1
|
Liu Q, Yu M, Lin Z, Wu L, Xia P, Zhu M, Huang B, Wu W, Zhang R, Li K, Zhu L, Wang Q. COL1A1-positive endothelial cells promote gastric cancer progression via the ANGPTL4-SDC4 axis driven by endothelial-to-mesenchymal transition. Cancer Lett 2025; 623:217731. [PMID: 40254092 DOI: 10.1016/j.canlet.2025.217731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/19/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Gastric cancer (GC) is an aggressive and heterogeneous disease with poor survival outcomes. The progression of GC involves complex, multi-step processes. Endothelial cells (ECs) play a crucial role in tumor angiogenesis, proliferation, invasion, and metastasis, particularly through the process of endothelial-to-mesenchymal transition (EndoMT). However, the specific role and mechanisms of EndoMT in gastric cancer remain unclear. Based on 6 GC single-cell RNA-sequencing (scRNA-seq) cohorts (samples = 97), we established an EndoMT-related gene signature, termed EdMTS. Leveraging this gene signature, ssGSEA was applied to calculate sample scores across multiple bulk RNA-seq datasets, which include information on immunotherapy, metastasis, GC progression, and survival. Moreover, we applied the Monocle2 method to calculate cell pseudotime and used CellChat to analyze interactions between malignant and EC cells. We verified the molecular mechanism by multiple immunofluorescence and cell function experiments. Findings In this study, we established a single-cell atlas of ECs in GC and identified a subpopulation of COL1A1+ ECs that play a critical role in tumor progression and metastasis. These COL1A1+ ECs were significantly associated with worse clinical outcomes in GC patients. Further analysis revealed that COL1A1+ ECs originated from lymphatic ECs and underwent EndoMT through the upregulation of CEBPB, driving tumor invasiveness. Moreover, COL1A1+ ECs interacted with malignant cells via ANGPTL4-SDC4 axis, enhancing invasion and migration. These findings provide a deeper understanding of the role of COL1A1+ ECs in GC progression and highlight potential therapeutic targets for disrupting the EndoMT process in these cells to provide a benefit for GC patients.
Collapse
Affiliation(s)
- Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Miao Yu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Peng Xia
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Wei Wu
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Qin Z, Li Y, Shao X, Li K, Bai Y, Wang B, Ma F, Shi W, Song L, Zhuang A, He F, Ding C, Yang W. HNF4A functions as a hepatocellular carcinoma oncogene or tumor suppressor depending upon the AMPK pathway activity status. Cancer Lett 2025; 623:217732. [PMID: 40254090 DOI: 10.1016/j.canlet.2025.217732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cancer cells frequently undergo energy metabolic stress induced by the increased dynamics of nutrient supply. Hepatocyte nuclear factor 4A (HNF4A) is a master transcription factor (TF) in hepatocytes that regulates metabolism and differentiation. However, the mechanism underlying how HNF4A functions in cancer progression remains unclear due to conflicting results observed in numerous studies. To address the roles of HNF4A in hepatocellular carcinoma (HCC), we investigated the regulatory functions of HNF4A in HCC cells under different glucose supply conditions. We found that HNF4A exhibited tumor-suppressive effects on the proliferation and migration of HCC cells in glucose-sufficient conditions and tumor-promotive effects on HCC cells in glucose-insufficient conditions. Further investigation revealed that this diverse function of HNF4A was dependent upon the AMPK pathway activity. Similarly, the prognosis predicted by HNF4A was also correlated with whether the AMPKa expression levels were low or high in clinical HCC patients. Multiomics approaches consisting of proteomics and ChIP-seq revealed that key HNF4A target genes, including NEDD4 and RPS6KA2, are involved in the diverse function of HNF4A in HCC in response to the AMPK activity status. Specifically, HNF4A could bind to the promoter region of NEDD4 and RPS6KA2, and upregulating their expression. Our study has demonstrated the relationship between and synergism of AMPK and HNF4A in the progression of HCC under diverse nutrient conditions.
Collapse
Affiliation(s)
- Zhaoyu Qin
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Yan Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Xiexiang Shao
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Kai Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Yihe Bai
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Bing Wang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Fahan Ma
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Wenhao Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lei Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Aojia Zhuang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Fuchu He
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Chen Ding
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
3
|
Li K, Wu L, Zhu L, Wang W, Chen Y, Ma Z, Zhang G, Gu M, Zhang H, Wu H. The Role of Herb-Partitioned Moxibustion in the Angiogenesis of Colitis-Associated Cancer in Rats. J Inflamm Res 2025; 18:6623-6639. [PMID: 40438180 PMCID: PMC12118578 DOI: 10.2147/jir.s518214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 05/08/2025] [Indexed: 06/01/2025] Open
Abstract
Purpose Angiogenesis in tumors is imperative to tumor growth. Our previous studies revealed that herb-partitioned moxibustion (HPM) could delay colitis-associated cancer (CAC), but the mechanism of the effects on the angiogenesis remains largely undiscovered. We aimed to investigate whether HPM delays CAC by inhibiting the angiogenesis with emergent three-dimensional (3D) imaging technologies. Materials and Methods The CAC model was induced by azoxymethane (AOM)/dextran sodium sulphate (DSS). The rats were randomly divided into normal, model and HPM groups. The tumorigenesis, number of tumors, and tumor diameter were observed. Immunohistochemistry or enzyme-linked immunosorbent assay (ELISA) was performed to assess the microvessel density (MVD), reactive oxygen species (ROS), hypoxia-inducible factor-1 alpha (HIF-1α), vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor receptor 1 (VEGFR1), interleukin-6 (IL-6), interleukin-1 beta (IL-1β) and tumor necrosis factor-alpha (TNF-α). The three-dimensional imaging of solvent-cleared organs with superior fluorescence-preserving capability (FDISCO) tissue clearing technique was used to clear colon tissues, and the platelet endothelial cells were stained and labelled with platelet endothelial cell adhesion molecule 1 (PECAM-1). Imaris software was used to perform 3D measurement and analysis of the colonic vascular architecture. Results The HPM group were found decreased in the colon tumor diameter, MVD, ROS, HIF-1α, VEGFA, VEGFR1, IL-6, IL-1β, and TNF-α in colon tissues compared with those in the model group. 3D imaging revealed that the number of vessels, number of branch points, and vessel branch level in the HPM group were lower than those in the model group. The number of branch points and vessel branch level were negatively correlated with the average vessel length. Conclusion HPM plays a role in inhibiting CAC angiogenesis. This study may provide new evidence at the macroscopic level of vascular architecture for HPM to inhibit the progression of CAC by FDISCO tissue clearing technique for 3D imaging.
Collapse
Affiliation(s)
- Kunshan Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Luyi Wu
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Lu Zhu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Wenjia Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yiyi Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhe Ma
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Guangtao Zhang
- Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Muen Gu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hanxiao Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Huangan Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
4
|
Xia W, Need E, Schiavone C, Singh N, Huang J, Goff M, Cave J, Gillespie DL, Jensen RL, Pagel MD, Dogra P, Shi S, Goel S. Image-guided targeting of mitochondrial metabolism sensitizes pediatric malignant rhabdoid tumors to low-dose radiotherapy. SCIENCE ADVANCES 2025; 11:eadv2930. [PMID: 40408469 PMCID: PMC12101499 DOI: 10.1126/sciadv.adv2930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/18/2025] [Indexed: 05/25/2025]
Abstract
Tumor hypoxia leads to radioresistance and markedly worse clinical outcomes for pediatric malignant rhabdoid tumors (MRTs). Our transcriptomics and bioenergetic profiling data reveal that mitochondrial oxidative phosphorylation is a metabolic vulnerability of MRT and can be exploited to overcome consumptive hypoxia by repurposing an FDA-approved antimalarial drug, atovaquone (AVO). We then establish the utility of oxygen-enhanced-multispectral optoacoustic tomography, a label-free, ionizing radiation-free imaging modality, to visualize and quantify spatiotemporal changes in tumor hypoxia in response to AVO. We show a potent but transient increase in tumor oxygenation upon AVO treatment that results in complete elimination of tumors in all tested mice when combined with 10-gray radiotherapy, a dose several times lower than the current clinic standard. Last, we use translational mathematical modeling for systematic evaluation of dosing regimens, administration timing, and therapeutic synergy in a virtual patient cohort. Together, our work establishes a framework for safe and pediatric patient-friendly image-guided metabolic radiosensitization of rhabdoid tumors.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Esther Need
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Carmine Schiavone
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiemin Huang
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Matthew Goff
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Joseph Cave
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - David L. Gillespie
- Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84132, USA
| | - Randy L. Jensen
- Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84132, USA
| | - Mark D. Pagel
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
5
|
Liu L, Song L, Liu T, Hui K, Hu C, Yang J, Pi X, Yan Y, Liu S, Zhang Y, Chen H, Cao Y, Zhou L, Qiao Y, Yu D, Yin C, Li X, Zhang C, Li D, Wang Z, Liu Z, Jiang X. Recombinant oncolytic virus NDV-anti-VEGFR2 enhances radiotherapy sensitivity in NSCLC by targeting VEGF signaling and impairing DNA repair. Gene Ther 2025:10.1038/s41434-025-00540-x. [PMID: 40382521 DOI: 10.1038/s41434-025-00540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 04/09/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Resistance to radiotherapy is a significant challenge in the clinical management of non-small cell lung cancer (NSCLC). This study investigates a novel multimodal therapeutic strategy that combines oncolytic Newcastle disease virus (NDV) with an anti-VEGFR2 single-chain variable fragment (NDV-anti-VEGFR2) to enhance radiosensitivity in NSCLC. We engineered NDV-anti-VEGFR2 and assessed its efficacy in sensitizing Calu-1 cells to radiation. In vitro results demonstrated that NDV-anti-VEGFR2 significantly inhibited tumor cell proliferation when combined with radiotherapy. In vivo experiments revealed that NDV-anti-VEGFR2, combined with radiation, achieved a tumor growth inhibition rate of 86.48%, surpassing the effects of NDV or radiation alone. Mechanistic investigations indicated that NDV-anti-VEGFR2 mitigated hypoxia by downregulating HIF-1α and impaired DNA repair pathways, as evidenced by reduced levels of RAD51 and γ-H2AX. These findings suggest that NDV-anti-VEGFR2 not only normalizes tumor vasculature but also enhances the cytotoxic effects of radiation by compromising DNA repair mechanisms. Collectively, our results support the clinical potential of NDV-anti-VEGFR2 combined with radiotherapy as a promising strategy to overcome radiotherapy resistance in NSCLC. Future studies in immunocompetent models are warranted to elucidate the immune-mediated effects of this innovative therapeutic approach.
Collapse
Affiliation(s)
- Liang Liu
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Liying Song
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Tianyan Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Kaiyuan Hui
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Chenxi Hu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Jiarui Yang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xuelei Pi
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yuanyuan Yan
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Shishi Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yating Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Hongna Chen
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yukai Cao
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Lihua Zhou
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Yun Qiao
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Dan Yu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chengkai Yin
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xu Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chenfeng Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Deshan Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhihang Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China.
| | - Xiaodong Jiang
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China.
| |
Collapse
|
6
|
Wu F, Lu Z, Que J, Ma S, Jiang L, Tang X, Zheng C, Zhou L, Huang Q, Zhang Y. The safety of combining Endostar with concurrent chemoradiotherapy for the treatment of locally advanced cervical cancer and the evaluation of its anti-angiogenic effects via transrectal contrast-enhanced ultrasound. Front Oncol 2025; 15:1514425. [PMID: 40406255 PMCID: PMC12095288 DOI: 10.3389/fonc.2025.1514425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Background In recent years, exploring the addition of angiogenesis inhibitors to chemoradiotherapy for locally advanced cervical cancer (LACC) has gained research interest. This study assessed the safety and anti-angiogenic effects of combining Endostar with concurrent chemoradiotherapy (CCRT) via transrectal contrast-enhanced ultrasound. Methods A total of 120 patients with locally advanced cervical cancer (LACC) were randomly allocated to two groups: CCRT combined with Endostar (CRT+E group, n = 60) and CCRT alone (CRT group, n = 60). Endostar was administered intravenously before radiotherapy and repeated for four cycles. All patients received platinum-based CCRT. Adverse events were monitored, and transrectal contrast-enhanced ultrasonography (CEUS) was conducted before, during, and after radiotherapy. Vascular malformation (VM) rates were calculated from tumor cross-sectional images, and quantitative analysis software measured peak intensity (PI), time to peak (TTP), and mean transit time (MTT) of tumor vessels. Results No significant differences were observed in hematological, hepatic, renal, gastrointestinal, or cardiac adverse reactions between the two groups (all P>0.05). In the CRT+E group, VM rates, TTP, and MTT significantly differed at three time points (with P values of 0.003, 0.002, and P<0.001, respectively), whereas the CRT group showed no significant changes (all P>0.05). Post-radiotherapy, statistically significant differences emerged between the CRT+E and CRT groups for VM rates (P = 0.027), MTT (P = 0.027), and TTP (P < 0.001), while PI showed no significant difference (65.67 ± 36.53 vs. 74.69 ± 61.21, P = 0.598). Conclusion The combination of Endostar with CCRT for locally advanced cervical cancer (LACC) demonstrated favorable safety and tolerability. Transrectal contrast-enhanced ultrasound (CEUS) effectively assessed tumor vascular normalization induced by Endostar during CCRT. Specifically, Endostar significantly reduced VM rates and shortened MTT, suggesting its potential to normalize tumor vasculature.
Collapse
Affiliation(s)
- Fang Wu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Zhouxue Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Jinting Que
- Department of Oncology, The First People’s Hospital of Qinzhou, Qinzhou, Guangxi, China
| | - Shanshan Ma
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Li Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Xiaobi Tang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Chengshan Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Li Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Qiufeng Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| | - Yong Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China
| |
Collapse
|
7
|
Harkos C, Hadjigeorgiou AG, Voutouri C, Kumar AS, Stylianopoulos T, Jain RK. Using mathematical modelling and AI to improve delivery and efficacy of therapies in cancer. Nat Rev Cancer 2025; 25:324-340. [PMID: 39972158 DOI: 10.1038/s41568-025-00796-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
Mathematical modelling has proven to be a valuable tool in predicting the delivery and efficacy of molecular, antibody-based, nano and cellular therapy in solid tumours. Mathematical models based on our understanding of the biological processes at subcellular, cellular and tissue level are known as mechanistic models that, in turn, are divided into continuous and discrete models. Continuous models are further divided into lumped parameter models - for describing the temporal distribution of medicine in tumours and normal organs - and distributed parameter models - for studying the spatiotemporal distribution of therapy in tumours. Discrete models capture interactions at the cellular and subcellular levels. Collectively, these models are useful for optimizing the delivery and efficacy of molecular, nanoscale and cellular therapy in tumours by incorporating the biological characteristics of tumours, the physicochemical properties of drugs, the interactions among drugs, cancer cells and various components of the tumour microenvironment, and for enabling patient-specific predictions when combined with medical imaging. Artificial intelligence-based methods, such as machine learning, have ushered in a new era in oncology. These data-driven approaches complement mechanistic models and have immense potential for improving cancer detection, treatment and drug discovery. Here we review these diverse approaches and suggest ways to combine mechanistic and artificial intelligence-based models to further improve patient treatment outcomes.
Collapse
Affiliation(s)
- Constantinos Harkos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas G Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Yang H, Ma Y, Zhang C, Leng Q, Cheng K, Zhao C, Cao D. Case Report: Combined PD-1 and tyrosine kinase blockade stabilizes refractory pancreatic cancer guided by the spatial structure of tumor immune microenvironment. Front Immunol 2025; 16:1547388. [PMID: 40376004 PMCID: PMC12078320 DOI: 10.3389/fimmu.2025.1547388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/10/2025] [Indexed: 05/18/2025] Open
Abstract
Pancreatic cancer is characterized by a poor prognosis and limited responsiveness to conventional therapies, presenting a substantial therapeutic challenge. Although chemotherapy remains the cornerstone of systemic treatment, options become scarce once frontline therapies fail. While targeted therapies and immunotherapies have emerged as potential alternatives, their efficacy in pancreatic cancer is not well established. As research advances, exploring the tumor immune microenvironment (TiME) of pancreatic cancer is crucial and holds significant potential for developing novel treatment strategies.We report a case of a pancreatic cancer patient who, after the failure of frontline and second-line treatments, was treated with a pioneering combination of targeted therapy and immunotherapy to modulate the unique TiME. The targeted agent, surufatinib, is a tyrosine kinase inhibitor (TKI) that targets vascular endothelial growth factor receptor (VEGFR) 1-3, fibroblast growth factor receptor 1 (FGFR1), and colony-stimulating factor 1 receptor (CSF-1R). The immunotherapy agent, toripalimab, is an immune checkpoint inhibitor targeting programmed cell death protein 1 (PD-1). Remarkably, the patient benefitted from this regimen, exhibiting stable disease, improved clinical symptoms, and prolonged progression-free survival. This case highlights the potential of personalized therapy in treating pancreatic cancer, particularly in patients with distinctive features of the TiME that may predict favorable responses to immunotherapy. Personalized strategies that consider the spatial structure and composition of the TiME may offer a promising avenue for achieving long-term progression-free survival in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Heqi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuhang Ma
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenyan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingqing Leng
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ke Cheng
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Dan Cao
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Bi X, Deng Y, Chu C, Wei M, Zhao J, Zhao J, Wang Y, Yin T, Gou J, He H, Tang X, Li G, Zhang Y. Precision-targeted explosion of biomimetic nanoparticles for the effective treatment of uveal melanoma. Int J Pharm 2025; 675:125543. [PMID: 40164415 DOI: 10.1016/j.ijpharm.2025.125543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/13/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, originating from the melanocytes within the uvea. Currently, the treatment of ocular tumors predominantly relies on conventional approaches such as brachytherapy and enucleation. Despite the limited pharmaceutical treatment options for uveal melanoma (UM), the effectiveness of ocular drug delivery is hindered by the ocular barrier to local drug administration and the complex tumor microenvironment (TME). In response, biomimetic low-density lipoprotein nanoparticles (LD-DPVP NPs) with active targeting capabilities were designed. This nanodrug system combined photosensitizer (verteporfin, VP) with the tumor vascular normalization drug (dexamethasone, DEX) to achieve low-toxicity, high-efficacy treatment of intraocular tumors. After intravenous injection, the nanoparticles selectively targeted the tumor site and induced VP to produce reactive oxygen species (ROS) that killed tumor cells under near-infrared laser stimulation. The produced ROS could also trigger the cleavage of the DEX prodrug (DPD) and rapid release of DEX via breakage of the thioether bond (TK). Additionally, DEX could modulate the TME, improving the delivery of nanoparticles to the tumor and further enhancing the efficacy of LD-DPVP NPs. We believe the biomimetic nanoparticles designed in this study have a potential clinical application value in inhibiting UM growth and provided a promising strategy for addressing other ocular malignancies.
Collapse
Affiliation(s)
- Xiaoshuang Bi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Yaxin Deng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Chenxiao Chu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Mingli Wei
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Jiansong Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Jiaqi Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Yuying Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 WenhuaRoad, shenyang 1100l6, Liaoning, China
| | - Tian Yin
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016 Liaoning, China
| | - JingXin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Guofei Li
- Shengjing Hospital of China Medical University, Department of Pharmacy, No. 36, Sanhao Street, Shenyang 110004, China.
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| |
Collapse
|
10
|
Cai Z, Meng K, Yu T, Xi Y, Yuan Z, Wang X, Wang C, Li L, Fu X. IFN-γ-mediated suppression of ANGPT2-Tie2 in endothelial cells facilitates tumor vascular normalization during immunotherapy. Front Immunol 2025; 16:1551322. [PMID: 40370455 PMCID: PMC12075545 DOI: 10.3389/fimmu.2025.1551322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Tumor angiogenesis is a critical biological hallmark of cancer, which involves multiple molecularly regulated signaling pathways, including the angiopoietin (ANGPT)-Tie2 and the vascular endothelial growth factor (VEGF) signaling pathways. Despite initial optimism, targeting tumor angiogenesis in the treatment of lung adenocarcinoma (LUAD) has been unsatisfactory. Currently, monotherapy with PD-1/PD-L1 inhibitors, or their combination with bevacizumab, is considered the standard therapeutic approach for LUAD. Recent studies have shown that immunotherapy suppresses tumor angiogenesis and facilitates vascular normalization. However, whether and how anti-PD-L1 therapy influences tumor vasculature remains unclear. Methods To investigate the impact of immunotherapy on the vasculature of LUAD, a mouse model of lung adenocarcinoma was established by subcutaneous implantation of Lewis lung carcinoma cells in vivo. The effects of different treatments on microvessel density and pericyte coverage were explored, and the expression of angiogenesis-related factors was analyzed. Furthermore, to explore the molecular mechanisms through which IFN-γ regulates tumor blood vessels during immunotherapy, we elucidated the specific mechanisms in vitro by means of techniques such as siRNA, ChIP, RT-qPCR, Western blot, and immunofluorescence. Finally, the effects of IFN-γ on the proliferation, migration, and angiogenic function of endothelial cells (ECs) were evaluated through CCK-8, Transwell, and HUVEC tube formation assays. Results Employing a mouse model of LUAD, we demonstrated that PD-L1 blockade therapy inhibits tumor angiogenesis and normalizes vasculature in an IFN-γ-signaling-dependent manner. Notably, anti-PD-L1 therapy reduced Tie2 and ANGPT2 expression, and these effects were reversed by the JAK1/2 inhibitor. Mechanistically, we demonstrated that IFN-γ inhibited Tie2 and ANGPT2 expression in ECs, and suppressed ANGPT2 gene transcription through the AKT-FOXO1 signaling pathway. Interestingly, IFN-γ-mediated activation of STAT1 exerts negative regulation by directly binding to the promoter regions of the ANGPT2 and TEK genes. Functionally, IFN-γ limits the migration, proliferation, and tube formation of ECs. Discussion In conclusion, our results revealed a novel mechanism wherein IFN-γ-mediated inhibition of ANGPT2-Tie2 facilitates vascular normalization during immunotherapy in LUAD, which performs an essential function in the antitumor efficacy of immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lequn Li
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Xiangning Fu
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| |
Collapse
|
11
|
Wang W, Yuan J, Zhu Y, Li R, Zhang J. Traditional Chinese medicine (TCM) enhances the therapeutic efficiency of a gemcitabine-loaded injectable hydrogel on postoperative breast cancer through modulating the microenvironment. J Mater Chem B 2025; 13:4864-4878. [PMID: 40171620 DOI: 10.1039/d4tb02776k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Local injection of the drug-loaded hydrogel at the surgery site is promising for postoperative breast cancer. However, the postoperative changes in the tumor microenvironment, such as inflammation, abnormal angiogenesis and hypoxia, inhibit drug perfusion and contribute to breast cancer recurrence (BCR). Normalizing the abnormal blood vessels can effectively improve perfusion and reduce hypoxia. Here, we encapsulated gemcitabine (GEM) in a PLGA-PEG-PLGA hydrogel (GEM-hydrogel) for local treatment of postoperative breast cancer. The GEM-hydrogel can be injected into the surgery cavity allowing sustained release of the drug. Meanwhile, traditional Chinese medicine (TCM) Shexiang Baoxin Pill (SBP) was given to normalize the blood vessels to enhance drug perfusion. The results suggest that the combination of SBP enhances the therapeutic efficiency of the GEM-hydrogel, inhibiting tumor recurrence. Mechanism studies reveal that SBP works by promoting PDGFB expression in macrophages, subsequently recruiting pericytes, and normalizing blood vessels, finally alleviating hypoxia. This study demonstrates that the combination of TCM and chemotherapeutics is promising for suppressing postoperative tumor recurrence.
Collapse
Affiliation(s)
- Wenxu Wang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jixiang Yuan
- Urology Centre, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200000, China
| | - Yuying Zhu
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ruixiang Li
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jiange Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
12
|
Zhou ZY, Bai N, Zheng WJ, Ni SJ. MultiOmics analysis of metabolic dysregulation and immune features in breast cancer. Int Immunopharmacol 2025; 152:114376. [PMID: 40054322 DOI: 10.1016/j.intimp.2025.114376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 02/09/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Metabolic disorders and diminished immune response are hallmark characteristics of tumors. However, limited studies have comprehensively integrated metabolic and immunological factors to evaluate or predict the prognosis of cancer patients. In this study, we utilized 72 metabolic pathway gene sets from the MsigDB database to conduct GSVA, univariate regression, and prognostic analyses on 247 breast cancer samples sourced from the TCGA and GEO databases. Consequently, five metabolic pathways with significant research value were identified. Based on these findings, unsupervised clustering was performed on the breast cancer samples to compare differences in gene expression, clinicopathological features, immune infiltration levels, and prognosis across different clusters. This process led to the identification of nine metabolism-related characteristic genes. Additionally, single-cell sequencing analysis was employed to assess the spatial expression patterns of these characteristic genes, revealing significantly higher expression indices in tumor cells compared to non-tumor cells. Subsequently, machine learning algorithms were applied to reconstruct metabolic risk models for evaluating the prognosis of breast cancer patients. The results indicated that the high metabolic risk group exhibited higher gene mutation scores, a greater proportion of unfavorable clinicopathological parameters, and lower chemokine and immune scores compared to the low-risk group. In conclusion, the metabolic risk model constructed using metabolism-related characteristic genes can accurately distinguish and predict the survival prognosis and immunotherapy outcomes of breast cancer patients, offering novel targets and insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Zuo-Yuan Zhou
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Nan Bai
- Medical school of Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Wen-Jie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| | - Su-Jie Ni
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
13
|
Hoeh AE, Chang JH, Mueller RS, Basche M, Fantin A, Sepetis A, De Rossi G, Dritsoula A, Ali RR, Turowski P, Moss SE, Greenwood J. LRG1 Alters Pericyte Phenotype and Compromises Vascular Maturation. Cells 2025; 14:593. [PMID: 40277918 PMCID: PMC12026257 DOI: 10.3390/cells14080593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/02/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Upregulation of leucine-rich alpha-2-glycoprotein-1 (LRG1) contributes to aberrant neovascularization in many different diseases. In contrast, LRG1 is not involved in developmental angiogenesis. Here, we investigated the vasculopathic properties of LRG1 by examining its effect on developing retinal blood vessels. By injecting recombinant protein or an expression vector into the mouse retina during vascular development, we showed that exogenous LRG1 reduces pericyte coverage and NG2 expression. It leads to diminished collagen IV sheathing, fewer adhesion and gap junctions, and reduced vessel calibre and vascular density. Moreover, in mouse retinae containing exogenous LRG1, the developing blood-retinal barrier remains more permeable with significantly higher numbers of transcytotic vesicles present in microvascular endothelial cells. These results reveal that exogeneous LRG1 is sufficient to interfere with the maturation of developing retinal vessels and drive vessel development towards a dysfunctional phenotype. These observations deliver further evidence that LRG1 is an angiopathic factor and highlight the therapeutic potential of blocking LRG1 in diseases characterized by pathogenic angiogenesis or vascular remodelling.
Collapse
Affiliation(s)
- Alexandra E. Hoeh
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Jui-Hsien Chang
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Ronja S. Mueller
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Mark Basche
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King’s College London, London SE1 9RT, UK
| | | | - Anastasios Sepetis
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Giulia De Rossi
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Athina Dritsoula
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Robin R. Ali
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King’s College London, London SE1 9RT, UK
| | - Patric Turowski
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Stephen E. Moss
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - John Greenwood
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| |
Collapse
|
14
|
Hongu T, Sarenqiqige, Shandan, Kusunoki H, Ishimura A, Suzuki T, Oskarsson T, Gotoh N. Permeable Lung Vasculature Creates Chemoresistant Endothelial Niche by Producing SERPINE1 at Breast Cancer Metastatic Sites. Cancer Sci 2025. [PMID: 40217581 DOI: 10.1111/cas.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 04/14/2025] Open
Abstract
Chemotherapy resistance remains a major obstacle for eradicating metastatic cancer cells in distant organs. We identified that endothelial cells (ECs) in the lungs, where breast cancer cells often metastasize, form a chemoresistant perivascular niche for disseminated breast cancer cells. By investigating the lung EC secretome activated by metastasis, we found that serine protease inhibitor family E member 1 (SERPINE1), encoded by Serpine1, is upregulated in metastasis-associated lung ECs. This upregulation shields cancer cells from paclitaxel-induced apoptosis and promotes cancer stem cell properties. Serpine1 expression appears to be driven by YAP-TEAD activation in lung ECs that lose cell-cell contact, a phenomenon associated with increased vascular permeability in lungs affected by metastasis. Crucially, pharmacological inhibition of SERPINE1 enhances the chemotherapy sensitivity of metastatic breast cancer cells in the lung. Overall, our findings underscore the pivotal role of the vascular niche, which produces SERPINE1, in conferring chemoresistance to breast cancer cells during metastatic progression in the lungs.
Collapse
Affiliation(s)
- Tsunaki Hongu
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| | - Sarenqiqige
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Shandan
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Hirokazu Kusunoki
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Akihiko Ishimura
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Takeshi Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Thordur Oskarsson
- Department of Molecular Oncology, H. Lee Moffit Cancer Center & Research Institute, Tampa, Florida, USA
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| |
Collapse
|
15
|
Torres-Mejia E, Weng S, Whittaker CA, Nguyen KB, Duong E, Yim L, Spranger S. Lung Cancer-Intrinsic SOX2 Expression Mediates Resistance to Checkpoint Blockade Therapy by Inducing Treg-Dependent CD8+ T-cell Exclusion. Cancer Immunol Res 2025; 13:496-516. [PMID: 39745382 PMCID: PMC11964848 DOI: 10.1158/2326-6066.cir-24-0184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 10/14/2024] [Accepted: 12/23/2024] [Indexed: 04/03/2025]
Abstract
Tumor cell-intrinsic signaling pathways can drastically affect the tumor immune microenvironment, promoting tumor progression and resistance to immunotherapy by excluding immune cell populations from the tumor. Several tumor cell-intrinsic pathways have been reported to modulate myeloid-cell and T-cell infiltration, creating "cold" tumors. However, clinical evidence suggests that excluding cytotoxic T cells from the tumor core also mediates immune evasion. In this study, we find that tumor cell-intrinsic SOX2 signaling in non-small cell lung cancer induces the exclusion of cytotoxic T cells from the tumor core and promotes resistance to checkpoint blockade therapy. Mechanistically, tumor cell-intrinsic SOX2 expression upregulates CCL2 in tumor cells, resulting in increased recruitment of regulatory T cells (Treg). CD8+ T-cell exclusion depended on Treg-mediated suppression of tumor vasculature. Depleting tumor-infiltrating Tregs via glucocorticoid-induced TNF receptor-related protein restored CD8+ T-cell infiltration and, when combined with checkpoint blockade therapy, reduced tumor growth. These results show that tumor cell-intrinsic SOX2 expression in lung cancer serves as a mechanism of immunotherapy resistance and provide evidence to support future studies investigating whether patients with non-small cell lung cancer with SOX2-dependent CD8+ T-cell exclusion would benefit from the depletion of glucocorticoid-induced TNFR-related protein-positive Tregs.
Collapse
Affiliation(s)
- Elen Torres-Mejia
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Sally Weng
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Wellesley College, Wellesley, MA 02481, USA
| | | | - Kim B. Nguyen
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ellen Duong
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139, USA
- Genentech, South San Francisco, CA 94080, USA
| | - Leon Yim
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
16
|
Pham AVQ, Na Y, Suk G, Yang C, Kang SM, Lee J, Choi H, Kim W, Chi SW, Han S, Choi HW, Kim H, Kim C. Identification of Tie2 as a sensor for reactive oxygen species and its therapeutic implication. Redox Biol 2025; 81:103555. [PMID: 39993340 PMCID: PMC11903958 DOI: 10.1016/j.redox.2025.103555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
Psoriasis is a chronic inflammatory disease characterized by hyperproliferation of keratinocytes and abnormal blood vessels. As hyperproliferation is driven by pro-inflammatory cytokines produced by activated immune cells, therapeutic strategies often target these cytokines to manage the disease. However, the role of abnormally developed blood vessels has often been overlooked in treatment approaches. In this study, we focused on blood vessels in psoriatic lesions and investigated the potential interplay between immune and endothelial cells by adopting imiquimod treated mice as in vivo model, together with various cell biological, biochemical, and structural analyses. We found that activated immune cells can generate reactive oxygen species, subsequently inducing oxidative stress in endothelial cells. Oxidative stress impairs endothelial cell layer integrity, thereby facilitating transendothelial migration of immune cells. Mechanistically, oxidative conditions inhibit Tie2 activation, potentially by modifying its cysteine residues, leading to deactivation of its vessel-stabilizing functions. Additionally, we demonstrated that reactivating Tie2 under such conditions could restore endothelial barrier function and alleviate the disease. These results suggest that Tie2 serves as a receptor that is directly responsive to oxidative environments, thereby modulating its kinase activity. Furthermore, we suggest that Tie2 reactivation is a promising alternative therapeutic approach for psoriasis.
Collapse
Affiliation(s)
- An Vuong Quynh Pham
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Yongwoo Na
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Gyeongseo Suk
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Chansik Yang
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - So Min Kang
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Joonha Lee
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Hongseo Choi
- Department of Molecular Science & Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Wook Kim
- Department of Molecular Science & Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Sung Wook Chi
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Sangyeul Han
- Ingenia Therapeutics, 34 Coolidge Ave. 2nd Floor, Watertown, MA, 02472, United States
| | - Hae Woong Choi
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Hyeonwoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea; KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Chungho Kim
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
17
|
Maruyama M, Torii R, Matsui H, Hayashi H, Ogawara KI, Higaki K. Repeated sequential administration of pegylated emulsion of SU5416 and liposomal paclitaxel enhances anti-tumor effect in 4T1 breast cancer-bearing mice. Eur J Pharm Biopharm 2025; 209:114663. [PMID: 39922508 DOI: 10.1016/j.ejpb.2025.114663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
To improve vascular normalization strategy for intractable triple-negative breast cancer 4T1, we examined the anti-tumor effects of repeated sequential administration of polyethylene glycol (PEG)-modified emulsion of SU5416 (PE-SU5416), a vascular endothelial growth factor (VEGF) receptor-2 kinase inhibitor, and PEG-modified liposomal paclitaxel (PL-PTX) in mice bearing 4T1 cells. Three sequential administrations (Seq×3) of PE-SU5416 and PL-PTX exhibited significantly higher anti-tumor activity than a single sequential administration (Seq×1). The tumor vasculatures were structurally normalized until after two PE-SU5416 (PE-SU5416×2) or sequential (Seq×2) administrations, while the improvement in vascular function, such as oxygen supply, blood flow, and PEG-liposomal distribution, was evident until after three administrations of PE-SU5416 (PE-SU5416×3) and Seq×3. Although some discrepancies between the structural and functional improvement in tumor vasculatures were observed after PE-SU5416×3 and Seq×3, cancer-associated fibroblasts (CAFs) and collagen levels were significantly reduced after PE-SU5416×2, PE-SU5416×3, Seq×2, and Seq×3, suggesting that a possible decrease in interstitial fluid pressure due to the reduction in CAFs and collagen would have compensated for vascular function. Furthermore, PE-SU5416×2, PE-SU5416×3, Seq×2, and Seq×3 significantly decreased tumor growth factor-β (TGF-β), an activator of CAFs, in tumor tissues, suggesting that the reduction in TGF-β levels by PE-SU5416 suppresses CAF activation.
Collapse
Affiliation(s)
- Masato Maruyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530 Japan.
| | - Reiya Torii
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530 Japan
| | - Hazuki Matsui
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530 Japan
| | - Hiroki Hayashi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530 Japan
| | - Ken-Ichi Ogawara
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558 Japan
| | - Kazutaka Higaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530 Japan
| |
Collapse
|
18
|
Chitoran E, Rotaru V, Stefan DC, Gullo G, Simion L. Blocking Tumoral Angiogenesis VEGF/VEGFR Pathway: Bevacizumab-20 Years of Therapeutic Success and Controversy. Cancers (Basel) 2025; 17:1126. [PMID: 40227654 PMCID: PMC11988089 DOI: 10.3390/cancers17071126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
The "angiogenesis switch"-defined as the active process by which solid tumors develop their own circulation-plays an important role in both tumoral growth and propagation. As the malignant tumor grows and reaches a critical size, the metabolic needs as a function of an ever-increasing distance to the nearest emergent blood vessel, can no longer be covered by the microenvironment of the peritumoral tissue. Although a relatively discrete process, the "angiogenic switch" acts as a limiting stage of tumoral development present from the avascular hyperplasia phase to the vascularized neoplastic phase, providing support for tumor expansion and metastasis. Over time, research has focused on blocking the angiogenetic pathways (such as VEGF/VEGFR signaling axis) leading to the development of targeted therapeutic agents such as Bevacizumab. Objectives: We conducted a review of the molecular principles of tumoral angiogenesis and we tried to follow the history of Bevacizumab from its first approval for human usage 20 years ago to current days, focusing on the impact this agent had in solid tumor therapy. A comprehensive review of clinical trials pertaining to Bevacizumab (from the era of the preclinic trials leading to approval for human usage, to the more recent randomized trial focusing on combination targeted therapy) further details the role of this drug. We aimed to establish if this ancient drug continues to have a place in modern oncology. Conclusions: Bevacizumab, one of the first drugs targeting tumoral microenvironment, remains one of the most important oncologic agents blocking the VEGF/VEGFR angiogenic pathway. otherwise, history of 20 years marked by numerous controversies (ranging from methodological errors of clinical trials to withdrawal of approval for human usage in breast cancer patients, from discussions about severe side effects to resistance to therapy and limited efficacity), Bevacizumab continues to provide an optimal therapeutic option for many solid tumors that previously had little to no means of treatment, improving otherwise bleak outcomes. Even in the era of personalized precision oncology, Bevacizumab continues to be a key element in many therapeutic regimens both as monotherapy and in combination with newer targeted agents.
Collapse
Affiliation(s)
- Elena Chitoran
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Vlad Rotaru
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Daniela-Cristina Stefan
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Laurentiu Simion
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| |
Collapse
|
19
|
Kane MA, Birmingham KG, Yeoman B, Patel N, Sperinde H, Molley TG, Beri P, Tuler J, Kumar A, Klein S, Zare S, Wallace A, Katira P, Engler AJ. Adhesion strength of tumor cells predicts metastatic disease in vivo. Cell Rep 2025; 44:115359. [PMID: 40049163 PMCID: PMC12014391 DOI: 10.1016/j.celrep.2025.115359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/26/2024] [Accepted: 02/06/2025] [Indexed: 03/29/2025] Open
Abstract
Although only a fraction of tumor cells contribute to metastatic disease, no prognostic biomarkers currently exist to identify these cells. We show that a physical marker-adhesion strength-predicts metastatic potential in a mouse breast cancer model and that it may stratify human disease. Cells disseminating from murine mammary tumors are weakly adherent, and, when pre-sorted by adhesion, primary tumors created from strongly adherent cells exhibit fewer lung metastases than weakly adherent cells do. We demonstrate that admixed cancer lines can be separated by label-free adhesive signatures. When applied to murine metastatic tumors, adhesion retrospectively predicts metastatic disease with 100% specificity, 85% sensitivity, and area under the curve (AUC) of 0.94. Cells from human reduction mammoplasties have a higher adhesion strength versus resected human tumors, which may also be stratified between invasive and more indolent cancers. Thus, highly metastatic cells may have a distinct physical phenotype that may be a predictive marker of clinical outcomes.
Collapse
Affiliation(s)
- Madison A Kane
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | | | - Benjamin Yeoman
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Department of Mechanical Engineering, San Diego State University, San Diego, CA 92182, USA
| | - Neal Patel
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Hayley Sperinde
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Thomas G Molley
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Pranjali Beri
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Jeremy Tuler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Aditya Kumar
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Sarah Klein
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Somaye Zare
- Department of Pathology, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92093, USA
| | - Anne Wallace
- Department of Surgery, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92093, USA
| | - Parag Katira
- Department of Mechanical Engineering, San Diego State University, San Diego, CA 92182, USA; Computational Science Research Center, San Diego State University, San Diego, CA 92182, USA
| | - Adam J Engler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Department of Pathology, UC San Diego, La Jolla, CA 92093, USA; Department of Surgery, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
20
|
Avnet S, Mizushima E, Severino B, Lipreri MV, Scognamiglio A, Corvino A, Baldini N, Cortini M. Antagonizing the S1P-S1P3 Axis as a Promising Anti-Angiogenic Strategy. Metabolites 2025; 15:178. [PMID: 40137142 PMCID: PMC11944055 DOI: 10.3390/metabo15030178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Angiogenesis, the process of new blood vessel formation, is critically regulated by a balance of pro- and anti-angiogenic factors. This process plays a central role in tumor progression and is modulated by tumor cells. Sphingosine-1-phosphate (S1P), a bioactive lipid signaling molecule acting via G-protein-coupled receptors (S1PR1-5), has emerged as a key mediator of vascular development and pathological angiogenesis in cancer. Consequently, targeting the S1P-S1PRs axis represents a promising strategy for antiangiogenic therapies. This study explores S1PR3 as a potential therapeutic target in osteosarcoma, the most common primary bone malignancy, which we have previously demonstrated to secrete S1P within the acidic tumor microenvironment. METHODS The effects of KRX-725-II and its derivatives, Tic-4-KRX-725-II and [D-Tic]4-KRX-725-II-pepducins acting as S1PR3 antagonists as allosteric modulators of GPCR activity-were tested on metastatic osteosarcoma cells (143B) for proliferation and migration inhibition. Anti-angiogenic activity was assessed using endothelial cells (HUVEC) through proliferation and tubulogenesis assays in 2D, alongside sprouting and migration analyses in a 3D passively perfused microfluidic chip. RESULTS S1PR3 inhibition did not alter osteosarcoma cell growth or migration. However, it impaired endothelial cell tubulogenesis up to 75% and sprouting up to 30% in respect to controls. Conventional 2D assays revealed reduced tubule nodes and length, while 3D microfluidic models demonstrated diminished sprouting area and maximum migration distance, indicating S1PR3's role in driving endothelial cell differentiation. CONCLUSIONS These findings highlight S1PR3 as a critical regulator of angiogenesis and posit its targeting as a novel anti-angiogenic strategy, particularly for aggressive, S1P-secreting tumors with pronounced metastatic potential and an acidic microenvironment.
Collapse
Affiliation(s)
- Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Emi Mizushima
- Department of Orthopaedic Surgery, School of Medicine, Sapporo Medical University, Sapporo 060-8543, Hokkaido, Japan;
| | - Beatrice Severino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Napoli, Italy; (B.S.); (A.S.); (A.C.)
| | - Maria Veronica Lipreri
- Biomedical Science, Technologies, and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Antonia Scognamiglio
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Napoli, Italy; (B.S.); (A.S.); (A.C.)
| | - Angela Corvino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Napoli, Italy; (B.S.); (A.S.); (A.C.)
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy;
- Biomedical Science, Technologies, and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Margherita Cortini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy;
| |
Collapse
|
21
|
Mohanty SS, Warrier S, Rangarajan A. Rethinking AMPK: A Reversible Switch Fortifying Cancer Cell Stress-Resilience. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2025; 98:33-52. [PMID: 40165808 PMCID: PMC11952127 DOI: 10.59249/jkbb6336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Stress adaptation is an evolutionarily conserved mechanism that promotes survival in the face of adverse conditions. AMP-activated protein kinase (AMPK) is a highly conserved energy-sensing kinase found in nearly all eukaryotic cells. It maintains energy homeostasis by promoting catabolism and inhibiting anabolism. In the context of cancer, the role of AMPK is controversial. It was initially touted as a tumor suppressor due to its association with Liver Kinase B1 (LKB1) (an upstream regulator and a known tumor suppressor) and ensuing growth-suppressive actions. However, emerging studies across a variety of cancer types unambiguously reveal AMPK's pro-survival and, thus, tumor-promoting activity, especially under cancer-associated stresses such as hypoxia, nutrient deprivation, oxidative stress, matrix detachment, and chemotherapy. In cancer cells, AMPK is activated in response to stress-induced increases in the levels of adenosine monophosphate (AMP), Ca2+, or reactive oxygen species (ROS). Upon activation, AMPK engages in metabolic rewiring and crosstalk with signaling molecules to mobilize resources toward survival while compromising proliferation. Here, we posit that AMPK is a non-genetic "reversible switch," allowing cancer cells' phenotype to switch to dormant, stem-like, and drug-resistant states, thereby enabling tumor cell survival, pathological progression, and therapy resistance. This review underscores the critical role of AMPK in driving cancer cell stress resilience and survival, advocating for the strategic use of AMPK inhibitors to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Shraddha S. Mohanty
- Department of Developmental Biology and Genetics, Indian Institute of Science,
Bengaluru, India
| | - Shweta Warrier
- Department of Developmental Biology and Genetics, Indian Institute of Science,
Bengaluru, India
| | - Annapoorni Rangarajan
- Department of Developmental Biology and Genetics, Indian Institute of Science,
Bengaluru, India
| |
Collapse
|
22
|
Moradi Kashkooli F, Mirala F, H H Tehrani M, Alirahimi M, Souri M, Golzaryan A, Kar S, Soltani M. Mechanical Forces in Tumor Growth and Treatment: Perspectives From Biology, Physics, Engineering, and Mathematical Modeling. WIREs Mech Dis 2025; 17:e70000. [PMID: 40170456 DOI: 10.1002/wsbm.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 04/03/2025]
Abstract
The progression of tumors is influenced by mechanical forces and biological elements, such as hypoxia and angiogenesis. Mechanical factors, including stress, pressure, interstitial fluid pressure, and cellular traction forces, compromise normal tissue architecture, augmenting stiffness and thus promoting tumor growth and invasion. The selective elimination of specific tumor components can reduce growth-induced mechanical stress, thereby improving therapeutic efficacy. Furthermore, stress-relief drugs have the potential in enhancing chemotherapy outcomes. In this setting, computational modeling functions as an essential tool for quantitatively elucidating the mechanical principles underlying tumor formation. These models can precisely replicate the impact of mechanical pressures on solid tumors, offering insight into the regulation of tumor behavior by these forces. Tumor growth produces mechanical forces, including compression, displacement, and deformation, leading to irregular stress patterns, expedited tumor advancement, and reduced treatment efficacy. This review analyzes the impact of mechanical forces on carcinogenesis and solid tumor proliferation, emphasizing the significance of stress alleviation in regulating tumor growth. Furthermore, we investigate the influence of mechanical forces on tumor dissemination and emphasize the promise of integrating computational modeling with force-targeted cancer therapies to improve treatment efficacy by tackling the fundamental mechanics of tumor proliferation.
Collapse
Affiliation(s)
| | - Fatemeh Mirala
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Masoud H H Tehrani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mahvash Alirahimi
- Department of Obstetrics & Gynecology, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Aryan Golzaryan
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Egaila, Kuwait
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
23
|
Chen L, Hao Y, Zhai T, Yang F, Chen S, Lin X, Li J. Single-cell Analysis Highlights Anti-apoptotic Subpopulation Promoting Malignant Progression and Predicting Prognosis in Bladder Cancer. Cancer Inform 2025; 24:11769351251323569. [PMID: 40018511 PMCID: PMC11866393 DOI: 10.1177/11769351251323569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/06/2025] [Indexed: 03/01/2025] Open
Abstract
Backgrounds Bladder cancer (BLCA) has a high degree of intratumor heterogeneity, which significantly affects patient prognosis. We performed single-cell analysis of BLCA tumors and organoids to elucidate the underlying mechanisms. Methods Single-cell RNA sequencing (scRNA-seq) data of BLCA samples were analyzed using Seurat, harmony, and infercnv for quality control, batch correction, and identification of malignant epithelial cells. Gene set enrichment analysis (GSEA), cell trajectory analysis, cell cycle analysis, and single-cell regulatory network inference and clustering (SCENIC) analysis explored the functional heterogeneity between malignant epithelial cell subpopulations. Cellchat was used to infer intercellular communication patterns. Co-expression analysis identified co-expression modules of the anti-apoptotic subpopulation. A prognostic model was constructed using hub genes and Cox regression, and nomogram analysis was performed. The tumor immune dysfunction and exclusion (TIDE) algorithm was applied to predict immunotherapy response. Results Organoids recapitulated the cellular and mutational landscape of the parent tumor. BLCA progression was characterized by mesenchymal features, epithelial-mesenchymal transition (EMT), immune microenvironment remodeling, and metabolic reprograming. An anti-apoptotic tumor subpopulation was identified, characterized by aberrant gene expression, transcriptional instability, and a high mutational burden. Key regulators of this subpopulation included CEBPB, EGR1, ELF3, and EZH2. This subpopulation interacted with immune and stromal cells through signaling pathways such as FGF, CXCL, and VEGF to promote tumor progression. Myofibroblast cancer-associated fibroblasts (mCAFs) and inflammatory cancer-associated fibroblasts (iCAFs) differentially contributed to metastasis. Protein-protein interaction (PPI) network analysis identified functional modules related to apoptosis, proliferation, and metabolism in the anti-apoptotic subpopulation. A 5-gene risk model was developed to predict patient prognosis, which was significantly associated with immune checkpoint gene expression, suggesting potential implications for immunotherapy. Conclusions We identified a distinct anti-apoptotic tumor subpopulation as a key driver of tumor progression with prognostic significance, laying the foundation for the development of new therapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Linhuan Chen
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Yangyang Hao
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Tianzhang Zhai
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Fan Yang
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Shuqiu Chen
- Department of Urology, Southeast University Zhongda Hospital, Nanjing, China
| | - Xue Lin
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Jian Li
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| |
Collapse
|
24
|
Yang S, Bai Z, Zhang F, Cui W, Bu P, Bai W, Xi Y. Expression and prognostic significance of CD93 in blood vessels in colorectal cancer: an immunohistochemical analysis of 134 cases. BMC Gastroenterol 2025; 25:84. [PMID: 39962383 PMCID: PMC11834615 DOI: 10.1186/s12876-025-03643-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE Tumor blood vessels are tortuous and dilated, contributing to the aberrant tumor microenvironment. CD93 is a newly reported transmembrane receptor, mainly expressed in tumor endothelial cells, that has demonstrated prognostic value in some cancer types. However, the role of CD93 in the vasculature of colorectal cancer (CRC) tissues and its prognostic significance remain unknown. It is therefore necessary to explore the effect of CD93 in patients with CRC. METHOD We detected the expression of CD93 in human CRC tissues using immunohistochemistry. We then examined the correlation between CD93 expression and clinicopathological factors in cancer tissues from 134 patients with CRC. RESULT CD93 expression levels were higher in CRC vessels than in vessels in adjacent normal tissues. Upregulation of CD93 was associated with tumor site and microsatellite instability. CD93 protein expression was positively related to macrophage infiltration in CRC. High expression of CD93 may indicate normalization of the tumor vasculature and was associated with better overall survival. CONCLUSION CD93 was highly expressed in CRC vessels and correlated with infiltration of immune cells. Our findings reveal that vascular normalization and patient prognosis can be predicted by detecting CD93 expression in CRC tumor tissues.
Collapse
Affiliation(s)
- Shuzhe Yang
- Second Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Zhongyuan Bai
- First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Fei Zhang
- Department of Pathology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hosipital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, No. 3, ZhiGongXinCun Street, Taiyuan, China
| | - Wei Cui
- Department of Pathology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hosipital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, No. 3, ZhiGongXinCun Street, Taiyuan, China
| | - Peng Bu
- Department of Pathology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hosipital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, No. 3, ZhiGongXinCun Street, Taiyuan, China
| | - Wenqi Bai
- Department of General Surgery Sciences, Cancer Hospital, Shanxi Province Cancer Hospital/ShanxiHospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to ShanxiMedical University, No. 3, ZhiGongXinCun Street, Taiyuan, China.
| | - Yanfeng Xi
- Department of Pathology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hosipital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, No. 3, ZhiGongXinCun Street, Taiyuan, China.
| |
Collapse
|
25
|
Zhao H, Jin S, Liu Y, Wang Q, Tan BSN, Wang S, Han WK, Niu X, Zhao Y. A Second Near-Infrared Window-Responsive Metal-Organic-Framework-Based Photosensitizer for Tumor Immunotherapy via Synergistic Ferroptosis and STING Activation. J Am Chem Soc 2025; 147:4871-4885. [PMID: 39854684 DOI: 10.1021/jacs.4c13241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Photodynamic therapy (PDT) holds promise as a cancer treatment modality due to its potential for enhanced therapy precision and safety. To enhance deep tissue penetration and minimize tissue adsorption and phototoxicity, developing photosensitizers activated by second near-infrared window (NIR-II) light shows significant potential. However, the efficacy of PDT is often impeded by tumor microenvironment hypoxia, primarily caused by irregular tumor vasculature. Fortunately, the stimulator of interferon genes (STING) pathway, known for immune activation, has been linked to vasculature normalization. In this study, we developed a nanoplatform (Fe-THBQ/SR) by loading a STING agonist (SR-717) into an iron-tetrahydroxy-1,4-benzoquinone (Fe-THBQ) metal-organic framework. Fe-THBQ was proven to be an effective NIR-II photosensitizer, generating numerous reactive oxygen species (ROS) under 1064 nm laser irradiation. These ROS downregulated heat shock protein expression, consequently promoting mild-photothermal therapy (mild-PTT), and facilitated ferroptosis by depleting glutathione (GSH)/glutathione peroxidase 4. Moreover, Fe-THBQ/SR released SR-717 upon GSH stimulation, synergizing with the ROS-mediated double-stranded DNA leakage to enhance STING activation. This process contributed to tumor vasculature normalization and hypoxia alleviation, thereby enhancing the PDT efficacy. Overall, we presented a versatile single-laser-triggered nanoplatform (Fe-THBQ/SR) for NIR-II PDT and NIR-II mild-PTT and simultaneously coupled it with the effective activation of STING to form a reinforcing cycle. These synergistic enhancements increased the immunogenicity of tumor cells, remodeled the immunosuppressive tumor microenvironment, increased T lymphocyte infiltration, and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Huan Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Shujuan Jin
- Senior Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Fuxin Road, No. 28, Haidian District, Beijing, 100853, China
| | - Yang Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Qian Wang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, China
| | - Brynne Shu Ni Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Shihuai Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
- Yunnan Key Laboratory for Micro/Nano Materials & Technology, School of Materials and Energy, Yunnan University, Kunming, Yunnan 650091, China
| | - Wang-Kang Han
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Xuping Niu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| |
Collapse
|
26
|
Ha H, Choi Y, Kim NH, Kim J, Jang J, Niepa THR, Tanaka M, Lee HY, Choi J. Lipid Nanoparticle Delivery System for Normalization of Tumor Microenvironment and Tumor Vascular Structure. Biomater Res 2025; 29:0144. [PMID: 39935791 PMCID: PMC11811622 DOI: 10.34133/bmr.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/07/2025] [Accepted: 01/18/2025] [Indexed: 02/13/2025] Open
Abstract
Tumors grow by receiving oxygen and nutrients from the surrounding blood vessels, leading to rapid angiogenesis. This results in functionally and structurally abnormal vasculature characterized by high permeability and irregular blood flow, causing hypoxia within the tumor microenvironment (TME). Hypoxia exacerbates the secretion of pro-angiogenic factors such as vascular endothelial growth factor (VEGF), further perpetuating abnormal vessel formation. This environment compromises the efficacy of radiotherapy, immunotherapy, and chemotherapy. In this study, we developed a pH-sensitive liposome (PSL) system, termed OD_PSL@AKB, to co-deliver oxygen (OD) and razuprotafib (AKB-9778) to tumors. This system rapidly responds to the acidic TME to alleviate hypoxia and inhibit VEGF secretion, restoring VE-cadherin expression in hypoxic endothelial cell/cancer cell cocultures. Our findings highlight the potential of OD_PSL@AKB in normalizing tumor vasculature and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Heejin Ha
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Department of Chemical Science and Engineering,
Institute of Science Tokyo, Kanagawa 226-8503, Japan
| | - Na-Hyeon Kim
- Department of Chemical Engineering,
Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Jiwon Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jaehee Jang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Tagbo H. R. Niepa
- Department of Chemical Engineering,
Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biomedical Engineering,
Carnegie Mellon University, Pittsburgh, PA, USA
| | - Masayoshi Tanaka
- Department of Chemical Science and Engineering,
Institute of Science Tokyo, Kanagawa 226-8503, Japan
| | - Hee-Young Lee
- Department of Chemical Engineering,
Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| |
Collapse
|
27
|
Wang F, Yu J, Lu X, Numata K, Ruan L, Zhang D, Liu X, Li X, Wan M, Zhang W, Zhang G. Relationship between contrast-enhanced ultrasound combined with ultrasound resolution microscopy imaging and histological features of hepatocellular carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04825-y. [PMID: 39928101 DOI: 10.1007/s00261-025-04825-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVES Using contrast-enhanced ultrasound (CEUS) and ultrasound resolution microscopy (URM) imaging, this study aimed to evaluate the relationship between microvascular parameters of small hepatocellular carcinoma (sHCC) (≤ 3 cm) and microscopic histological features, which include vessels encapsulating tumour clusters (VETC), microvascular invasion (MVI), and histological grade. METHODS Sixteen patients with solitary resected sHCC were prospectively enrolled. CEUS and URM were performed one week before resection. All "ratio" refers to comparisons between the active area (where CEUS microbubble show visible motion track by URM) and the entire lesion. Blood vessel complexity (ratio), blood vessel density (ratio), area (ratio), flow velocity, blood vessel diameter, and perfusion index ("flow velocity" × "vessel ratio") were analysed using URM. The relationships between URM parameters and microscopic histological features (MVI, VETC, and histological grade) were analysed. RESULTS There were 5 (31.3%), 8 (50%), and 7 (43.7%) cases of poorly differentiated, MVI-positive, and VETC-positive HCC, respectively. The mean velocity of the entire lesion was higher in the poorly differentiated group than that in the moderately differentiated group (p = 0.026). The complexity ratio (MVI-positive: 1.07 ± 0.03, MVI-negative: 1.03 ± 0.02, p = 0.012), area ratio (MVI-positive: 0.63 ± 0.18, MVI-negative: 0.39 ± 0.16, p = 0.017), and perfusion index (MVI-positive: 8.67 ± 1.88, MVI-negative: 6.42 ± 0.94, p = 0.009) were greater in MVI-positive HCCs. The density ratio (VETC-positive: 1.30 ± 0.19, VETC-negative: 1.10 ± 0.05, p = 0.006) was larger in VETC-positive HCCs. CONCLUSION Higher blood flow velocity and area of HCC lesions, and higher blood vessel complexity and density may be related to microscopic histological features. This relationship might provide a strategy of using URM for preoperative non-invasive diagnostic VETC, MVI, and histological grade in the future.
Collapse
Affiliation(s)
- Feiqian Wang
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Jingtong Yu
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Baoji Hospital of Traditional Chinese Medicine, Baoji, China
| | - Xingqi Lu
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Baoji Hospital of Traditional Chinese Medicine, Baoji, China
| | - Kazushi Numata
- Yokohama City University Medical Center, Yokohama, Japan
| | - Litao Ruan
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dong Zhang
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Liu
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaojing Li
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | | | - Wenbin Zhang
- VINNO Technology Company Limited, Jiangsu, China
| | - Guanjun Zhang
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
28
|
Wu C, Shi L, Deng Y, Chen H, Lu Y, Xiong X, Yin X. Bufalin Regulates STAT3 Signaling Pathway to Inhibit Corneal Neovascularization and Fibrosis After Alkali Burn in Rats. Curr Eye Res 2025; 50:139-147. [PMID: 39356002 DOI: 10.1080/02713683.2024.2408392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/17/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE Bufalin (BU) is a bioactive ingredient extracted from the skin and parotid venom glands of Bufo raddei, which can effectively inhibit angiogenesis. The aim of this study was to investigate whether BU could affect corneal neovascularization (CoNV). METHODS A rat CoNV model (right eye) was constructed by administration of NaOH, and the left eye served as a control. Corneal damage scores of rats were detected. Hematoxylin & eosin, TUNEL, and Masson staining examined pathological changes, apoptosis, and fibrosis of corneal tissues. Immunohistochemistry and western blotting assessed the expression of proteins. RESULTS BU intervention resulted in a significant reduction in corneal inflammatory cells, repair of corneal epithelial hyperplasia, significant reduction in stromal edema, and reduction in vascular proliferation. BU can inhibit corneal neovascularization. CONCLUSION This study demonstrated that BU inhibits CoNV, fibrosis, and inflammation by modulating the STAT3 signaling pathway, elucidating the intrinsic mechanism of its protective effect. BU has great potential in the treatment of CoNV caused by corneal alkali burns.
Collapse
Affiliation(s)
- Chao Wu
- Department of Ophthalmology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Lu Shi
- Department of Ophthalmology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Yan Deng
- Department of Ophthalmology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Hongping Chen
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang, China
| | - Ying Lu
- Department of Ophthalmology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Xiaoyan Xiong
- Department of Ophthalmology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Xiaolong Yin
- Department of Ophthalmology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| |
Collapse
|
29
|
Magdalena JB, Justyna C, Joanna C, Ryszard S, Alina D, Dorota SL, Ewelina P, Sybilla M, Tomasz C. Normalization of tumor vasculature by imiquimod: proposal for a new anticancer therapeutic indication for a TLR7 agonist. Cancer Immunol Immunother 2025; 74:90. [PMID: 39891776 PMCID: PMC11787066 DOI: 10.1007/s00262-025-03943-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
Imiquimod (IMQ), a drug from aminoquinoline group, is the toll-like receptor 7 (TLR7) agonist. It acts as an immunostimulant and radio-sensitizing agent. IMQ stimulates both innate and adaptive immune response. Despite studies conducted, there are no unambiguous data showing how IMQ affects the condition of tumor blood vessels. Tumor vasculature plays the main role in tumor progression. Formation of abnormal blood vessels increases area of hypoxia which recruits suppressor cells, blocks tumor infiltration by CD8+ T lymphocytes, inhibits efficacy of chemoterapeutic drug and leads to cancer relapse. Normalization is a type of therapy targeted at abnormal tumor blood vessels. Here, we demonstrated that 50 µg of IMQ inhibits the growth of melanoma tumors more efficiently, compared to other tested doses and the control group. Dose escalation did not improve the therapeutic antitumor potential of TLR7 agonist. A dose of 50 µg of IMQ most effectively reduced tumor blood vessel density. Imiquimod normalized tumor vasculature both structurally (by reducing vessel tortuosity and increasing pericyte coverage) and functionally (by improving tumor perfusion) in a dose-dependent manner. Hypoxia regions in tumors of treated mice were significantly reduced after IMQ administration. A dose of 50 µg of IMQ had also the greatest impact on the changes in tumor-infiltrating T lymphocytes levels. TLR7 agonist inhibited angiogenesis in treated mice. Functional vascular normalization by IMQ increases the effectiveness of low dose of doxorubicin. Higher dose of IMQ showed worse effects than lower doses including decreased tumor perfusion, increased tumor hypoxia and immunosuppression. This knowledge may help to optimize the combination of the selected IMQ dose with e.g. chemotherapy or radiotherapy to elicit synergistic effect in cancer treatment. To conclude, we outline IMQ repurposing as a vascular normalizing agent. Our research results may contribute to expanding the therapeutic indications for the use of IMQ in anticancer therapy in the future.
Collapse
Affiliation(s)
- Jarosz-Biej Magdalena
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland.
| | - Czapla Justyna
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Ciepła Joanna
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Smolarczyk Ryszard
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Drzyzga Alina
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Sprus-Lipka Dorota
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Pilny Ewelina
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Matuszczak Sybilla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Cichoń Tomasz
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| |
Collapse
|
30
|
Shahedi F, Naseri S, Momennezhad M, Zare H. MR Imaging Techniques for Microenvironment Mapping of the Glioma Tumors: A Systematic Review. Acad Radiol 2025:S1076-6332(25)00066-2. [PMID: 39894708 DOI: 10.1016/j.acra.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/18/2025] [Accepted: 01/19/2025] [Indexed: 02/04/2025]
Abstract
RATIONALE AND OBJECTIVES The tumor microenvironment (TME) is a critical regulator of cancer progression, metastasis, and treatment response. Currently, various imaging approaches exist to assess the pathophysiological features of the TME. This systematic review provides an overview of magnetic resonance imaging (MRI) methods used in clinical practice to characterize the pathophysiological features of the gliomas TME. METHODS This review involved a systematic comprehensive search of original open-access articles reporting the clinical use of MR imaging in glioma patients of all ages in the PubMed, Scopus, and Web of Science databases between January 2010 and December 2023. We restricted our research to papers published in the English language. RESULTS A total of 1137 studies were preliminarily identified through electronic database searches. After duplicate studies were removed, 44 studies met the eligibility criteria. The glioma TME was accompanied by alterations in metabolism, pH, vascularity, oxygenation, and extracellular matrix components, including tumor-associated macrophages, and sodium concentration. CONCLUSION Multiparametric MRI is capable of noninvasively assessing the pathophysiological features and tumor-supportive niches of the TME, which is in line with its application in personalized medicine.
Collapse
Affiliation(s)
- Fateme Shahedi
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (F.S., S.N., M.M., H.Z.)
| | - Shahrokh Naseri
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (F.S., S.N., M.M., H.Z.)
| | - Mahdi Momennezhad
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (F.S., S.N., M.M., H.Z.)
| | - Hoda Zare
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (F.S., S.N., M.M., H.Z.); Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran (H.Z.).
| |
Collapse
|
31
|
Yang Z, Xiao H, Li X, Wei Z, Kang M, Wang R, Huang J. Antiangiogenic therapy with recombinant human endostatin may improve blood perfusion of cervical node with necrosis in nasopharyngeal carcinoma patients: a preliminary study by using contrast-enhanced ultrasound. Front Oncol 2025; 15:1521762. [PMID: 39959670 PMCID: PMC11825469 DOI: 10.3389/fonc.2025.1521762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/13/2025] [Indexed: 02/18/2025] Open
Abstract
Background The cervical node with necrosis (CNN) is an important poor prognostic factor for nasopharyngeal carcinoma (NPC) patients. The tumor microenvironment of the CNN has severely insufficient blood perfusion, thus leading to hypoxia and reducing the effect of radiotherapy (RT) and chemotherapy. By using contrast-enhanced ultrasound (CEUS) as a monitoring method, we conducted this study to assess whether antiangiogenic therapy (AT) with recombinant human endostatin (RHES) may improve blood perfusion of the CNN. Materials and methods Fifteen NPC patients with CNN were enrolled and underwent CEUS the day before and day 5 after AT with RHES initiation, respectively. By analyzing the variations of CEUS parameters of CNN, such as peak intensity (PI), time to peak (TTP), and mean transit time (MTT) at different time points, we evaluate the impact of AT with RHES on blood perfusion of CNN. Results The PI of day 5 after AT was significantly enhanced compared to the PI of the day before AT [-44.94 ± 4.72 (dB) vs. -50.33 ± 6.85 (dB), p < 0.001]. The TTP of day 5 after AT became dramatically shorter than the TTP of the day before AT [19.48 ± 3.63 (s) vs. 24.19 ± 6.93 (s), p = 0.031]. The MTT of day 5 after AT became obviously shorter than the MTT of the day before AT [28.08 ± 3.03 (s) vs. 33.76 ± 6.20 (s), p = 0.001]. Conclusion These results revealed that the blood volume and the blood flow velocity in the microvessels of the CNN increased after AT, indicating that AT with RHES may improve blood perfusion in the CNN of NPC, thus providing valuable insights for the clinical application of AT combined with RT and/or chemotherapy in NPC patients with CNN. Moreover, CEUS as a noninvasive and real-time monitoring method may be suitable for clinically evaluating tumor blood perfusion changes.
Collapse
Affiliation(s)
- Zhendong Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huimin Xiao
- Department of Oncology, Rui-Kang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Xigui Li
- Department of Ultrasonography, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhuxin Wei
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Min Kang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rensheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jianyuan Huang
- Department of Ultrasonography, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
32
|
Sweeney A, Langley A, Xavierselvan M, Shethia RT, Solomon P, Arora A, Mallidi S. Vascular regional analysis unveils differential responses to anti-angiogenic therapy in pancreatic xenografts through macroscopic photoacoustic imaging. Theranostics 2025; 15:2649-2671. [PMID: 39990229 PMCID: PMC11840746 DOI: 10.7150/thno.99361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/17/2024] [Indexed: 02/25/2025] Open
Abstract
Background: Amongst the various imaging techniques that provide surrogate tumor radiographic indications to aid in planning, monitoring, and predicting outcomes of therapy, ultrasound-guided photoacoustic imaging (US-PAI) is a promising non-ionizing modality based on endogenous blood (hemoglobin) and blood oxygen saturation (StO₂) contrast. Adaptation of US-PAI to the clinical realm requires macroscopic system configurations for adequate depth visualization. Methods: Here we present a vascular regional analysis (VRA) methodology of obtaining areas of low and high vessel density regions within the tumor (LVD and HVD respectively) by frequency domain filtering of macroscopic PA images. In this work, we evaluated the various vascular and oxygenation profiles of different murine xenografts of pancreatic cancer (AsPC-1, MIA PaCa-2, and BxPC-3) that have varying levels of angiogenic potentials and investigated the effects of receptor tyrosine kinase inhibitor (sunitinib) on the tumor microvessel density and StO₂. Results: The administration of sunitinib resulted in transient deoxygenation and reduction in vessel density within 72 h in two (AsPC-1 and MIA PaCa-2) of the three tumor types. Utilizing VRA, the regional change in StO2 (∆StO2) revealed the preferential targeting of sunitinib in LVD regions in only the AsPC-1 tumors. We also identified the presence of vascular normalization (validated through immunohistochemistry) in the sunitinib treated AsPC-1 tumors at day 8 post-treatment where a significant increases in HVD ∆StO2 (~20%) were seen following the 72-hour time point, indicative of improved vessel flow and functionality. Treated AsPC-1 vasculature displayed increased maturity and functionality compared to non-treated tumors on day 8, while these same metrics showed no conclusive evidence of vascular normalization in MIA PaCa-2 or BxPC-3 tumors. Conclusion: Overall, VRA as a tool to monitor treatment response allowed us to identify time points of vascular remodeling, highlighting its ability to provide insights into the tumor microenvironment for sunitinib treatment and other anti-angiogenic therapies.
Collapse
Affiliation(s)
- Allison Sweeney
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Andrew Langley
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Marvin Xavierselvan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Ronak T. Shethia
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Patrick Solomon
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Aayush Arora
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Srivalleesha Mallidi
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
33
|
Chen Y, Jin H, Wen W, Xu Y, Zhang X, Yang J, Wang Y. Targeting RhoA expression with formononetin and salvianolic acid B to mitigate pancreatic cancer-associated endothelial cells changes. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118711. [PMID: 39181286 DOI: 10.1016/j.jep.2024.118711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/08/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE According to the theory of Qi and blood in Traditional Chinese Medicine (TCM), the combination of Qi-reinforcing herbs and blood-activating herbs has a synergistic effect in improving blood stasis syndrome, especially in tumor treatment. The classic "Radix Astragali - Salvia miltiorrhiza" duo exemplifies this principle, renowned for invigorating Qi and activating blood flow, employed widely in tumor therapies. Our prior research underscores the potent inhibition of pancreatic tumor xenografts by the combination of Formononetin (from Radix Astragali) and Salvianolic acid B (from Salvia miltiorrhiza) in vitro. However, it remains unclear whether this combination can inhibit the abnormal vascularization of pancreatic tumors to achieve its anti-cancer effect. AIM OF THE STUDY Abnormal vasculature, known to facilitate tumor growth and metastasis. Strategies to normalize tumor-associated blood vessels provide a promising avenue for anti-tumor therapy. This study aimed to unravel the therapeutic potential of Formononetin combined with Salvianolic acid B (FcS) in modulating pancreatic cancer's impact on endothelial cells, illuminate the underlying mechanisms that govern this therapeutic interaction, thereby advancing strategies to normalize tumor vasculature and combat cancer progression. MATERIALS AND METHODS A co-culture system involving Human Umbilical Vein Endothelial Cells (HUVECs) and PANC-1 cells was established to investigate the potential of targeting abnormal vasculature as a novel anti-tumor therapeutic strategy. We systematically compared HUVEC proliferation, migration, invasion, and lumenogenesis in both mono- and co-culture conditions with PANC-1 (H-P). Subsequently, FcS treatment of the H-P system was evaluated for its anti-angiogenic properties. Molecular docking was utilized to predict the interactions between Formononetin and Salvianolic acid B with RhoA, and the post-treatment expression of RhoA in HUVECs was assessed. Furthermore, we utilized shRhoA lentivirus to elucidate the role of RhoA in FcS-mediated effects on HUVECs. In vivo, a zebrafish xenograft tumor model was employed to assess FcS's anti-tumor potential, focusing on cancer cell proliferation, migration, apoptosis, and vascular development. RESULTS FcS treatment demonstrated a significant, dose-dependent inhibition of PANC-1-induced alterations in HUVECs, including proliferation, migration, invasion, and tube formation capabilities. Molecular docking analyses indicated potential interactions between FcS and RhoA. Further, FcS treatment was found to downregulate RhoA expression and modulated the PI3K/AKT signaling pathway in PANC-1-induced HUVECs. Notably, the phenotypic inhibitory effects of FcS on HUVECs were attenuated by RhoA knockdown. In vivo zebrafish studies validated FcS's anti-tumor activity, inhibiting cancer cell proliferation, metastasis, and vascular sprouting, while promoting tumor cell apoptosis. CONCLUSIONS This study underscores the promising potential of FcS in countering pancreatic cancer-induced endothelial alterations. FcS exhibits pronounced anti-abnormal vasculature effects, potentially achieved through downregulation of RhoA and inhibition of the PI3K/Akt signaling pathway, thereby presenting a novel therapeutic avenue for pancreatic cancer management.
Collapse
Affiliation(s)
- Yan Chen
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China
| | - Hangbin Jin
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Weiwei Wen
- Department of Dermatology, Third People's Hospital of Hangzhou, Hangzhou, China
| | - Ying Xu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Yu Wang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China.
| |
Collapse
|
34
|
Li G, Gao J, Ding P, Gao Y. The role of endothelial cell-pericyte interactions in vascularization and diseases. J Adv Res 2025; 67:269-288. [PMID: 38246244 PMCID: PMC11725166 DOI: 10.1016/j.jare.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Endothelial cells (ECs) and pericytes (PCs) are crucial components of the vascular system, with ECs lining the inner layer of blood vessels and PCs surrounding capillaries to regulate blood flow and angiogenesis. Intercellular communication between ECs and PCs is vital for the formation, stability, and function of blood vessels. Various signaling pathways, such as the vascular endothelial growth factor/vascular endothelial growth factor receptor pathway and the platelet-derived growth factor-B/platelet-derived growth factor receptor-β pathway, play roles in communication between ECs and PCs. Dysfunctional communication between these cells is associated with various diseases, including vascular diseases, central nervous system disorders, and certain types of cancers. AIM OF REVIEW This review aimed to explore the diverse roles of ECs and PCs in the formation and reshaping of blood vessels. This review focused on the essential signaling pathways that facilitate communication between these cells and investigated how disruptions in these pathways may contribute to disease. Additionally, the review explored potential therapeutic targets, future research directions, and innovative approaches, such as investigating the impact of EC-PCs in novel systemic diseases, addressing resistance to antiangiogenic drugs, and developing novel antiangiogenic medications to enhance therapeutic efficacy. KEY SCIENTIFIC CONCEPTS OF REVIEW Disordered EC-PC intercellular signaling plays a role in abnormal blood vessel formation, thus contributing to the progression of various diseases and the development of resistance to antiangiogenic drugs. Therefore, studies on EC-PC intercellular interactions have high clinical relevance.
Collapse
Affiliation(s)
- Gan Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
35
|
Heriz MH, Razzak Mahmood AA, Tahtamouni LH, Al-Sakhen MF, Kanaan SI, Saleh KM, Yasin SR. New Carbothioamide and Carboxamide Derivatives of 3-Phenoxybenzoic Acid as Potent VEGFR-2 Inhibitors: Synthesis, Molecular Docking, and Cytotoxicity Assessment. Curr Cancer Drug Targets 2025; 25:412-430. [PMID: 38747227 DOI: 10.2174/0115680096307334240429050730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/16/2024] [Accepted: 03/31/2024] [Indexed: 04/12/2025]
Abstract
INTRODUCTION/BACKGROUND Because of the well-established link between angiogenesis and tumor development, the use of antiangiogenic therapeutics, such as those targeting VEGFR-2, presents a promising approach to cancer treatment. In the current study, a set of five hydrazine-1- carbothioamide (compounds 3a-e) and three hydrazine-1-carboxamide derivatives (compounds 4a-c) were successfully synthesized from 3-phenoxybenzoic acid. These compounds were specially created as antiproliferative agents with the goal of targeting cancer cells by inhibiting VEGFR- 2 tyrosine kinase. MATERIALS AND METHODS The new derivatives were synthesized by conventional organic methods, and their structure was versified by IR, 1HNMR, 13CNMR, and mass spectroscopy. In silico investigation was carried out to identify the compounds' target, molecular similarity, ADMET, and toxicity profile. The cytotoxic activity of the prepared compounds was evaluated in vitro against three human cancer cell lines (DLD1 colorectal adenocarcinoma, HeLa cervical cancer, and HepG2 hepatocellular carcinoma). The effects of the leading compound on cell cycle progression and apoptosis induction were investigated by flow cytometry, and the specific apoptotic pathway triggered by the treatment was evaluated by RT-PCR and immunoblotting. Finally, the inhibitory activities of the new compounds against VEGFR-2 was measured. RESULTS The designed derivatives exhibited comparable binding positions and interactions to the VEGFR-2 binding site to that of sorafenib (a standard VEGFR-2 tyrosine kinase inhibitor), as determined by molecular docking analysis. Compound 4b was the most cytotoxic compound, achieving the lowest IC50 against HeLa cells. Compound 4b, a strong representative of the synthesized series, induced cell cycle arrest at the G2/M phase, increased the proportion of necrotic and apoptotic HeLa cells, and activated caspase 3. The EC50 value of compound 4b against VEGFR-2 kinase activity was comparable to sorafenib's. CONCLUSION Overall, the findings suggest that compound 4b has a promising future as a starting point for the development of new anticancer drugs.
Collapse
Affiliation(s)
- Mohammad Hamza Heriz
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Al-Zahraa University for Women, Karbala-Baghdad Street, Karbala, Iraq
| | - Ammar A Razzak Mahmood
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad, Bab-Almoudam, 10001, Baghdad, Iraq
| | - Lubna H Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Mai F Al-Sakhen
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Sana I Kanaan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Khaled M Saleh
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Salem R Yasin
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
36
|
Liu Y, Yuan L, Lin Z, Huixian M, Huangyang M, Cheng W. The serum LDH level and KELIM scores are potential predictors of a benefit from bevacizumab first-line therapy for patients with advanced ovarian cancer. Clin Transl Oncol 2025; 27:340-350. [PMID: 38904923 DOI: 10.1007/s12094-024-03569-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVE The survival benefit of first-line treatment with bevacizumab in advanced ovarian cancer patients are multifaceted. In our study, we aimed to identify potential markers of bevacizumab efficacy to help predict which patients would experience survival benefits. METHODS This was a retrospective analysis of 114 patients examined from January 1, 2015, to March 1, 2023, and data on clinical, biological, and imaging variables, such as ascites, serum LDH, and CA125, were extracted from electronic medical records. We performed a correlation analysis and principal component analysis to investigate correlations among variables and reduce their dimensionality. Then, univariate and multivariate Cox proportional hazards regression analyses were used to identify the predictors of progression-free survival. RESULTS Favorable KELIM score (≥ 1, HR 0.376, 95% CI [0.202-0.700], p = 0.002), which indicated better chemosensitivity, and lower LDH levels (≤ 210 U/L, HR 38.73, 95% CI [6.108-245.6], p < 0.001) were found to be independent predictors of a treatment benefit with bevacizumab in patients with advanced ovarian cancer. Regardless of LDH level, patients with favorable KELIM scores had a higher progression-free survival (PFS) benefit (p = 0.18). Among patients with unfavorable KELIM scores, those with higher LDH levels had the lowest PFS benefit (median: 11.5 months, p = 0.0059). CONCLUSION Patients with poor chemosensitivity and low LDH levels are more likely to benefit from first-line bevacizumab treatment. The combination of the two markers can be a helpful predictor of patients who are most likely to benefit from treatment and a guide for treatment decisions-making. Retrospectively registered: 2020-MD-371, 2020.10.12.
Collapse
Affiliation(s)
- Yi Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Lin Yuan
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Zhang Lin
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Miao Huixian
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Meng Huangyang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
37
|
Gonçalves M, Warwas KM, Meyer M, Schwartz-Albiez R, Bulbuc N, Zörnig I, Jäger D, Momburg F. Reversal of Endothelial Cell Anergy by T Cell-Engaging Bispecific Antibodies. Cancers (Basel) 2024; 16:4251. [PMID: 39766150 PMCID: PMC11674949 DOI: 10.3390/cancers16244251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Objectives: Reduced expression of adhesion molecules in tumor vasculature can limit infiltration of effector T cells. To improve T cell adhesion to tumor endothelial cell (EC) antigens and enhance transendothelial migration, we developed bispecific, T-cell engaging antibodies (bsAb) that activate T cells after cross-linking with EC cell surface antigens. Methods: Recombinant T-cell stimulatory anti-VEGFR2-anti-CD3 and costimulatory anti-TIE2-anti-CD28 or anti-PD-L1-anti-CD28 bsAb were engineered and expressed. Primary lines of human umbilical vein endothelial cells (HUVEC) that constitutively express VEGFR2 and TIE2 growth factor receptors and PD-L1, but very low levels of adhesion molecules, served as models for anergic tumor EC. Results: In cocultures with HUVEC, anti-VEGFR2-anti-CD3 bsAb increased T cell binding and elicited rapid T cell activation. The release of proinflammatory cytokines TNF-α, IFN-γ, and IL-6 was greatly augmented by the addition of anti-TIE2-anti-CD28 or anti-PD-L1-anti-CD28 costimulatory bsAb. Concomitantly, T cell-released cytokines upregulated E-selectin, ICAM1, and VCAM1 adhesion molecules on HUVEC. HUVEC cultured in breast cancer cell-conditioned medium to mimic the influence of tumor-secreted factors were similarly activated by T cell-engaging bsAb. Migration of T cells in transwell assays was significantly increased by anti-VEGFR2-anti-CD3 bsAb. The combination with costimulatory anti-TIE2-anti-CD28 bsAb augmented activation and proliferation of migrated T cells and their cytotoxic capacity against spheroids of the MCF-7 breast cancer cell line seeded in the lower transwell chamber. Conclusions: T cells activated by anti-VEGFR2-anti-CD3 and costimulatory EC-targeting bsAb can reverse the energy of quiescent EC in vitro, resulting in improved T cell migration through an EC layer.
Collapse
Affiliation(s)
- Márcia Gonçalves
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.G.)
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Karsten M. Warwas
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.G.)
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marten Meyer
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.G.)
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Reinhard Schwartz-Albiez
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Nadja Bulbuc
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Dirk Jäger
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.G.)
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Frank Momburg
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.G.)
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
38
|
Xia W, Goff M, Schiavone C, Singh N, Huang J, Need E, Cave J, Gillespie DL, Jensen RL, Pagel MD, Dogra P, Shi S, Goel S. Image-Guided Targeting of Mitochondrial Metabolism Sensitizes Pediatric Malignant Rhabdoid Tumors to Low Dose Radiotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607364. [PMID: 39211061 PMCID: PMC11361026 DOI: 10.1101/2024.08.09.607364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Tumor hypoxia leads to radioresistance and markedly worse clinical outcomes for pediatric malignant rhabdoid tumors (MRT). Our transcriptomics and bioenergetic profiling data reveal that mitochondrial oxidative phosphorylation (OXPHOS) is a metabolic vulnerability of MRT and can be exploited to overcome consumptive hypoxia by repurposing an FDA-approved anti-malarial drug, Atovaquone (AVO). We then establish the utility of Oxygen-Enhanced-Multispectral Optoacoustic Tomography (OE-MSOT), a label-free, ionizing radiation-free imaging modality, to visualize and quantify spatiotemporal changes in tumor hypoxia in response to AVO. We show a potent but transient increase in tumor oxygenation upon AVO treatment which results in complete elimination of tumors in all tested mice when combined with 10 Gy radiotherapy, a dose several times lower than the current clinic standard. Finally, we use translational mathematical modeling for systematic evaluation of dosing regimens, administration timing, and therapeutic synergy in a virtual clinical patient population. Together, our work establishes a framework for safe and pediatric patient-friendly image-guided metabolic radiosensitization of rhabdoid tumors.
Collapse
|
39
|
Zhu S, Jin G, He X, Li Y, Xu F, Guo H. Mechano-assisted strategies to improve cancer chemotherapy. Life Sci 2024; 359:123178. [PMID: 39471901 DOI: 10.1016/j.lfs.2024.123178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
Chemotherapy remains a cornerstone in cancer treatment; however, its effectiveness is frequently undermined by the development of drug resistance. Recent studies underscores the pivotal role of the tumor mechanical microenvironment (TMME) and the emerging field of mechanical nanomedicine in tackling chemo-resistance. This review offers an in-depth analysis of mechano-assisted strategies aimed at mitigating chemo-resistance through the modification of the TMME and the refinement of mechanical nanomedicine delivery systems. We explore the potential of targeting abnormal tumor mechanical properties as a promising avenue for enhancing the efficacy of cancer chemotherapy, which offers novel directions for advancing future cancer therapies, especially from the mechanomedicine perspective.
Collapse
Affiliation(s)
- Shanshan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Xiaocong He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuan Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, PR China.
| |
Collapse
|
40
|
Qin H, Yan H, Zhang X, Huang Z, Chen Y, Zhang Y, Xiang S, Zhang Y, Yang N, Zeng L. Octreotide plus IBI-318 plus anlotinib in the treatment of multiple neuroendocrine metastases of unknown primary lesions: a case report. Front Oncol 2024; 14:1390299. [PMID: 39723389 PMCID: PMC11668696 DOI: 10.3389/fonc.2024.1390299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 11/04/2024] [Indexed: 12/28/2024] Open
Abstract
Background The second-line treatment of neuroendocrine tumors (NETs) of unknown primary origin remains uncertain. This report presented a patient who received octreotide plus IBI-318 plus anlotinib as a second-line treatment for multiple metastatic NETs of unknown primary lesions after the failure of octreotide plus everolimus. Case presentation A 32-year-old male patient presented with elevated CEA (197.83 ng/ml) without specific symptoms. A contrast-enhanced computed tomography (CT) scan showed multiple metastatic lymph nodes and multiple low-density nodules in the liver of undetermined nature. A right supraclavicular lymph node biopsy indicated NET, but the primary tumor origin remained unknown. PD-L1 expression was negative in tumor tissue according to immunohistochemistry. Immunofluorescence indicated the CD4+ T cells, CD8+ T cells, and Treg cells were gathered around blood vessels, with only a few infiltrating lymphocytes in the tumor tissue. Treatment with octreotide (30 mg/28 d) plus everolimus (5 mg qd) led to disease progression after three cycles. Treatment was changed to octreotide (30 mg/28 d) plus IBI318 (400 mg/28 d) plus anlotinib (10 mg/1-14 d/q3w), leading to partial remission, which was sustained up to the last follow-up (June 20, 2023), with a PFS of 11 months. The patient experienced no treatment-related adverse reactions. Conclusions Octreotide plus IBI318 plus anlotinib achieved benefits in a patient with advanced NETs of unknown primary lesions after first-line treatment failure, even though with low PD-L1 expression. This case suggests that combining SSAs, TKIs and PD-1/PD-L1 inhibitors could be an alternative second-line treatment for patients with advanced, well-differentiated NETs.
Collapse
Affiliation(s)
- Haoyue Qin
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Huan Yan
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xing Zhang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhe Huang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yangqian Chen
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuda Zhang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Siqi Xiang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongchang Zhang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The Cancer Center, The Second People's Hospital of Hunan Province/The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Liang Zeng
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
41
|
Kim CG, Jose J, Hay MP, Choi PJ. Novel Prodrug Strategies for the Treatment of Tuberculosis. Chem Asian J 2024; 19:e202400944. [PMID: 39179514 PMCID: PMC11613820 DOI: 10.1002/asia.202400944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024]
Abstract
The emergence of drug-resistant strains of Mycobacterium tuberculosis (M.tb), the causative agent of tuberculosis, is on the rise and increasing antimicrobial resistance is a global threat. This phenomenon necessitates new drug design methods such as a prodrug strategy to develop novel antitubercular agents. The prodrug strategy is a viable and useful means to improve the absorption, distribution, metabolism, excretion and toxicity (ADMET) profiles of pharmacologically active agents. Granulomas are a pathological hallmark of M.tb infection and bear a remarkable resemblance to the tumour microenvironment, including regions of hypoxia. The hypoxic environment observed in the two structures offer an exceptional opportunity to deliver antitubercular agents selectively in a similar manner to hypoxia activated prodrugs in cancer therapy. Nitroimidazoles have been studied extensively as bioactivated prodrugs of cancer, and their suitability as substrates for mammalian reductases highlight their huge potential. This review will discuss the mechanism of action and resistance mechanisms of the current prodrugs used for the treatment of tuberculosis. It will also highlight the potential advantages and challenges of using hypoxia activated prodrugs as a viable strategy to target latent M.tb in hypoxic regions of granulomas.
Collapse
Affiliation(s)
- Christine G. Kim
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| | - Jiney Jose
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| | - Michael P. Hay
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| | - Peter J. Choi
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| |
Collapse
|
42
|
Lin Z, Assaraf YG, Kwok HF. Peptides for microbe-induced cancers: latest therapeutic strategies and their advanced technologies. Cancer Metastasis Rev 2024; 43:1315-1336. [PMID: 39008152 DOI: 10.1007/s10555-024-10197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
Cancer is a significant global health concern associated with multiple distinct factors, including microbial and viral infections. Numerous studies have elucidated the role of microorganisms, such as Helicobacter pylori (H. pylori), as well as viruses for example human papillomavirus (HPV), hepatitis B virus (HBV), and hepatitis C virus (HCV), in the development of human malignancies. Substantial attention has been focused on the treatment of these microorganism- and virus-associated cancers, with promising outcomes observed in studies employing peptide-based therapies. The current paper provides an overview of microbe- and virus-induced cancers and their underlying molecular mechanisms. We discuss an assortment of peptide-based therapies which are currently being developed, including tumor-targeting peptides and microbial/viral peptide-based vaccines. We describe the major technological advancements that have been made in the design, screening, and delivery of peptides as anticancer agents. The primary focus of the current review is to provide insight into the latest research and development in this field and to provide a realistic glimpse into the future of peptide-based therapies for microbe- and virus-induced neoplasms.
Collapse
Affiliation(s)
- Ziqi Lin
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Instituteof Technology, Haifa, 3200003, Israel
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
43
|
Zhang Y, Numata K, Imajo K, Uojima H, Funaoka A, Komiyama S, Ogushi K, Chuma M, Irie K, Kokubu S, Yoneda M, Kobayashi T, Hidaka H, Fukushima T, Kobayashi S, Morimoto M, Kagawa T, Hattori N, Watanabe T, Iwase S, Maeda S. Lenvatinib radiofrequency ablation sequential therapy offers survival benefits for patients with unresectable hepatocellular carcinoma at intermediate stage and the liver reserve of Child-Pugh A category: A multicenter study. Hepatol Res 2024; 54:1174-1192. [PMID: 38953838 DOI: 10.1111/hepr.14089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 07/04/2024]
Abstract
AIM This study aims to evaluate the efficacy and safety of lenvatinib radiofrequency ablation (RFA) sequential therapy for certain hepatocellular carcinoma (HCC) patients. METHODS One hundred and nineteen patients with unresectable HCC in the intermediate stage with Child-Pugh A were retrospectively recruited in a multicenter setting. Those in the lenvatinib RFA sequential therapy group received lenvatinib initially, followed by RFA and the retreatment with lenvatinib. The study compared overall survival (OS), progression-free survival (PFS), tumor response, and adverse events (AEs) between patients undergoing sequential therapy and lenvatinib monotherapy. RESULTS After propensity score matching, 25 patients on sequential therapy and 50 on monotherapy were evaluated. Independent factors influencing OS were identified as sequential therapy, modified albumin-bilirubin (mALBI) grade, and relative dose intensity (%) with hazard ratios (HRs) of 0.381 (95% confidence interval [CI], 0.186-0.782), 2.220 (95% CI, 1.410-3.493), and 0.982 (95% CI, 0.966-0.999), respectively. Stratified analysis based on mALBI grades confirmed the independent influence of treatment strategy across all mALBI grades for OS (HR, 0.376; 95% CI, 0.176-0.804). Furthermore, sequential therapy was identified as an independent factor of PFS (HR, 0.382; 95% CI, 0.215-0.678). Sequential therapy significantly outperformed monotherapy on survival benefits (OS: 38.27 vs. 18.96 months for sequential therapy and monotherapy, respectively, p = 0.004; PFS: 13.80 vs. 5.32 months for sequential therapy and monotherapy, respectively, p < 0.001). Sequential therapy was significantly associated with complete response by modified Response Evaluation Criteria in Solid Tumors (odds ratio, 63.089). Ten of 119 patients experienced grade 3 AEs, with no AE beyond grade 3 observed. CONCLUSION Lenvatinib RFA sequential therapy might offer favorable tolerability and potential prognostic improvement compared to lenvatinib monotherapy.
Collapse
Affiliation(s)
- Ying Zhang
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Medical Ultrasound, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Kazushi Numata
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Kento Imajo
- Department of Gastroenterology, Shin-Yurigaoka General Hospital, Kawasaki, Japan
- Minimally Invasive Surgical and Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | - Haruki Uojima
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Akihiro Funaoka
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Satoshi Komiyama
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Katsuaki Ogushi
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Makoto Chuma
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Kuniyasu Irie
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shigehiro Kokubu
- Department of Gastroenterology, Shin-Yurigaoka General Hospital, Kawasaki, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hisashi Hidaka
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Taito Fukushima
- Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Satoshi Kobayashi
- Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Manabu Morimoto
- Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Nobuhiro Hattori
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tsunamasa Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shigeru Iwase
- Department of Gastroenterology, Fujisawa City Hospital, Fujisawa, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
44
|
Gu D, Yu H, Ding N, Xu J, Qian P, Zhu J, Jiang M, Tao H, Zhu X. A phase II study of anlotinib plus whole brain radiation therapy for patients with NSCLC with multiple brain metastases. Ann Med 2024; 56:2401618. [PMID: 39445449 PMCID: PMC11616763 DOI: 10.1080/07853890.2024.2401618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/12/2024] [Accepted: 08/29/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Whole brain radiotherapy (WBRT) is the mainstay of treatment for patients with non-small cell lung cancer (NSCLC) with multiple brain metastases (BMs); however, the BRAIN study showed that the efficacy of WBRT is unsatisfactory. This prospective phase II study aimed to evaluate the efficacy and safety of WBRT combined with anlotinib, a novel anti-angiogenic multi-target tyrosine kinase inhibitor (TKI), in patients with multiple BMs (>3) from advanced NSCLC. METHODS Patients with advanced NSCLC with multiple BMs who had received two or more lines of treatment were eligible for enrolment into this study. All patients were treated with anlotinib (8-12 mg, QD, on days 1-14 of a 21-day cycle) combined with WBRT (DT 30 Gy/12 F), followed by maintenance therapy with anlotinib until disease progression or treatment intolerance. The primary endpoint of this study was the intracranial progression-free survival (iPFS). The secondary endpoints were intracranial objective response rate (iORR), intracranial disease control rate (iDCR), overall survival (OS) and treatment safety. RESULTS Between May 2019 and January 2021, 28 patients were enrolled, all of whom were evaluable for efficacy and safety. The median age was 57.7 years, and 46.4% were male. Twenty-five patients had adenocarcinoma (89.3%), six had EGFR mutations (21.4%) and two had ALK mutations (7.1%). The median iPFS was 11.1 months (95% confidence interval (CI): 5.4-16.8 months) and the median OS was 13.4 months (95% CI: 5.2-21.6 months). The iORR was 71.4% (six complete responses + 14 partial responses). The most frequently observed adverse events (AEs) were hypertension (71.4%), fatigue (64.3%), anorexia (46.4%), and foot and hand skin reactions (25.0%). No patients developed ≥ grade 4 AEs. No intracranial haemorrhages occurred during treatment. Dose adjustment due to AEs occurred in 17.9% of patients. CONCLUSIONS Anlotinib combined with WBRT is effective and well-tolerated in patients with NSCLC with multiple BMs.
Collapse
Affiliation(s)
- Dayong Gu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Hongliang Yu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Naixin Ding
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Jianhua Xu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Pudong Qian
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Jun Zhu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Ming Jiang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Hua Tao
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| | - Xiangzhi Zhu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, People’s Republic of China
| |
Collapse
|
45
|
Hamada Y, Tanoue K, Arigami T, Yamakuchi M, Okawa M, Matsushita D, Takenouchi K, Yamada S, Maywar DN, Nakayama C, Oyama Y, Higashi S, Fujisaki C, Hozaka Y, Kita Y, Hashiguchi T, Ohtsuka T. The Vascular Endothelial Growth Factor-A121/Vascular Endothelial Growth Factor-A165 Ratio as a Predictor of the Therapeutic Response to Immune Checkpoint Inhibitors in Gastric Cancer. Cancers (Basel) 2024; 16:3958. [PMID: 39682145 PMCID: PMC11640175 DOI: 10.3390/cancers16233958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES The response rate to immune checkpoint inhibitor (ICI) therapy is limited. Further, there is a need to discover biomarkers to predict therapeutic efficacy. The vascular endothelial growth factor (VEGF) is strongly associated with intra-tumoral immunity; however, its utility as a marker remains unknown. Therefore, our objectives were to examine the isoforms of VEGF and determine whether VEGF levels predict ICI efficacy. METHODS Levels of VEGF isoforms VEGF-A121 and VEGF-A165 were measured in stored serum samples obtained from 30 patients with advanced or recurrent gastric cancer who received nivolumab monotherapy at Kagoshima University Hospital, and the association with prognosis and treatment efficacy was retrospectively analyzed. RESULTS The serum levels of the total VEGF, VEGF-A121, and VEGF-A165 were not significantly associated with prognosis. However, the ratio of VEGF-A121/VEGF-A165 (VEGF-A121/165) exhibited a statistically significant (p = 0.0088) difference in progression-free survival (PFS) with the low-ratio group having a 67-day prolonged median PFS time. Under univariable analysis, only VEGF-A121/165 values exhibited reduced progression-free survival with statistical significance. When comparing treatment responses in the low (n = 15) and high (n = 15) serum VEGF-A-121/165 groups, RECIST evaluation was 3 to 0 for complete response (CR), 2 to 0 for partial response (PR), 3 to 2 for stable disease (SD), and 3 to 10 for progressive disease (PD). Patients with clinically unsettled PR or SD were classified as non-CR/non-PD (4 vs. 3), with a disease control rate of 80% vs. 33%. CONCLUSIONS The serum VEGF-A121/165 ratio may represent a new, easily measured biomarker for predicting the therapeutic response to ICIs.
Collapse
Affiliation(s)
- Yuki Hamada
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
| | - Kiyonori Tanoue
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Takaaki Arigami
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Masashi Okawa
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Daisuke Matsushita
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
| | - Kazunori Takenouchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | | | - Drew N. Maywar
- Department of Electrical and Computer Engineering Technology, Rochester Institute of Technology, Rochester, NY 14623, USA
| | - Chieri Nakayama
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
| | - Yoko Oyama
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Sadayuki Higashi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Chieko Fujisaki
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Yuto Hozaka
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
| | - Yoshiaki Kita
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
| | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Takao Ohtsuka
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan (T.A.)
| |
Collapse
|
46
|
Deng H, Xu Q, Li XT, Huang X, Liu JY, Yan R, Quan ZS, Shen QK, Guo HY. Design, synthesis, and evaluation of antitumor activity in Pseudolaric acid B Azole derivatives: Novel and potent angiogenesis inhibitor via regulation of the PI3K/AKT and MAPK mediated HIF-1/VEGF signaling pathway. Eur J Med Chem 2024; 278:116813. [PMID: 39226705 DOI: 10.1016/j.ejmech.2024.116813] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Tumor proliferation and metastasis are intricately linked to blood vessel formation, with vascular endothelial growth factor (VEGF) playing a pivotal role in orchestrating angiogenesis throughout tumor progression. Pseudolaric acid B (PAB) has emerged as a potent inhibitor of tumor cell proliferation, migration, and angiogenesis. In efforts to enhance its efficacy, 37 derivatives of PAB were synthesized and assessed for their capacity to suppress VEGF secretion in SiHa cells under hypoxic conditions. Notably, majority of these derivatives exhibited significant inhibition of VEGF protein secretion without inducing cytotoxicity. Among them, compound M2 displayed the most potent inhibitory activity, with an IC50 value of 0.68 μM, outperforming the lead compound PAB (IC50 = 5.44 μM). Compound M2 not only curbed the migration and angiogenesis of HUVECs under hypoxic conditions but also hindered the invasion of SiHa cells. Mechanistic investigations unveiled that compound M2 may impede the accumulation and nuclear translocation of hypoxia-inducible factor 1α (HIF-1α) in SiHa cells, thereby downregulating VEGF expression. This inhibitory effect on HIF-1α was corroborated by experiments utilizing the protease inhibitor MG-132 and protein synthesis inhibitor CHX, indicating that compound M2 diminishes HIF-1α levels by reducing its synthesis. Furthermore, compound M2 was observed to modulate the PI3K/AKT/mTOR and MAPK signaling pathways in tumor cells, thereby regulating HIF-1α translation and synthesis. In vivo studies demonstrated that compound M2 exhibited low toxicity and effectively curbed tumor growth. Immunohistochemistry analyses validated that compound M2 effectively suppressed the expression of HIF-1α and VEGF in tumor tissues, underscoring its potential as a promising therapeutic agent for targeting tumor angiogenesis.
Collapse
Affiliation(s)
- Hao Deng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China
| | - Qian Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China
| | - Xiao-Ting Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China
| | - Xing Huang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China
| | - Jin-Ying Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China
| | - Rui Yan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China.
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China.
| | - Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China.
| |
Collapse
|
47
|
Qiu X, Li S, Fan T, Zhang Y, Wang B, Zhang B, Zhang M, Zhang L. Advances and prospects in tumor infiltrating lymphocyte therapy. Discov Oncol 2024; 15:630. [PMID: 39514075 PMCID: PMC11549075 DOI: 10.1007/s12672-024-01410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy in adoptive T-cell therapy (ACT) has already caused durable regression in a variety of cancer types due to T-cell persistence, clinical activity, and duration of objective response and safety. TILs are composed of polyclonal effector T-cells specific to heterogenetic tumor antigens, reasonably providing a promising means for tumor therapy. In addition, their expansion in vitro can release them from the suppressive tumor microenvironment. Even though significant advances have been made in the procedure of TIL therapy, from TIL isolation, modification, expansion, and infusion back to the patient to target the tumor, strategy optimization is always ongoing to overcome drawbacks such as a complex process, options for the lineage differentiation status of TILs, and sufficient trafficking of TILs to the tumor. In this review, we summarize the current advances of TIL therapy, raise problem-based optimization strategies, and provide future perspectives on next-generation TIL therapy as a potential avenue for enhancing cell-based immunotherapy.
Collapse
Affiliation(s)
- Xu Qiu
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Shengjun Li
- Clinical Laboratory, Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Tianyu Fan
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Taian City Central Hospital, Taian, Shandong, China
| | - Yue Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Bin Wang
- The Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Bei Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Mingzhe Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
48
|
Yang C, Zhang ZM, Zhao ZP, Wang ZQ, Zheng J, Xiao HJ, Xu H, Liu H, Yang L. Radiomic analysis based on magnetic resonance imaging for the prediction of VEGF expression in hepatocellular carcinoma patients. Abdom Radiol (NY) 2024; 49:3824-3833. [PMID: 38896246 PMCID: PMC11519187 DOI: 10.1007/s00261-024-04427-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE The purpose of this study was to investigate the ability of radiomic characteristics of magnetic resonance images to predict vascular endothelial growth factor (VEGF) expression in hepatocellular carcinoma (HCC) patients. METHODS One hundred and twenty-four patients with HCC who underwent fat-suppressed T2-weighted imaging (FS-T2WI) and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) one week before surgical resection were enrolled in this retrospective study. Immunohistochemical analysis was used to evaluate the expression level of VEGF. Radiomic features were extracted from the axial FS-T2WI, DCE-MRI (arterial phase and portal venous phase) images of axial MRI. Least absolute shrinkage and selection operator (LASSO) and stepwise regression analyses were performed to select the best radiomic features. Multivariate logistic regression models were constructed and validated using tenfold cross-validation. Receiver operating characteristic (ROC) curve analysis, calibration curve analysis and decision curve analysis (DCA) were employed to evaluate these models. RESULTS Our results show that there were 94 patients with high VEGF expression and 30 patients with low VEGF expression among the 124 HCC patients. The FS-T2WI, DCE-MRI and combined MRI radiomics models had AUCs of 0.8713, 0.7819, and 0.9191, respectively. There was no significant difference in the AUC between the FS-T2WI radiomics model and the DCE-MRI radiomics model (p > 0.05), but the AUC for the combined model was significantly greater than the AUCs for the other two models (p < 0.05) according to the DeLong test. The combined model had the greatest net benefit according to the DCA results. CONCLUSION The radiomic model based on multisequence MR images has the potential to predict VEGF expression in HCC patients. The combined model showed the best performance.
Collapse
Affiliation(s)
- Cui Yang
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Ze-Ming Zhang
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Zhang-Ping Zhao
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Zhi-Qing Wang
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Jing Zheng
- Medical Imaging Key Laboratory of Sichuan Province, Science and Technology Innovation Center, Interventional Medical Center, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, P. R. China
| | - Hua-Jing Xiao
- Department of Pathology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Hong Xu
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Hui Liu
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Lin Yang
- Medical Imaging Key Laboratory of Sichuan Province, Science and Technology Innovation Center, Interventional Medical Center, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, P. R. China.
| |
Collapse
|
49
|
Wang Z, Su Y, Zhao L, Liu W, Zhang J, Yang W, Li H, Feng M, Wang H, Song Z. Pan-Cancer Single-Cell Transcriptomic Analysis Reveals Divergent Expression of Embryonic Proangiogenesis Gene Modules in Tumorigenesis. Cancer Med 2024; 13:e70373. [PMID: 39526457 PMCID: PMC11551789 DOI: 10.1002/cam4.70373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/15/2024] [Accepted: 09/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Angiogenesis is indispensable for the sustained survival and progression of both embryonic development and tumorigenesis. This intricate process is tightly regulated by a multitude of pro-angiogenic genes. The presence of gene modules facilitating angiogenesis has been substantiated in both embryonic development and the context of tumor proliferation. However, it remains unresolved whether the pro-angiogenic gene modules expressed during embryonic development also exist in tumors. METHODS This study performed a pan-cancer single-cell RNA sequencing (scRNA-seq) analysis on samples from 332 patients across seven cancer types: thyroid carcinoma, lung cancer, breast cancer, hepatocellular carcinoma, colorectal cancer, ovarian carcinoma, and prostate adenocarcinoma. Data processing was carried out using the Seurat R package, with rigorous quality control to filter high-quality cells and mitigate batch effects across datasets. We used principal component analysis (PCA), shared nearest neighbor graph-based clustering, and Uniform Manifold Approximation and Projection (UMAP) to visualize cell types and identify distinct cell clusters. Myeloid cell subpopulations were further analyzed for the expression of embryonic pro-angiogenic gene modules (EPGM) and tumor pro-angiogenic gene modules (TPGM). RESULTS The analysis identified nine major cell types within the tumor microenvironment, with myeloid cells consistently exhibiting elevated expression of both tumor pro-angiogenic gene modules (TPGM) and EPGM across all tumor types. In particular, myeloid cells, including macrophages and monocytes, showed high EPGM expression, indicating an active role of embryonic pro-angiogenesis pathways in tumors. A subset analysis revealed 20 distinct myeloid subtypes with varying EPGM and TPGM expression across different cancers. Treatment and disease stage influenced these gene expressions, with certain subtypes, such as HSPAhi/STAT1+ macrophages in breast cancer, displaying reduced pro-angiogenic gene activity post-treatment. CONCLUSION This study provides evidence that tumors may exploit EPGM to enhance vascularization and support sustained growth, as evidenced by the elevated EPGM expression in tumor-associated myeloid cells. The consistent presence of EPGM in TAMs across multiple cancer types suggests a conserved mechanism wherein tumors harness embryonic angiogenic pathways to facilitate their progression. Distinct EPGM expression patterns in specific myeloid cell subsets indicate potential therapeutic targets, particularly in cases where EPGM activation contributes to resistance against anti-angiogenic therapies. These findings shed new light on the molecular mechanisms underlying tumor angiogenesis and highlight the prognostic relevance of EPGM expression in cancer, underscoring its potential as a biomarker for clinical applications.
Collapse
Affiliation(s)
- Zeshuai Wang
- College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
- Maternal‐Fetal Medicine Institute, Department of Obstetrics and Gynecology, Shenzhen Baoan Women’s and Children’s HospitalShenzhen UniversityShenzhenChina
| | - Yiyi Su
- College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
- Maternal‐Fetal Medicine Institute, Department of Obstetrics and Gynecology, Shenzhen Baoan Women’s and Children’s HospitalShenzhen UniversityShenzhenChina
| | - Lisha Zhao
- Maternal‐Fetal Medicine Institute, Department of Obstetrics and Gynecology, Shenzhen Baoan Women’s and Children’s HospitalShenzhen UniversityShenzhenChina
| | - Wei Liu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsGuangdong Medical UniversityDongguanChina
| | - Jiaqi Zhang
- Key Laboratory of Quantitative Synthetic BiologyShenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Wei Yang
- Maternal‐Fetal Medicine Institute, Department of Obstetrics and Gynecology, Shenzhen Baoan Women’s and Children’s HospitalShenzhen UniversityShenzhenChina
| | - Hanjie Li
- Key Laboratory of Quantitative Synthetic BiologyShenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Mingqian Feng
- College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Hao Wang
- Maternal‐Fetal Medicine Institute, Department of Obstetrics and Gynecology, Shenzhen Baoan Women’s and Children’s HospitalShenzhen UniversityShenzhenChina
| | - Zhuo Song
- Maternal‐Fetal Medicine Institute, Department of Obstetrics and Gynecology, Shenzhen Baoan Women’s and Children’s HospitalShenzhen UniversityShenzhenChina
| |
Collapse
|
50
|
Xiao G, Huang X, Huang T, Chen Z, Huang Y, Huang R, Wang X. Hepatitis B virus X protein differentially regulates the angiogenesis of Hepatocellular Carcinoma through p53-VEGF axis according to glucose levels. Ann Hepatol 2024; 29:101543. [PMID: 39216627 DOI: 10.1016/j.aohep.2024.101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION AND OBJECTIVES Blood glucose fluctuates severely in the diabetes (DM) and tumor microenvironment. Our previous works have found Hepatitis B virus X protein (HBx) differentially regulated metastasis and apoptosis of hepatoma cells depending on glucose concentration. We here aimed to explore whether HBx played dual roles in the angiogenesis of hepatocellular carcinoma varying on different glucose levels. MATERIALS AND METHODS We collected conditioned medium from HBx-overexpressing cells cultured with two solubilities of glucose, and then applied to EA.hy926 cells. Alternatively, a co-culture cell system was established with hepatoma cells and EA.hy926 cells. We analyzed the angiogenesis of EA.hy926 cells with CCK8, wound-healing, transwell-migartion and tube formation experiment. ELISA was conducted to detect the secretion levels of angiogenesis-related factors. siRNAs were used to detect the P53-VEGF axis. RESULTS HBx expressed in hepatoma cells suppressed VEGF secretion, and subsequently inhibited the proliferation, migration and tube formation of EA.hy926 cells in a high glucose condition, while attenuating these in the lower glucose condition. Furthermore, the p53-VEGF axis was required for the dual role of HBx in angiogenesis. Additionally, HBx mainly regulated the nuclear p53. CONCLUSIONS These data suggest that the dual roles of HBx confer hepatoma cells to remain in a glucose-rich environment and escape from the glucose-low milieu through tumor vessels, promoting liver tumor progression overall. We exclusively revealed the dual role of HBx on the angiogenesis of liver tumors, which may shed new light on the mechanism and management strategy of HBV- and DM-related hepatocellular carcinoma.
Collapse
Affiliation(s)
- Guitao Xiao
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, 105, Jiuyibei Road, Xin Luo, Longyan, Fujian 364000, PR China
| | - Xiaoyun Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Fujian Medical University Cancer Center, Fujian Medical University, 1, Xuefubei Road, Minhou, Fuzhou, Fujian 350001, PR China
| | - Tingxuan Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China
| | - Zhixin Chen
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China
| | - Yuehong Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China
| | - Rongfeng Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Fujian Medical University Cancer Center, Fujian Medical University, 1, Xuefubei Road, Minhou, Fuzhou, Fujian 350001, PR China.
| | - Xiaozhong Wang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China; Fujian Medical University Cancer Center, Fujian Medical University, 1, Xuefubei Road, Minhou, Fuzhou, Fujian 350001, PR China.
| |
Collapse
|