首页 > 最新文献

Molecular Cancer最新文献

英文 中文
Microbiome-targeted nanoplatforms and engineering approaches in breast cancer therapy 靶向微生物组的纳米平台和乳腺癌治疗的工程方法
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-30 DOI: 10.1186/s12943-025-02456-x
Zufa Sabeel, Zhao Yang
Breast cancer (BC) presents persistent therapeutic challenges due to tumor heterogeneity, therapy resistance, and immune dysfunction, further compounded by emerging evidence of microbiome involvement in disease progression. Pathogenic bacteria such as Fusobacterium nucleatum have been implicated in modulating immune evasion, remodeling the extracellular matrix, and influencing therapy outcomes. Conventional microbiome-modulating strategies (e.g., antibiotics, probiotics) suffer from non-specificity and potential dysbiosis. Nanomaterial (NM)-based microbiome modulation offers a transformative alternative by enabling: targeted elimination of tumor-associated pathogens (e.g., Gal/GalNAc-functionalized nanoparticles for F. nucleatum), bacterial membrane-coated nanoplatforms for precision antimicrobial delivery, and multifunctional nanosystems that modulate the tumor microenvironment (TME) while preserving beneficial microbiota. This review explores NM-based microbiome modulation across BC contexts, including localized drug delivery, immunomodulation, and microbial niche editing. Highlighted strategies include lipid-based antimicrobial nanoparticles (NP), metallic NPs enhancing therapy sensitivity, and Traditional Chinese Medicine (TCM)-inspired nanoformulations for microbiome balance. Synergies with chemotherapy, immunotherapy, and radiotherapy are evaluated, alongside preclinical findings demonstrating improved tumor control and microbiome resilience. Finally, challenges in clinical translation, including toxicity, immunogenicity, and scalability, are discussed, with future directions emphasizing smart nanosystems capable of microbiome-responsive, biomarker-guided, and immune-integrated interventions in BC therapy.
由于肿瘤异质性、治疗耐药性和免疫功能障碍,乳腺癌(BC)呈现出持续的治疗挑战,微生物组参与疾病进展的新证据进一步加剧了这一挑战。致病菌如核梭杆菌参与调节免疫逃避、重塑细胞外基质和影响治疗结果。传统的微生物组调节策略(如抗生素、益生菌)存在非特异性和潜在的生态失调。基于纳米材料(NM)的微生物组调节提供了一种变革性的替代方案,通过实现:靶向消除肿瘤相关病原体(例如,针对具核梭菌的Gal/ galnac功能化纳米颗粒),用于精确抗菌递送的细菌膜包被纳米平台,以及在保存有益微生物群的同时调节肿瘤微环境(TME)的多功能纳米系统。这篇综述探讨了在BC环境下基于纳米颗粒的微生物组调节,包括局部药物传递、免疫调节和微生物生态位编辑。重点强调的策略包括基于脂质的抗菌纳米颗粒(NP),增强治疗敏感性的金属纳米颗粒,以及用于微生物组平衡的中药纳米配方。与化疗、免疫治疗和放疗的协同作用进行了评估,同时临床前研究结果显示肿瘤控制和微生物组恢复能力得到改善。最后,讨论了临床转化的挑战,包括毒性、免疫原性和可扩展性,未来的方向是强调能够在BC治疗中对微生物组反应、生物标志物引导和免疫整合干预的智能纳米系统。
{"title":"Microbiome-targeted nanoplatforms and engineering approaches in breast cancer therapy","authors":"Zufa Sabeel, Zhao Yang","doi":"10.1186/s12943-025-02456-x","DOIUrl":"https://doi.org/10.1186/s12943-025-02456-x","url":null,"abstract":"Breast cancer (BC) presents persistent therapeutic challenges due to tumor heterogeneity, therapy resistance, and immune dysfunction, further compounded by emerging evidence of microbiome involvement in disease progression. Pathogenic bacteria such as Fusobacterium nucleatum have been implicated in modulating immune evasion, remodeling the extracellular matrix, and influencing therapy outcomes. Conventional microbiome-modulating strategies (e.g., antibiotics, probiotics) suffer from non-specificity and potential dysbiosis. Nanomaterial (NM)-based microbiome modulation offers a transformative alternative by enabling: targeted elimination of tumor-associated pathogens (e.g., Gal/GalNAc-functionalized nanoparticles for F. nucleatum), bacterial membrane-coated nanoplatforms for precision antimicrobial delivery, and multifunctional nanosystems that modulate the tumor microenvironment (TME) while preserving beneficial microbiota. This review explores NM-based microbiome modulation across BC contexts, including localized drug delivery, immunomodulation, and microbial niche editing. Highlighted strategies include lipid-based antimicrobial nanoparticles (NP), metallic NPs enhancing therapy sensitivity, and Traditional Chinese Medicine (TCM)-inspired nanoformulations for microbiome balance. Synergies with chemotherapy, immunotherapy, and radiotherapy are evaluated, alongside preclinical findings demonstrating improved tumor control and microbiome resilience. Finally, challenges in clinical translation, including toxicity, immunogenicity, and scalability, are discussed, with future directions emphasizing smart nanosystems capable of microbiome-responsive, biomarker-guided, and immune-integrated interventions in BC therapy.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"108 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145397456","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}
引用次数: 0
Context-dependent impact of type I interferon signaling in cancer I型干扰素信号在癌症中的环境依赖性影响
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-30 DOI: 10.1186/s12943-025-02495-4
Ainhoa Ruiz-Iglesias, Emma Guilbaud, Lorenzo Galluzzi, Santos Mañes
Type I interferon (IFN) has long been known as a critical component of the molecular machinery that first responds to viral infection in multicellular eukaryotes. More recently, type I IFN signaling has also emerged as a common process in the microenvironment of naturally developing neoplasms, as well as tumors responding to (immuno)therapy. In this setting, robust, acute but ultimately resolving type I IFN responses appear to support natural and therapy-driven cancer immunosurveillance by a number of mechanisms, including an accrued propensity of malignant cells to arrest their proliferation and undergo apoptotic cell death, as well as broad immunostimulatory effects on CD8+ cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, dendritic cells (DCs) and tumor-associated macrophages (TAMs). Conversely, weak, indolent and ultimately non-resolving type I IFN responses de facto facilitate tumor progression, not only by promoting stemness in malignant cells (which is associated with increased metastatic dissemination and resistance to therapy), but also by favoring the establishment of a highly immunosuppressive lymphoid and myeloid tumor microenvironment. Here, we provide a critical discussion of the context-dependent impact of type I IFN signaling on cancer progression and response to treatment, focusing on the tumor-intrinsic and extrinsic factors that may account for such a heterogeneity.
I型干扰素(IFN)长期以来一直被认为是多细胞真核生物中首先对病毒感染作出反应的分子机制的关键组成部分。最近,I型IFN信号也在自然发展的肿瘤微环境中作为一个共同的过程出现,以及肿瘤对(免疫)治疗的反应。在这种情况下,强大、急性但最终解决的I型IFN反应似乎通过多种机制支持自然和治疗驱动的癌症免疫监测,包括恶性细胞阻止其增殖和凋亡细胞死亡的累积倾向,以及对CD8+细胞毒性T淋巴细胞(ctl)、自然杀伤细胞(NK)细胞、树突状细胞(DCs)和肿瘤相关巨噬细胞(tam)的广泛免疫刺激作用。相反,弱的、惰性的和最终不解决的I型IFN反应事实上促进肿瘤进展,不仅通过促进恶性细胞的干性(这与转移性传播增加和对治疗的抵抗有关),而且还有利于建立高度免疫抑制的淋巴和髓系肿瘤微环境。在这里,我们对I型IFN信号对癌症进展和治疗反应的环境依赖性影响进行了关键讨论,重点关注可能导致这种异质性的肿瘤内在和外在因素。
{"title":"Context-dependent impact of type I interferon signaling in cancer","authors":"Ainhoa Ruiz-Iglesias, Emma Guilbaud, Lorenzo Galluzzi, Santos Mañes","doi":"10.1186/s12943-025-02495-4","DOIUrl":"https://doi.org/10.1186/s12943-025-02495-4","url":null,"abstract":"Type I interferon (IFN) has long been known as a critical component of the molecular machinery that first responds to viral infection in multicellular eukaryotes. More recently, type I IFN signaling has also emerged as a common process in the microenvironment of naturally developing neoplasms, as well as tumors responding to (immuno)therapy. In this setting, robust, acute but ultimately resolving type I IFN responses appear to support natural and therapy-driven cancer immunosurveillance by a number of mechanisms, including an accrued propensity of malignant cells to arrest their proliferation and undergo apoptotic cell death, as well as broad immunostimulatory effects on CD8+ cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, dendritic cells (DCs) and tumor-associated macrophages (TAMs). Conversely, weak, indolent and ultimately non-resolving type I IFN responses de facto facilitate tumor progression, not only by promoting stemness in malignant cells (which is associated with increased metastatic dissemination and resistance to therapy), but also by favoring the establishment of a highly immunosuppressive lymphoid and myeloid tumor microenvironment. Here, we provide a critical discussion of the context-dependent impact of type I IFN signaling on cancer progression and response to treatment, focusing on the tumor-intrinsic and extrinsic factors that may account for such a heterogeneity.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"54 1","pages":""},"PeriodicalIF":37.3,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145397320","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}
引用次数: 0
Advances in engineering immune-tumor microenvironments on-a-chip: integrative microfluidic platforms for immunotherapy and drug discovery. 芯片上工程免疫肿瘤微环境的进展:免疫治疗和药物发现的集成微流控平台。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-28 DOI: 10.1186/s12943-025-02479-4
Farnaz Dabbagh Moghaddam,Ali Anvar,Ehsan Ilkhani,Delara Dadgar,Maedeh Rafiee,Najmeh Ranjbaran,Pejman Mortazavi,Seyed Majid Ghoreishian,Yun Suk Huh,Pooyan Makvandi
The design and application of microfluidic immune system-on-a-chip (ISOC) technology have played a critical role in cancer immunology and drug discovery over the past decades. The system provides a highly controlled and physiologically relevant platform for studying immune responses and therapeutic interventions. Emerging trends in 3D bioprinting, organoid fusion, and multi-organ systems-on-a-chip further expand the capabilities of ISOC by enabling systemic immune interactions and modeling of the tumor microenvironment. Despite these advances, scalability, standardization, and long-term immune cell viability remain significant challenges that must be addressed to fully harness the potential of ISOC in clinical applications. Overall, ISOC represents a transformative tool in cancer research, offering innovative solutions for immunotherapy trials, biomarker discovery, and precision medicine. Therefore, in this study, the role of ISOC in cancer immunotherapy was investigated, focusing on its ability to recapitulate primary and secondary immune functions, model immune-tumor interactions, and enhance screening and optimization of immune-based therapies. Device design and modeling strategies were also discussed, demonstrating how ISOC platforms simulate dynamic immune cell activity, cytokine signaling, and antigen presentation to improve drug efficacy assessments. The application of ISOC technology in drug discovery and its potential to accelerate clinical trials and develop personalized immunotherapy were further explored.
在过去的几十年里,微流控免疫系统芯片(ISOC)技术的设计和应用在癌症免疫学和药物发现方面发挥了关键作用。该系统为研究免疫反应和治疗干预提供了一个高度可控和生理相关的平台。3D生物打印、类器官融合和芯片上多器官系统的新兴趋势通过实现系统免疫相互作用和肿瘤微环境建模,进一步扩展了ISOC的能力。尽管取得了这些进展,但为了充分利用ISOC在临床应用中的潜力,可扩展性、标准化和长期免疫细胞活力仍然是必须解决的重大挑战。总的来说,ISOC代表了癌症研究的变革性工具,为免疫治疗试验、生物标志物发现和精准医学提供了创新的解决方案。因此,本研究探讨了ISOC在肿瘤免疫治疗中的作用,重点研究了ISOC在原发性和继发性免疫功能概括、免疫-肿瘤相互作用模型以及增强免疫治疗筛选和优化的能力。还讨论了设备设计和建模策略,展示了ISOC平台如何模拟动态免疫细胞活性、细胞因子信号传导和抗原呈递,以改善药物疗效评估。进一步探讨了ISOC技术在药物发现中的应用及其在加速临床试验和开发个性化免疫治疗方面的潜力。
{"title":"Advances in engineering immune-tumor microenvironments on-a-chip: integrative microfluidic platforms for immunotherapy and drug discovery.","authors":"Farnaz Dabbagh Moghaddam,Ali Anvar,Ehsan Ilkhani,Delara Dadgar,Maedeh Rafiee,Najmeh Ranjbaran,Pejman Mortazavi,Seyed Majid Ghoreishian,Yun Suk Huh,Pooyan Makvandi","doi":"10.1186/s12943-025-02479-4","DOIUrl":"https://doi.org/10.1186/s12943-025-02479-4","url":null,"abstract":"The design and application of microfluidic immune system-on-a-chip (ISOC) technology have played a critical role in cancer immunology and drug discovery over the past decades. The system provides a highly controlled and physiologically relevant platform for studying immune responses and therapeutic interventions. Emerging trends in 3D bioprinting, organoid fusion, and multi-organ systems-on-a-chip further expand the capabilities of ISOC by enabling systemic immune interactions and modeling of the tumor microenvironment. Despite these advances, scalability, standardization, and long-term immune cell viability remain significant challenges that must be addressed to fully harness the potential of ISOC in clinical applications. Overall, ISOC represents a transformative tool in cancer research, offering innovative solutions for immunotherapy trials, biomarker discovery, and precision medicine. Therefore, in this study, the role of ISOC in cancer immunotherapy was investigated, focusing on its ability to recapitulate primary and secondary immune functions, model immune-tumor interactions, and enhance screening and optimization of immune-based therapies. Device design and modeling strategies were also discussed, demonstrating how ISOC platforms simulate dynamic immune cell activity, cytokine signaling, and antigen presentation to improve drug efficacy assessments. The application of ISOC technology in drug discovery and its potential to accelerate clinical trials and develop personalized immunotherapy were further explored.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"51 1","pages":"271"},"PeriodicalIF":37.3,"publicationDate":"2025-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145381074","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}
引用次数: 0
IFITM3-MET interaction drives osimertinib resistance through AKT pathway activation in EGFR-mutant non-small cell lung cancer. 在egfr突变的非小细胞肺癌中,IFITM3-MET相互作用通过AKT通路激活驱动奥西替尼耐药。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-28 DOI: 10.1186/s12943-025-02493-6
Ritsu Ibusuki,Eiji Iwama,Atsushi Shimauchi,Hiromu Kawano,Shun Mizusaki,Satoshi Nakamura,Yui Miyazaki,Yu Inutsuka,Mikiko Hashisako,Taishi Harada,Yuko Tsuchiya-Kawano,Hirono Tsutsumi,Takayuki Nakanishi,Noriaki Nakagaki,Yuichiro Koga,Shinichi Kimura,Shun Mashimoto,Daisuke Shibahara,Kohei Otsubo,Yasuto Yoneshima,Kentaro Tanaka,Yoshinao Oda,Isamu Okamoto
BACKGROUNDDespite an initial favorable response of EGFR-mutant non-small cell lung cancer (NSCLC) to osimertinib, an EGFR tyrosine kinase inhibitor (TKI), resistance to this drug inevitably develops. Whereas genetic mechanisms for such acquired resistance have been identified, the molecular mediators of resistance induction have remained unclear.METHODSTo identify factors that mediate induction of osimertinib resistance, we studied clinical samples from individuals with EGFR-mutant NSCLC as well as cell lines including PC-9 and H1975. Methods adopted included transcriptomics analysis and immunohistochemistry of pretreatment NSCLC specimens, spatial transcriptomics analysis, a cell viability assay, immunofluorescence and quantitative PCR analysis, RNA sequencing, immunoblot analysis, comprehensive proteomics analysis by mass spectrometry, co-immunoprecipitation and proximity ligation assays, and a mouse xenograft tumor model.RESULTSTranscriptomics analysis of pretreatment clinical specimens identified IFITM3 (interferon-induced transmembrane protein 3) as a gene specifically upregulated in patients with a poor response to osimertinib treatment. Immunohistochemistry confirmed that patients with IFITM3-positive tumors experienced a shorter progression-free survival on osimertinib treatment. Spatial transcriptomics and other analyses further revealed that IFITM3 expression in tumor cells was increased in response to cytokines derived from the tumor microenvironment (TME) during osimertinib treatment. IFITM3 was found to promote the development of osimertinib resistance in NSCLC cell lines through interaction with MET and activation of the AKT signaling pathway. Furthermore, combined treatment with a MET inhibitor suppressed the development of osimertinib resistance in a mouse xenograft tumor model.CONCLUSIONSOur findings reveal that upregulation of IFITM3 driven by TME cytokines represents a previously unrecognized mechanism of osimertinib resistance, and they suggest that targeting of the IFITM3-MET axis may improve EGFR-TKI treatment outcome for EGFR-mutant NSCLC.
背景:尽管EGFR突变的非小细胞肺癌(NSCLC)对奥西替尼(一种EGFR酪氨酸激酶抑制剂(TKI))最初有良好的反应,但不可避免地会产生对这种药物的耐药性。虽然这种获得性耐药的遗传机制已经确定,但诱导耐药的分子介质仍然不清楚。方法为了确定介导奥西替尼耐药的因素,我们研究了egfr突变的非小细胞肺癌个体以及PC-9和H1975细胞系的临床样本。采用的方法包括预处理NSCLC标本的转录组学和免疫组织化学分析、空间转录组学分析、细胞活力测定、免疫荧光和定量PCR分析、RNA测序、免疫印迹分析、质谱综合蛋白质组学分析、共免疫沉淀和近端结扎试验以及小鼠异种移植肿瘤模型。结果对预处理临床标本进行转录组学分析,发现干扰素诱导的跨膜蛋白3 (IFITM3)基因在对奥希替尼治疗反应不佳的患者中特异性上调。免疫组织化学证实,ifitm3阳性肿瘤患者接受奥西替尼治疗后的无进展生存期较短。空间转录组学和其他分析进一步显示,在奥西替尼治疗期间,肿瘤细胞中IFITM3的表达增加,以响应肿瘤微环境(TME)衍生的细胞因子。IFITM3通过与MET相互作用和激活AKT信号通路,促进NSCLC细胞系对奥西替尼的耐药。此外,与MET抑制剂联合治疗可以抑制小鼠异种移植肿瘤模型中奥西替尼耐药性的发展。我们的研究结果表明,由TME细胞因子驱动的IFITM3上调代表了一种以前未被认识的奥西替尼耐药机制,并且他们表明靶向IFITM3- met轴可能改善EGFR-TKI治疗egfr -突变型NSCLC的结果。
{"title":"IFITM3-MET interaction drives osimertinib resistance through AKT pathway activation in EGFR-mutant non-small cell lung cancer.","authors":"Ritsu Ibusuki,Eiji Iwama,Atsushi Shimauchi,Hiromu Kawano,Shun Mizusaki,Satoshi Nakamura,Yui Miyazaki,Yu Inutsuka,Mikiko Hashisako,Taishi Harada,Yuko Tsuchiya-Kawano,Hirono Tsutsumi,Takayuki Nakanishi,Noriaki Nakagaki,Yuichiro Koga,Shinichi Kimura,Shun Mashimoto,Daisuke Shibahara,Kohei Otsubo,Yasuto Yoneshima,Kentaro Tanaka,Yoshinao Oda,Isamu Okamoto","doi":"10.1186/s12943-025-02493-6","DOIUrl":"https://doi.org/10.1186/s12943-025-02493-6","url":null,"abstract":"BACKGROUNDDespite an initial favorable response of EGFR-mutant non-small cell lung cancer (NSCLC) to osimertinib, an EGFR tyrosine kinase inhibitor (TKI), resistance to this drug inevitably develops. Whereas genetic mechanisms for such acquired resistance have been identified, the molecular mediators of resistance induction have remained unclear.METHODSTo identify factors that mediate induction of osimertinib resistance, we studied clinical samples from individuals with EGFR-mutant NSCLC as well as cell lines including PC-9 and H1975. Methods adopted included transcriptomics analysis and immunohistochemistry of pretreatment NSCLC specimens, spatial transcriptomics analysis, a cell viability assay, immunofluorescence and quantitative PCR analysis, RNA sequencing, immunoblot analysis, comprehensive proteomics analysis by mass spectrometry, co-immunoprecipitation and proximity ligation assays, and a mouse xenograft tumor model.RESULTSTranscriptomics analysis of pretreatment clinical specimens identified IFITM3 (interferon-induced transmembrane protein 3) as a gene specifically upregulated in patients with a poor response to osimertinib treatment. Immunohistochemistry confirmed that patients with IFITM3-positive tumors experienced a shorter progression-free survival on osimertinib treatment. Spatial transcriptomics and other analyses further revealed that IFITM3 expression in tumor cells was increased in response to cytokines derived from the tumor microenvironment (TME) during osimertinib treatment. IFITM3 was found to promote the development of osimertinib resistance in NSCLC cell lines through interaction with MET and activation of the AKT signaling pathway. Furthermore, combined treatment with a MET inhibitor suppressed the development of osimertinib resistance in a mouse xenograft tumor model.CONCLUSIONSOur findings reveal that upregulation of IFITM3 driven by TME cytokines represents a previously unrecognized mechanism of osimertinib resistance, and they suggest that targeting of the IFITM3-MET axis may improve EGFR-TKI treatment outcome for EGFR-mutant NSCLC.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"21 1","pages":"272"},"PeriodicalIF":37.3,"publicationDate":"2025-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145381075","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}
引用次数: 0
Correction: ZEB1 transcriptionally regulated carbonic anhydrase 9 mediates the chemoresistance of tongue cancer via maintaining intracellular pH. 更正:ZEB1转录调节的碳酸酐酶9通过维持细胞内pH介导舌癌的化疗耐药。
IF 33.9 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-28 DOI: 10.1186/s12943-025-02504-6
Guopei Zheng, Cong Peng, Xiaoting Jia, Yixue Gu, Zhijie Zhang, Yingen Deng, Chengkun Wang, Nan Li, Jiang Yin, Xiaorong Liu, Minying Lu, Hailin Tang, Zhimin He
{"title":"Correction: ZEB1 transcriptionally regulated carbonic anhydrase 9 mediates the chemoresistance of tongue cancer via maintaining intracellular pH.","authors":"Guopei Zheng, Cong Peng, Xiaoting Jia, Yixue Gu, Zhijie Zhang, Yingen Deng, Chengkun Wang, Nan Li, Jiang Yin, Xiaorong Liu, Minying Lu, Hailin Tang, Zhimin He","doi":"10.1186/s12943-025-02504-6","DOIUrl":"10.1186/s12943-025-02504-6","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"24 1","pages":"270"},"PeriodicalIF":33.9,"publicationDate":"2025-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12560301/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145391637","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}
引用次数: 0
Safe delivery of a highly toxic anthracycline derivative through liposomal nanoformulation achieves complete cancer regression. 通过脂质体纳米配方安全递送高毒性蒽环类衍生物,实现完全的癌症消退。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 DOI: 10.1186/s12943-025-02444-1
András Füredi,Szilárd Tóth,Kristóf Hegedüs,Pál T Szabó,Anikó Gaál,Gergő Barta,Lívia N Naszályi,Krisztina Kiss,Kata Bölcskei,Zoltán Szeltner,Eszter Bajtai,Balázs Gombos,Dániel Kiss,Mihály T Cserepes,Attila Kiss,Peter Pokreisz,Lukas Kenner,Sandra Högler,Csaba Magyar,Jamie D Cowles,Agnes Csiszar,József Tóvári,Dávid Szüts,Zsuzsanna Helyes,Zoltán Varga,Gábor Mező,Gergely Szakács
BACKGROUNDChemotherapy remains the cornerstone of cancer treatment despite its well-documented challenges, including toxic side effects and drug resistance. Here, we demonstrate that a novel, highly toxic, daunosamine-modified derivative of daunorubicin (2-pyrrolino-daunorubicin, PyDau) can be safely administered to mice when encapsulated in liposome.METHODSPyDau was synthesized from daunorubicin in a one-step reaction. Its increased in vitro cytotoxicity was confirmed across 42 human cell lines representing 12 cancer types, including multidrug resistant cells. The activity profile of this new derivative was analyzed in the context of 13 commonly used cancer drugs across a panel of lymphoblast cell lines missing individual components of DNA-repair enzymes. To enable in vivo application, PyDau was encapsulated in pegylated liposome, resulting in liposomal PyDau (LiPyDau). In vivo efficacy of LiPyDau was evaluated in three allograft models (melanoma, breast, lung), a xenograft model (uterine sarcoma), a patient-derived xenograft model (lung), and a genetically engineered mouse model of mammary cancer, including two models of drug resistance.RESULTSWhile PyDau exhibited up to 1000-fold greater cytotoxicity than daunomycin and doxorubicin against cancer cell lines, its in vivo application was hindered by an extremely narrow therapeutic window. Liposomal nanoformulation mitigated the limiting toxicity, allowing LiPyDau to be tested in preclinical allograft and xenograft mouse models. LiPyDau demonstrated robust efficacy across all models including multidrug-resistant cancer, completely eradicating tumors in a genetically engineered mouse model of triple-negative breast cancer. LiPyDau exerts its anticancer effect through a unique mechanism involving the crosslinking of complementary DNA strands, resulting in irreversible DNA damage.CONCLUSIONLiposomal formulations of extremely cytotoxic anthracycline analogs, such as LiPyDau, represent a promising and highly effective therapeutic approach for combating drug resistant cancer.
背景:化疗仍然是癌症治疗的基石,尽管它有很多挑战,包括毒副作用和耐药性。在这里,我们证明了一种新的,高毒性的,丹诺胺修饰的柔红霉素衍生物(2-吡咯利诺-柔红霉素,PyDau)可以安全地给药于脂质体小鼠。方法以柔红霉素为原料,一步法合成spydau。在代表12种癌症类型的42种人类细胞系(包括多药耐药细胞)中证实了其体外细胞毒性的增加。在缺少dna修复酶单个组分的淋巴母细胞系中,在13种常用抗癌药物的背景下分析了这种新衍生物的活性谱。为了使体内应用,PyDau被封装在聚乙二醇化脂质体中,形成脂质体PyDau (LiPyDau)。LiPyDau在三种异体移植模型(黑色素瘤、乳腺、肺)、异种移植模型(子宫肉瘤)、患者来源的异种移植模型(肺)和基因工程小鼠乳腺癌模型(包括两种耐药模型)中进行了体内疗效评估。结果PyDau对癌细胞的细胞毒性比道诺霉素和阿霉素高1000倍,但其在体内的应用受到极窄的治疗窗口的阻碍。脂质体纳米制剂减轻了局限性毒性,使LiPyDau能够在临床前同种异体移植和异种移植小鼠模型中进行测试。LiPyDau在包括耐多药癌症在内的所有模型中显示出强大的疗效,在基因工程小鼠三阴性乳腺癌模型中完全根除肿瘤。LiPyDau通过一种独特的机制发挥其抗癌作用,该机制涉及互补DNA链的交联,导致不可逆的DNA损伤。结论极具细胞毒性的蒽环类药物脂质体制剂,如LiPyDau,是治疗耐药癌症的有效途径。
{"title":"Safe delivery of a highly toxic anthracycline derivative through liposomal nanoformulation achieves complete cancer regression.","authors":"András Füredi,Szilárd Tóth,Kristóf Hegedüs,Pál T Szabó,Anikó Gaál,Gergő Barta,Lívia N Naszályi,Krisztina Kiss,Kata Bölcskei,Zoltán Szeltner,Eszter Bajtai,Balázs Gombos,Dániel Kiss,Mihály T Cserepes,Attila Kiss,Peter Pokreisz,Lukas Kenner,Sandra Högler,Csaba Magyar,Jamie D Cowles,Agnes Csiszar,József Tóvári,Dávid Szüts,Zsuzsanna Helyes,Zoltán Varga,Gábor Mező,Gergely Szakács","doi":"10.1186/s12943-025-02444-1","DOIUrl":"https://doi.org/10.1186/s12943-025-02444-1","url":null,"abstract":"BACKGROUNDChemotherapy remains the cornerstone of cancer treatment despite its well-documented challenges, including toxic side effects and drug resistance. Here, we demonstrate that a novel, highly toxic, daunosamine-modified derivative of daunorubicin (2-pyrrolino-daunorubicin, PyDau) can be safely administered to mice when encapsulated in liposome.METHODSPyDau was synthesized from daunorubicin in a one-step reaction. Its increased in vitro cytotoxicity was confirmed across 42 human cell lines representing 12 cancer types, including multidrug resistant cells. The activity profile of this new derivative was analyzed in the context of 13 commonly used cancer drugs across a panel of lymphoblast cell lines missing individual components of DNA-repair enzymes. To enable in vivo application, PyDau was encapsulated in pegylated liposome, resulting in liposomal PyDau (LiPyDau). In vivo efficacy of LiPyDau was evaluated in three allograft models (melanoma, breast, lung), a xenograft model (uterine sarcoma), a patient-derived xenograft model (lung), and a genetically engineered mouse model of mammary cancer, including two models of drug resistance.RESULTSWhile PyDau exhibited up to 1000-fold greater cytotoxicity than daunomycin and doxorubicin against cancer cell lines, its in vivo application was hindered by an extremely narrow therapeutic window. Liposomal nanoformulation mitigated the limiting toxicity, allowing LiPyDau to be tested in preclinical allograft and xenograft mouse models. LiPyDau demonstrated robust efficacy across all models including multidrug-resistant cancer, completely eradicating tumors in a genetically engineered mouse model of triple-negative breast cancer. LiPyDau exerts its anticancer effect through a unique mechanism involving the crosslinking of complementary DNA strands, resulting in irreversible DNA damage.CONCLUSIONLiposomal formulations of extremely cytotoxic anthracycline analogs, such as LiPyDau, represent a promising and highly effective therapeutic approach for combating drug resistant cancer.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"57 1","pages":"269"},"PeriodicalIF":37.3,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145374017","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}
引用次数: 0
Orchestrating movement: the role of Caveolin-1 in migration and metastasis. 协调运动:小窝蛋白-1在迁移和转移中的作用。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-23 DOI: 10.1186/s12943-025-02469-6
Jiri Navratil,Martina Raudenska,Monika Kratochvilova,Jan Balvan,Yoav David Shaul,Michal Masarik
{"title":"Orchestrating movement: the role of Caveolin-1 in migration and metastasis.","authors":"Jiri Navratil,Martina Raudenska,Monika Kratochvilova,Jan Balvan,Yoav David Shaul,Michal Masarik","doi":"10.1186/s12943-025-02469-6","DOIUrl":"https://doi.org/10.1186/s12943-025-02469-6","url":null,"abstract":"","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"54 1","pages":"267"},"PeriodicalIF":37.3,"publicationDate":"2025-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145339187","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}
引用次数: 0
Efferocytosis: the art of cellular clearance and novel perspectives in disease therapy. Efferocytosis:细胞清除的艺术和疾病治疗的新观点。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-23 DOI: 10.1186/s12943-025-02484-7
Gege Li,Jiashuai Xu,Xiaohan Tian,Jingyi Xiao,Junqi Long,Yining Chen,Wenzhi Shen,Shuangtao Zhao
Efferocytosis, the process of apoptotic cell clearance, is a fundamental biological mechanism for maintaining tissue homeostasis. However, its role in disease pathogenesis is often oversimplified, neglecting a critical knowledge gap: how the single process could drive opposing pathological outcomes. This review provides a comprehensive analysis centered on the functional duality of efferocytosis. By synthesizing evidence across a spectrum of human pathologies-from atherosclerosis and neurodegeneration to cancer-we establish a core paradigm: impaired efferocytosis is a central pathogenic driver in chronic inflammatory and autoimmune diseases, leading to unresolved inflammation. Conversely, the hijacking of efferocytosis by tumors fosters an immunosuppressive microenvironment, facilitating immune evasion. This dichotomy presents a significant therapeutic conundrum, as enhancing efferocytosis benefits inflammatory conditions but exacerbates cancer. By dissecting these context-dependent mechanisms, we argue that the future of efferocytosis-based medicine hinges on developing targeted, disease-specific strategies to safely harness this powerful biological process.
Efferocytosis是凋亡细胞清除的过程,是维持组织稳态的基本生物学机制。然而,其在疾病发病机制中的作用往往被过度简化,忽视了一个关键的知识缺口:单一的过程如何驱动相反的病理结果。本文综述了以卵泡细胞增多症的功能二重性为中心的综合分析。通过综合人类病理谱的证据——从动脉粥样硬化、神经退行性变到癌症——我们建立了一个核心范式:efferocytosis受损是慢性炎症和自身免疫性疾病的核心致病驱动因素,导致未解决的炎症。相反,肿瘤对efferocytosis的劫持促进了免疫抑制微环境,促进了免疫逃避。这种二分法提出了一个重要的治疗难题,因为增强efferocytosis有利于炎症条件,但加剧了癌症。通过剖析这些依赖于环境的机制,我们认为基于efferocysis的医学的未来取决于开发有针对性的,疾病特异性的策略来安全地利用这一强大的生物过程。
{"title":"Efferocytosis: the art of cellular clearance and novel perspectives in disease therapy.","authors":"Gege Li,Jiashuai Xu,Xiaohan Tian,Jingyi Xiao,Junqi Long,Yining Chen,Wenzhi Shen,Shuangtao Zhao","doi":"10.1186/s12943-025-02484-7","DOIUrl":"https://doi.org/10.1186/s12943-025-02484-7","url":null,"abstract":"Efferocytosis, the process of apoptotic cell clearance, is a fundamental biological mechanism for maintaining tissue homeostasis. However, its role in disease pathogenesis is often oversimplified, neglecting a critical knowledge gap: how the single process could drive opposing pathological outcomes. This review provides a comprehensive analysis centered on the functional duality of efferocytosis. By synthesizing evidence across a spectrum of human pathologies-from atherosclerosis and neurodegeneration to cancer-we establish a core paradigm: impaired efferocytosis is a central pathogenic driver in chronic inflammatory and autoimmune diseases, leading to unresolved inflammation. Conversely, the hijacking of efferocytosis by tumors fosters an immunosuppressive microenvironment, facilitating immune evasion. This dichotomy presents a significant therapeutic conundrum, as enhancing efferocytosis benefits inflammatory conditions but exacerbates cancer. By dissecting these context-dependent mechanisms, we argue that the future of efferocytosis-based medicine hinges on developing targeted, disease-specific strategies to safely harness this powerful biological process.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"69 1","pages":"268"},"PeriodicalIF":37.3,"publicationDate":"2025-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145339407","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}
引用次数: 0
Novel molecular mechanisms of FLT3 deregulation: from the acute myeloid leukemia experience to therapeutic insights in acute lymphoblastic leukemia. FLT3解除调控的新分子机制:从急性髓性白血病经验到急性淋巴细胞白血病的治疗见解。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-22 DOI: 10.1186/s12943-025-02455-y
F Lo Schiavo,C Salvesi,M Jandoubi,F Pirini,J Garbetta,G Martinelli,G Simonetti,A Ferrari
Fms-like tyrosine kinase 3 (FLT3), a class III receptor tyrosine kinase essential for hematopoiesis, is a well-established oncogenic driver in acute myeloid leukemia (AML). Canonical internal tandem duplications (ITD) and tyrosine kinase domain (TKD) mutations inform prognosis and guide targeted therapy. Recent evidence highlights FLT3 as a critical oncogenic hub in acute lymphoblastic leukemia (ALL), where its alterations extend beyond ITD/TKD mutations to include non-canonical mutations with only partially explored functional implications. Moreover, recently discovered regulatory mechanisms, mostly acting on the FLT3 locus, drive FLT3 overexpression in ALL, including transcriptional regulation by rearranged ZNF384, epigenetic modifications, novel circular-RNA URAD::FLT3 fusions, and 13q12.2 deletions leading to enhancer hijacking and topologically associated domain (TAD)-boundary disruptions. The impact of these alterations on leukemogenesis and the possibility to target them in ALL subtypes is discussed here. Data from the Functional Omics Resource of Acute Lymphoblastic Leukemia (FORALL) across B- and T-ALL cell line subtypes drug screening, and from preclinical and clinical evidence reveals a variable efficacy in FLT3-mutated and FLT3-overexpressing ALL subtypes, supporting a molecularly guided treatment approach. Building on the success of FLT3 inhibitors in mutated AML and in light of the emerging results in patients lacking FLT3-ITD and in FLT3-like AML cases, presenting with a gene expression pattern similar to FLT3-mutated ones despite the absence of mutations, we discuss their potential in ALL and we consider novel therapeutic strategies, including new FLT3 inhibitors, antibody-based approaches, FLT3 CAR-T therapy, and synergistic drug combinations, such as FLT3 and BCL2 inhibition. These new insights reviewed here may redefine FLT3 as a pan-leukemic target, with ALL-specific activation mechanisms offering unique therapeutic windows. The implementation of FLT3 expression profiling and full-coding mutation screening in ALL (and in AML) diagnostics could unlock precision medicine approaches. By bridging the AML experience with ALL innovations, this review outlines a roadmap for FLT3-targeted therapies and combination strategies, underscoring the urgency of biomarker-driven clinical trials to optimize FLT3-directed interventions in acute leukemias.
fms样酪氨酸激酶3 (FLT3)是造血所必需的III类受体酪氨酸激酶,是急性髓性白血病(AML)中公认的致癌驱动因子。典型内串联重复(ITD)和酪氨酸激酶结构域(TKD)突变影响预后和指导靶向治疗。最近的证据表明,FLT3在急性淋巴细胞白血病(ALL)中是一个关键的致癌中心,其改变超出了ITD/TKD突变,包括非典型突变,仅部分探索了功能意义。此外,最近发现的调控机制,主要作用于FLT3位点,驱动FLT3在ALL中的过表达,包括重排ZNF384的转录调控,表观遗传修饰,新型环状rna URAD::FLT3融合,13q12.2缺失导致增强子劫持和拓扑相关结构域(TAD)边界破坏。本文讨论了这些改变对白血病发生的影响以及在ALL亚型中靶向它们的可能性。来自急性淋巴母细胞白血病(FORALL)功能组学资源(Functional Omics Resource of Acute Lymphoblastic Leukemia, FORALL)跨B-和T-ALL细胞系亚型药物筛选,以及临床前和临床证据的数据显示,flt3突变和flt3过表达的ALL亚型的疗效不同,支持分子引导治疗方法。基于FLT3抑制剂在突变AML中的成功,以及在缺乏FLT3- itd的患者和FLT3样AML病例中出现的新结果,尽管没有突变,但表现出与FLT3突变相似的基因表达模式,我们讨论了它们在ALL中的潜力,并考虑了新的治疗策略,包括新的FLT3抑制剂,基于抗体的方法,FLT3 CAR-T治疗,以及协同药物组合,如FLT3和BCL2抑制。这里回顾的这些新见解可能重新定义FLT3作为泛白血病靶点,all特异性激活机制提供了独特的治疗窗口。在ALL(和AML)诊断中实施FLT3表达谱分析和全编码突变筛查可以开启精准医学方法。通过将AML经验与ALL创新相结合,本综述概述了flt3靶向治疗和联合策略的路线图,强调了生物标志物驱动的临床试验的紧迫性,以优化flt3导向的急性白血病干预措施。
{"title":"Novel molecular mechanisms of FLT3 deregulation: from the acute myeloid leukemia experience to therapeutic insights in acute lymphoblastic leukemia.","authors":"F Lo Schiavo,C Salvesi,M Jandoubi,F Pirini,J Garbetta,G Martinelli,G Simonetti,A Ferrari","doi":"10.1186/s12943-025-02455-y","DOIUrl":"https://doi.org/10.1186/s12943-025-02455-y","url":null,"abstract":"Fms-like tyrosine kinase 3 (FLT3), a class III receptor tyrosine kinase essential for hematopoiesis, is a well-established oncogenic driver in acute myeloid leukemia (AML). Canonical internal tandem duplications (ITD) and tyrosine kinase domain (TKD) mutations inform prognosis and guide targeted therapy. Recent evidence highlights FLT3 as a critical oncogenic hub in acute lymphoblastic leukemia (ALL), where its alterations extend beyond ITD/TKD mutations to include non-canonical mutations with only partially explored functional implications. Moreover, recently discovered regulatory mechanisms, mostly acting on the FLT3 locus, drive FLT3 overexpression in ALL, including transcriptional regulation by rearranged ZNF384, epigenetic modifications, novel circular-RNA URAD::FLT3 fusions, and 13q12.2 deletions leading to enhancer hijacking and topologically associated domain (TAD)-boundary disruptions. The impact of these alterations on leukemogenesis and the possibility to target them in ALL subtypes is discussed here. Data from the Functional Omics Resource of Acute Lymphoblastic Leukemia (FORALL) across B- and T-ALL cell line subtypes drug screening, and from preclinical and clinical evidence reveals a variable efficacy in FLT3-mutated and FLT3-overexpressing ALL subtypes, supporting a molecularly guided treatment approach. Building on the success of FLT3 inhibitors in mutated AML and in light of the emerging results in patients lacking FLT3-ITD and in FLT3-like AML cases, presenting with a gene expression pattern similar to FLT3-mutated ones despite the absence of mutations, we discuss their potential in ALL and we consider novel therapeutic strategies, including new FLT3 inhibitors, antibody-based approaches, FLT3 CAR-T therapy, and synergistic drug combinations, such as FLT3 and BCL2 inhibition. These new insights reviewed here may redefine FLT3 as a pan-leukemic target, with ALL-specific activation mechanisms offering unique therapeutic windows. The implementation of FLT3 expression profiling and full-coding mutation screening in ALL (and in AML) diagnostics could unlock precision medicine approaches. By bridging the AML experience with ALL innovations, this review outlines a roadmap for FLT3-targeted therapies and combination strategies, underscoring the urgency of biomarker-driven clinical trials to optimize FLT3-directed interventions in acute leukemias.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"82 1","pages":"266"},"PeriodicalIF":37.3,"publicationDate":"2025-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145339262","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}
引用次数: 0
Ex vivo modelling of human colorectal cancer liver metastasis by normothermic machine perfusion. 常温机器灌注造人大肠癌肝转移的体外模型。
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-21 DOI: 10.1186/s12943-025-02430-7
Manuel Trebo,Thomas Maurer,Felix J Krendl,Stefan Salcher,Agnieszka Martowicz,Theresa Hautz,Sieghart Sopper,Arno Amann,Benno Cardini,Lukas H Poelsler,Anna Mair,Julia Hofmann,Andras T Meszaros,Martin Hermann,Michael Günther,Steffen Ormanns,Zlatko Trajanoski,Stefan Schneeberger,Dominik Wolf,Rupert Oberhuber,Andreas Pircher
BACKGROUNDColorectal cancer liver metastasis (CRLM) is associated with poor survival, primarily due to acquired therapy resistance. While novel therapies arise, translation is limited by the lack of tumor models accurately representing dynamic microenvironmental interplay. Here, we show that ex vivo normothermic machine perfusion (NMP) offers a novel preclinical framework to study the intratumoral dynamics of CRLM biology.METHODSSix resected metastatic human livers were preserved for two days and subjected to multi-omic profiling of serially sampled adjacent liver and metastatic tissue using single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST). Tissue integrity was assessed and cross-validated by immunofluorescence (IF), high-resolution respirometry (HRR) and flow-cytometry.RESULTSNMP was successfuly applied to metastatic livers with minimal surgical adaptations, preserving both intrinsic hepatic properties and tissue viability over an extended duration. Single-cell and spatial mapping confirmed preservation of CRLM phenotypic properties and demonstrated high clinical translatability by applicability of the intrinsic epithelial consensus molecular subtypes to metastasis. Spatially deconvoluted pathway activities reflected functional tissue-microenvironments. Transcriptomic profiles - including those of tumor-associated myeloid cells - were preserved during NMP. Finally, we demonstrate tumor-associated myeloid cell persistence as a driver of disease progression and poor survival in colorectal cancer.CONCLUSIONOur findings represent the basis for future innovative applications adopting NMP in the context of CRLM, providing a new preclinical tumor model avenue.
结直肠癌肝转移(CRLM)与低生存率相关,主要是由于获得性治疗抵抗。虽然出现了新的治疗方法,但由于缺乏准确代表动态微环境相互作用的肿瘤模型,翻译受到限制。在这里,我们表明体外恒温机器灌注(NMP)为研究CRLM生物学的肿瘤内动力学提供了一个新的临床前框架。方法将6个切除的转移性人肝脏保存2天,并使用单细胞RNA测序(scRNA-seq)和空间转录组学(ST)对连续取样的邻近肝脏和转移组织进行多组学分析。通过免疫荧光(IF)、高分辨率呼吸仪(HRR)和流式细胞术评估和交叉验证组织完整性。结果snmp成功应用于转移性肝脏,手术适应性最小,在较长时间内保持肝脏固有特性和组织活力。单细胞和空间定位证实了CRLM表型特性的保存,并通过固有上皮一致分子亚型对转移的适用性证明了高临床可翻译性。空间解卷积通路活动反映了功能性组织微环境。转录组谱-包括肿瘤相关的髓样细胞-在NMP期间被保留。最后,我们证明了肿瘤相关的骨髓细胞持久性是结直肠癌疾病进展和低生存率的驱动因素。结论本研究结果为未来在CRLM中采用NMP的创新应用奠定了基础,为临床前肿瘤模型的建立提供了新的途径。
{"title":"Ex vivo modelling of human colorectal cancer liver metastasis by normothermic machine perfusion.","authors":"Manuel Trebo,Thomas Maurer,Felix J Krendl,Stefan Salcher,Agnieszka Martowicz,Theresa Hautz,Sieghart Sopper,Arno Amann,Benno Cardini,Lukas H Poelsler,Anna Mair,Julia Hofmann,Andras T Meszaros,Martin Hermann,Michael Günther,Steffen Ormanns,Zlatko Trajanoski,Stefan Schneeberger,Dominik Wolf,Rupert Oberhuber,Andreas Pircher","doi":"10.1186/s12943-025-02430-7","DOIUrl":"https://doi.org/10.1186/s12943-025-02430-7","url":null,"abstract":"BACKGROUNDColorectal cancer liver metastasis (CRLM) is associated with poor survival, primarily due to acquired therapy resistance. While novel therapies arise, translation is limited by the lack of tumor models accurately representing dynamic microenvironmental interplay. Here, we show that ex vivo normothermic machine perfusion (NMP) offers a novel preclinical framework to study the intratumoral dynamics of CRLM biology.METHODSSix resected metastatic human livers were preserved for two days and subjected to multi-omic profiling of serially sampled adjacent liver and metastatic tissue using single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST). Tissue integrity was assessed and cross-validated by immunofluorescence (IF), high-resolution respirometry (HRR) and flow-cytometry.RESULTSNMP was successfuly applied to metastatic livers with minimal surgical adaptations, preserving both intrinsic hepatic properties and tissue viability over an extended duration. Single-cell and spatial mapping confirmed preservation of CRLM phenotypic properties and demonstrated high clinical translatability by applicability of the intrinsic epithelial consensus molecular subtypes to metastasis. Spatially deconvoluted pathway activities reflected functional tissue-microenvironments. Transcriptomic profiles - including those of tumor-associated myeloid cells - were preserved during NMP. Finally, we demonstrate tumor-associated myeloid cell persistence as a driver of disease progression and poor survival in colorectal cancer.CONCLUSIONOur findings represent the basis for future innovative applications adopting NMP in the context of CRLM, providing a new preclinical tumor model avenue.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":"101 1","pages":"264"},"PeriodicalIF":37.3,"publicationDate":"2025-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145338655","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}
引用次数: 0
期刊
Molecular Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1