Published in last 50 years
Articles published on Xenograft Model
- New
- Research Article
- 10.1038/s41417-025-00981-x
- Nov 4, 2025
- Cancer gene therapy
- Tianyan Liu + 13 more
Newcastle disease virus (NDV) is a promising oncolytic virus, yet requires further optimization. In this study, we engineered an F-gene-chimeric NDV expressing human Interleukin 2 (hIL-2) to enhance the oncolytic efficacy of the NDV Clone30 strain. This recombinant virus, designated ovNDV-28, was then produced in suspension-cultured HEK293 cells. The therapeutic potential of ovNDV-28 was evaluated across multiple cancer cell lines, as well as in the HuH-7 xenograft and B16-F0 syngeneic models. Both in vitro and in vivo results demonstrated that ovNDV-28 significantly improved tumor growth suppression compared to the wild-type NDV. Flow cytometry revealed notable increases in tumor-infiltrating CD3⁺CD4⁺ T cells, CD3⁺CD8⁺ T cells, and CD3⁻CD49b⁺ cells, along with elevated expression levels of IFN-γ, TNF-α, perforin, and Granzyme B within tumor tissue. Comprehensive toxicological assessments conducted on B16-F0 tumor-bearing mice involved intratumoral administration of ovNDV-28 at doses of 1.12 × 10⁶ or 1.46 × 10⁷ PFU/mouse every other day for 14 days. No ovNDV-28-related biochemical, hematological, or histopathological abnormalities were observed. The virus was detected in tumor tissue, mesenteric lymph nodes, abdominal adipose tissue, brain, and biceps femoris, without evidence of blood circulation or viral shedding. This study systematically demonstrates the efficacy, safety, and pharmacokinetics of ovNDV-28, supporting its potential for clinical translation.
- New
- Research Article
- 10.1158/1535-7163.mct-25-0652
- Nov 4, 2025
- Molecular cancer therapeutics
- Jacqueline H Starrett + 14 more
Despite recent advances in the treatment of fibroblast growth factor receptor 3 (FGFR3)-altered metastatic urothelial carcinoma, there is no approved precision therapy that selectively targets FGFR3 while sparing other FGFR isoforms. Dabogratinib (TYRA-300)-a rationally designed selective FGFR3 inhibitor-was evaluated in vitro and in vivo. We also report three patient cases from the ongoing first-in-human, phase I/II SURF301 study (NCT05544552). Dabogratinib elicited a dose-dependent reduction in downstream signaling across three bladder cancer cell lines harboring an FGFR3 fusion, mutation, or gatekeeper resistance mutation. In a xenograft model driven by an FGFR3S249C-activating mutation, dabogratinib treatment resulted in dose-dependent tumor growth inhibition with tumor regression observed at the highest doses. These preclinical findings are supported by the three case reports from the SURF301 study, which demonstrate early clinical activity in patients with advanced metastatic urothelial carcinoma with an FGFR3 fusion or activating mutation.
- New
- Research Article
- 10.17305/bb.2025.11251
- Nov 3, 2025
- Biomolecules & biomedicine
- Bingqing Shi + 8 more
Histone methylation dysregulation is a crucial epigenetic driver of lung carcinogenesis; however, the role of lysine-specific demethylase 3A (KDM3A) in non-small cell lung cancer (NSCLC) remains inadequately understood. In this study, we established NSCLC cell models with both KDM3A overexpression and knockdown to investigate its functional impact. In vitroassays demonstrated that KDM3A depletion increased histone H3 lysine 9 dimethylation (H3K9me2), suppressed cell proliferation, and impaired migration and invasion by attenuating epithelial-mesenchymal transition (EMT) and the expression of matrix metalloproteinase-9 (MMP-9). Conversely, KDM3A overexpression led to reduced H3K9me2 levels, activated EMT, and enhanced metastatic potential. Mechanistically, KDM3A decreased H3K9me2 occupancy at the promoters of VIM and MMP-9, thus upregulating their expression. Additionally, KDM3A downregulated E-cadherin by activating the p-STAT3 pathway. In vivo, KDM3A knockdown significantly inhibited tumor growth in xenograft models. Clinical analyses revealed elevated KDM3A expression in metastatic NSCLC tissues, with a negative correlation between KDM3A and H3K9me2, and a positive association between KDM3A and FOXP3. These findings establish KDM3A as an epigenetic modulator of NSCLC progression through H3K9me2-dependent regulation of EMT and metastatic pathways, highlighting its therapeutic potential for NSCLC treatment.
- New
- Research Article
- 10.1186/s13008-025-00168-8
- Nov 3, 2025
- Cell Division
- Yucheng Guo + 6 more
BackgroundNon-small cell lung cancer (NSCLC) is a major subtype of lung cancer, with high mortality and limited treatment approaches. This paper explores the function of TAL1 in NSCLC progression and glycolysis and its mechanism.MethodsBioinformatics analysis screened out TAL1 and the upstream and downstream molecules. MTT, EdU, wound healing assay, Transwell assay, and TUNEL were utilized to detect the malignant phenotype of A549 and H460 cells. Western blot analysis was conducted to detect the expression of the proliferation-associated protein (Ki67), EMT-associated proteins (E-cadherin, N-cadherin), and glycolysis-associated proteins (GLUT1, LDHA, and PDK1). Cellular metabolism assays detected changes in glucose metabolites. A xenograft model was constructed, and the mouse tumor weight and volumes were measured periodically. Dual-luciferase assays and ChIP assays were performed to authenticate the transcriptional regulation of TAL1 on PKM2 and the relationship between DNMT3B and TAL1.ResultsTAL1 was lowly expressed in NSCLC, and TAL1 overexpression prevented the proliferation, migration, and invasion and elevated apoptosis. TAL1 inhibited PKM2 transcription, and overexpression of PKM2 reversed the trend of overexpression of TAL1 and promoted glycolysis. DNMT3B inhibited TAL1 expression through methylation modification. DNMT3B overexpression facilitated NSCLC cell growth and promoted glycolysis, and further overexpression of TAL1 reversed this trend. In vivo experiments showed that overexpression of TAL1 inhibited NSCLC progression, while combined overexpression of PKM2 promoted NSCLC progression. Overexpression of DNMT3B promoted NSCLC progression, and combined knockdown of PKM2 inhibited NSCLC progression.ConclusionDNMT3B activates glycolysis and promotes NSCLC progression by mediating methylation modification of TAL1 and inducing PKM2 transcription.
- New
- Research Article
- 10.1084/jem.20250256
- Nov 3, 2025
- The Journal of experimental medicine
- Yumei Cheng + 15 more
The only approved systemic treatments for gastrointestinal stromal tumors (GISTs) are KIT/PDGFRA-directed tyrosine kinase inhibitors (TKIs), which eventually lead to the development of secondary polyclonal resistance mutations. Complementary treatment strategies are urgently needed. Using transcriptomic profiling, CRISPR screens, and chemical screens, we identify aurora kinase B (AURKB) as a previously less recognized therapeutic vulnerability to advanced GISTs. AURKB is frequently overexpressed in high-risk and metastatic GISTs but not in low-/intermediate-risk GISTs across our two patient cohorts, with FOXM1 responsible for AURKB overexpression. Genetic depletion of AURKB inhibits GIST proliferation and growth in vitro and in vivo. Mechanistically, our mass spectrometry-based proteomics screen further reveals that AURKB binds to and stabilizes ATAD2 via the ubiquitin-proteasome system, enhancing chromatin accessibility for DNA damage repair genes. Notably, AURKB inhibitors demonstrate potent efficacy in multiple preclinical GIST cell models and xenograft models at safe doses, overcoming TKI resistance. Our comprehensive approaches define unique AURKB-ATAD2 dependency in GISTs and identify non-receptor tyrosine kinase therapeutic strategies for clinical translation.
- New
- Research Article
- 10.3724/abbs.2025154
- Nov 3, 2025
- Acta biochimica et biophysica Sinica
- Jin Ji + 7 more
Mitochondrial dysfunction is closely related to tumor development. Adenine nucleotide translocator 1 (ANT1), which promotes ADP/ATP translocation across the inner mitochondrial membrane, is an important protein involved in mitochondrial function and plays a role in a variety of diseases, including cancers. However, its role in colorectal cancer (CRC) progression remains poorly understood. This study aims to explore the potential role of ANT1 in CRC and its relationship with mitophagy. Through immunohistochemical analysis, we find that ANT1 expression is significantly higher in the tumor tissues of CRC patients than in adjacent normal tissues and that its overexpression is associated with poor prognosis. Further experiments demonstrate that ANT1 knockdown significantly inhibits CRC cell proliferation, migration, and invasion and leads to mitochondrial dysfunction, increased ROS production, and apoptosis by suppressing mitophagy. Mechanistically, ANT1 knockdown downregulates the PINK1/Parkin pathway, thereby inhibiting mitophagy activity. Notably, PINK1 overexpression partially rescues the cellular dysfunction induced by ANT1 knockdown, suggesting a potential role for PINK1 in reversing the suppression of mitophagy. In vivo xenograft models also show that ANT1 knockdown markedly inhibits tumor growth. In conclusion, ANT1 may play a critical role in CRC progression by regulating mitophagy, providing a basis for its potential as a therapeutic target.
- New
- Research Article
- 10.1186/s12967-025-07174-y
- Nov 3, 2025
- Journal of Translational Medicine
- Nana Li + 11 more
Metastasis is the most common cause of colorectal cancer (CRC)-related death. Neutrophil extracellular traps (NETs) promote tumor progression and distant metastasis. This study aimed to explore the role of NETs in CRC liver metastasis. Through analysis of publicly available single-cell transcriptome sequencing databases, in vitro experiments and nude mouse liver xenograft model experiments, we revealed that NETs promote CRC metastatic progression. Using scRNA-Seq technology, we showed that NETs marker expression was higher in metastatic lesions than in primary tumors. NET marker expression was high in colorectal cancer tissues and correlated with advanced tumor pathological grade. In addition, treatment with NETs enhanced the proliferation, migration and invasion of CRC cells in vitro by inducing EMT, as indicated by downregulation of E-cadherin and upregulation of N-cadherin and Vimentin. Cell–cell communication analysis revealed that NETs are related to the PI3K/AKT pathway and regulate the expression of LDHA, a key enzyme in glucose metabolism. In vitro, treatment with NETs promoted LDHA production and cell invasion and migration in CRC, while knockdown of LDHA suppressed EMT. Further, inhibition of LDHA expression or NET formation effectively inhibited NET-induced liver metastasis. In summary, this study elucidates the mechanism by which NETs regulate LDHA expression to promote CRC liver metastasis.Supplementary InformationThe online version contains supplementary material available at 10.1186/s12967-025-07174-y.
- New
- Research Article
- 10.1158/0008-5472.can-25-1435
- Nov 3, 2025
- Cancer research
- Changying Li + 12 more
Pancreatic adenocarcinoma (PAAD) remains one of the most lethal malignancies, primarily due to its robust resistance to genotoxic therapies, such as chemotherapy and radiotherapy. Understanding the mechanisms underlying this resistance is essential to improve clinical outcomes. Here, we identified trimethylguanosine synthase 1 (TGS1), previously known for its role in RNA modification, as a critical mediator of homologous recombination (HR) repair that specifically contributes to resistance in PAAD. TGS1 was significantly overexpressed in PAAD tissues, correlating strongly with advanced disease stages, therapy resistance, and poor patient prognosis. Following DNA damage, ATM kinase phosphorylated TGS1 at serine residues S389 and S531, which mediated its direct interaction with BRCA1 and subsequent recruitment of BRCA1 to DNA damage sites. The phosphorylation-dependent interaction enhanced HR repair efficiency, enabling cancer cells to survive genotoxic stress. Depletion or pharmacological inhibition of TGS1 induced HR deficiency and markedly enhanced sensitivity to DNA-damaging agents, particularly PARP inhibitors, in PAAD cell lines in vitro and in both cell line-derived and patient-derived xenograft models in vivo. Collectively, these findings uncover an ATM-TGS1-BRCA1 signaling axis that promotes DNA repair and resistance to genotoxic therapies in pancreatic cancer, positioning TGS1 as a promising predictive biomarker and therapeutic target to enhance treatment efficacy.
- New
- Research Article
- 10.1158/2326-6066.cir-25-0149
- Nov 3, 2025
- Cancer immunology research
- Stacey Van Pelt + 8 more
Chimeric antigen receptor (CAR) T-cell therapy has demonstrated remarkable efficacy against hematologic malignancies but has struggled to achieve comparable success in solid tumors. A key obstacle in solid tumors is the extracellular matrix (ECM), which impedes CAR T-cell infiltration. In clinical trials, neuroblastoma has shown responsiveness to GD2-directed CAR T-cell therapy; however, the failure of GD2.CAR T cells to effectively clear bulky disease-characterized by dense ECM-highlights the critical challenge of infiltration. In this study, we demonstrate that GD2.CAR T cells exhibit a unique infiltration restriction compared with other CAR T cells and endogenous T cells. A separate analysis of clinical datasets identified MMP7 and SPP1 [which encodes osteopontin (OPN)] as candidate genes to improve the infiltration of GD2.CAR T cells as these were upregulated in tumor-infiltrating leukocytes. MMP-7 and OPN overexpression enhanced CAR T-cell extravasation and interstitial movement in ECM-dense environments in vitro. Overexpression of either OPN or MMP-7 significantly improved tumor infiltration in a xenograft model of neuroblastoma. This resulted in improved tumor control and a survival extension in OPN-GD2.CAR T cell-treated mice compared with unmodified GD2.CAR T cells. OPN overexpression did not increase off-target infiltration into healthy tissues or promote tumor metastasis, highlighting its potential for safe therapeutic application. Our study provides a framework for further exploration of gene modifications to improve CAR T-cell infiltration in solid tumors and identifies OPN as a candidate to explore in this regard. See related Spotlight by Gasparetto and Chiarle, p. 1698.
- New
- Research Article
- 10.1002/mc.70059
- Nov 2, 2025
- Molecular carcinogenesis
- Lu Li + 6 more
M2-like tumor-associated macrophages (TAMs) exert immunosuppressive and protumorigenic functions in hepatocellular carcinoma (HCC). In this study, we explored the function and mechanism of Type IIA topoisomerase (TOP2A) in TAM recruitment and M2 polarization in the HCC microenvironment. The IGF2BP3-TOP2A relationship was confirmed by RIP, MeRIP, luciferase, and mRNA stability assays. Coculture experiments using a transwell system were performed to analyze the impact on the migration, CD206+ cell population, and M2-related marker expression in THP-1-differentiated macrophages (THP-1-M0). Xenograft models were constructed to evaluate TOP2A's role in tumor growth. Expression analysis was performed by quantitative PCR (qPCR), immunoblotting, and immunohistochemical staining. Increased TOP2A expression was associated with advanced tumor stage and worse outcomes in HCC. IGF2BP3 was upregulated and positively correlated with TOP2A expression in HCC samples. TOP2A depletion reduced THP-1-M0 migration and M2 polarization in vitro and attenuated xenograft growth by suppressing TAM infiltration and M2 polarization in vivo. Mechanistically, IGF2BP3 recognized METTL3-catalyzed m6A sites to increase the stability and expression of TOP2A mRNA. TOP2A re-expression abolished IGF2BP3 knockdown-driven suppression of THP-1-M0 migration and M2 polarization. Moreover, TOP2A depletion decreased CCL2 production and YAP1 activation. CCL2 reconstruction or the Hippo pathway inhibitor XMU-MP-1 reversed TOP2A knockdown-driven suppression of THP-1-M0 migration and M2 polarization. Our findings identify the IGF2BP3/TOP2A axis as a master regulator of TAM recruitment and polarization in HCC via IGF2BP3-m6A-dependent TOP2A stabilization to facilitate YAP1-mediated CCL2 upregulation, providing novel strategies to overcome immunosuppression and combat HCC.
- New
- Research Article
- 10.1002/mc.70060
- Nov 2, 2025
- Molecular carcinogenesis
- Tianming Chen + 3 more
Prostate cancer (PCa) is one of the most common genitourinary malignancies in men worldwide. As a 5-methylcytosine (m5C) methyltransferase, NSUN2 has been implicated in regulating PCa progression. This study aimed to investigate the role of NSUN2 in PCa and elucidate its underlying mechanisms. The biological behaviors of PCa cells were assessed using Cell Counting Kit-8, EdU incorporation, and Transwell assays. The expression levels of relevant RNAs were determined via quantitative real-time PCR. The interaction between NSUN2 and YES proto-oncogene 1 (YES1) was examined through methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP), and dual-luciferase reporter assays. Results showed that NSUN2 was elevated in PCa, and its downregulation suppressed cell viability, proliferation, migration, and invasion. Mechanistically, NSUN2 interacted with YES1 and stabilized its mRNA by promoting m5C modification on YES1. The oncogenic role of NSUN2 was further confirmed in xenograft models in vivo. In conclusion, our study demonstrated that NSUN2 facilitated malignant proliferation and migration of PCa cells by enhancing YES1 mRNA stability via m5C modification. These findings suggested that both NSUN2 and YES1 may serve as potential therapeutic targets for PCa, offering new strategies for treatment.
- New
- Research Article
- 10.2967/jnumed.125.269707
- Nov 1, 2025
- Journal of Nuclear Medicine
- Feifei Liu + 22 more
Prostate-specific membrane antigen (PSMA)-directed radiopharmaceutical therapies continue to improve treatment outcomes in patients with metastatic castration-resistant prostate cancer. Here, we report the invitro and invivo characterization of a PSMA-targeted therapy (ADVC001) specifically designed for targeted α-therapy with 212Pb. Methods: The binding affinity to PSMA was determined by PSMA enzymatic assays and by radioligand binding assays using PSMA-high prostate cancer (PC) cells. In vitro cytotoxicity against PC cell lines with high and medium PSMA expression was evaluated using clonogenic, metabolic, and imaging-based cytotoxic assays. Pharmacokinetics and biodistribution were assessed using PSMA-high subcutaneous tumor xenografts. In vivo single-dose and multidose efficacy was assessed in subcutaneous PC xenograft models expressing various levels of PSMA. Results: A high binding affinity to PSMA was observed for ADVC001 with nanomolar inhibitory concentration of 50% values. In cellular assays, [212Pb]Pb-ADVC001 (212Pb-ADVC001 hereafter for simplicity) exhibited specific cytotoxic activity against PSMA-expressing cells with nanomolar effective concentration of 50% values. In vivo biodistribution of 212Pb-ADVC001 in the PC3-PIP xenograft model revealed rapid and persistent tumor uptake, fast renal clearance, and low retention in normal tissues. Single-dose efficacy studies of 212Pb-ADVC001 (0.46 MBq) showed improved survival compared with [177Lu]Lu-PSMA-I&T (177Lu-PSMA-I&T hereafter for simplicity) (20 MBq) treatment. In a multidose experiment, 2 doses of 212Pb-ADVC001 (0.5 MBq) significantly increased median survival (86 d vs. 45.5 d, P < 0.05) compared with 2 doses of 177Lu-PSMA-I&T (15 MBq). Treatment with 212Pb-ADVC001 (0.5 MBq) after initial 177Lu-PSMA-I&T (15 MBq) relapse showed an enhanced survival benefit (59.5 d). In a C4-2 xenograft model with medium-level PSMA expression, single doses of 0.3, 0.8, and 1.1 MBq of 212Pb-ADVC001 significantly extended median survival to 34, 57, and 62.5 d, compared with untreated cohorts (16 d). All treatments were well tolerated. Conclusion: The preclinical results support the clinical development of 212Pb-ADVC001 as a targeted α-therapy for the treatment of patients with PC.
- New
- Research Article
- 10.1016/j.cancergen.2025.10.104
- Nov 1, 2025
- Cancer genetics
- Haohui Li + 10 more
Expression and clinical significance of the imprinted gene PHLDA2 in colorectal cancer.
- New
- Research Article
- 10.1016/j.cellsig.2025.112064
- Nov 1, 2025
- Cellular signalling
- Qiaozhi Jin + 4 more
LncRNA ZNF295-AS1 modulates nasopharyngeal carcinoma progression via the miR-762/HDAC6 axis-mediated autophagy.
- New
- Research Article
- 10.1016/j.nucmedbio.2025.109092
- Nov 1, 2025
- Nuclear medicine and biology
- Christopher T Hensley + 11 more
L-5-[11C]-glutamine PET of breast cancer: Preclinical studies in mouse models.
- New
- Research Article
- 10.1016/j.ejmech.2025.118020
- Nov 1, 2025
- European journal of medicinal chemistry
- Ru Wang + 8 more
Discovery of potent and selective STAT3 inhibitors against triple-negative breast cancer.
- New
- Research Article
- 10.1016/j.biopha.2025.118714
- Nov 1, 2025
- Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie
- Eunji Kim + 7 more
Cetuximab-mediated antibody-dependent cell-mediated cytotoxicity enhances anti-tumor efficacy of patient-derived natural killer cells in pancreatic cancer.
- New
- Research Article
- 10.1016/j.cellsig.2025.112054
- Nov 1, 2025
- Cellular signalling
- Yiwei Lin + 6 more
GLI2/Parkin-mediated mitophagy promotes pazopanib resistance in clear cell renal cell carcinoma.
- New
- Research Article
- 10.1016/j.cellsig.2025.111987
- Nov 1, 2025
- Cellular signalling
- Shuangde Liu + 4 more
High expression of CHEK2 promotes tumor progression in kidney renal clear cell carcinoma through targeting TEAD4 signaling pathway.
- New
- Research Article
- 10.1016/j.cellsig.2025.112047
- Nov 1, 2025
- Cellular signalling
- Xin Yang + 6 more
RBM39 silence suppresses esophageal cancer proliferation and metastasis via FANCD2 mRNA destabilization.