Pub Date : 2025-12-01Epub Date: 2025-09-03DOI: 10.1002/1878-0261.70121
Johan Staaf, Daniel Ehinger, Hans Brunnström, Mats Jönsson, Frida Rosengren, Marija Kotevska, Anna Karlsson, Mattias Aine, Christian Frezza, Maria Planck, Elsa Arbajian
Squamous cell lung carcinoma (SqCC) is the second most common histological subtype of lung cancer. Besides tumor-initiating and promoting DNA, RNA, and epigenetic alterations, aberrant cell metabolism is a hallmark of carcinogenesis. This study aimed to identify SqCC-specific key regulators that could eventually be used as new anticancer targets. Transcriptional and metabolomic data were gathered for a cohort of resected lung cancers. SqCC-specific differentially expressed genes were integrated with metabolic data. Findings were validated in cohorts of tumors, normal specimens, and cell lines. In situ protein expression of SLC6A8 was investigated. Differential gene expression analysis identified a subset of SqCC-specific genes with metabolic functions through the Reactome database, and/or correlated to specific metabolites through GEMs models. Metabolic profiling identified seven SqCC-specific metabolites, of which increased creatine levels, in particular, matched to SqCC-specific expression of SLC6A8. Expression of the gene appeared tumor cell-associated. Elevated creatine levels and overexpression of its transporter SLC6A8 appear a distinct metabolic feature of SqCC. Considering ongoing clinical trials in other malignancies, exploring SLC6A8 inhibition in SqCC appears motivated based on a metabolic addiction hypothesis.
{"title":"Multi-omic profiling of squamous cell lung cancer identifies metabolites and related genes associated with squamous cell carcinoma.","authors":"Johan Staaf, Daniel Ehinger, Hans Brunnström, Mats Jönsson, Frida Rosengren, Marija Kotevska, Anna Karlsson, Mattias Aine, Christian Frezza, Maria Planck, Elsa Arbajian","doi":"10.1002/1878-0261.70121","DOIUrl":"10.1002/1878-0261.70121","url":null,"abstract":"<p><p>Squamous cell lung carcinoma (SqCC) is the second most common histological subtype of lung cancer. Besides tumor-initiating and promoting DNA, RNA, and epigenetic alterations, aberrant cell metabolism is a hallmark of carcinogenesis. This study aimed to identify SqCC-specific key regulators that could eventually be used as new anticancer targets. Transcriptional and metabolomic data were gathered for a cohort of resected lung cancers. SqCC-specific differentially expressed genes were integrated with metabolic data. Findings were validated in cohorts of tumors, normal specimens, and cell lines. In situ protein expression of SLC6A8 was investigated. Differential gene expression analysis identified a subset of SqCC-specific genes with metabolic functions through the Reactome database, and/or correlated to specific metabolites through GEMs models. Metabolic profiling identified seven SqCC-specific metabolites, of which increased creatine levels, in particular, matched to SqCC-specific expression of SLC6A8. Expression of the gene appeared tumor cell-associated. Elevated creatine levels and overexpression of its transporter SLC6A8 appear a distinct metabolic feature of SqCC. Considering ongoing clinical trials in other malignancies, exploring SLC6A8 inhibition in SqCC appears motivated based on a metabolic addiction hypothesis.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3806-3820"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688162/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993011","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: 2025-07-11DOI: 10.1002/1878-0261.70023
Livia Concetti, Manuel Scimeca, Julia Bischof, Jonathan Woodsmith, Massimiliano Agostini, Cristina Fiorani, Yufang Shi, Eleonora Candi, Gerry Melino, Alessandro Mauriello, Giuseppe S Sica
Colorectal cancer (CRC) is a heterogenous disease with distinct biological and clinical subgroups, each with different prognoses and responses to therapy. In this case report, taking inspiration from a case of locally advanced CRC with serine/threonine-protein kinase B-raf (BRAF) V600E mutation, we highlight an atypical consensus molecular subtype 1 (CMS1). Deep multi-omic analyses showed a limited expression of programmed cell death protein 1 (PD-1) and reduced T-cell infiltration, including CD8+ and natural killer (NK) cells, in the analyzed CMS1 tumor. In parallel, a reduced activation of the JAK/STAT pathway was detected, suggesting a lack of clinical response to immunotherapy with checkpoint inhibitors. Furthermore, the finding of up-regulated expression of WEE1 G2 checkpoint kinase (WEE1), checkpoint kinase 1 (CHK1), and checkpoint kinase 2 (CHK2), poly(ADP-ribose) polymerase (PARP), and heat shock protein 90 (HSP90) suggests a potential alternative therapeutic approach using inhibitors of the cell cycle, HSP90, or PARP in combination with conventional chemotherapy, targeted agents, or immunotherapy. This paradigmatic case should stimulate a regular deep omics analysis to improve precision medicine. We therefore suggest that full mutational and expression profiling analyses of CRC subtypes should be undertaken to improve therapeutic strategies in CRC treatment.
{"title":"Multi-omic characterization of consensus molecular subtype 1 (CMS1) colorectal cancer with dampened immune response improves precision medicine.","authors":"Livia Concetti, Manuel Scimeca, Julia Bischof, Jonathan Woodsmith, Massimiliano Agostini, Cristina Fiorani, Yufang Shi, Eleonora Candi, Gerry Melino, Alessandro Mauriello, Giuseppe S Sica","doi":"10.1002/1878-0261.70023","DOIUrl":"10.1002/1878-0261.70023","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is a heterogenous disease with distinct biological and clinical subgroups, each with different prognoses and responses to therapy. In this case report, taking inspiration from a case of locally advanced CRC with serine/threonine-protein kinase B-raf (BRAF) V600E mutation, we highlight an atypical consensus molecular subtype 1 (CMS1). Deep multi-omic analyses showed a limited expression of programmed cell death protein 1 (PD-1) and reduced T-cell infiltration, including CD8<sup>+</sup> and natural killer (NK) cells, in the analyzed CMS1 tumor. In parallel, a reduced activation of the JAK/STAT pathway was detected, suggesting a lack of clinical response to immunotherapy with checkpoint inhibitors. Furthermore, the finding of up-regulated expression of WEE1 G2 checkpoint kinase (WEE1), checkpoint kinase 1 (CHK1), and checkpoint kinase 2 (CHK2), poly(ADP-ribose) polymerase (PARP), and heat shock protein 90 (HSP90) suggests a potential alternative therapeutic approach using inhibitors of the cell cycle, HSP90, or PARP in combination with conventional chemotherapy, targeted agents, or immunotherapy. This paradigmatic case should stimulate a regular deep omics analysis to improve precision medicine. We therefore suggest that full mutational and expression profiling analyses of CRC subtypes should be undertaken to improve therapeutic strategies in CRC treatment.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3486-3498"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688173/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144961673","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: 2025-04-01DOI: 10.1002/1878-0261.70019
Rebecca M Schlösser, Florian Krumbach, Eyleen Corrales, Geoffroy Andrieux, Christian Preisinger, Franziska Liss, Alexandra Golzmann, Melanie Boerries, Kerstin Becker, Ruth Knüchel, Stefan Garczyk, Bernhard Lüscher
Epigenetic regulators, such as the SWI/SNF complex, with important roles in tissue development and homeostasis, are frequently mutated in cancer. ARID1A, a subunit of the SWI/SNF complex, is mutated in approximately 20% of all bladder tumors; however, the consequences of this remain poorly understood. Finding truncations to be the most common mutation, we generated loss- and gain-of-function models to conduct RNA-Seq, interactome analyses, Omni-ATAC-Seq, and functional studies to characterize ARID1A-affected pathways potentially suitable for the treatment of ARID1A-deficient bladder cancers. We observed decreased cell proliferation and deregulation of stress-regulated pathways, including DNA repair, in ARID1A-deficient cells. Furthermore, ARID1A was linked to alternative splicing and translational regulation on RNA and interactome levels. ARID1A deficiency drastically reduced the accessibility of chromatin, especially around introns and distal enhancers, in a functional enrichment analysis. Less accessible chromatin areas were mapped to pathways such as cell proliferation and DNA damage response. Indeed, the G2/M checkpoint appeared impaired after DNA damage in ARID1A-deficient cells. Together, our data highlight the broad impact of ARID1A loss and the possibility of targeting proliferative and DNA repair pathways for treatment.
{"title":"Multidimensional OMICs reveal ARID1A orchestrated control of DNA damage, splicing, and cell cycle in normal-like and malignant urothelial cells.","authors":"Rebecca M Schlösser, Florian Krumbach, Eyleen Corrales, Geoffroy Andrieux, Christian Preisinger, Franziska Liss, Alexandra Golzmann, Melanie Boerries, Kerstin Becker, Ruth Knüchel, Stefan Garczyk, Bernhard Lüscher","doi":"10.1002/1878-0261.70019","DOIUrl":"10.1002/1878-0261.70019","url":null,"abstract":"<p><p>Epigenetic regulators, such as the SWI/SNF complex, with important roles in tissue development and homeostasis, are frequently mutated in cancer. ARID1A, a subunit of the SWI/SNF complex, is mutated in approximately 20% of all bladder tumors; however, the consequences of this remain poorly understood. Finding truncations to be the most common mutation, we generated loss- and gain-of-function models to conduct RNA-Seq, interactome analyses, Omni-ATAC-Seq, and functional studies to characterize ARID1A-affected pathways potentially suitable for the treatment of ARID1A-deficient bladder cancers. We observed decreased cell proliferation and deregulation of stress-regulated pathways, including DNA repair, in ARID1A-deficient cells. Furthermore, ARID1A was linked to alternative splicing and translational regulation on RNA and interactome levels. ARID1A deficiency drastically reduced the accessibility of chromatin, especially around introns and distal enhancers, in a functional enrichment analysis. Less accessible chromatin areas were mapped to pathways such as cell proliferation and DNA damage response. Indeed, the G2/M checkpoint appeared impaired after DNA damage in ARID1A-deficient cells. Together, our data highlight the broad impact of ARID1A loss and the possibility of targeting proliferative and DNA repair pathways for treatment.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3784-3805"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688176/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143764461","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: 2025-08-27DOI: 10.1002/1878-0261.70038
Ruba Al-Abdulla, Simone Venz, Ruslan Al-Ali, Martin Wendlandt, Mandy Radefeldt, Elke Krüger
Proteasomes are involved in the maintenance of cellular protein homeostasis and the control of numerous cellular pathways. Single proteasome genes or subunits have been identified as important players in cancer development and progression without considering the proteasome as a multisubunit protease. We here conducted a comprehensive pan-cancer analysis encompassing transcriptional, epigenetic, mutational landscapes, pathway enrichments, and survival outcomes linked to the 20S proteasome core complex. The impact of proteasome gene expression on patient survival exhibited a cancer type-dependent pattern. Increased proteasome expression correlated with elevated activation of oncogenic pathways, such as DNA repair, MYC-controlled gene networks, MTORC1 signalling, oxidative phosphorylation, as well as metabolic pathways including glycolysis and fatty acid metabolism. Accordingly, potential loss of function variants of proteasome subunit genes are associated with improved patient survival. The TCGA-derived outcomes were further supported by gene expression analysis of THP-1 cells. Our study highlighted the importance of studying the proteasome as an enzymatic functional unit rather than separated subunits.
{"title":"Expression and DNA methylation of 20S proteasome subunits as prognostic and resistance markers in cancer.","authors":"Ruba Al-Abdulla, Simone Venz, Ruslan Al-Ali, Martin Wendlandt, Mandy Radefeldt, Elke Krüger","doi":"10.1002/1878-0261.70038","DOIUrl":"10.1002/1878-0261.70038","url":null,"abstract":"<p><p>Proteasomes are involved in the maintenance of cellular protein homeostasis and the control of numerous cellular pathways. Single proteasome genes or subunits have been identified as important players in cancer development and progression without considering the proteasome as a multisubunit protease. We here conducted a comprehensive pan-cancer analysis encompassing transcriptional, epigenetic, mutational landscapes, pathway enrichments, and survival outcomes linked to the 20S proteasome core complex. The impact of proteasome gene expression on patient survival exhibited a cancer type-dependent pattern. Increased proteasome expression correlated with elevated activation of oncogenic pathways, such as DNA repair, MYC-controlled gene networks, MTORC1 signalling, oxidative phosphorylation, as well as metabolic pathways including glycolysis and fatty acid metabolism. Accordingly, potential loss of function variants of proteasome subunit genes are associated with improved patient survival. The TCGA-derived outcomes were further supported by gene expression analysis of THP-1 cells. Our study highlighted the importance of studying the proteasome as an enzymatic functional unit rather than separated subunits.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3729-3749"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688171/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144961683","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: 2025-09-19DOI: 10.1002/1878-0261.70126
Michela De Meo, Marco Siringo, Alessandro Vici, Ann Zeuner, Orietta Gandini, Paola Gazzaniga, Chiara Nicolazzo
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality, with metastatic CRC (mCRC) posing significant challenges due to tumor heterogeneity and resistance to therapy. Circulating tumor cells (CTC) and circulating hybrid cells (CHC) detected via liquid biopsies have emerged as promising biomarkers for monitoring disease progression. This study aimed to evaluate the prognostic utility of automated CTC enumeration using the ACCEPT software compared to a manual method and assess the potential clinical relevance of CHC in mCRC. A retrospective analysis of CellSearch® images from 67 mCRC patients was conducted, correlating CTC and CHC counts with progression-free survival and overall survival (OS). Automated enumeration demonstrated improved accuracy and reduced variability, confirming the prognostic significance of CTC counts for OS. However, CHC enumeration showed no significant association with clinical outcomes, suggesting sporadic detection rather than consistent prognostic value. These findings underscore the reliability of automated CTC enumeration in mCRC prognosis while highlighting the need for further research into the biological and clinical roles of CHC.
{"title":"The ACCEPTance of automation: refining circulating tumor cells enumeration for improved metastatic colorectal cancer prognosis.","authors":"Michela De Meo, Marco Siringo, Alessandro Vici, Ann Zeuner, Orietta Gandini, Paola Gazzaniga, Chiara Nicolazzo","doi":"10.1002/1878-0261.70126","DOIUrl":"10.1002/1878-0261.70126","url":null,"abstract":"<p><p>Colorectal cancer (CRC) remains a leading cause of cancer-related mortality, with metastatic CRC (mCRC) posing significant challenges due to tumor heterogeneity and resistance to therapy. Circulating tumor cells (CTC) and circulating hybrid cells (CHC) detected via liquid biopsies have emerged as promising biomarkers for monitoring disease progression. This study aimed to evaluate the prognostic utility of automated CTC enumeration using the ACCEPT software compared to a manual method and assess the potential clinical relevance of CHC in mCRC. A retrospective analysis of CellSearch<sup>®</sup> images from 67 mCRC patients was conducted, correlating CTC and CHC counts with progression-free survival and overall survival (OS). Automated enumeration demonstrated improved accuracy and reduced variability, confirming the prognostic significance of CTC counts for OS. However, CHC enumeration showed no significant association with clinical outcomes, suggesting sporadic detection rather than consistent prognostic value. These findings underscore the reliability of automated CTC enumeration in mCRC prognosis while highlighting the need for further research into the biological and clinical roles of CHC.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3651-3664"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688169/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145086565","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: 2025-03-25DOI: 10.1002/1878-0261.70032
Paula Díez, Pablo Juanes-Velasco, Marina L García-Vaquero, Conrad Droste, Alicia Landeira-Viñuela, Miguel Alcoceba, Helena Fidalgo-Gómez, Sara Misiego-Herrero, Almudena Navarro-Bailón, Mónica Baile, José M Bastida, Jose Manuel Sanchez-Santos, Rafael Góngora, Julia Almeida, Marcos Gonzalez-Diaz, Alberto Orfao, Javier De Las Rivas, Manuel Fuentes
B-cell chronic lymphocytic leukemia (B-CLL) is characterized by highly heterogeneous genomic alterations and altered signaling pathways, with limited studies on its proteome. Our study presents a comprehensive analysis of the proteome and phosphoproteome in B-CLL and CLL-like monoclonal B-cell lymphocytosis (MBL) primary cells. Using high-resolution mass spectrometry, we identified 2970 proteins and 316 phosphoproteins across five tumor samples, including 55 newly identified phosphopeptides (ProteomeXchange-PXD005997). Our multifaceted approach also integrated protein microarrays and western blotting for further data validation in a new patient cohort of 14 patients. Despite sharing 73% of their proteomes, the phosphoproteomes varied significantly among samples, independent of cytogenetic alterations and immunoglobulin heavy variable cluster (IGHV) mutational status. We identified common functional hallmarks in B-CLL and MBL phosphoproteomes, notably tonic signaling (low-level, constitutive signaling) of the B-cell antigen-specific receptor (BCR) and nuclear factor NF-kappa-B (NF-kβ)/signal transducer and activator of transcription 3 (STAT3) pathways. Nine phosphoproteins involved in BCR signaling were further validated, showing a high correlation with early disease stages. Our study advances the field by providing a detailed perspective on the proteome and phosphoproteome of B-CLL cells, revealing signaling pathways crucial for disease development and progression. Integrating diverse proteomics techniques and identifying novel phosphopeptides offers new insights into CLL biology, potentially informing future therapeutic strategies and biomarker development for early diagnosis and personalized treatment.
b细胞慢性淋巴细胞白血病(B-CLL)的特点是高度异质性的基因组改变和信号通路改变,对其蛋白质组的研究有限。我们的研究全面分析了B-CLL和cll样单克隆b细胞淋巴细胞增多症(MBL)原代细胞的蛋白质组和磷酸化蛋白质组。利用高分辨率质谱技术,我们在5个肿瘤样本中鉴定出2970种蛋白和316种磷酸化蛋白,其中包括55种新鉴定的磷酸化肽(ProteomeXchange-PXD005997)。我们的多方面方法还整合了蛋白质微阵列和western blotting,在14例新患者队列中进一步验证数据。尽管有73%的蛋白质组是相同的,但磷蛋白质组在不同样本之间存在显著差异,不受细胞遗传学改变和免疫球蛋白重变量簇(IGHV)突变状态的影响。我们发现了B-CLL和MBL磷酸化蛋白质组的共同功能特征,特别是b细胞抗原特异性受体(BCR)和核因子nf - κ b (NF-kβ)/信号换能器和转录激活器3 (STAT3)途径的滋补信号(低水平的组成信号)。9个参与BCR信号传导的磷酸化蛋白被进一步验证,显示与早期疾病阶段高度相关。我们的研究为B-CLL细胞的蛋白质组和磷酸化蛋白质组提供了详细的视角,揭示了疾病发生和进展的关键信号通路,从而推动了该领域的发展。整合多种蛋白质组学技术和鉴定新的磷酸肽为CLL生物学提供了新的见解,可能为未来的治疗策略和早期诊断和个性化治疗的生物标志物开发提供信息。
{"title":"Tonic signaling of the B-cell antigen-specific receptor is a common functional hallmark in chronic lymphocytic leukemia cell phosphoproteomes at early disease stages.","authors":"Paula Díez, Pablo Juanes-Velasco, Marina L García-Vaquero, Conrad Droste, Alicia Landeira-Viñuela, Miguel Alcoceba, Helena Fidalgo-Gómez, Sara Misiego-Herrero, Almudena Navarro-Bailón, Mónica Baile, José M Bastida, Jose Manuel Sanchez-Santos, Rafael Góngora, Julia Almeida, Marcos Gonzalez-Diaz, Alberto Orfao, Javier De Las Rivas, Manuel Fuentes","doi":"10.1002/1878-0261.70032","DOIUrl":"10.1002/1878-0261.70032","url":null,"abstract":"<p><p>B-cell chronic lymphocytic leukemia (B-CLL) is characterized by highly heterogeneous genomic alterations and altered signaling pathways, with limited studies on its proteome. Our study presents a comprehensive analysis of the proteome and phosphoproteome in B-CLL and CLL-like monoclonal B-cell lymphocytosis (MBL) primary cells. Using high-resolution mass spectrometry, we identified 2970 proteins and 316 phosphoproteins across five tumor samples, including 55 newly identified phosphopeptides (ProteomeXchange-PXD005997). Our multifaceted approach also integrated protein microarrays and western blotting for further data validation in a new patient cohort of 14 patients. Despite sharing 73% of their proteomes, the phosphoproteomes varied significantly among samples, independent of cytogenetic alterations and immunoglobulin heavy variable cluster (IGHV) mutational status. We identified common functional hallmarks in B-CLL and MBL phosphoproteomes, notably tonic signaling (low-level, constitutive signaling) of the B-cell antigen-specific receptor (BCR) and nuclear factor NF-kappa-B (NF-kβ)/signal transducer and activator of transcription 3 (STAT3) pathways. Nine phosphoproteins involved in BCR signaling were further validated, showing a high correlation with early disease stages. Our study advances the field by providing a detailed perspective on the proteome and phosphoproteome of B-CLL cells, revealing signaling pathways crucial for disease development and progression. Integrating diverse proteomics techniques and identifying novel phosphopeptides offers new insights into CLL biology, potentially informing future therapeutic strategies and biomarker development for early diagnosis and personalized treatment.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3706-3728"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688175/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143701067","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: 2025-07-25DOI: 10.1002/1878-0261.70100
Cenk Celik, Shi Pan, Eloise Withnell, Hou Wang Lam, Maria Secrier
Spatial transcriptomics (ST) has emerged as a powerful tool to map gene expression patterns to the local tissue structure in cancer, enabling unprecedented insights into cellular heterogeneity and tumour microenvironments. As the technology matures, developing new, spatially informed analytical frameworks will be essential to fully leverage its potential to elucidate the complex organisation and emerging properties of cancer tissues. Here, we highlight key challenges in cancer spatial transcriptomics, focusing on three emerging topics: (a) defining cell states, (b) delineating cellular niches and (c) integrating spatial data with other modalities that can pave the way towards clinical translation. We discuss multiple analytical approaches that are currently implemented or could be adapted in the future in order to tackle these challenges, including classical biostatistics methods as well as methods inherited from geospatial analytics or artificial intelligence. In the rapidly expanding landscape of ST, such methodologies lay the foundation for biological discoveries that conceptualise cancer as an evolving system of interconnected niches.
{"title":"Decrypting cancer's spatial code: from single cells to tissue niches.","authors":"Cenk Celik, Shi Pan, Eloise Withnell, Hou Wang Lam, Maria Secrier","doi":"10.1002/1878-0261.70100","DOIUrl":"10.1002/1878-0261.70100","url":null,"abstract":"<p><p>Spatial transcriptomics (ST) has emerged as a powerful tool to map gene expression patterns to the local tissue structure in cancer, enabling unprecedented insights into cellular heterogeneity and tumour microenvironments. As the technology matures, developing new, spatially informed analytical frameworks will be essential to fully leverage its potential to elucidate the complex organisation and emerging properties of cancer tissues. Here, we highlight key challenges in cancer spatial transcriptomics, focusing on three emerging topics: (a) defining cell states, (b) delineating cellular niches and (c) integrating spatial data with other modalities that can pave the way towards clinical translation. We discuss multiple analytical approaches that are currently implemented or could be adapted in the future in order to tackle these challenges, including classical biostatistics methods as well as methods inherited from geospatial analytics or artificial intelligence. In the rapidly expanding landscape of ST, such methodologies lay the foundation for biological discoveries that conceptualise cancer as an evolving system of interconnected niches.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3465-3485"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688177/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144715150","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}
Nongenetic adaptive resistance to chemotherapy, driven by transcriptional rewiring, is emerging as a significant mechanism in tumor survival. In this study we combined longitudinal transcriptomics with temporal pattern analysis to investigate patient-specific mechanisms underlying acquired resistance in breast cancer. Matched tumor biopsies (pretreatment, posttreatment, and adjacent normal) were collected from breast cancer patients who received neoadjuvant chemotherapy. Transcriptomes were analyzed by longitudinal gene-pattern classification to track patient-specific gene expression alterations that occur during treatment. Our findings reveal that resistance-associated genes were already dysregulated in primary tumors, suggesting the presence of a preexisting drug-tolerant state. While each patient displayed unique resistance-associated gene rewiring, these alterations converged into a limited number of dysregulated functional modules. Notably, patients receiving the same treatment exhibited distinct rewiring of genes and pathways, revealing parallel, individualized routes to resistance. In conclusion, we propose that tumor cells survive chemotherapy by sustaining or amplifying a preexisting drug-tolerant state that circumvents drug action. We suggest that individualized "chemoresistome maps" could identify cancer vulnerabilities and inform personalized therapeutic strategies to overcome or prevent resistance.
{"title":"Chemoresistome mapping in individual breast cancer patients unravels diversity in dynamic transcriptional adaptation.","authors":"Maya Dadiani, Gilgi Friedlander, Gili Perry, Nora Balint-Lahat, Shlomit Gilad, Dana Morzaev-Sulzbach, Anjana Shenoy, Noa Bossel Ben-Moshe, Anya Pavlovsky, Rinat Bernstein-Molho, Eytan Domany, Iris Barshack, Tamar Geiger, Bella Kaufman, Einav Nili Gal-Yam","doi":"10.1002/1878-0261.70030","DOIUrl":"10.1002/1878-0261.70030","url":null,"abstract":"<p><p>Nongenetic adaptive resistance to chemotherapy, driven by transcriptional rewiring, is emerging as a significant mechanism in tumor survival. In this study we combined longitudinal transcriptomics with temporal pattern analysis to investigate patient-specific mechanisms underlying acquired resistance in breast cancer. Matched tumor biopsies (pretreatment, posttreatment, and adjacent normal) were collected from breast cancer patients who received neoadjuvant chemotherapy. Transcriptomes were analyzed by longitudinal gene-pattern classification to track patient-specific gene expression alterations that occur during treatment. Our findings reveal that resistance-associated genes were already dysregulated in primary tumors, suggesting the presence of a preexisting drug-tolerant state. While each patient displayed unique resistance-associated gene rewiring, these alterations converged into a limited number of dysregulated functional modules. Notably, patients receiving the same treatment exhibited distinct rewiring of genes and pathways, revealing parallel, individualized routes to resistance. In conclusion, we propose that tumor cells survive chemotherapy by sustaining or amplifying a preexisting drug-tolerant state that circumvents drug action. We suggest that individualized \"chemoresistome maps\" could identify cancer vulnerabilities and inform personalized therapeutic strategies to overcome or prevent resistance.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3665-3684"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688168/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144030030","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: 2025-05-19DOI: 10.1002/1878-0261.70053
Yang Song, Ting Liu, Qishan Hao, Qiuyun Fang, Xiaoyuan Gong, Yan Li, Zheng Tian, Hui Wei, Min Wang, Jianxiang Wang, Tao Cheng, Yingchang Mi
B-cell acute lymphoblastic leukemia (B-ALL) is a highly heterogeneous disease with a challenging prognosis, particularly in adult patients. We enrolled 88 adult B-ALL patients with transcriptomic and mutation profiles for classification system identification, and a comprehensive system for B-ALL patients (COMBAT) was developed. COMBAT stratified patients into three cohorts: (1) COMBAT1, characterized by high stem/myeloid antigen expression, low immune infiltration, high infiltration of endothelial cells, and hypo-CIMP (CpG island methylator phenotype); (2) COMBAT2, defined as an inflamed subtype with immune exhaustion, moderate myeloid antigen expression, and hypo-CIMP; and (3) COMBAT3, marked by proliferative profiles with MYC pathway activation and hypomethylation at enhancer regions in patients characterized by CIMP. The molecular features of the three COMBATs were verified in two external cohorts, the GSE34861 (N = 194) and GSE66005 (N = 109) datasets. In univariate analysis, only COMBAT classification presented significance for OS, and patients of COMBAT3 presented significantly superior survival than COMBAT1/2 in Ph-negative ALL. Ph-negative ALL patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) in the COMBAT3 group showed better overall survival (OS) than those in the COMBAT1-2 groups (estimated 3-year OS: 100% vs. 65.6%, P = 0.034), suggesting a prognostic benefit of this subtype. In summary, the COMBAT system redefines the characteristics of adult B-ALL subtypes and guides the selection of allo-HSCT for Ph-negative patients.
b细胞急性淋巴细胞白血病(B-ALL)是一种高度异质性的疾病,预后具有挑战性,特别是在成人患者中。我们招募了88名具有转录组学和突变谱的成年B-ALL患者进行分类系统鉴定,并开发了B-ALL患者的综合系统(COMBAT)。COMBAT将患者分层分为三个队列:(1)COMBAT1,其特征是高干/髓样抗原表达,低免疫浸润,高内皮细胞浸润,低cimp (CpG岛甲基化表型);(2) COMBAT2,定义为具有免疫衰竭、中度髓系抗原表达和低cimp的炎症亚型;(3)在CIMP患者中,以MYC通路激活和增强子区低甲基化的增殖谱为特征的COMBAT3。在两个外部队列GSE34861 (N = 194)和GSE66005 (N = 109)数据集中验证了这三种COMBATs的分子特征。在单变量分析中,只有COMBAT分类对OS有意义,而在ph阴性ALL中,COMBAT3患者的生存期明显优于COMBAT1/2患者。接受同种异体造血干细胞移植(alloo - hsct)的ph阴性ALL患者在COMBAT3组的总生存率(OS)高于COMBAT1-2组(估计3年OS: 100% vs. 65.6%, P = 0.034),表明该亚型具有预后益处。总之,COMBAT系统重新定义了成人B-ALL亚型的特征,并指导了ph阴性患者的同种异体造血干细胞移植的选择。
{"title":"Comprehensive omics-based classification system in adult patients with B-cell acute lymphoblastic leukemia.","authors":"Yang Song, Ting Liu, Qishan Hao, Qiuyun Fang, Xiaoyuan Gong, Yan Li, Zheng Tian, Hui Wei, Min Wang, Jianxiang Wang, Tao Cheng, Yingchang Mi","doi":"10.1002/1878-0261.70053","DOIUrl":"10.1002/1878-0261.70053","url":null,"abstract":"<p><p>B-cell acute lymphoblastic leukemia (B-ALL) is a highly heterogeneous disease with a challenging prognosis, particularly in adult patients. We enrolled 88 adult B-ALL patients with transcriptomic and mutation profiles for classification system identification, and a comprehensive system for B-ALL patients (COMBAT) was developed. COMBAT stratified patients into three cohorts: (1) COMBAT1, characterized by high stem/myeloid antigen expression, low immune infiltration, high infiltration of endothelial cells, and hypo-CIMP (CpG island methylator phenotype); (2) COMBAT2, defined as an inflamed subtype with immune exhaustion, moderate myeloid antigen expression, and hypo-CIMP; and (3) COMBAT3, marked by proliferative profiles with MYC pathway activation and hypomethylation at enhancer regions in patients characterized by CIMP. The molecular features of the three COMBATs were verified in two external cohorts, the GSE34861 (N = 194) and GSE66005 (N = 109) datasets. In univariate analysis, only COMBAT classification presented significance for OS, and patients of COMBAT3 presented significantly superior survival than COMBAT1/2 in Ph-negative ALL. Ph-negative ALL patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) in the COMBAT3 group showed better overall survival (OS) than those in the COMBAT1-2 groups (estimated 3-year OS: 100% vs. 65.6%, P = 0.034), suggesting a prognostic benefit of this subtype. In summary, the COMBAT system redefines the characteristics of adult B-ALL subtypes and guides the selection of allo-HSCT for Ph-negative patients.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3578-3595"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688170/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144102049","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: 2025-10-05DOI: 10.1002/1878-0261.70132
Justyna Topa, Julia Richert, Tomasz Stokowy, Alicja Staśczak, Mariusz Szajewski, Maciej Ciesielski, Petra M Grešner, Bartłomiej Tomasik, Łukasz Arcimowicz, Agnieszka Stankiewicz, Grażyna Suchodolska, Elżbieta Senkus, Wiesław Kruszewski, Anna J Żaczek, Aleksandra Markiewicz
Epithelial-mesenchymal transition (EMT) generates heterogeneity in circulating tumor cells (CTCs), affecting their biological properties and hampering their detection. This limits our understanding of the mechanisms underlying hematogenous dissemination, especially in early breast cancer (BC), where CTCs are rare. Here, we aimed to detect CTCs with different EMT statuses from BC patients. CTCs in blood samples from 107 BC patients were evaluated using immunomagnetic depletion and multi-marker immunofluorescence (EpCAM, E-cadherin, MCAM, cell surface vimentin, CD31, CD45), followed by single-cell transcriptomics. CTCs were detected in 51.9% of therapy-naïve early BC cases, with 3.8% showing only epithelial CTCs (eCTCs), 5.8% epithelial-mesenchymal (emCTCs), 26.0% mesenchymal (mCTCs), and 16.3% mixed phenotypes. CTC heterogeneity was more frequent in triple-negative (86%) than in luminal BC (17%, P = 0.008). Lymph node involvement strongly predicted dissemination of all CTC phenotypes, while tumor size correlated with mCTC abundance. Single-cell RNA sequencing revealed downregulation of ribosomal genes and translation inhibition in CTCs with mesenchymal features, linked to mTORC1 signaling. Findings were also validated in an independent dataset, highlighting vulnerabilities in CTCs during dissemination.
{"title":"Characterizing epithelial-mesenchymal transition-linked heterogeneity in breast cancer circulating tumor cells at a single-cell level.","authors":"Justyna Topa, Julia Richert, Tomasz Stokowy, Alicja Staśczak, Mariusz Szajewski, Maciej Ciesielski, Petra M Grešner, Bartłomiej Tomasik, Łukasz Arcimowicz, Agnieszka Stankiewicz, Grażyna Suchodolska, Elżbieta Senkus, Wiesław Kruszewski, Anna J Żaczek, Aleksandra Markiewicz","doi":"10.1002/1878-0261.70132","DOIUrl":"10.1002/1878-0261.70132","url":null,"abstract":"<p><p>Epithelial-mesenchymal transition (EMT) generates heterogeneity in circulating tumor cells (CTCs), affecting their biological properties and hampering their detection. This limits our understanding of the mechanisms underlying hematogenous dissemination, especially in early breast cancer (BC), where CTCs are rare. Here, we aimed to detect CTCs with different EMT statuses from BC patients. CTCs in blood samples from 107 BC patients were evaluated using immunomagnetic depletion and multi-marker immunofluorescence (EpCAM, E-cadherin, MCAM, cell surface vimentin, CD31, CD45), followed by single-cell transcriptomics. CTCs were detected in 51.9% of therapy-naïve early BC cases, with 3.8% showing only epithelial CTCs (eCTCs), 5.8% epithelial-mesenchymal (emCTCs), 26.0% mesenchymal (mCTCs), and 16.3% mixed phenotypes. CTC heterogeneity was more frequent in triple-negative (86%) than in luminal BC (17%, P = 0.008). Lymph node involvement strongly predicted dissemination of all CTC phenotypes, while tumor size correlated with mCTC abundance. Single-cell RNA sequencing revealed downregulation of ribosomal genes and translation inhibition in CTCs with mesenchymal features, linked to mTORC1 signaling. Findings were also validated in an independent dataset, highlighting vulnerabilities in CTCs during dissemination.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"3685-3705"},"PeriodicalIF":4.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12688172/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145228674","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}