth inhibition in an in vivo patient-derived xenograft model. These results suggest that targeting metabolic stress pathways via ATF4 activation presents a novel therapeutic approach for chordoma, warranting further clinical investigation.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145636117","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kristin Løge Aanestad, Marie Austdal, Oddmund Nordgård, Gunnar Mellgren, Satu Oltedal, Marie L Austbø, Ylva H Vignes, Thomas Helland, Kristin Jonsdottir, Tone H Lende, Emilius A M Janssen, Bjørnar Gilje, Kjersti Tjensvoll
Breast cancer is known for late recurrences, yet current follow-up lacks radiological or blood-based monitoring for systemic relapse. This study evaluated circulating tumor DNA (ctDNA) monitoring for early detection of systemic relapse after curative treatment. In this case-control study of 70 patients with operable breast cancer (35 with relapse and 35 without relapse), blood samples were collected every 6-12 months during a median 8.3-year follow-up. ctDNA was analyzed by targeted DNA sequencing using Oncomine™ Breast cfDNA Research Assay v2, and results were compared to genetic analysis of tumor and metastasis biopsies. ctDNA was detected at relapse in 19 of 35 (54%) patients with disease relapse and preceded clinical or radiological relapse detection in 17, with a median lead time of 10.3 months. In 13 (68%) patients, there was concordance with tumor mutations, and in seven patients, there was also concordance with metastasis. Among the relapse-free patients, seven were ctDNA-positive postsurgery, and only one of them had a match among the tumor variants. These findings suggest serial ctDNA analysis may enable earlier detection of systemic relapse in patients with operable breast cancer.
乳腺癌以晚期复发而闻名,但目前的随访缺乏对全身复发的放射学或血液监测。本研究评估了循环肿瘤DNA (ctDNA)监测对治愈治疗后全身复发的早期检测。在这项70例可手术乳腺癌患者(35例复发和35例未复发)的病例对照研究中,每6-12个月采集一次血液样本,中位随访8.3年。使用Oncomine™Breast cfDNA Research Assay v2进行靶向DNA测序,分析ctDNA,并将结果与肿瘤和转移活检的遗传分析进行比较。35例疾病复发患者中有19例(54%)在复发时检测到ctDNA, 17例在临床或放射学复发前检测到ctDNA,中位提前期为10.3个月。13例(68%)患者与肿瘤突变一致,7例患者与转移一致。在无复发患者中,7例术后ctdna阳性,其中只有1例与肿瘤变异相匹配。这些发现表明,序列ctDNA分析可能有助于早期发现可手术乳腺癌患者的全身复发。
{"title":"Monitoring of circulating tumor DNA allows early detection of disease relapse in patients with operable breast cancer.","authors":"Kristin Løge Aanestad, Marie Austdal, Oddmund Nordgård, Gunnar Mellgren, Satu Oltedal, Marie L Austbø, Ylva H Vignes, Thomas Helland, Kristin Jonsdottir, Tone H Lende, Emilius A M Janssen, Bjørnar Gilje, Kjersti Tjensvoll","doi":"10.1002/1878-0261.70170","DOIUrl":"https://doi.org/10.1002/1878-0261.70170","url":null,"abstract":"<p><p>Breast cancer is known for late recurrences, yet current follow-up lacks radiological or blood-based monitoring for systemic relapse. This study evaluated circulating tumor DNA (ctDNA) monitoring for early detection of systemic relapse after curative treatment. In this case-control study of 70 patients with operable breast cancer (35 with relapse and 35 without relapse), blood samples were collected every 6-12 months during a median 8.3-year follow-up. ctDNA was analyzed by targeted DNA sequencing using Oncomine™ Breast cfDNA Research Assay v2, and results were compared to genetic analysis of tumor and metastasis biopsies. ctDNA was detected at relapse in 19 of 35 (54%) patients with disease relapse and preceded clinical or radiological relapse detection in 17, with a median lead time of 10.3 months. In 13 (68%) patients, there was concordance with tumor mutations, and in seven patients, there was also concordance with metastasis. Among the relapse-free patients, seven were ctDNA-positive postsurgery, and only one of them had a match among the tumor variants. These findings suggest serial ctDNA analysis may enable earlier detection of systemic relapse in patients with operable breast cancer.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145636161","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Simon Broad, Mahmood Hachim, Tom Bertin, Karolina Penderecka, Rifat Hamoudi, Saif Khan, Rui Henrique, Natalie Bergmoser, Manit Arya, Kalle Sipila, Matteo Vietri Rudan, Asif Muneer, Aamir Ahmed
Penile carcinoma is a rare malignancy in developed countries but is more common in South America and East Africa. The small number of cases means there are limited resources to investigate disease pathogenesis. This report describes a method of generating primary cell lines from freshly isolated human penile tissue using a clonal expansion approach on mitotically inactivated fibroblasts. Matched normal and penile cancer cell lines from two patients were generated and characterised. Molecular karyotyping and targeted sequencing were performed to compare their genomic landscape. Gains in 8q13.3, 10q23.2, 10q25.1, 10q26.13,12p13.33, 20q13.33, Xq21.1 or losses in Yq11.23 were consistent in both tumour cell lines. Gains in 8q13.3 and Xq21.1 cytobands positively correlated with changes in the expression of nearby genes. The top 20 differentially expressed genes are involved in immune responses like interferon alpha/beta signalling. Additionally, there was an increase in integrin β1, transglutaminase 1, keratins 5, 10, 14 and 16, and a decrease in involucrin protein expression. The cell lines described in this study can provide an invaluable platform for new insights and testing of therapies for penile carcinoma.
{"title":"Molecular characterisation of human penile carcinoma and generation of paired epithelial primary cell lines.","authors":"Simon Broad, Mahmood Hachim, Tom Bertin, Karolina Penderecka, Rifat Hamoudi, Saif Khan, Rui Henrique, Natalie Bergmoser, Manit Arya, Kalle Sipila, Matteo Vietri Rudan, Asif Muneer, Aamir Ahmed","doi":"10.1002/1878-0261.70156","DOIUrl":"https://doi.org/10.1002/1878-0261.70156","url":null,"abstract":"<p><p>Penile carcinoma is a rare malignancy in developed countries but is more common in South America and East Africa. The small number of cases means there are limited resources to investigate disease pathogenesis. This report describes a method of generating primary cell lines from freshly isolated human penile tissue using a clonal expansion approach on mitotically inactivated fibroblasts. Matched normal and penile cancer cell lines from two patients were generated and characterised. Molecular karyotyping and targeted sequencing were performed to compare their genomic landscape. Gains in 8q13.3, 10q23.2, 10q25.1, 10q26.13,12p13.33, 20q13.33, Xq21.1 or losses in Yq11.23 were consistent in both tumour cell lines. Gains in 8q13.3 and Xq21.1 cytobands positively correlated with changes in the expression of nearby genes. The top 20 differentially expressed genes are involved in immune responses like interferon alpha/beta signalling. Additionally, there was an increase in integrin β1, transglutaminase 1, keratins 5, 10, 14 and 16, and a decrease in involucrin protein expression. The cell lines described in this study can provide an invaluable platform for new insights and testing of therapies for penile carcinoma.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145605123","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Elena Benidovskaya, Joséphine Deneft, Marc Van den Eynde
Immunotherapy has revolutionized cancer treatment; yet, a subset of patients with microsatellite instability-high (MSI-H) tumors fails to respond to treatment despite their elevated tumor mutational burden and immunogenic potential. In a recent study, Xu et al. uncover a key mechanism of immune evasion in MSI-H tumors mediated by the exonuclease TREX1, which degrades cytosolic DNA and suppresses activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING)-type I interferon pathway. Loss of TREX1 restores cytosolic DNA sensing, promotes CD8+ T and NK cell infiltration, and enhances antitumor immunity. These findings highlight TREX1 as a potential therapeutic target to overcome resistance to immune checkpoint blockade.
{"title":"TREX1, a predator for treating MSI-H tumors?","authors":"Elena Benidovskaya, Joséphine Deneft, Marc Van den Eynde","doi":"10.1002/1878-0261.70173","DOIUrl":"https://doi.org/10.1002/1878-0261.70173","url":null,"abstract":"<p><p>Immunotherapy has revolutionized cancer treatment; yet, a subset of patients with microsatellite instability-high (MSI-H) tumors fails to respond to treatment despite their elevated tumor mutational burden and immunogenic potential. In a recent study, Xu et al. uncover a key mechanism of immune evasion in MSI-H tumors mediated by the exonuclease TREX1, which degrades cytosolic DNA and suppresses activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING)-type I interferon pathway. Loss of TREX1 restores cytosolic DNA sensing, promotes CD8<sup>+</sup> T and NK cell infiltration, and enhances antitumor immunity. These findings highlight TREX1 as a potential therapeutic target to overcome resistance to immune checkpoint blockade.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145605066","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Christoffer Trier Maansson, Anders Lade Nielsen, Boe Sandahl Sorensen
Liquid biopsies containing circulating tumor DNA (ctDNA) are important biomarkers across several forms of cancer. The detection of mutations in cell-free DNA (cfDNA) indicates the presence of ctDNA. However, unsatisfactory ctDNA mutation sensitivities, issues with sequencing errors, and clonal hematopoiesis variants have limited the clinical utility of mutation-based ctDNA assays. Recently, a new avenue of cfDNA assays has been developed, focusing on cfDNA epigenetics. Here, we outline the recent advancements in cfDNA epigenetics, focusing on cfDNA methylation, fragmentomics, and post-translational modifications (PTMs) of circulating nucleosomes. We present various methylation strategies concerning ctDNA detection and tissue of origin (TOO) analyses. cfDNA fragmentomics focuses on cfDNA fragment lengths, fragment end motifs, and nucleosome positioning to infer gene expression and estimate the ctDNA fraction. Lastly, we discuss the development of cell-free chromatin immunoprecipitation of circulating nucleosomes with PTMs. This method has been implemented to detect tumor gene expression, TOO, and treatment resistance. Combining the epigenetic features of cfDNA will expand the utility of liquid biopsies to give a more comprehensive insight into tumor biology, treatment response, and resistance.
{"title":"Liquid biopsy epigenetics: establishing a molecular profile based on cell-free DNA.","authors":"Christoffer Trier Maansson, Anders Lade Nielsen, Boe Sandahl Sorensen","doi":"10.1002/1878-0261.70145","DOIUrl":"https://doi.org/10.1002/1878-0261.70145","url":null,"abstract":"<p><p>Liquid biopsies containing circulating tumor DNA (ctDNA) are important biomarkers across several forms of cancer. The detection of mutations in cell-free DNA (cfDNA) indicates the presence of ctDNA. However, unsatisfactory ctDNA mutation sensitivities, issues with sequencing errors, and clonal hematopoiesis variants have limited the clinical utility of mutation-based ctDNA assays. Recently, a new avenue of cfDNA assays has been developed, focusing on cfDNA epigenetics. Here, we outline the recent advancements in cfDNA epigenetics, focusing on cfDNA methylation, fragmentomics, and post-translational modifications (PTMs) of circulating nucleosomes. We present various methylation strategies concerning ctDNA detection and tissue of origin (TOO) analyses. cfDNA fragmentomics focuses on cfDNA fragment lengths, fragment end motifs, and nucleosome positioning to infer gene expression and estimate the ctDNA fraction. Lastly, we discuss the development of cell-free chromatin immunoprecipitation of circulating nucleosomes with PTMs. This method has been implemented to detect tumor gene expression, TOO, and treatment resistance. Combining the epigenetic features of cfDNA will expand the utility of liquid biopsies to give a more comprehensive insight into tumor biology, treatment response, and resistance.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145564644","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Megha Bhardwaj, Clara Frick, Ben Schöttker, Bernd Holleczek, Hermann Brenner
Screening heavy smokers by low-dose computed tomography (LDCT) can reduce lung cancer (LC) mortality, but defining the population that benefits most, a prerequisite for cost-effective screening, is challenging. In order to contribute to a more nuanced risk stratification of high-risk target populations, we developed and validated a blood-based protein marker model for LC. A two-stage design was implemented in this study, and the derivation set comprised 18 868 participants from the UK Biobank, which included 200 incident LC cases identified at 6 years of follow-up. The independent validation set included 101 LC cases identified at 6 years of follow-up. A total of 2025 protein markers measured by proximity extension assays available for both datasets were used for analysis. A risk prediction algorithm by least absolute shrinkage and selection operator regression with bootstrap method was developed in the derivation set and then externally evaluated in the independent validation set. The risk discriminatory performance of the protein marker model was compared with the established PLCOm2012 model, USPSTF 2020 guidelines and trial criteria used in different LDCT trials. The protein marker model comprising of four protein biomarkers-CEACAM5, CXCL17, MMP12, and WFDC2-outperformed the PLCOm2012 model, and the areas under the receiver operating curve (AUCs) for the protein marker model in the derivation and validation sets were 0.814 [95% confidence interval (95% CI), 0.785-0.843] and 0.814 (95% CI, 0.756-0.873), respectively. The addition of the protein marker model to the PLCOm2012 model increased the AUCs up to 0.056 and 0.057 and yielded up to 16 and 12 percentage points higher sensitivities to identify future LC cases compared to the LDCT trial criteria, in the derivation and validation sets, respectively. The protein marker model improves the selection of high LC risk individuals for LDCT screening and thereby enhances screening efficacy.
{"title":"Next-generation proteomics improves lung cancer risk prediction.","authors":"Megha Bhardwaj, Clara Frick, Ben Schöttker, Bernd Holleczek, Hermann Brenner","doi":"10.1002/1878-0261.70166","DOIUrl":"https://doi.org/10.1002/1878-0261.70166","url":null,"abstract":"<p><p>Screening heavy smokers by low-dose computed tomography (LDCT) can reduce lung cancer (LC) mortality, but defining the population that benefits most, a prerequisite for cost-effective screening, is challenging. In order to contribute to a more nuanced risk stratification of high-risk target populations, we developed and validated a blood-based protein marker model for LC. A two-stage design was implemented in this study, and the derivation set comprised 18 868 participants from the UK Biobank, which included 200 incident LC cases identified at 6 years of follow-up. The independent validation set included 101 LC cases identified at 6 years of follow-up. A total of 2025 protein markers measured by proximity extension assays available for both datasets were used for analysis. A risk prediction algorithm by least absolute shrinkage and selection operator regression with bootstrap method was developed in the derivation set and then externally evaluated in the independent validation set. The risk discriminatory performance of the protein marker model was compared with the established PLCO<sub>m2012</sub> model, USPSTF 2020 guidelines and trial criteria used in different LDCT trials. The protein marker model comprising of four protein biomarkers-CEACAM5, CXCL17, MMP12, and WFDC2-outperformed the PLCO<sub>m2012</sub> model, and the areas under the receiver operating curve (AUCs) for the protein marker model in the derivation and validation sets were 0.814 [95% confidence interval (95% CI), 0.785-0.843] and 0.814 (95% CI, 0.756-0.873), respectively. The addition of the protein marker model to the PLCO<sub>m2012</sub> model increased the AUCs up to 0.056 and 0.057 and yielded up to 16 and 12 percentage points higher sensitivities to identify future LC cases compared to the LDCT trial criteria, in the derivation and validation sets, respectively. The protein marker model improves the selection of high LC risk individuals for LDCT screening and thereby enhances screening efficacy.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145564662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mangesh A Thorat, Marc Arbyn, David Baldwin, Xavier Castells, Solveig Hofvind, Urska Ivanus, Carlo Senore, Esther Toes-Zoutendijk, Carlijn van der Aalst, Carlos Canelo-Aybar, Fiorella Karina Fernández-Sáenz, Ariadna Feliu, Hajo Zeeb, Andre L Carvalho, Erica D'Souza, David Ritchie, Carolina Espina, Iris Lansdorp-Vogelaar, Andrea DeCensi
Several medical therapies modify the risk of developing certain cancers in an individual. The aim of this paper was to provide the scientific justification for the 5th edition of the European Code Against Cancer (ECAC5) recommendation on the use of hormone replacement therapy (HRT) and other drugs used at population scale, such as hormonal contraceptives and aspirin. HRT modifies the risk of developing certain cancers in an individual. Except for vaginal oestrogens, all forms of HRT are associated with an increased breast cancer risk; the risk of serous ovarian cancer and endometrial cancer may also be increased. Despite such an increase in cancer risk, HRT often remains the only option for the management of certain menopausal symptoms for the restoration of quality of life and mental health. Therefore, the ECAC5 recommends using HRT for bothersome menopausal symptoms only after a thorough discussion with a healthcare professional and limiting its use for as short a duration as possible. On review of up-to-date evidence for hormonal contraceptives and aspirin, the ECAC5 Working Group elected not to make a recommendation for the average-risk general population regarding the use of these therapies.
{"title":"European Code Against Cancer, 5th edition - hormone replacement therapy, other common medical therapies and cancer.","authors":"Mangesh A Thorat, Marc Arbyn, David Baldwin, Xavier Castells, Solveig Hofvind, Urska Ivanus, Carlo Senore, Esther Toes-Zoutendijk, Carlijn van der Aalst, Carlos Canelo-Aybar, Fiorella Karina Fernández-Sáenz, Ariadna Feliu, Hajo Zeeb, Andre L Carvalho, Erica D'Souza, David Ritchie, Carolina Espina, Iris Lansdorp-Vogelaar, Andrea DeCensi","doi":"10.1002/1878-0261.70158","DOIUrl":"https://doi.org/10.1002/1878-0261.70158","url":null,"abstract":"<p><p>Several medical therapies modify the risk of developing certain cancers in an individual. The aim of this paper was to provide the scientific justification for the 5th edition of the European Code Against Cancer (ECAC5) recommendation on the use of hormone replacement therapy (HRT) and other drugs used at population scale, such as hormonal contraceptives and aspirin. HRT modifies the risk of developing certain cancers in an individual. Except for vaginal oestrogens, all forms of HRT are associated with an increased breast cancer risk; the risk of serous ovarian cancer and endometrial cancer may also be increased. Despite such an increase in cancer risk, HRT often remains the only option for the management of certain menopausal symptoms for the restoration of quality of life and mental health. Therefore, the ECAC5 recommends using HRT for bothersome menopausal symptoms only after a thorough discussion with a healthcare professional and limiting its use for as short a duration as possible. On review of up-to-date evidence for hormonal contraceptives and aspirin, the ECAC5 Working Group elected not to make a recommendation for the average-risk general population regarding the use of these therapies.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145540541","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}