Metastatic progression, driven by the dissemination of circulating tumor cells (CTCs) through the bloodstream, remains the leading cause of cancer-related death. A rare subset of CTCs, characterized by tumor-initiating properties and phenotypic plasticity, plays a pivotal role in the formation of distant metastases. The ability of these cells to survive in the circulation, evade the immune surveillance, and establish secondary tumors underscores their biological significance. However, CTC extreme rarity and heterogeneity pose major challenges for their in-depth functional characterization. Disseminated tumor cells (DTCs) are cells that have extravasated and persist in distant organ niches, often in a dormant state, and represent a complementary and equally critical component of metastatic progression. Their capacity to remain quiescent for prolonged periods before reactivation highlights the need to study both CTCs and DTCs to fully understand metastasis initiation and relapse. Recent advances in CTC isolation and culture have led to the development of patient-derived CTC lines and CTC-derived xenograft animal models, offering unprecedented opportunities to investigate metastatic seeding, therapeutic resistance and tumor evolution. CTC- and DTC-based models provide valuable insights into the biology of CTCs from different cancer types, revealing key molecular drivers of metastasis formation and potential therapeutic targets. In this review, we summarize the state-of-the-art methodologies for establishing CTC- and DTC-based models and evaluate their contribution to understand tumor progression and response to treatments. We discuss the current challenges in generating and maintaining these models, including the influence of hypoxic conditions, enrichment strategies, and culture medium optimization. Then, we highlight their potential applications in precision oncology, particularly for biomarker discovery and for preclinical drug testing.
{"title":"Reconstructing the metastatic journey: functional circulating tumor cells and disseminated tumor cells based models for translational oncology.","authors":"Caroline Sanglier, Laure Cayrefourcq, Catherine Alix-Panabières","doi":"10.1186/s13046-025-03617-y","DOIUrl":"https://doi.org/10.1186/s13046-025-03617-y","url":null,"abstract":"<p><p>Metastatic progression, driven by the dissemination of circulating tumor cells (CTCs) through the bloodstream, remains the leading cause of cancer-related death. A rare subset of CTCs, characterized by tumor-initiating properties and phenotypic plasticity, plays a pivotal role in the formation of distant metastases. The ability of these cells to survive in the circulation, evade the immune surveillance, and establish secondary tumors underscores their biological significance. However, CTC extreme rarity and heterogeneity pose major challenges for their in-depth functional characterization. Disseminated tumor cells (DTCs) are cells that have extravasated and persist in distant organ niches, often in a dormant state, and represent a complementary and equally critical component of metastatic progression. Their capacity to remain quiescent for prolonged periods before reactivation highlights the need to study both CTCs and DTCs to fully understand metastasis initiation and relapse. Recent advances in CTC isolation and culture have led to the development of patient-derived CTC lines and CTC-derived xenograft animal models, offering unprecedented opportunities to investigate metastatic seeding, therapeutic resistance and tumor evolution. CTC- and DTC-based models provide valuable insights into the biology of CTCs from different cancer types, revealing key molecular drivers of metastasis formation and potential therapeutic targets. In this review, we summarize the state-of-the-art methodologies for establishing CTC- and DTC-based models and evaluate their contribution to understand tumor progression and response to treatments. We discuss the current challenges in generating and maintaining these models, including the influence of hypoxic conditions, enrichment strategies, and culture medium optimization. Then, we highlight their potential applications in precision oncology, particularly for biomarker discovery and for preclinical drug testing.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145967759","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-13DOI: 10.1186/s13046-026-03643-4
Aslihan Yavas, Leon Boshoven, Kai Horny, Sebastian Haensch, Wolfgang Goering, Martin Schlensog, Lena Haeberle, Irene Esposito
Background: Pancreatic ductal adenocarcinoma (PDAC) is known to develop through a stepwise progression from precursor lesions, such as pancreatic intraepithelial neoplasias (PanIN). An alternative carcinogenic pathway has been proposed via transformation of acinar cells, with development of acinar-ductal metaplasia (ADM) and atypical flat lesions (AFL). Defining the characteristics of PDAC precursors is crucial to better understand PDAC carcinogenesis.
Methods: 15 KC (Ptf1aCre/+, KrasLSLG12D/+) and 15 KPC-like mice (Ptf1aCre/+, KrasLSLG12D/+, Trp53LoxP/LoxP, referred as fKPC hereafter) were sacrificed at different time points. A meticulous morphological evaluation was performed to define different lesion types. Multiplex immunofluorescence staining was applied to define the characteristics of the immune and stromal microenvironment of the lesions. To investigate the association between the genetic alterations and the components of the microenvironment, all lesion types were subjected to next-generation sequencing (NGS) using a 20 genes-panel.
Results: AFL showed a trend towards a more intense immune cell infiltration compared to PanIN and ADM. AFL had higher number of CD4+ helper T cells, FOXP3+ regulatory T cells, and CD19+ B cells than all other analyzed lesions. They displayed more CD8+ cytotoxic T cells and FOXP3+ cells than PDAC, while peripheral and central PDAC tissues tended to be infiltrated by macrophages in higher frequency. In addition, αSMA-expressing myofibroblastic cancer-associated fibroblasts were tendentially more frequent in AFL than other lesions. PDAC appeared to have higher CXCL12 expression and more common CD109+ cells than other lesions. In NGS analysis, none of the lesions in fKPC mice revealed additional coding mutations, while the preneoplastic lesions in 7 KC mice showed variable coding alterations in 16 different genes. The most frequently affected genes were Arid1a, Rnf43, and Pik3ca. PDAC precursors in KC mice showed more dense infiltration of adaptive immune cells than in fKPC mice, supporting the immunosuppressive role of Trp53 alterations.
Conclusions: Our study highlights the unique immunological and stromal features of AFL. Moreover, reinforcing their potential as precursor lesions, ADM and AFL exhibit variable alterations in the genes that have a critical role in PDAC carcinogenesis.
{"title":"Immunological and molecular insights into acinar-ductal metaplasia and atypical flat lesions as precursor lesions of pancreatic ductal adenocarcinoma.","authors":"Aslihan Yavas, Leon Boshoven, Kai Horny, Sebastian Haensch, Wolfgang Goering, Martin Schlensog, Lena Haeberle, Irene Esposito","doi":"10.1186/s13046-026-03643-4","DOIUrl":"https://doi.org/10.1186/s13046-026-03643-4","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic ductal adenocarcinoma (PDAC) is known to develop through a stepwise progression from precursor lesions, such as pancreatic intraepithelial neoplasias (PanIN). An alternative carcinogenic pathway has been proposed via transformation of acinar cells, with development of acinar-ductal metaplasia (ADM) and atypical flat lesions (AFL). Defining the characteristics of PDAC precursors is crucial to better understand PDAC carcinogenesis.</p><p><strong>Methods: </strong>15 KC (Ptf1a<sup>Cre/+</sup>, Kras<sup>LSLG12D/+</sup>) and 15 KPC-like mice (Ptf1a<sup>Cre/+</sup>, Kras<sup>LSLG12D/+</sup>, Trp53<sup>LoxP/LoxP</sup>, referred as fKPC hereafter) were sacrificed at different time points. A meticulous morphological evaluation was performed to define different lesion types. Multiplex immunofluorescence staining was applied to define the characteristics of the immune and stromal microenvironment of the lesions. To investigate the association between the genetic alterations and the components of the microenvironment, all lesion types were subjected to next-generation sequencing (NGS) using a 20 genes-panel.</p><p><strong>Results: </strong>AFL showed a trend towards a more intense immune cell infiltration compared to PanIN and ADM. AFL had higher number of CD4<sup>+</sup> helper T cells, FOXP3<sup>+</sup> regulatory T cells, and CD19<sup>+</sup> B cells than all other analyzed lesions. They displayed more CD8<sup>+</sup> cytotoxic T cells and FOXP3<sup>+</sup> cells than PDAC, while peripheral and central PDAC tissues tended to be infiltrated by macrophages in higher frequency. In addition, αSMA-expressing myofibroblastic cancer-associated fibroblasts were tendentially more frequent in AFL than other lesions. PDAC appeared to have higher CXCL12 expression and more common CD109<sup>+</sup> cells than other lesions. In NGS analysis, none of the lesions in fKPC mice revealed additional coding mutations, while the preneoplastic lesions in 7 KC mice showed variable coding alterations in 16 different genes. The most frequently affected genes were Arid1a, Rnf43, and Pik3ca. PDAC precursors in KC mice showed more dense infiltration of adaptive immune cells than in fKPC mice, supporting the immunosuppressive role of Trp53 alterations.</p><p><strong>Conclusions: </strong>Our study highlights the unique immunological and stromal features of AFL. Moreover, reinforcing their potential as precursor lesions, ADM and AFL exhibit variable alterations in the genes that have a critical role in PDAC carcinogenesis.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145967737","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-09DOI: 10.1186/s13046-025-03634-x
Sofia Titah, Aurélie Guillemette, Clara Lewuillon, Faruk Azam Shaik, Céline Berthon, Laure Goursaud, Meryem Tardivel, Antonino Bongiovanni, Paul Chauvet, Nathalie Jouy, Pauline Peyrouze, Meyling Cheok, Carine Brinster, Salomon Manier, Mehmet Çagatay Tarhan, Loïc Lemonnier, Bruno Quesnel, Yasmine Touil
<p><strong>Background: </strong>Acute myeloid leukemia (AML) remains a high-risk hematologic malignancy due to frequent relapse and therapeutic resistance. Although induction therapy can achieve cytological remission, a fraction of leukemic cells (minimal residual disease, MRD) persists within the protective bone marrow (BM) microenvironment. MRD is heterogeneous and may include subclones with intrinsic survival features present before therapy. Among these, rare BM-resident leukemic cells (BMresLC) may represent pre-adapted precursors of MRD, maintained in a low-proliferative (Ki67<sup>low</sup>) or quiescent state. We previously showed that calcium signaling through ORAI1-dependent store-operated calcium entry (SOCE) contributes not only to AML stemness and drug resistance but also to the regulation of the G0-G1 cell-cycle transition and the emergence of slow-cycling leukemic cells. With this study, we have characterized the stemness and calcium signature of BMresLC before any therapeutic intervention. Our results, beyond further characterizing a population of cells rarely studied, could thus pave the way to new therapeutic opportunities combining current treatments with the targeting of relevant pathways highlighted by our work.</p><p><strong>Methods: </strong>A patient-derived xenograft (PDX) model in NSG (NOD/SCID/IL2Rγ<sup>null</sup>) mice was used to localize, isolate, and characterize human BMresLC. Whole-bone clearing and 3D-Imaris imaging enabled spatial localization of rare leukemic cells. Flow cytometry and qPCR assessed cell-cycle status, immunophenotype (CD34, CD38, TIM-3, PD-L1, Ki67), stemness, and calcium-signaling components (ORAI1-3, STIM1-2, NFATc1-4). SOCE was measured using Indo-1 assays. Comparative analyses were performed against diagnostic AML cells, public MRD RNA-seq datasets, and prognosis-stratified patient cohorts.</p><p><strong>Results: </strong>BMresLC displayed an immune-evasive immunophenotype and contained a small fraction of Ki67<sup>neg</sup> quiescent cells, but were not enriched in fully quiescent cells. Instead, they predominantly exhibited a Ki67<sup>low</sup> slow-cycling profile, consistent with a low-proliferative persistent state. Transcriptional analysis revealed overexpression of stemness-associated genes and selective downregulation of calcium-signaling components ORAI1, ORAI2, STIM2, and NFATc1/c4, consistent with a SOCE-suppressed calcium signature. Functional assays confirmed reduced calcium influx. Compared with post-therapy MRD datasets, BMresLC showed some stemness and immune-evasion traits but displayed a distinct pre-therapeutic calcium signature, suggesting that it represents an early, persistent state preceding full MRD remodeling. Prognostic subgroup analysis further showed that BMresLC calcium and stemness profiles partially recapitulate features of adverse-risk AML, including differences in CD34, CD38, PD-L1, MMRN1, LAPTM4B, NFATc2, and STIM2 expression.</p><p><strong>Conclusions: </stro
{"title":"Pre-therapeutic bone marrow-resident leukemic cells in acute myeloid leukemia exhibit a distinct dysregulated calcium signature and stem-like profile reflecting minimal residual disease precursors.","authors":"Sofia Titah, Aurélie Guillemette, Clara Lewuillon, Faruk Azam Shaik, Céline Berthon, Laure Goursaud, Meryem Tardivel, Antonino Bongiovanni, Paul Chauvet, Nathalie Jouy, Pauline Peyrouze, Meyling Cheok, Carine Brinster, Salomon Manier, Mehmet Çagatay Tarhan, Loïc Lemonnier, Bruno Quesnel, Yasmine Touil","doi":"10.1186/s13046-025-03634-x","DOIUrl":"https://doi.org/10.1186/s13046-025-03634-x","url":null,"abstract":"<p><strong>Background: </strong>Acute myeloid leukemia (AML) remains a high-risk hematologic malignancy due to frequent relapse and therapeutic resistance. Although induction therapy can achieve cytological remission, a fraction of leukemic cells (minimal residual disease, MRD) persists within the protective bone marrow (BM) microenvironment. MRD is heterogeneous and may include subclones with intrinsic survival features present before therapy. Among these, rare BM-resident leukemic cells (BMresLC) may represent pre-adapted precursors of MRD, maintained in a low-proliferative (Ki67<sup>low</sup>) or quiescent state. We previously showed that calcium signaling through ORAI1-dependent store-operated calcium entry (SOCE) contributes not only to AML stemness and drug resistance but also to the regulation of the G0-G1 cell-cycle transition and the emergence of slow-cycling leukemic cells. With this study, we have characterized the stemness and calcium signature of BMresLC before any therapeutic intervention. Our results, beyond further characterizing a population of cells rarely studied, could thus pave the way to new therapeutic opportunities combining current treatments with the targeting of relevant pathways highlighted by our work.</p><p><strong>Methods: </strong>A patient-derived xenograft (PDX) model in NSG (NOD/SCID/IL2Rγ<sup>null</sup>) mice was used to localize, isolate, and characterize human BMresLC. Whole-bone clearing and 3D-Imaris imaging enabled spatial localization of rare leukemic cells. Flow cytometry and qPCR assessed cell-cycle status, immunophenotype (CD34, CD38, TIM-3, PD-L1, Ki67), stemness, and calcium-signaling components (ORAI1-3, STIM1-2, NFATc1-4). SOCE was measured using Indo-1 assays. Comparative analyses were performed against diagnostic AML cells, public MRD RNA-seq datasets, and prognosis-stratified patient cohorts.</p><p><strong>Results: </strong>BMresLC displayed an immune-evasive immunophenotype and contained a small fraction of Ki67<sup>neg</sup> quiescent cells, but were not enriched in fully quiescent cells. Instead, they predominantly exhibited a Ki67<sup>low</sup> slow-cycling profile, consistent with a low-proliferative persistent state. Transcriptional analysis revealed overexpression of stemness-associated genes and selective downregulation of calcium-signaling components ORAI1, ORAI2, STIM2, and NFATc1/c4, consistent with a SOCE-suppressed calcium signature. Functional assays confirmed reduced calcium influx. Compared with post-therapy MRD datasets, BMresLC showed some stemness and immune-evasion traits but displayed a distinct pre-therapeutic calcium signature, suggesting that it represents an early, persistent state preceding full MRD remodeling. Prognostic subgroup analysis further showed that BMresLC calcium and stemness profiles partially recapitulate features of adverse-risk AML, including differences in CD34, CD38, PD-L1, MMRN1, LAPTM4B, NFATc2, and STIM2 expression.</p><p><strong>Conclusions: </stro","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-09DOI: 10.1186/s13046-025-03632-z
Patricia Rider, Anna Tutusaus, Carlos Cuño-Gómiz, Flavia Savino, Aida Marsal, Neus Llarch, Gemma Iserte, Anna Colell, Pablo García de Frutos, Tania Hernáez-Alsina, Marco Sanduzzi-Zamparelli, Montserrat Marí, María Reig, Albert Morales
{"title":"Immune remodeling via mitochondria-dependent STING activation enhances cabozantinib response in hepatocellular carcinoma.","authors":"Patricia Rider, Anna Tutusaus, Carlos Cuño-Gómiz, Flavia Savino, Aida Marsal, Neus Llarch, Gemma Iserte, Anna Colell, Pablo García de Frutos, Tania Hernáez-Alsina, Marco Sanduzzi-Zamparelli, Montserrat Marí, María Reig, Albert Morales","doi":"10.1186/s13046-025-03632-z","DOIUrl":"https://doi.org/10.1186/s13046-025-03632-z","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935915","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-08DOI: 10.1186/s13046-025-03631-0
Lavinia Ghiani, Simona Citro, Alessandro Medda, Mirko Doni, Farkhondeh Ghoryani, Roberta Noberini, Ottavio Croci, Fausto Maffini, Claudia Miccolo, Laura Monteleone, Marta Tagliabue, Rita De Berardinis, Stefano Campaner, Tiziana Bonaldi, Mohssen Ansarin, Susanna Chiocca
{"title":"NSD2 upregulation is driven by high-risk HPV E6/E7 and disrupts epithelial differentiation in HPV-associated head and neck cancer.","authors":"Lavinia Ghiani, Simona Citro, Alessandro Medda, Mirko Doni, Farkhondeh Ghoryani, Roberta Noberini, Ottavio Croci, Fausto Maffini, Claudia Miccolo, Laura Monteleone, Marta Tagliabue, Rita De Berardinis, Stefano Campaner, Tiziana Bonaldi, Mohssen Ansarin, Susanna Chiocca","doi":"10.1186/s13046-025-03631-0","DOIUrl":"https://doi.org/10.1186/s13046-025-03631-0","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935899","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-08DOI: 10.1186/s13046-025-03626-x
Vittoria Matafora, Alice Elhagh, Alessandra Morelli, Laura Tronci, Angela Cattaneo, Camilla Conti, Francesca Casagrande, Francesco Farris, Angela Bachi
{"title":"BACE2 tunes lipid uptake through lipid transporters shedding supporting cancer cell proliferation.","authors":"Vittoria Matafora, Alice Elhagh, Alessandra Morelli, Laura Tronci, Angela Cattaneo, Camilla Conti, Francesca Casagrande, Francesco Farris, Angela Bachi","doi":"10.1186/s13046-025-03626-x","DOIUrl":"https://doi.org/10.1186/s13046-025-03626-x","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935911","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}