{"title":"Balancing between cuproplasia and copper-dependent cell death: molecular basis and clinical implications of ATOX1 in cancer.","authors":"Justyna Suwara, Mariusz L Hartman","doi":"10.1186/s13046-025-03486-5","DOIUrl":"10.1186/s13046-025-03486-5","url":null,"abstract":"<p><p>Human antioxidant protein 1 (ATOX1) is an essential regulator of copper homeostasis in cells. By interacting with other proteins involved in controlling the intracellular levels of cuprous ions (Cu<sup>+</sup>), ATOX1 contributes to the import, export, and subcellular distribution of Cu<sup>+</sup> as it functions within the CTR1-ATOX1-ATP7A/ATP7B axis. For this reason, ATOX1 plays a key role in preventing copper toxicity. Since copper ions have been shown to regulate the activity of a subset of other signaling proteins, ATOX1 can support cell proliferation, migration, and survival. Notably, ATOX1 is the only identified copper chaperone that has transcription factor activity. In this respect, CCND1, MDC1, NCF1, PPA2, and SOD3 have been experimentally validated as transcriptional targets of ATOX1 in distinct types of cells. The multifaceted actions of ATOX1 indicate that its dysregulation can lead to changes in the activity of crucial signaling pathways associated with diverse disorders, including cancer. Indeed, ATOX1 levels are frequently increased in cancer as demonstrated in multiple studies and supported by data available in GEPIA. ATOX1 has been implicated in cancer biology because of its role in the proliferation and metastatic spread of cancer cells and protection from oxidative stress. Additionally, ATOX1 may impact the drug response and resistance of cancer cells by influencing detoxification mechanisms as demonstrated for platinum-based therapies. In turn, the role of ATOX1 in the susceptibility of cancer cells to targeted therapies and immunotherapy remains elusive. This, however, should be a direction of further research considering the recent advances in understanding the complex role of copper in cancer cells, which can be associated with either protumorigenic effects (cuproplasia) or the induction of novel copper-dependent regulated cell death (cuproptosis) to combat cancer cells. Therefore, the disruption of ATOX1-mediated processes could be beneficial for the efficacy of anticancer therapies, although this possibility should be treated with caution because of the dual role of copper in cancer. Moreover, the prognostic value of ATOX1 expression for the clinical outcome of cancer patients needs to be clarified. In this review, we summarize the current state of knowledge about ATOX1 in cancer focusing on its molecular aspects and potential clinical implications.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"222"},"PeriodicalIF":12.8,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144735029","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}
{"title":"Comparing Xenium 5K and Visium HD data from identical tissue slide at a pathological perspective.","authors":"Mengping Long, Taobo Hu, Weixin Wang, Junshun Gao, Nan Wang, Mats Nilsson","doi":"10.1186/s13046-025-03479-4","DOIUrl":"10.1186/s13046-025-03479-4","url":null,"abstract":"<p><p>Recent advancements in spatial transcriptomics have been largely triggered by two high-resolution technologies: Visium-HD and Xenium in-situ. While sequencing-based Visium HD features a refined bin size of 2 µm and transcriptome wide coverage, Xenium in-situ is a targeted imaging-based detection technology with sub-micron resolution. Herein we use a publicly available lung dataset which contains Visium-HD and Xenium-5K data generated on identical tissue slides to make a bona-fide technical comparison aligned with thorough pathological annotations. Whilst Visium-HD offers a broader gene coverage for detection and likely detects more tumor subclones, Xenium-5K achieves comparable results when robust clustering algorithms are applied. Importantly, from the pathological point of view, the single-cell segmentation accuracy is essential when analyzing irregularly shaped cells, where Xenium may be in favor. At the opposite side, although Xenium-5K based cell segmentation to delineate immune cells, normal lung, and vasculature at cell resolution is decent, it relies on fluorescent signals for transcript detection, which is challenging in heavily pigmented tissues such as melanoma or dust-laden alveolar macrophages, an application scenario for which Visium HD may stand out. From this perspective, pathological derived factors are the prior consideration for selecting an appropriate ST approach under difference research settings including cancer.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"219"},"PeriodicalIF":12.8,"publicationDate":"2025-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12298044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144719047","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rui Guan, Ce Li, Ruijie Jiao, Jingao Li, Ran Wei, Chen Feng, Shengda Cao, Ye Qian, Jugao Fang, Jun Liu, Wenming Li, Dongmin Wei, Dapeng Lei
{"title":"MRPL21-PARP1 axis promotes cisplatin resistance in head and neck squamous cell carcinoma by inhibiting autophagy through the PI3K/AKT/mTOR signaling pathway.","authors":"Rui Guan, Ce Li, Ruijie Jiao, Jingao Li, Ran Wei, Chen Feng, Shengda Cao, Ye Qian, Jugao Fang, Jun Liu, Wenming Li, Dongmin Wei, Dapeng Lei","doi":"10.1186/s13046-025-03482-9","DOIUrl":"10.1186/s13046-025-03482-9","url":null,"abstract":"<p><strong>Background: </strong>Head and neck squamous cell carcinoma (HNSCC) constitutes a major clinical challenge that severely affects patient survival. Mitochondrial ribosomal protein (MRP) family plays an important role in energy metabolism by participating in mitochondrial oxidative phosphorylation. However, their roles in HNSCC and the underlying mechanisms are still unclear.</p><p><strong>Methods: </strong>Single-cell analysis highlighted MRPL21 as a notable biomarker of HNSCC. Human HNSCC tissues, cell lines, and xenograft models in nude mice were used to explore the expression and function of MRPL21. The mass spectrometry was performed to analyze the potential binding targets of MRPL21. In vitro and in vivo experiments were performed to evaluate the effect of MRPL21 on autophagy and cisplatin resistance. The inhibitory actions of siMRPL21 nanodelivery systems on HNSCC progression were also evaluated in vivo.</p><p><strong>Results: </strong>Clinically, relatively high expression level of MRPL21 was associated with poor prognosis in HNSCC patients, and overexpression of MRPL21 significantly promoted HNSCC tumorigenesis, metastasis, and cisplatin resistance. Mechanistically, MRPL21 upregulated mitochondrial oxidative phosphorylation (OXPHOS) and increased PARylation level, inhibited autophagy through activating the downstream PI3K/AKT/mTOR signaling pathway, and ultimately led to tumor progression and cisplatin resistance in HNSCC.</p><p><strong>Conclusion: </strong>We conclude that MRPL21 is a novel biomarker and therapeutic target of HNSCC progression and cisplatin resistant, which may provide a new approach for overcoming cisplatin resistance in HNSCC patients.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"221"},"PeriodicalIF":12.8,"publicationDate":"2025-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12297673/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144719049","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Copy number amplification of TTPAL promotes cholesterol biosynthesis and esophageal squamous cell carcinoma progression via elevating NSUN2-mediated m5C modification of SREBP2 mRNA.","authors":"Shan Huang, Yuanyuan Liu, Manyu Zhao, Tao Wang, Lihua Mao, Ting Wang, Chunyuan Guo, Wentao Huang, Zimei Peng, Zhen Zhang, Rui Jiang, Xinrui Ma, Nimei Shen, Jun Rao, Xing Wang, Zhi Zheng, Lixiao Chen","doi":"10.1186/s13046-025-03483-8","DOIUrl":"10.1186/s13046-025-03483-8","url":null,"abstract":"<p><p>Alterations in copy number are crucial genetic events in the development of esophageal squamous cell carcinoma (ESCC). Here, we show that Tocopherol alpha transfer protein-like (TTPAL) is highly amplified and frequently overexpressed in human ESCC. Using Ttpal-KO mouse mode, we demonstrate that TTPAL promotes ESCC cell proliferation and accelerates tumor development by driving cholesterol biosynthesis. Mechanistically, TTPAL upregulates a key transcription factor in cholesterol biosynthesis-sterol regulatory element-binding transcription factor (SREBP2) in ESCC cells. TTPAL interacts with the RNA methyltransferase NSUN2 and relieves the ubiquitination of NSUN2, protecting NSUN2 from proteasome-mediated degradation. In turn, NSUN2 catalyzes the m5C modification of SREBP2 mRNA, and then the m5C modified SREBP2 mRNA binds to the m5C reader protein-ALYREF to enhance its stability, thereby increasing SREBP2 expression. Moreover, we validate the efficacy of cholesterol biosynthesis inhibitor simvastatin in ESCC with high TTPAL expression. Overall, our results uncover a novel function of TTPAL in regulating SREBP2 expression, revealed a previously unknown TTPAL/NSUN2/SREBP2 pathway that promotes cholesterol biosynthesis in ESCC cells, and identified sensitively to cholesterol biosynthesis inhibitor simvastatin.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"220"},"PeriodicalIF":12.8,"publicationDate":"2025-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12297799/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144719048","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Unraveling resistance mechanisms to the novel nucleoside analog RX-3117 in lung cancer: insights into DNA repair, cell cycle dysregulation and targeting PKMYT1 for improved therapy.","authors":"Mahrou Vahabi, Geng Xu, Dzjemma Sarkisjan, Btissame El Hassouni, Giulia Mantini, Valentina Donati, Bing Wang, Giulia Lencioni, Richard J Honeywell, Dongmei Deng, Sabrina Strano, Godefridus J Peters, Giovanni Blandino, Elisa Giovannetti","doi":"10.1186/s13046-025-03470-z","DOIUrl":"10.1186/s13046-025-03470-z","url":null,"abstract":"<p><strong>Background: </strong>Nucleoside analogues are crucial in treating non-small cell lung cancer (NSCLC), but resistance hampers patient outcomes. The cytidine analogue RX-3117 shows promise in gemcitabine-resistant cancers, yet mechanisms underlying acquired resistance to this drug remain unexplored. This study includes a comprehensive investigation into RX-3117 resistance mechanisms by leveraging new preclinical models and cutting-edge genomic tools, including a CRISPR-Cas9 knockout screen and transcriptomics.</p><p><strong>Methods: </strong>NSCLC cell lines A549 and SW1573 were exposed to stepwise increasing concentrations of RX-3117 to establish stable resistant subclones, confirmed by SRB and clonogenic assays. Intracellular RX-3117 nucleotide levels were measured via LC/MS-MS, prompting the evaluation and modulation of the expression of key metabolic enzymes by Western blot and siRNA. A CRISPR-Cas9 screen identified genes whose loss increased RX-3117 sensitivity, while RNA-sequencing with differential expression analyses revealed resistance-related pathways, further investigated through cell cycle distribution, knock-out, and ELISA assays.</p><p><strong>Results: </strong>Resistant clones exhibited decreased accumulation of RX-3117 nucleotides, which however, was not associated to reduced expression of activation enzymes (UCK2, UMPK, CMPK, NME1/NDPK, RR1 and RR2). Instead, increased expression was observed in certain DNA repair and deactivation enzymes (NT5C3) but pharmacological inhibition and silencing of the latter did not circumvent resistance. Remarkably, a comprehensive approach with CRISPR-Cas9 screen highlighted DNA-repair and cell cycle determinants as key sensitizing genes. XL-PCR and RNA-sequencing confirmed aberrations in DNA-repair and pathways involved in cell cycle regulation. Knock-out and pharmacological inhibition validated the role of PKMYT1, a protein kinase involved in G2/M transition and genomic stability. RX-3117-resistant A549 cells showed enhanced sensitivity to the PKMYT1 inhibitor lunresertib and its synergism with RX-3117, suggesting further studies, especially in patients with high PKMYT1 expression who have significantly shorter survival rates, as observed in public databases and validated in an internal cohort of NSCLC patients.</p><p><strong>Conclusion: </strong>By integrating CRISPR-Cas9 with functional assays and transcriptomics, our study established a framework for decoding resistance mechanisms and highlights potential therapeutic strategies to enhance RX-3117 efficacy in NSCLC. We demonstrated for the first time that aberrant DNA repair and cell cycle dysregulation led resistance, identifying PKMYT1 as a promising target.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"217"},"PeriodicalIF":11.4,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12288264/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144700242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Angela Galardi, Valentina Fogazzi, Claudia Tottone, Marta Giussani, Serenella M Pupa, Giulia Cosentino, Marilena V Iorio
{"title":"\"Small extracellular vesicles: messengers at the service of breast cancer agenda in the primary and distant microenvironments\".","authors":"Angela Galardi, Valentina Fogazzi, Claudia Tottone, Marta Giussani, Serenella M Pupa, Giulia Cosentino, Marilena V Iorio","doi":"10.1186/s13046-025-03471-y","DOIUrl":"10.1186/s13046-025-03471-y","url":null,"abstract":"<p><p>Breast cancer (BC) remains a leading cause of cancer-related mortality in women, with complex mechanisms driving its initiation, progression, and resistance to therapy. In recent years, the tumor microenvironment (TME) has gained attention for its critical role in shaping tumor behavior, where small extracellular vesicles (small EVs) have emerged as key mediators of intercellular communication. These vesicles carry a diverse cargo of proteins, lipids, DNA, and various non-coding RNAs-such as miR-21, miR-155, and miR-1246-mirroring the molecular status of their originating cells. This review highlights the roles of small EVs in immune modulation, stromal remodelling, and metastatic niche formation, emphasizing their contribution to therapy resistance and immune evasion. We discuss recent updates on EV biogenesis, characterisation and isolation techniques, such as ultracentrifugation, immunoaffinity and microfluidic systems. We also critically evaluate their potential for clinical application and how well they conform to the MISEV2023 guidelines. Furthermore, we examine small EVs as diagnostic tools in liquid biopsies and compare them with conventional methods such as mammography and tissue biopsies. We also discuss organotropism mediated by small EV cargo (e.g., integrins α6β4, αvβ5) and the diagnostic potential of protein and lipid signatures (e.g., PD-L1, CD63, and exosomal lipidomics). Therapeutically, we explore engineered small EVs for drug delivery, gene modulation, and immune activation, addressing challenges of targeting efficiency, in vivo stability, immunogenicity, and clinical scalability. The review discusses ongoing clinical trials involving small EVs in BC and highlights key translational gaps between preclinical advances and clinical implementation. Finally, we explores how integrating artificial intelligence, single-cell transcriptomics, and multi-omics approaches can help overcome major challenges such as small EV heterogeneity and tracking limitations. Crucially, this integration enables a more tailored understanding of each patient's tumor biology, reducing therapeutic failures by guiding more personalized and effective treatment strategies. Overall, small EVs represent a transformative tool in precision oncology, contingent on resolving key challenges in their clinical translation.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"216"},"PeriodicalIF":11.4,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12278669/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144683463","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sophie G Shifman, Jennifer L O'Connor, Daniel P Radin, Aryan Sharma, Laura Infante, Francesca Ferraresso, Christian J Kastrup, Daniel A Lawrence, Stella E Tsirka
{"title":"Targeting autophagy and plasminogen activator inhibitor-1 increases survival and remodels the tumor microenvironment in glioblastoma.","authors":"Sophie G Shifman, Jennifer L O'Connor, Daniel P Radin, Aryan Sharma, Laura Infante, Francesca Ferraresso, Christian J Kastrup, Daniel A Lawrence, Stella E Tsirka","doi":"10.1186/s13046-025-03473-w","DOIUrl":"10.1186/s13046-025-03473-w","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GBM), the most common and aggressive type of primary brain tumor, engages multiple survival mechanisms, including autophagy. GBM exploits both degradative and secretory autophagy pathways to support tumor growth and limit the efficacy of standard-of-care treatments. We have previously shown that lucanthone, a blood-brain barrier permeable autophagy inhibitor, reduces tumor burden. However, although lucanthone-treated tumors are significantly smaller in size, they are not completely obliterated, suggesting compensatory survival mechanisms. A critical factor for GBM survival is communication with the tumor microenvironment (TME), which can be programmed by glioma cells to support growth and immunosuppression. Plasminogen activator inhibitor-1 (PAI-1), a secreted serine protease inhibitor, has been implicated in the progression of several cancers, including GBM, and has been shown to be modulated by autophagy in other cancers. The role of PAI-1 in GBM, namely its relationship with intracellular autophagy dysregulation and extracellular TME as a mechanism of tumor survival, remains incompletely understood.</p><p><strong>Methods: </strong>Murine glioma models were established using intracranial injection of GL261 cells in C57BL/6 mice, followed by autophagy inhibition with intraperitoneal lucanthone and/or PAI-1 inhibition with MDI-2268 chow, and tumors were assessed by immunohistochemistry. In culture, glioma cell lines were challenged with MDI-2268, lucanthone, mitoxantrone, or siRNA-LNPs targeting PAI-1, and assessed by MTT assay, q-RT-PCR, ELISA, invasion assay, immunoblot, and immunocytochemistry. Lysosomal markers and transient transfection with fluorescent vesicular proteins were utilized to evaluate PAI-1 intracellular localization via confocal microscopy. Synergy was analyzed using the HSA model in Combenefit, and statistical analyses included t-tests, ANOVA, and log-rank tests for survival.</p><p><strong>Results: </strong>Lucanthone treatment increased intracellular PAI-1 and autophagy markers while reducing active extracellular PAI-1. PAI-1 colocalized with lysosomal markers, suggesting impaired secretory autophagy. PAI-1 inhibition reduced glioma cell viability and invasion. Combination therapy with lucanthone and MDI-2268 drastically decreased tumor volume, prolonged survival, and promoted a pro-inflammatory state in the tumor microenvironment.</p><p><strong>Conclusions: </strong>Our findings suggest that PAI-1 may be a compensatory survival mechanism in GBM after autophagy inhibition, and that dual targeting of autophagy and PAI-1 disrupts tumor progression and enhances anti-tumor immunity, providing promising evidence for targeting this axis.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"214"},"PeriodicalIF":11.4,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12275254/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144668841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rui Wang, Claudia Gilbert, Houda Tahiri, Chun Yang, Solange Landreville, Pierre Hardy
{"title":"MiR-181a-driven downregulation of cholesterol biosynthesis through SREBP2 inhibition suppresses uveal melanoma metastasis.","authors":"Rui Wang, Claudia Gilbert, Houda Tahiri, Chun Yang, Solange Landreville, Pierre Hardy","doi":"10.1186/s13046-025-03459-8","DOIUrl":"10.1186/s13046-025-03459-8","url":null,"abstract":"<p><strong>Background: </strong>uveal melanoma (UM) is the most common primary intraocular tumor in adults, with metastasis being the leading cause of death. However, effective treatments for metastatic UM remain limited. Emerging evidence suggests that cholesterol metabolism plays a role in cancer progression, but its impact on UM metastasis is not well understood.</p><p><strong>Methods: </strong>we investigated the effects of miR-181a on UM metastasis using multiple UM cell lines and a suprachoroidal injection mouse model. Functional assays, including migration, invasion, and cancer stem-like cell (CSC) formation, were performed. The target of miR-181a was identified through bioinformatics, luciferase assays, and western blotting. Cholesterol levels were measured, and in vitro and in vivo studies assessed the therapeutic potential of combining miR-181a with crizotinib.</p><p><strong>Results: </strong>miR-181a significantly decreases UM cell migration, invasion, and metastasis. Mechanistically, miR-181a was found to target sterol regulatory element-binding protein 2 (SREBP2), thereby inhibiting cholesterol biosynthesis. This decrease in cholesterol levels hindered reduced epithelial-to-mesenchymal transition (EMT) and led to a decline in cancer stem-like cell (CSC) populations in UM. Furthermore, elevated cholesterol or overexpression of SREBP2 abrogated the anti-metastatic effects of miR-181a. Additionally, a combination of miR-181a and crizotinib significantly inhibited metastasis, both in vitro and in vivo.</p><p><strong>Conclusions: </strong>miR-181a inhibits UM metastasis by targeting SREBP2 and reducing cholesterol biosynthesis. Its combination with crizotinib may provide a promising therapeutic strategy for metastatic UM.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"215"},"PeriodicalIF":11.4,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12275384/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144668838","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Devayani Machiraju, Christian H Ziener, Elena Clementi, Francisco García-Asencio, Jennifer Hüllein, Jasmin Richter, Bénédicte Lenoir, Melanie Wiecken, Daniel Hübschmann, Dirk Jäger, Jessica C Hassel
{"title":"Monitoring soluble cMET and ctDNA in metastatic uveal melanoma patients to track early disease progression on immunotherapies.","authors":"Devayani Machiraju, Christian H Ziener, Elena Clementi, Francisco García-Asencio, Jennifer Hüllein, Jasmin Richter, Bénédicte Lenoir, Melanie Wiecken, Daniel Hübschmann, Dirk Jäger, Jessica C Hassel","doi":"10.1186/s13046-025-03451-2","DOIUrl":"10.1186/s13046-025-03451-2","url":null,"abstract":"<p><strong>Background: </strong>Metastatic uveal melanoma (mUM) is a rare malignancy and is different from metastatic cutaneous melanoma (mCM) in tumor characteristics and efficacy to immunotherapies. Tumor-specific biomarkers are required for mUM patients to monitor early disease progression on immunotherapies.</p><p><strong>Methods: </strong>We investigated clinical characteristics such as liver tumor burden and routine blood tumor markers, including lactate dehydrogenase (LDH) and transaminases in patients with mUM and with liver metastasized cutaneous melanoma (LmCM), treated with immune checkpoint inhibitors (ICIs) between May 2013-February 2024. In addition, we analyzed soluble cMET (scMET) in serum samples from these patients along with a cohort of mCM patients without liver metastases (nLmCM) using ELISA. Circulating tumor DNA (ctDNA) in the plasma was analyzed using digital droplet PCR (ddPCR) in mUM patients receiving immunotherapies. scMET, ctDNA, and LDH combination was used to simultaneously monitor disease progression in ICI and tebentafusp-receiving mUM patients.</p><p><strong>Results: </strong>Sixty-nine patients with mUM and seventy-six patients with LmCM were treated with either anti-PD1 monotherapy (n = 69, 48%) or ipi + nivo combination therapy (n = 76, 52%). Irrespective of the type of melanoma and type of immunotherapy, patients with liver metastasis size greater than 8cm experienced rapid disease progression. ICI-treated mUM patients with increased LDH, aspartate aminotransferase (AST), alanine transaminase (ALT), scMET, ctDNA, and rapidly growing tumors were significantly associated with treatment resistance and shorter progression-free and overall survival (p < 0.05). scMET (AUC: 0.82) outperforms LDH (AUC: 0.77) and S100 (0.68) in predicting one-year overall survival in these patients. A validation set with LmCM and nLmCM patient samples showed that increased scMET is likely a mUM-specific feature and does not predict ICI outcomes in LmCM or nLmCM patients (p > 0.05). Moreover, monitoring ctDNA and scMET in mUM patients under ICIs or tebentafusp treatment revealed the potential for early detection of disease progression.</p><p><strong>Conclusion: </strong>Soluble cMET might serve as a tumor-specific biomarker to predict clinical outcomes in mUM patients. A combinational assessment of scMET and ctDNA in mUM patients' blood offers a highly sensitive potential approach to monitor early disease progression under immunotherapies with ICI or tebentafusp.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"213"},"PeriodicalIF":11.4,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12275281/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144668839","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}