2025-04-20 2025, Volume 15 Issue 4

  • Select all
  • RESEARCH ARTICLE
    Yuchao Zhang , Asma Khanniche , Yizhe Li , Zhenchuan Wu , Hailong Wang , Hongyu Zhang , Xiaoxue Li , Landian Hu , Xiangyin Kong
    2025, 15(4): e70139. https://doi.org/10.1002/ctm2.70139

    Background: The IFN-γ cytokine plays a dual role in anti-tumor immunity, enhancing immune defense against cancer cells while promoting tumor survival and progression. Its influence on prognosis and therapeutic responses across cancer types remains unclear.

    Objective: This study aimed to perform a pan-cancer analysis of IFN-γ response genes to determine their prognostic significance and evaluate their impact on clinical outcomes and anti-PD1 immunotherapy responses.

    Methods: Using multiple datasets, 46 IFN-γ response genes were identified as prognostic for disease-specific survival, and their expression was used to construct the IFN-γ Response Gene Network Signature (IFGRNS) score. The prognostic and therapeutic relevance of the IFGRNS score was assessed across cancer types, considering tumor pathology, genomic alterations, tumor mutation burden, and microenvironment. Single-cell transcriptomic analysis identified cellular contributors, and a murine pancreatic cancer (PAN02) model was used to validate findings with anti-PD1 therapy.

    Results: The IFGRNS score emerged as a robust prognostic indicator of survival, with higher scores correlating with worse outcomes in most cancer types. The prognostic significance of the score was influenced by factors such as cancer type, tumor pathology, and the tumor microenvironment. Single-cell analysis revealed that myeloid cells, particularly the M2 macrophage subtype, demonstrated high levels of IFGRNS expression, which was associated with tumor progression. A negative correlation was observed between the IFGRNS score and outcomes to anti-PD1 immunotherapy in urologic cancers, where patients with higher scores showed worse prognosis and lower response rates to therapy. Experimental validation in the PAN02 murine model confirmed that anti-PD1 therapy significantly reduced tumor size and IFGRNS expression in M2 macrophages, supporting the clinical findings.

    Conclusions: The IFGRNS score is a novel prognostic indicator for survival and therapeutic responses in cancer. These findings underline the complexity of IFN-γ signaling and suggest potential applications for the IFGRNS score in cancer diagnosis, prognosis, and immunotherapy.Novelty & impact statements: IFN-γ response genes play a significant role in tumour biology, yet comprehensive analysis across various cancers is limited. This study identifies a novel prognostic biomarker, the IFGRNS score, which is elevated in myeloid lineage cells and correlates with survival across multiple cancers. The IFGRNS score is also associated with tumour pathology, immune microenvironment, and immunotherapy response, highlighting its diagnostic and therapeutic potential in cancer management.

  • RESEARCH ARTICLE
    Manuela Mancini , Sara De Santis , Cecilia Monaldi , Fausto Castagnetti , Miriam Iezza , Alessandra Iurlo , Daniele Cattaneo , Sara Galimberti , Marco Cerrano , Isabella Capodanno , Massimiliano Bonifacio , Maura Rossi , Claudio Agostinelli , Manja Meggendorfer , Torsten Haferlach , Michele Cavo , Gabriele Gugliotta , Simona Soverini
    2025, 15(4): e70163. https://doi.org/10.1002/ctm2.70163

    The SETD2 tumour suppressor encodes a histone methyltransferase that specifically trimethylates histone H3 on lysine 36 (H3K36me3), a key histone mark implicated in the maintenance of genomic integrity among other functions. We found that SETD2 protein deficiency, mirrored by H3K36me3 deficiency, is a nearly universal event in advanced-phase chronic myeloid leukemia (CML) patients. Similarly, K562 and KCL22 cell lines exhibited markedly reduced or undetectable SETD2/H3K36me3 levels, respectively. This resulted from altered SETD2 protein turnover rather than mutations or transcriptional downregulation, and proteasome inhibition led to the accumulation of hyper-ubiquitinated SETD2 and to H3K36me3 rescue suggesting that a functional SETD2 protein is produced but abnormally degraded. We demonstrated that phosphorylation by Aurora-A kinase and ubiquitination by MDM2 plays a key role in the proteasome-mediated degradation of SETD2. Moreover, we found that SETD2 and H3K36me3 loss impinges on the activation and proficiency of homologous recombination and mismatch repair. Finally, we showed that proteasome and Aurora-A kinase inhibitors, acting via SETD2/H3K36me3 rescue, are effective in inducing apoptosis and reducing clonogenic growth in cell lines and primary cells from advanced-phase patients. Taken together, our results point to SETD2/H3K36me3 deficiency as a mechanism, already identified by our group in systemic mastocytosis, that is reversible, druggable, and BCR::ABL1-independent, able to cooperate with BCR::ABL1 in driving genetic instability in CML.

  • LETTER TO THE JOURNAL
    Yan Zhao , Jing Dai , Angwei Gong , Sheng Jin , Chengjian Guan , Keke Wang , Qianli Ma , Haijuan Hu , Yuming Wu , Bing Xiao
    2025, 15(4): e70269. https://doi.org/10.1002/ctm2.70269
  • LETTER TO THE JOURNAL
    Garima Joshi , Garima Verma , Simrandeep Kaur , Ellango Ramasamy , Navya Chauhan , Savita Singh , Pradipta Jana , Zaigham Abbas Rizvi , Pallavi Kshetrapal , Shinjini Bhatnagar , Anil Kumar Pandey , Amit Awasthi , Bhabatosh Das , Prasenjit Guchhait
    2025, 15(4): e70275. https://doi.org/10.1002/ctm2.70275
  • REVIEW
    Jiaxin Wan , Zhi Sun , Xueqiong Feng , Peipei Zhou , Mateus T. N. Macho , Zhouyang Jiao , Hui Cao , Chuang Zhang , Rijin Lin , Xiaowen Zhang , Mengyan Fan , Nan Zhang , Jiamei Zhang , Huixiang Liu , Jing Li , Sheng Guan
    2025, 15(4): e70277. https://doi.org/10.1002/ctm2.70277

    Atherosclerosis is a chronic inflammatory condition of the arteries, marked by the development of plaques within the arterial intima. The rupture of unstable plaques can lead to thrombosis, downstream vessel occlusion and serious clinical events. The composition of atherosclerotic plaques is complex and highly heterogeneous, posing challenges for their study. The current pathology and histological subtype classification of plaques may fail to fully encompass the microscopic molecular components in the tissue, the disease progress in various stages of atherosclerosis and the potential mechanism of plaque rupture. However, spatial mapping of the heterogeneity in plaque tissue components can enhance our understanding of these lesions. Despite the considerable progress made by traditional omics in the field of disease research, and its status as an indispensable technology, there remain inherent limitations in the investigation of minute molecular. In recent years, spatial omics techniques have advanced significantly, enabling the visualisation and analysis of specific components within plaques that may serve as causal targets associated with disease progression. The effective application of spatial omics in both research and clinical settings represents a promising area for further exploration. This review focuses on the recent advancements and findings related to spatial omics in the study of extracranial carotid atherosclerotic cerebrovascular disease. Spatial omics analysis of atherosclerotic plaques can facilitate the detection of biomarkers with diagnostic significance or potential relevance to disease, offering new methods and insights into the diagnosis of atherosclerosis and its complications.

  • RESEARCH ARTICLE
    Thomas McLarnon , Steven Watterson , Sean McCallion , Eamonn Cooper , Andrew R. English , Ying Kuan , David S. Gibson , Elaine K. Murray , Frank McCarroll , Shu-Dong Zhang , Anthony J. Bjourson , Taranjit Singh Rai
    2025, 15(4): e70279. https://doi.org/10.1002/ctm2.70279

    Background: Senescence associated secretory phenotype (SASP) contributes to age-related pathology, however the role of SASP in Chronic Kidney Disease (CKD) is unclear. Here, we employ a variety of omic techniques to show that senescence signatures can separate CKD patients into distinct senescence endotypes (Sendotype).

    Methods: Using specific numbers of senescent proteins, we clustered CKD patients into two distinct sendotypes based on proteomic expression. These clusters were evaluated with three independent criteria assessing inter and intra cluster distances. Differential expression analysis was then performed to investigate differing proteomic expression between sendotypes.

    Results: These clusters accurately stratified CKD patients, with patients in each sendotype having different clinical profiles. Higher expression of these proteins correlated with worsened disease symptomologies. Biological signalling pathways such as TNF, Janus kinase-signal transducers and activators of transcription (JAK-STAT) and NFKB were differentially enriched between patient sendotypes, suggesting potential mechanisms driving the endotype of CKD.

    Conclusion: Our work reveals that, combining clinical features with SASP signatures from CKD patients may help predict whether a patient will have worsening or stable renal trajectory. This has implications for the CKD clinical care pathway and will help clinicians stratify CKD patients accurately.

  • INVITED LETTER
    Ignacio Melero , Kathrin Klar , Eugen Leo
    2025, 15(4): e70280. https://doi.org/10.1002/ctm2.70280
  • RESEARCH ARTICLE
    Weijiao Xu , Haitang Yang , Ke Xu , Anshun Zhu , Sean R. R. Hall , Yunxuan Jia , Baicheng Zhao , Enshuo Zhang , Gang Liu , Jianlin Xu , Thomas M. Marti , Ren-Wang Peng , Patrick Dorn , Yongliang Niu , Xufeng Pan , Yajuan Zhang , Feng Yao
    2025, 15(4): e70281. https://doi.org/10.1002/ctm2.70281

    Background: The heterogeneity of cancer-associated fibroblasts (CAFs) has become a crucial focus in understanding cancer biology and treatment response, revealing distinct subpopulations with specific roles in tumor pathobiology. CAFs have also been shown to promote resistance in lung cancer cells to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs). However, the specific CAF subsets responsible for driving tumor advancement and resistance to EGFR-TKIs in lung adenocarcinoma (LUAD) remain poorly understood.

    Methods: We integrate multiple scRNA-seq datasets to identify cell subclusters most relevant to tumor stage, patient survival, and EGFR–TKIs response. Additionally, in vitro and in vivo experiments, clinical tissue sample immunohistochemistry and patient plasma ELISA experiments are performed to validate key findings in independent LUAD cohorts.

    Results: By analyzing multiple scRNA-seq and spatial transcriptomic datasets, we identified a unique subset of CXCL14+ myofibroblastic CAFs (myCAFs), emerging during the early differentiation phase of pan-cancer invasiveness-associated THBS2+ POSTN+ COL11A1+ myCAFs. Notably, plasma levels of CXCL14 in LUAD patients correlate significantly with tumor stage. Mechanistically, this subset enhances tumor aggressiveness through epithelial-to-mesenchymal transition and angiogenesis. Among standard treatment regimens, transitional CXCL14+ myCAFs specifically confer resistance to EGFR-TKIs, while showing no significant impact on chemotherapy or immunotherapy outcomes. Through a pharmacological screen of FDA-approved drugs, we identified Filgotinib as an effective agent to counteract the EGFR-TKIs resistance conferred by this CAF subset.

    Conclusions: In summary, our study highlights the role of the differentiated stage from transitional CXCL14+ myCAFs to invasiveness-associated myCAFs in driving tumor progression and therapy resistance in LUAD, positioning Filgotinib as a promising targeted therapy for this process. These insights may enhance patient stratification and inform precision treatment strategies in LUAD.

  • RESEARCH ARTICLE
    Yuanbo Hu , Chenbin Chen , Kezhi Lin , Xinya Tong , Tingting Huang , Tianle Qiu , Xietao Chen , Jun Xu , Wangkai Xie , Xiangwei Sun , Shiyu Feng , Mingdong Lu , Zhiguang Zhao , Xiaodong Chen , Xiangyang Xue , Xian Shen
    2025, 15(4): e70282. https://doi.org/10.1002/ctm2.70282

    NSUN2, a major methyltransferase that catalyzes m5C methylation in eukaryotes, is known to be implicated in the development of multiple cancers. However, its role in colorectal cancer (CRC) and the related molecular mechanisms have yet to be sufficiently determined. Here, we conducted an analysis of public database (722 CRC patients) and two distinct cohorts from our centre (1559 CRC patients), which revealed that NSUN2 is upregulated in CRC and correlates with unfavourable prognosis. Our analyses also showed that NSUN2 promotes the proliferation and metastasis capabilities of CRC cells. Intriguingly, NSUN2 was found to promote CRC via an m5C-independent mechanism, which has not been previously reported. Overexpression of both wild-type and m5C enzymatic-dead mutant NSUN2 upregulated and activated the ErbB-STAT3 signalling pathway. We also found that both wild-type and the m5C enzymatic-dead mutant NSUN2 closely interacted with CUL4B. Silencing of CUL4B effectively inhibited the m5C-independent function of NSUN2. Moreover, overexpression of NSUN2 enhanced the sensitivity of CRC cells to lapatinib. Taken together, our findings revealed a novel m5C-independent mechanism for NSUN2 in the malignancy and lapatinib sensitivity of CRC via activation of the CUL4B/ErbB-STAT3 pathway, which provides a potential therapeutic strategy for patients with CRC.

  • LETTER TO THE JOURNAL
    Jinghan Huang , Anson C. M. Chow , Nelson L. S. Tang , Sheung Chi Phillip Yam
    2025, 15(4): e70283. https://doi.org/10.1002/ctm2.70283
  • LETTER TO THE JOURNAL
    Wenting Wang , Miao Yang , Xue Zhang , Jiayuan Chen , Shaowei Qiu , Bingcheng Liu , Yingchang Mi , Jianxiang Wang , Hui Wei
    2025, 15(4): e70284. https://doi.org/10.1002/ctm2.70284
  • LETTER TO THE JOURNAL
    Marta Barber , Ariadna Boloix , Alfonso Parrilla , Mariana Köber , Laia Avilés-Domínguez , Nora Ventosa , Lidia del Carmen Ramírez-Morales , Asunción Perez-Benavente , Antonio Gil-Moreno , Eva Colàs , Juan Morote , Miguel F. Segura , Olga Méndez , Anna Santamaria
    2025, 15(4): e70285. https://doi.org/10.1002/ctm2.70285
  • RESEARCH ARTICLE
    Kang Li , Yuxuan Yi , Rongsong Ling , Caihua Zhang , Zhihui Zhang , Yue Wang , Ganping Wang , Jie Chen , Maosheng Cheng , Shuang Chen
    2025, 15(4): e70286. https://doi.org/10.1002/ctm2.70286

    Oesophageal squamous cell carcinoma (OSCC) represents a highly aggressive malignancy with limited therapeutic options and poor prognosis. This study uncovers PCIF1 as a critical driver of OSCC progression via m6Am RNA modification, leading to translational repression of the tumour suppressor MTF2. Our results demonstrate that PCIF1 selectively suppresses MTF2 translation, activating oncogenic pathways that promote OSCC growth. In vitro and in vivo models confirm that PCIF1 knockdown reduces OSCC progression, whereas MTF2 knockdown counteracts this effect, highlighting the importance of the PCIF1-MTF2 axis. Clinical analyses further reveal that high PCIF1 expression and low MTF2 expression correlate with advanced tumour stage, poor treatment response and decreased overall survival. Furthermore, in a preclinical mouse model, PCIF1 knockout enhanced the efficacy of anti-PD1 immunotherapy, reducing tumour burden and improving histological outcomes. Notably, flow cytometry analysis indicated that PCIF1 primarily exerts its effects through tumour-intrinsic mechanisms rather than direct modulation of the immune microenvironment, distinguishing its mode of action from PD1 blockade. These findings establish PCIF1 and MTF2 as promising prognostic markers and therapeutic targets for OSCC, offering new avenues for treatment strategies and patient stratification.

  • RESEARCH ARTICLE
    Matias A. Bustos , Kelly K. Chong , Yoko Koh , SooMin Kim , Eleanor Ziarnik , Romela I. Ramos , Gianna Jimenez , David L. Krasne , Warren M. Allen , Timothy G. Wilson , Dave S. B. Hoon
    2025, 15(4): e70288. https://doi.org/10.1002/ctm2.70288

    Background: Nomograms or comparable techniques can be used to determine which patients with prostate cancer (PCa) will benefit from extended pelvic lymph node dissection (ePLND). While nomograms help guide clinical decisions, ∼80% of the patients undergo unnecessary ePLND. This pilot study aims to identify both transcriptomic mRNA and microRNA (miR) signatures in primary PCa tumours that are associated with the presence of lymph node metastasis (LNM).

    Methods: Primary PCa tumours obtained from 88 patients (pathologically diagnosed as N0 [pN0, n = 44] or as N1 [pN1, n = 44]) were profiled using two different probe-based captured direct assays based on next-generation sequencing and targeting 19398 mRNA transcripts (human transcriptome panel [HTP] dataset) and 2083 miRs (miRs whole-transcriptome assay [WTA] dataset). The TCGA-PRAD (pN0 [n = 382] and pN1 [n = 70]) and GSE220095 (pN0 [n = 138] and pN1 [n = 17]) datasets were used for validation using bioinformatic analyses.

    Results: A four-mRNA signature (CHRNA2, NPR3, VGLL3 and PAH) was found in primary tumour tissue samples from pN1 PCa patients, and then it was validated using the TCGA-PRAD and GSE220095 datasets. Adding serum prostate-specific antigen (PSA) values to the four-gene signature increased the performance to identify pN1 (HTP [AUC = .8487, p = 2.18e-09], TCGA-PRAD [AUC = .7150, p = 8.66e-08] and GSE220095 datasets [AUC = .8772, p = 4.09e-07]). Paired miR analyses showed that eight miRs were significantly upregulated in primary PCa that were pN1 (p < .01). The eight-miR signature performance increased when adding PSA (WTA dataset [AUC = .8626, p = 4.66e-10]) or Grade group (WTA dataset [AUC = .8689, p = 2e-10]). When combining the miR/mRNA signatures (miR-663b, CHRNA2 and PAH) with PSA levels, it showed the best performance to distinguish pN1 from pN0 PCa patients.

    Conclusion: This study found miR/mRNA signatures in primary PCa tumours that in combination with serum PSA levels may complement nomograms for better detection of PCa patients with LNM and triage patients into better surgical decision-making.

  • RESEARCH ARTICLE
    Yan Bai , Xinyu Zhan , Qing Zhu , Xingyue Ji , Yingying Lu , Yiyun Gao , Fei Li , Zhu Guan , Haoming Zhou , Zhuqing Rao
    2025, 15(4): e70289. https://doi.org/10.1002/ctm2.70289

    Background: The lung is the organ most commonly affected by sepsis. Additionally, acute lung injury (ALI) resulting from sepsis is a major cause of death in intensive care units. Macrophages are essential for maintaining normal lung physiological functions and are implicated in various pulmonary diseases. An essential autophagy protein, autophagy-related protein 16-like 1 (ATG16L1), is crucial for the inflammatory activation of macrophages.

    Methods: ATG16L1 expression was measured in lung from mice with sepsis. ALI was induced in myeloid ATG16L1-, NLRP3- and STING-deficient mice by intraperitoneal injection of lipopolysaccharide (LPS, 10 mg/kg). Using immunofluorescence and flow cytometry to assess the inflammatory status of LPS-treated bone marrow-derived macrophages (BMDMs). A co-culture system of BMDMs and MLE-12 cells was established in vitro.

    Results: Myeloid ATG16L1-deficient mice exhibited exacerbated septic lung injury and a more intense inflammatory response following LPS treatment. Mechanistically, ATG16L1-deficient macrophages exhibited impaired LC3B lipidation, damaged mitochondria and reactive oxygen species (ROS) accumulation. These abnormalities led to the activation of NOD-like receptor family pyrin domain-containing protein 3 (NLRP3), subsequently enhancing proinflammatory response. Overactivated ATG16L1-deficient macrophages aggravated the damage to alveolar epithelial cells and enhanced the release of double-stranded DNA (dsDNA), thereby promoting STING activation and subsequent NLRP3 activation in macrophages, leading to positive feedback activation of macrophage NLRP3 signalling. Scavenging mitochondrial ROS or inhibiting STING activation effectively suppresses NLRP3 activation in macrophages and alleviates ALI. Furthermore, overexpression of myeloid ATG16L1 limits NLRP3 activation and reduces the severity of ALI.

    Conclusions: Our findings reveal a new role for ATG16L1 in regulating macrophage NLRP3 feedback activation during sepsis, suggesting it as a potential therapeutic target for treating sepsis-induced ALI.

  • EDITORIAL
    Matteo Barberis , Jinkun Xie
    2025, 15(4): e70290. https://doi.org/10.1002/ctm2.70290

    Single-cell omics has emerged as a powerful tool for elucidating cellular heterogeneity in health and disease. Parallel advances in artificial intelligence (AI), particularly in pattern recognition, feature extraction and predictive modelling, now offer unprecedented opportunities to translate these insights into clinical applications. Here, we propose single-cell -omics-based Disease Predictor through AI (scDisPreAI), a unified framework that leverages AI to integrate single-cell -omics data, enabling robust disease and disease-stage prediction, alongside biomarker discovery. The foundation of scDisPreAI lies in assembling a large, standardised database spanning diverse diseases and multiple disease stages. Rigorous data preprocessing, including normalisation and batch effect correction, ensures that biological rather than technical variation drives downstream models. Machine learning pipelines or deep learning architectures can then be trained in a multi-task fashion, classifying both disease identity and disease stage. Crucially, interpretability techniques such as SHapley Additive exPlanations (SHAP) values or attention weights pinpoint the genes most influential for these predictions, highlighting biomarkers that may be shared across diseases or disease stages. By consolidating predictive modelling with interpretable biomarker identification, scDisPreAI may be deployed as a clinical decision assistant, flagging potential therapeutic targets for drug repurposing and guiding tailored treatments. In this editorial, we propose the technical and methodological roadmap for scDisPreAI and emphasises future directions, including the incorporation of multi-omics, standardised protocols and prospective clinical validation, to fully harness the transformative potential of single-cell AI in precision medicine.

  • RESEARCH ARTICLE
    Na Zhao , Guojian Wang , Shuang Long , Xiaofan Lv , Xinze Ran , Junping Wang , Yongping Su , Tao Wang
    2025, 15(4): e70291. https://doi.org/10.1002/ctm2.70291

    Background: Effective control of inflammation is crucial for the healing of cutaneous wounds, but the molecular mechanisms governing inflammation resolution during wound closure are still not yet clear. Here, we describe a homeostatic mechanism that facilitates the inflammation resolution by timely regulating the targeted proteases activities through antiprotease Spink7 (serine peptidase inhibitor, kazal type 7).

    Methods: The expression pattern of Spink7 was investigated by quantitative RT-PCR, immunohistochemistry (IHC) and in situ hybridization. In both Spink7 knockdown and knockout models, quantitative comparisons were made between the healing rate of wounds and histopathological morphometric analysis. Microarrays, multiple chemokine assays, IHC, immunofluorescence, protease activity measurement were performed to explore the underlying mechanisms of Spink7 knockout in impaired wound healing. Radiation-wound combined injury (R-W-CI) model was employed to evaluate the therapeutic effects of Spink7 manipulation.

    Results: Our study demonstrates that Spink7 is significantly upregulated in the differentiated epidermal granular keratinocytes of proliferative phase during murine wound closure. Both local knockdown of Spink7 levels in wounds using siRNA gel and systemic knockout of Spink7 using KO mice resulted in delayed wound closure with sustained neutrophil infiltration. Loss of Spink7 leads to augmented inflammatory responses, increased production of multiple chemokines/cytokines, and impaired M2 polarization of macrophages in wound healing. Furthermore, loss of Spink7 results in elevated proteolytic activities of uPA, MMP2/9 and KLK5/7 in proliferative phase. However, inhibiting KLK5/7 downstream PAR2 activation exacerbates the phenotype of KO mice. In R-W-CI model, further significant induction of Spink7 is observed in wounds with insufficient inflammatory response. Local suppression of Spink7 promotes wound healing in the R-W-CI model by augmenting inflammation.

    Conclusions: Maintaining an endogenous balance between Spink7 and its target proteases is a crucial checkpoint for regulating inflammation resolution during healing. Therefore, manipulating levels of Spink7 might be an effective treatment for impaired wounds caused by inflammatory dysregulation.

  • LETTER TO THE JOURNAL
    Yanwen Chen , Lisha Zhao , Shuo Cai , Yuchen Zou , Weiwei Tang , Bin Li
    2025, 15(4): e70292. https://doi.org/10.1002/ctm2.70292
  • INVITED LETTER
    Yahui Zhu , Donglin Wei , Michael Karin , Li Gu
    2025, 15(4): e70293. https://doi.org/10.1002/ctm2.70293
  • EDITORIAL
    Qiqing Yang , Ce Guo , Long Zhang
    2025, 15(4): e70294. https://doi.org/10.1002/ctm2.70294

    Recent advancements in immunometabolism have highlighted the critical role of metabolite sensors in regulating immune responses. Metabolites such as lactate, succinate, itaconate, and β-hydroxybutyrate influence immune cell function by interacting with specific sensors. These metabolites act as signaling molecules, linking cellular metabolic changes to immune responses. Lactate, a metabolite commonly produced under hypoxic conditions, has emerged as a major regulator of innate immunity. Key enzymes, including AARS1 and AARS2, function as intracellular lactate sensors, catalyzing lactylation on proteins like cGAS, which plays a central role in DNA sensing and immune activation. The lactylation of cGAS inhibits its activity, modulating immune responses by balancing inflammation and immune tolerance. Metabolite sensors, like MCT1, also contribute to immune modulation, particularly in cancer and chronic inflammatory diseases. Therapeutically, targeting these sensors offers potential for restoring immune function, especially in cancer immunotherapy. However, challenges in specificity, off-target effects, and long-term safety require further investigation. This article explores the emerging role of metabolite sensors in immune regulation, with a focus on lactate sensors, and outlines potential therapeutic strategies to enhance immune responses in metabolic diseases.

  • INVITED LETTER
    Xiaoxue Zhou , Linghui Zeng , Fangfang Zhou
    2025, 15(4): e70296. https://doi.org/10.1002/ctm2.70296
  • RESEARCH ARTICLE
    Qin Sun , Ran Teng , Qiankun Shi , Yun Liu , Xing Cai , Bin Yang , Quan Cao , Chang Shu , Xu Mei , Weiqi Zeng , Bingxue Hu , Junyi Zhang , Haibo Qiu , Ling Liu
    2025, 15(4): e70297. https://doi.org/10.1002/ctm2.70297

    Background: Accurate pathogen identification is critical for managing sepsis. However, traditional microbiological methods are time-consuming and exhibit limited sensitivity, particularly with blood samples. Metagenomic sequencing of plasma or whole blood was highly affected by the proportion of host nucleic acid.

    Methods: We developed a Probe-Capture Metagenomic assay and established a multicentre prospective cohort to assess its clinical utility. In this study, 184 blood samples from patients suspected of sepsis were sent for blood culture and Probe-Capture Metagenomic sequencing before using antibiotics. The pathogen-positive rate and auxiliary abilities in diagnosis were compared among Probe-Capture Metagenomics, blood culture and real-time PCR (RT-PCR). Antibiotic therapy adjustments were based on the identification of pathogens, and changes in the Sequential Organ Failure Assessment (SOFA) score were monitored on days 0, 3 and 7 of admission.

    Results: A total of 184 sepsis patients were enrolled, with a mean age of 66 years (range 56–74). The Probe-Capture Metagenomics method, confirmed by RT-PCR, demonstrated a significantly higher pathogen detection rate than blood culture alone (51.6% vs. 17.4%, p < .001). When combining the results of blood culture and RT-PCR, Probe-Capture Metagenomics achieved a concordance rate of 91.8% (169/184), with a sensitivity of 100% and specificity of 87.1%. In terms of clinical impact, antibiotic therapy was adjusted for 64 patients (34.8%) based on the results from Probe-Capture Metagenomics, and 41 patients (22.3%) showed a > 2-point decrease in SOFA score following antibiotic adjustments.

    Conclusion: Probe-Capture Metagenomics significantly enhances the ability of pathogen detection compared with traditional metagenomics. Compared to blood culture and RT-PCR in sepsis patients, it leads to improved antibiotic treatment and better patient outcomes. This study, for the first time, evaluates the clinical impact of metagenomic sequencing by integrating antibiotic adjustments and SOFA score changes, indicating that approximately one-fifth of sepsis patients benefit from this advanced diagnostic approach.

    Trial registration: This study has been registered in clinical trials (clinicaltrials.gov) on 30 November 2018, and the registration number is NCT03760315.

  • RESEARCH ARTICLE
    Xiucheng Yang , Shanchao Hong , Tao Yan , Mingzhao Liu , Mingyao Liu , Jin Zhao , Bingqing Yue , Di Wu , Jingbo Shao , Man Huang , Jingyu Chen
    2025, 15(4): e70298. https://doi.org/10.1002/ctm2.70298

    Background: The limited donor lung pool for lung transplantation (LTx) is largely due to concerns over ischaemia–reperfusion injury (IRI), a major cause of primary graft dysfunction (PGD). NLRP3 inflammasome activation is known to play a pivotal role in the onset of IRI. While human umbilical cord mesenchymal stromal cell-derived extracellular vesicles (hucMSC-EVs) have shown potential in reducing acute lung injury, their effects on NLRP3 activation in the context of LTx remain unclear.

    Methods: In this study, engineered hucMSC-EVs were delivered via nebulisation to mitigate IRI in rat LTx models. We utilised both a rat orthotopic LTx model and a cell cold preservation reperfusion model to evaluate the therapeutic efficacy of hucMSC-EVs. Bulk-RNA sequencing, single-cell sequencing analysis, immunofluorescence and Western blot techniques were employed to assess NLRP3 inflammasome activation and inflammation.

    Results: Nebulised hucMSC-EVs were efficiently internalised by alveolar macrophages (AMs), significantly reducing lung injury and improving oxygenation in the LTx models. Mechanistically, the engineered hucMSC-EVs, which enhance the expression of miR-146a, can more effectively suppress the activation of the NLRP3 inflammasome by targeting the IRAK1/TRAF6/NF-κB pathway, resulting in decreased levels of IL-1β, IL-18 and other inflammatory cytokines. These findings highlight the potential of miR-146a-modified EVs in modulating innate immune responses to alleviate IRI.

    Conclusion: Our results demonstrate that nebulised delivery of engineered hucMSC-EVs effectively mitigates IRI in LTx by inhibiting NLRP3 inflammasome activation. This innovative approach presents a promising strategy for enhancing donor lung preservation and improving post-transplant outcomes in LTx.

  • REVIEW
    Ri Hong , Puguang Yu , Xiaoli Zhang , Peng Su , Hongyuan Liang , Dan Dong , Xuesong Wang , Kefeng Wang
    2025, 15(4): e70299. https://doi.org/10.1002/ctm2.70299

    Urological tumours are a type of neoplasms that significantly jeopardise human life and wellbeing. Cancer-associated fibroblasts (CAFs), serving as the primary component of the stromal cellular milieu, form a diverse cellular cohort that exerts substantial influence on tumourigenesis and tumour progression. In this review, we summarised the literatures regarding the functions of CAFs in the urinary tumour microenvironment (TME). We primarily examined the multifaceted activities of CAFs in the TME of urological system tumours, including inhibiting tumour immunity, remodelling the extracellular matrix, promoting tumour growth, metastasis, drug resistance and their clinical applications. We also discussed potential future directions for leveraging artificial intelligence in CAFs research.

  • LETTER TO THE JOURNAL
    Zhe Wang , Zhiyan Liu , Guangyan Mu , Qiufen Xie , Shuang Zhou , Zining Wang , Yimin Cui , Qian Xiang
    2025, 15(4): e70300. https://doi.org/10.1002/ctm2.70300
  • ERRATUM
    Zijuan Wu , Wei Zhang , Luqiao Wang , Jiayan Leng , Yongle Li , Zhou Fan , Mengtao Zhan , Lei Cao , Yongning Jiang , Yan Jiang , Bing Sun , Jianxin Fu , Jianyong Li , Wenyu Shi , Hui Jin
    2025, 15(4): e70301. https://doi.org/10.1002/ctm2.70301
  • EDITORIAL
    Xiaojun Yan , Wanxin Duan , Xiangdong Wang
    2025, 15(4): e70303. https://doi.org/10.1002/ctm2.70303

    Marine biomedicine is an important field in oceanology and bio-ecosystem and has evolved significantly alongside advances in biotechnology and growing understanding of marine life. In this perspective, we propose a refined concept of clinical marine biomedicine, with a clear mission to establish an emerging discipline that bridges marine biomedicine and clinical practice. The exploration of marine-origin sources should be emphasised, with a strong focus on the identification, validation and development of human disease-specific diagnostics and target-oriented pharmaceutics. The perspective headlines some of critical components, including marine-oriented human evolution and development, humanised marine-based models, biomarker innovation and validation, marine microbiomes and metabolites, and target nutrition and therapy. We envision that clinical marine biomedicine will become a crucial pillar clinical molecular medicine, contributing to the improvement of human health and the prognosis of patient.

  • RESEARCH ARTICLE
    Yu Zhang , Ke Qiu , Jiayuan Ai , Maosen Xu , Binhan Wang , Aqu Alu , Chunjun Ye , Xiya Huang , Yu Zhang , Yingqiong Zhou , Zhiruo Song , Jie Shi , Yishan Lu , Yuquan Wei , Jianjun Ren , Yu Zhao , Ping Cheng , Xiawei Wei
    2025, 15(4): e70305. https://doi.org/10.1002/ctm2.70305

    Background: High-risk human papillomavirus (HPV), especially HPV16, is closely correlated with certain cancers. E6 and E7 proteins of HPV16 play critical roles in oncogenesis, making them optimal targets for treating HPV-associated cancers. Here, we engineered an innovative vaccine, Ad-E6/7-HR, designed to evoke immune responses through the incorporation of self-assembling heptad-repeat 1 (HR1) and HR2 originated from Severe acute respiratory syndrome coronavirus 2.

    Methods: Ad-E6/7-HR was constructed utilising a replication-defective human adenovirus serotype 5 vector and evaluated its immunogenicity and therapeutic efficacy in murine models. We verified the antitumour efficacy of the vaccine in TC-1 subcutaneous and pulmonary models. Flow cytometry, enzyme-linked immunospot assay, and immunofluorescence staining were used to assess the cellular immunogenicity of Ad-E6/7-HR.

    Results: Ad-E6/7-HR induced robust immune responses, significantly increasing antigen-specific CD8+ T cells. The vaccine also enhanced memory T-cell generation and induced potent cytokine secretion, as exemplified by interferon-γ and tumour necrosis factor-α. Ad-E6/7-HR conferred complete protection against tumour growth in the prophylactic model. In therapeutic settings, Ad-E6/7-HR significantly reduced tumour size and improved survival. Furthermore, Ad-E6/7-HR reshaped the tumour microenvironment by increased CD8+ T-cell recruitment and reduced immunosuppressive cells, like myeloid-derived suppressor cells and M2 macrophages, thereby enhancing antitumour immunity.

    Conclusions: By targeting HPV16 E6 and E7 proteins and leveraging the self-assembling HR1 and HR2 sequences to enhance immune responses, Ad-E6/7-HR represented a promising candidate for preventing and treating HPV-associated cancers. Further clinical investigation is warranted to evaluate its potential in human trials.

  • REVIEW
    Lvying Wu , Lingfeng Zhu , Jin Chen
    2025, 15(4): e70306. https://doi.org/10.1002/ctm2.70306

    Background: Chimeric antigen receptor (CAR)-engineered cell therapies have made significant progress in haematological cancer treatment. This success has motivated researchers to investigate its potential applications in non-cancerous diseases, with substantial strides already made in this field.

    Main Body: This review summarises the latest research on CAR-engineered cell therapies, with a particular focus on CAR-T cell therapy for non-cancerous diseases, including but not limited to infectious diseases, autoimmune diseases, cardiac diseases and immune-mediated disorders in transplantation. Additionally, the review discusses the current obstacles that need to be addressed for broader clinical applications.

    Conclusion: With ongoing research and continuous improvements, CAR-engineered cell therapy holds promise as a potent tool for treating various diseases in the future.

  • INVITED LETTER
    Kai Pang , Zhongtao Zhang
    2025, 15(4): e70307. https://doi.org/10.1002/ctm2.70307
  • LETTER TO THE JOURNAL
    Xiaowei Zhang , Ting Zhao , Biqiang Zheng , Wangjun Yan , Yong Chen , Yu Xu , Chunmeng Wang , Junhua Zhang , Jian Wang , Lin Yu , Xin Liu , Zhiguo Luo
    2025, 15(4): e70308. https://doi.org/10.1002/ctm2.70308
  • RESEARCH ARTICLE
    Rong Shen , Wei-Guo Cao , Li Wang , Ling-Shuang Sheng , Yi-Lun Zhang , Wen Wu , Peng-Peng Xu , Shu Cheng , Meng-Ke Liu , Yan Dong , Yue Wang , Xiang-Qin Weng , Xu-Feng Jiang , Qi Song , Hong-Mei Yi , Lei Li , Sheng Chen , Zi-Xun Yan , Wei-Li Zhao
    2025, 15(4): e70310. https://doi.org/10.1002/ctm2.70310

    Background: CD19 chimeric antigen receptor (CAR) T-cell therapy is a potential treatment for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). The combination of targeted therapeutic strategies, particularly bruton tyrosine kinase inhibitor zanubrutinib and programmed death-1 inhibitor tislelizumab, may improve clinical outcomes and modulate the tumour microenvironment (TME).

    Methods: We studied patients with R/R LBCL who received response-adapted zanubrutinib plus tislelizumab upon CD19 CAR T-cell therapy between June 2021 and March 2023. Patients were treated with zanubrutinib daily from leukapheresis to day 28 post-infusion; those achieving complete response continued zanubrutinib monotherapy for 3 months, while partial responders received combined zanubrutinib for 3 months and tislelizumab for up to 2 years. We evaluated the overall response rate (ORR), complete response rate (CRR), progression-free survival (PFS), overall survival (OS), and safety. DNA sequencing and RNA sequencing were performed on available tumour samples to analyse genetic aberrations and TME characteristics.

    Results: A total of 54 patients with LBCL were included, with a median follow-up of 23.6 months. The ORR at day 28, month 3, and month 6 were 94% (CRR 66%), 87% (CRR 80%), and 80% (CRR 76%), respectively. The 2-year PFS and 2-year OS rates were 68% and 76%, respectively. Median PFS and median OS were not reached. Grade ≥ 3 cytokine release syndrome occurred in 9% of patients, with no grade ≥ 3 neurotoxicity observed. Genomic and transcriptomic data indicated that this regimen was effective across genetic subtypes and abrogated T-cell exhaustion within the TME. However, tumour-infiltrating M2 macrophages with dysregulated lipid metabolism were associated with poor clinical outcome.

    Conclusions: Response-adapted zanubrutinib and tislelizumab potentially enhances the efficacy of CAR T-cell therapy with a favourable safety profile in R/R LBCL, effectively counteracting T-cell exhaustion. Future studies should focus on targeting M2 macrophages by reprogramming lipid metabolism to further attenuate the immunosuppressive TME.

  • RESEARCH ARTICLE
    Kaiwen Yu , Xiang Li , Xin Shi , Ruogu Li , Min Zhang
    2025, 15(4): e70311. https://doi.org/10.1002/ctm2.70311

    Background: Inflammation and endothelial apoptosis are implicated in the advancement of atherosclerosis. EEPD1 holds a pivotal position in the repair of DNA damage and contributes to the progression of multiple cancers. However, the role of EEPD1 in cardiovascular diseases needs to be explored further, especially in atherosclerosis.

    Methods: We constructed EEPD1 and ApoE (apolipoprotein E)-deficient mice to assess how EEPD1 influences endothelial inflammation and apoptosis within atherosclerotic plaques. High-throughput RNA sequencing of human aortic endothelial cell groups treated with siCon+TNFα and siEEPD1+TNFα identified notable disparities in the MAPK pathway between groups. Chromatin immunoprecipitation and luciferase reporter assay confirmed that KLF4 directly regulates EEPD1.

    Results: Further examination of gene expression data revealed elevated EEPD1 concentrations in atherosclerotic plaques of patients, which findings were corroborated in the aortas of ApoE−/− mice. Present study demonstrated that adhesion molecule expression, endothelial apoptosis, aortic root plaques and macrophage accumulation were markedly ameliorated in EEPD1−/−ApoE−/− mice compared to WT ApoE−/− mice. Functional analysis revealed that increase in EEPD1 promotes ERK phosphorylation and significantly increases endothelial apoptosis and inflammation in atherosclerosis, which was abrogated by inhibition of ERK phosphorylation. We found KLF4 to be the transcription repressor of EEPD1 through luciferase assay and chromatin immunoprecipitation, and KLF4 inhibition abrogated the amelioration of endothelial apoptosis and inflammation caused by EEPD1 deletion.

    Conclusions: Collectively, this study revealed that EEPD1 deletion can lead to amelioration of atherosclerosis through the KLF4-EEPD1-ERK axis. Hence, targeting EEPD1 could be a promising therapeutic strategy for patients with atherosclerosis.

  • REVIEW
    Shuaixi Yang , Yingshuai Fang , Yangcheng Ma , Fuqi Wang , Yuhang Wang , Jiachi Jia , Yabing Yang , Weipeng Sun , Quanbo Zhou , Zhen Li
    2025, 15(4): e70313. https://doi.org/10.1002/ctm2.70313

    Angiogenesis, as a core marker of cancer survival and growth, is integral to the processes of tumour growth, invasion and metastasis. In recent years, targeted angiogenesis treatment strategies have gradually become an important direction in cancer treatment. Single-cell sequencing technology can provide new insights into targeted angiogenesis by providing a deeper understanding of the heterogeneity of tumour endothelial cells and exploring the interactions between endothelial cells and surrounding cells in the tumour microenvironment. Here, we systematically review the research progress in endothelial cell pathophysiology and its endothelial‒mesenchymal transition and illustrate the heterogeneity of endothelial cells from a single-cell perspective. Finally, we examine the contributions of different cell types within the tumour microenvironment in relation to tumour angiogenesis, as well as the latest progress and strategies in targeted angiogenesis therapy, hoping to provide useful insights into the clinical application of antiangiogenic treatment. Furthermore, a summary of the present progress in the development of potential angiogenesis inhibitors and the ongoing clinical trials for combination therapies is provided.

  • RESEARCH ARTICLE
    Lan Weiss , Michele Carrer , Alyaa Shmara , Angela Martin , Hong Yin , Pallabi Pal , Cheng Cheng , Lac Ta , Victoria Boock , Yasamin Fazeli , Mindy Chang , Marvin Paguio , Jonathan Lee , Howard Yu , John Weiss , Tamar R Grossman , Nina Raben , Paymaan Jafar-Nejad , Virginia Kimonis
    2025, 15(4): e70314. https://doi.org/10.1002/ctm2.70314

    Pompe disease (PD) is a progressive myopathy caused by the aberrant accumulation of glycogen in skeletal and cardiac muscle resulting from the deficiency of the enzyme acid alpha-glucosidase (GAA). Administration of recombinant human GAA as enzyme replacement therapy (ERT) works well in alleviating the cardiac manifestations of PD but loses sustained benefit in ameliorating the skeletal muscle pathology. The limited efficacy of ERT in skeletal muscle is partially attributable to its inability to curb the accumulation of new glycogen produced by the muscle enzyme glycogen synthase 1 (GYS1). Substrate reduction therapies aimed at knocking down GYS1 expression represent a promising avenue to improve Pompe myopathy. However, finding specific inhibitors for GYS1 is challenging given the presence of the highly homologous GYS2 in the liver. Antisense oligonucleotides (ASOs) are chemically modified oligomers that hybridise to their complementary target RNA to induce their degradation with exquisite specificity. In the present study, we show that ASO-mediated Gys1 knockdown in the Gaa−/− mouse model of PD led to a robust reduction in glycogen accumulation in skeletal muscle. In addition, combining Gys1 ASO with ERT slightly further reduced glycogen content in muscle, eliminated autophagic buildup and lysosomal dysfunction, and improved motor function in Gaa−/− mice. Our results provide a strong foundation for validation of the use of Gys1 ASO, alone or in combination with ERT, as a therapy for PD. We propose that early administration of Gys1 ASO in combination with ERT may be the key to preventative treatment options in PD.

  • LETTER TO THE JOURNAL
    Congcong Jiao , Yuxin Zhao , Yang Shao , Haoshen Feng , Cong Ma , Xiangnan Hao , Xiaomei Liu , Junjun Luan , Xu Yang , Hua Zhou
    2025, 15(4): e70317. https://doi.org/10.1002/ctm2.70317