{"title":"Demethoxycurcumin suppresses HK2-mediated glycolysis by targeting PTEN/Akt signaling.","authors":"Jinzhuang Liao, Shiming Tan, Shuangze Han, Ruirui Wang, Pengfei Guo, Wei Zheng, Xinfang Yu, Wei Li","doi":"10.1038/s41417-025-00972-y","DOIUrl":"https://doi.org/10.1038/s41417-025-00972-y","url":null,"abstract":"<p><p>Aerobic glycolysis is a hallmark of tumor cells, with the expression of glycolytic enzymes often being upregulated in many cancers, leading to enhanced metabolic activity. Among the key rate-limiting enzymes in this process, Hexokinase 2 (HK2) plays a crucial role in sustaining the biological activities of human cancers. Therefore, HK2 is considered a potential therapeutic target, although effective targeted drugs for oral squamous cell carcinoma (OSCC) treatment are currently lacking. To confirm potent anti-tumor agents that inhibit HK2 expression, we screened a library of 639 natural products and discovered that Demethoxycurcumin (Deme) was the most effective anti-tumor agent via inhibiting HK2-mediated glycolysis in OSCC cells, inducing intrinsic apoptosis. Mechanistically, Deme enhanced the interaction between USP13 and PTEN, leading to the stabilization of PTEN and consequent downregulation of HK2 via the PTEN/Akt/HK2 pathway. Notably, HK2 overexpression reversed the inhibitory effect of Deme on OSCC cells. Furthermore, at the tumor-inhibitory dose, Deme showed no effect on non-tumor HaCat cells. In vivo, Deme significantly suppressed tumor growth without apparent toxicity to vital organs. Together, these data suggest that Deme is a promising and safe anti-tumor compound that downregulates HK2 expression, providing a potential therapeutic strategy for OSCC treatment.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145211892","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"New insights into the classification of the RAC1 P29S hotspot mutation in melanoma as an oncogene.","authors":"Amin Mirzaiebadizi, Mohammad Reza Ahmadian","doi":"10.1038/s41417-025-00965-x","DOIUrl":"https://doi.org/10.1038/s41417-025-00965-x","url":null,"abstract":"<p><p>The RAC1<sup>P29S</sup> hotspot mutation, which is prevalent in melanoma, drives tumorigenesis by promoting the persistent activation of RAC1. This mutation enhances molecular interactions, and hyperactivates key signaling pathways, making RAC1<sup>P29S</sup> a promising target for cancer therapy. This study provides a comprehensive biochemical and cell-based characterization of RAC1<sup>P29S</sup>, as well as comparisons with wild-type RAC1 and the T17N and F28L mutants. The P29S substitution significantly impairs nucleotide binding while accelerating intrinsic nucleotide exchange. While it minimally affects regulation by guanosine dissociation inhibitor 1 (GDI1), RAC1<sup>P29S</sup> exhibits reduced activation via DBL family guanine nucleotide exchange factors (GEFs) but retains effective activation by dedicator of cytokinesis 2 (DOCK2). Importantly, the P29S mutation severely impairs GTPase-activating protein-stimulated GTP hydrolysis, which most likely contributes to RAC1<sup>P29S</sup> hyperactivation by prolonging its GTP-bound active form. This mutation displays a stronger binding affinity for the IQ motif-containing GTPase-activating protein 1 (IQGAP1) than for the p21-activated kinase 1 (PAK1), indicating altered effector interactions that modulate downstream signaling spatially. These biochemical findings are consistent with the fact that RAC1<sup>P29S</sup> predominantly adopts an active GTP-bound state under serum-starved conditions. IGR1 human melanoma cells harboring endogenous RAC1<sup>P29S</sup> exhibit persistent RAC1<sup>P29S</sup>•GTP accumulation, even without upstream GEF activation. Furthermore, the pharmacological inhibition of DOCK2 with CPYPP significantly reduces RAC1<sup>P29S</sup> activation in these cells, which confirms the pivotal role of DOCK2 in sustaining RAC1<sup>P29S</sup>-driven signaling. Overexpression of RAC1<sup>P29S</sup> activates key oncogenic pathways, including ERK1/2 and p38 MAPK, highlighting its role as a constitutively active driver mutation. Together, these results imply that targeting upstream regulators such as DOCK2 and downstream effectors, such as IQGAP1, could be effective therapeutic strategies for counteracting RAC1<sup>P29S</sup>-mediated melanoma progression and resistance to targeted therapies. A model of RAC1<sup>P29S</sup> activation and signaling in cancer cells. RAC1<sup>P29S</sup> remains in an inactive GDP-bound state in the cytoplasm where GDI1 prevents its membrane association. Upon stimulation, GEFs, primarily DOCK2, activate RAC1<sup>P29S</sup> by promoting GDP-GTP exchange, facilitating its transition to the active GTP-bound state and initiating downstream signaling. RAC1<sup>P29S</sup> binds preferentially to IQGAP1 over PAK1, reflecting a shift in effector interactions. IQGAP1 acts as a scaffolding protein, spatially modulating RAC1<sup>P29S</sup>-driven signaling and amplifying its effects. Under normal conditions, GAPs such as p50GAP regulate RAC1 by accelerating G","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145205782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dhana Sekhar Reddy Bandi, Sujith Sarvesh, Jeremy Foote, Doug Welsch, Changde Cheng, Mehmet Akce, Ganji Purnachandra Nagaraju, Bassel F El-Rayes
{"title":"Paricalcitol and hydroxychloroquine modulates extracellular matrix and enhance chemotherapy efficacy in pancreatic cancer.","authors":"Dhana Sekhar Reddy Bandi, Sujith Sarvesh, Jeremy Foote, Doug Welsch, Changde Cheng, Mehmet Akce, Ganji Purnachandra Nagaraju, Bassel F El-Rayes","doi":"10.1038/s41417-025-00967-9","DOIUrl":"10.1038/s41417-025-00967-9","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis and limited therapeutic options. In a previous publication, our group defined some of the mechanisms that vitamin D analogue paricalcitol (P) and hydroxychloroquine (H) potentiated the effects of gemcitabine-based chemotherapy in PDAC. Based on this, we hypothesized that PH may potentiate 5-fluorouracil (5FU) and Oxaliplatin-based chemotherapy, and this may involve a novel mechanism of extracellular matrix (ECM) modulation. The combination of PH with 5FU+Oxaliplatin significantly increased the cell death, apoptosis, and S-phase cell cycle arrest as compared to untreated or 5FU + Oxaliplatin-treated MIA PaCa-2, HPAC and KPC cell lines. In vivo, the combination therapy inhibited PDAC growth and altered the immune landscape by activating T and NK cells. Proteomic analysis revealed significant reduction in ECM proteins, specifically integrin beta-4 (ITGB4). Confirmation of the role of ITGB4 was performed through genetic knockdown of ITGB4, which led ECM inhibition. In conclusion, the combination of PH significantly enhances the efficacy of Oxaliplatin and 5FU. We identified a new mechanism of action of PH through inhibiting ITGB4, leading to ECM modulation. These results suggest that the combination of PH with cytotoxic chemotherapy should be tested in PDAC clinical trials.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145181782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Bacillus Calmette-Guerin induces CD8+ T cell infiltration and suppresses tumor progression in microsatellite stable colorectal cancer by downregulating ARID1A.","authors":"Zhiyue Xie, Nan Peng, Zhihua Pan, Yun Feng, Yihan Wu, Yansheng Yang, Rui Li, Liang Zhao","doi":"10.1038/s41417-025-00964-y","DOIUrl":"https://doi.org/10.1038/s41417-025-00964-y","url":null,"abstract":"<p><p>Neoadjuvant immunotherapy demonstrates limited efficacy in microsatellite-stable (MSS) colorectal cancer (CRC). In vivo observations reveal that Bacillus Calmette-Guérin (BCG) can inhibit the progression of MSS-CRC and downregulate ARID1A in both in vivo and in vitro settings. Through the analysis of clinical samples, in vivo and in vitro models, and bioinformatics, we found that the low expression of ARID1A promotes tumor growth in vitro; however, in vivo, it enhances CD8+ T cell infiltration in MSS-CRC tissues while inhibiting tumor growth. Further investigation revealed that BCG downregulates ARID1A via the TLR4/NF-κB pathway, leading to the downregulation of MLH1 and PMS2 and subsequent alterations in MMR function in MSS-CRC. This cascade enhances antigen presentation, promotes CD8+ T cell infiltration, and contributes to tumor suppression.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145102562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lin Ma, Meng Xu, Shaoxian Xu, Xueyan Guo, Wei Zong, Xi Zhao, Zi Yang, Guisheng Liu, Lin Shen
{"title":"HMGB1 downregulates DDX3 to activate the MAPK pathway, promoting the progression of colorectal cancer.","authors":"Lin Ma, Meng Xu, Shaoxian Xu, Xueyan Guo, Wei Zong, Xi Zhao, Zi Yang, Guisheng Liu, Lin Shen","doi":"10.1038/s41417-025-00963-z","DOIUrl":"https://doi.org/10.1038/s41417-025-00963-z","url":null,"abstract":"<p><p>High mobility group box 1 (HMGB1) has been implicated in the development of various cancers, but its role in colorectal cancer (CRC) remains poorly understood. This study investigated the role of HMGB1 in CRC progression, particularly through its interaction with DEAD-box helicase 3 (DDX3), which, as demonstrated by our previous research, regulates CRC via the MAPK pathway. We analysed HMGB1 expression in CRC using public databases and tissue microarrays and detected significantly higher expression in CRC tissues than in normal tissues, which was associated with poor prognosis. HMGB1 expression was knocked down in the SW480 and HCT116 cell lines using siRNA and lentiviral vectors, and this knockdown inhibited CRC cell proliferation, migration, invasion, and adhesion, as confirmed by both in vitro and in vivo experiments. Molecular analyses revealed reduced phosphorylation of Erk1/2, c-Jun, and Elk1, along with decreased β-catenin and Snail expression and increased E-cadherin expression. Coimmunoprecipitation assay results further confirmed the interaction between HMGB1 and DDX3. These findings suggest that HMGB1 is an oncogene in CRC that promotes tumour progression through the MAPK pathway by downregulating DDX3. These findings highlight HMGB1 as a potential therapeutic target in CRC.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145102626","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Gene editing in cancer therapy: overcoming drug resistance and enhancing precision medicine.","authors":"Hyeonjeong Park, Suyeun Yu, Taeyoung Koo","doi":"10.1038/s41417-025-00959-9","DOIUrl":"https://doi.org/10.1038/s41417-025-00959-9","url":null,"abstract":"<p><p>The CRISPR system has revolutionized cancer gene therapy, offering unparalleled precision in genetic manipulation for targeted oncogene disruption, mutation correction, and immune system modulation. This breakthrough tool has demonstrated remarkable potential in overcoming drug resistance, enhancing chemotherapy sensitivity, and improving immunotherapy strategies such as CRISPR-engineered CAR-T cells. Additionally, oncolytic virus-mediated CRISPR delivery has emerged as a novel approach for tumor-specific gene editing, minimizing off-target effects. The rapid transition of CRISPR-based cancer therapeutics from preclinical research to clinical trials underscores its therapeutic potential. This review explores the latest advancements in CRISPR applications for cancer therapy, including gene knockout, base editing for mutation correction, and integration with immune and viral therapies. Despite significant progress, challenges such as off-target effects, immune responses, and delivery limitations remain key hurdles. We discuss current strategies to enhance CRISPR safety and efficacy, emphasizing its potential for personalized cancer treatment.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145084587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Heng Cao, Jiaqi Ye, Xiaojiao Li, Yao Si, Xin Jin, Shengfeng Xiong, Teng Ji, Yi Ding, Ding Ma, Qinglei Gao, Xiaoli Wang, Zhoutong Dai, Fei Li
{"title":"A novel GM-CSF-encoding oncolytic adenovirus induces profound autophagy and promotes viral replication to enhance anti-tumor efficacy.","authors":"Heng Cao, Jiaqi Ye, Xiaojiao Li, Yao Si, Xin Jin, Shengfeng Xiong, Teng Ji, Yi Ding, Ding Ma, Qinglei Gao, Xiaoli Wang, Zhoutong Dai, Fei Li","doi":"10.1038/s41417-025-00962-0","DOIUrl":"https://doi.org/10.1038/s41417-025-00962-0","url":null,"abstract":"<p><p>Granulocyte-macrophage colony-stimulating factor (GM-CSF) acts as a double-edged sword in cancer by enhancing both anti- and pro-tumorigenic immune cells. In this study, two oncolytic adenoviruses were engineered to modulate GM-CSF expression using different strategies: one with the CMV promoter (oAd-CMV-GM-CSF) and the other using the endogenous viral E3 promoter (oAd-GM-CSF). The impacts of these modifications on transgene expression, cytotoxicity, viral replication, and apoptosis were assessed both in vitro and in vivo. The results demonstrated that oAd-CMV-GM-CSF produced significantly lower GM-CSF levels than oAd-GM-CSF, interestingly oAd-CMV-GM-CSF exhibited increased cytotoxicity and apoptosis compared to oAd-GM-CSF and control groups. The further study showed oAd-CMV-GM-CSF induced profound autophagy through the activation of the Janus kinase 2/Signal Transducer and Activator of Transcription 2 (JAK2/STAT2) signaling pathway. The use of autophagy and JAK-2 inhibitors, Chloroquine (CQ) and AG-490, respectively, significantly mitigated the apoptosis induced by oAd-CMV-GM-CSF. In addition, oAd-CMV-GM-CSF presented a faster viral replication and production of more active progeny virus than oAd-GM-CSF, which could be inhibited by CQ. oAd-CMV-GM-CSF augments propagation of the progeny viruses and induces immunogenic cell death(ICD) in A549 and PANC-1 cells. In vivo oAd-CMV-GM-CSF had stronger anti-tumor effect than oAd-GM-CSF in immunodeficient model and immune-competent model. Our findings indicate that oAd-CMV-GM-CSF induces more profound autophagy and promoting viral replication to enhance the anti-tumor efficacy.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145079825","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Myla Hudson, Robert H Newman, Checo J Rorie, Bryan L Holloman, Howard L Kaufman, Samuel D Rabkin, Joseph Graves, Dipongkor Saha
{"title":"Promoting the therapeutic potential of interleukin-7 (IL-7) by expression in viral vectors.","authors":"Myla Hudson, Robert H Newman, Checo J Rorie, Bryan L Holloman, Howard L Kaufman, Samuel D Rabkin, Joseph Graves, Dipongkor Saha","doi":"10.1038/s41417-025-00960-2","DOIUrl":"https://doi.org/10.1038/s41417-025-00960-2","url":null,"abstract":"<p><p>Interleukin 7 (IL-7) is an immunostimulatory cytokine essential for T cell development, proliferation, and maintenance. While IL-7 generates antitumor immunity, systemic IL-7 has not consistently produced strong anticancer effects. Achieving therapeutic cytokine concentrations in tumors often requires high systemic doses, leading to toxicity. To address this, localized cytokine expression within the tumor microenvironment (TME) has gained interest. One such approach involves cytokine expression by oncolytic viruses (OVs) that selectively replicate in cancerous cells while sparing 'normal' cells. Additionally, non-replicative viral vectors have become valuable tools for sustaining cytokine expression in the TME, inducing antitumor effects through non-lytic mechanisms. To effectively harness IL-7's antitumor potential, both oncolytic and non-lytic viruses have been engineered to express IL-7, either alone or in combination with other immunomodulators, such as IL-12, IL-15, B7-1, or CCL19. Despite promising advancements, no comprehensive review exists on IL-7 expression in virus-based immunotherapy for cancer. Therefore, this manuscript aims to (i) summarize studies on viral IL-7 expression alone or with other immunomodulators, (ii) discuss the associated immune mechanisms of action, and (iii) explore opportunities for co-expressing IL-7 with other key cytokines to optimize immunovirotherapy strategies for cancer.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145074437","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hanan Elimam, Abdullah F Radwan, Nadine H El Said, Nourhan Elfar, Mai A Abd-Elmawla, Nora M Aborehab, Khloud Nassar, Osama A Mohammed, Ahmed S Doghish
{"title":"Long non-coding RNAs and signaling networks in non-small cell lung cancer: mechanistic insights into tumor pathogenesis.","authors":"Hanan Elimam, Abdullah F Radwan, Nadine H El Said, Nourhan Elfar, Mai A Abd-Elmawla, Nora M Aborehab, Khloud Nassar, Osama A Mohammed, Ahmed S Doghish","doi":"10.1038/s41417-025-00950-4","DOIUrl":"https://doi.org/10.1038/s41417-025-00950-4","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality globally, largely attributable to its molecular heterogeneity and resistance to current therapeutic modalities. Dysregulation of key intracellular signaling pathways, including EGFR, PI3K/AKT/mTOR, JAK/STAT, and p53, plays a central role in NSCLC pathogenesis, driving tumor initiation, progression, metastasis, and therapeutic resistance. Increasing evidence highlights long non-coding RNAs (lncRNAs) as critical regulatory molecules within these signaling networks. Aberrant lncRNA expression contributes to oncogenic signaling, modulates the tumor microenvironment, and promotes hallmark cancer traits such as uncontrolled proliferation, evasion of apoptosis, metastasis, and chemoresistance. This review synthesizes contemporary findings on the molecular mechanisms by which lncRNAs influence major oncogenic cascades in NSCLC. Both oncogenic and tumor-suppressive lncRNAs are examined, with an emphasis on their functional interplay with signaling mediators and their contributions to tumor biology. Moreover, the clinical relevance of lncRNAs as diagnostic and prognostic biomarkers is explored, alongside emerging therapeutic strategies designed to target lncRNA-mediated dysregulation. Approaches such as antisense oligonucleotides, RNA interference, and CRISPR/Cas9-based gene modulation offer promising avenues for therapeutic intervention. This review provides a comprehensive framework for understanding the roles of lncRNAs in NSCLC and supports the advancement of lncRNA-targeted precision medicine strategies in lung cancer management.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145069068","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Integration of humanized ROBO1 CAR in PD-1 locus in natural killer cells delivers synergistic tumor-killing effect against non-small cell lung cancer.","authors":"Jia-Hao Tao, Jun Zhang, Chun-Yan Tang, Jia-Xi Duan, Wen-Jing Zhong, Chen-Yu Zhang, Yu-Biao Liu, Jin Ling, Hua-Shun Li, Yong Zhou, Cha-Xiang Guan","doi":"10.1038/s41417-025-00957-x","DOIUrl":"https://doi.org/10.1038/s41417-025-00957-x","url":null,"abstract":"<p><p>Lung cancer is the most common cancer and one of the leading causes of cancer-related deaths in the world, however, the treatment of non-small cell lung cancer (NSCLC) is still limited, and it is a clinically urgent problem. ROBO1 is an important surface receptor on tumor cells, but the role of humanized chimeric antigen receptor (CAR) modified natural killer (NK) cells targeting ROBO1 in NSCLC is rarely explored. Furthermore, the role of PD-1 in NK cell killing tumor cells remains controversial. In this study, we identified the expression pattern of ROBO1 in lung squamous cell carcinoma (LUSC) by searching biological information databases. We constructed hROBO1-CAR-NK-92 cells and performed functional identification.We inserted the hROBO1-CAR at the PD-1 locus and performed functional detection in vitro and in vivo. The results showed that ROBO1 expression was significantly increased in LUSC. After inserting the hROBO1-CAR sequence at the PD-1 locus, the PD-1-KO-hROBO1-CAR-NK-92 cells had the best long-term killing ability and cytokine secretion ability, and had a significant inhibitory effect on tumor growth in the mouse xenograft model. We also observed that the long-term killing ability of PD-1-KO-hROBO1-CAR-NK-92 cells was achieved by inhibiting cell senescence via knocking out PD-1. These studies proposed ROBO1 as a key target for CAR-NK therapy in NSCLC and integrated hROBO1 CAR in PD-1 locus in NK cells, resulting in synergistic tumor killing effects in NSCLC, presenting a new treatment strategy for solid tumor treatment.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145005929","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}