351
|
Suzuki H, Mishra S, Paul S, Hoshida Y. Molecular and immune landscape of hepatocellular carcinoma for therapeutic development. JOURNAL OF LIVER CANCER 2025; 25:9-18. [PMID: 39639434 PMCID: PMC7617546 DOI: 10.17998/jlc.2024.12.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with an estimated 750,000 deaths in 2022. Recent emergence of molecular targeted agents and immune checkpoint inhibitors and their combination therapies have been transforming HCC care, but their prognostic impact in advanced-stage disease remains unsatisfactory. In addition, their application to early-stage disease is still an unmet need. Omics profiling studies have elucidated recurrent and heterogeneously present molecular aberrations involved in pro-cancer tumor (immune) microenvironment that may guide therapeutic strategies. Recurrent aberrations such somatic mutations in TERT promoter and TP53 have been regarded undruggable, but recent studies have suggested that these may serve as new classes of therapeutic targets. HCC markers such as alpha-fetoprotein, glypican-3, and epithelial cell adhesion molecule have also been explored as therapeutic targets. These molecular features may be utilized as biomarkers to guide the application of new approaches as companion biomarkers to maximize therapeutic benefits in patients who are likely to benefit from the therapies, while minimizing unnecessary harm in patients who will not respond. The explosive number of new agents in the pipelines have posed challenges in their clinical testing. Novel clinical trial designs guided by predictive biomarkers have been proposed to enable their efficient and cost-effective evaluation. These new developments collectively facilitate clinical translation of personalized molecular-targeted therapies in HCC and substantially improve prognosis of HCC patients.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sumit Mishra
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Subhojit Paul
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
352
|
Chantre-justino M, Silvestre RT, De Castro TL, Luz E, Pinheiro RDCES, Caruso A, Lopes ACDS, Meohas W, Alves G, Ornellas MHF. Genetic profiling of osteosarcoma in an adolescent using a next‑generation sequencing panel and Sanger sequencing: A case report and review of the literature. Biomed Rep 2025; 22:42. [PMID: 39810900 PMCID: PMC11729137 DOI: 10.3892/br.2025.1920] [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: 06/11/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor affecting adolescents and young adults and it usually occurs in the long bones of the extremities. The detection of cancer-related genetic alterations has a growing effect in guiding diagnosis, prognosis and targeted therapies. However, little is known about the molecular aspects involved in the etiology and progression of OS, which limits options for targeted therapies. The present study described a case of an adolescent patient (16-years-old) who was diagnosed with conventional central OS in the right distal femur without the evidence of pulmonary metastases; the patient was treated with surgery and adjuvant chemotherapy. Genetic alterations in resected tumor tissue were investigated via next-generation sequencing (NGS) technology using a targeted NGS panel. Sanger sequencing was also performed to investigate somatic and germline TP53 mutations (exons 4-8). NGS analysis revealed an intratumor heterogeneity signature in OS tumor, including several single nucleotide variants identified in genes encoding tyrosine kinase proteins. No PCR products for TP53 exon 5 were detected in the tumor sample by PCR analysis prior to Sanger sequencing, suggesting a significant deletion in this exon. Sanger sequencing analysis revealed the missense variant TP53 c.712T>A (p.Cys238Ser) in tumor tissue sample, thus reinforcing the role of TP53 somatic mutations in OS development. Additionally, the TP53 c.215C>G (p.Pro72Arg) germline missense variant was identified in the peripheral blood sample. In conclusion, the findings provided new information on genetic aspects that may contribute to OS development, especially in pediatric patients.
Collapse
Affiliation(s)
- Mariana Chantre-justino
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Rafaele Tavares Silvestre
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Thiago Luz De Castro
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Eliane Luz
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Rafael De Castro E Silva Pinheiro
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Anabela Caruso
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Ana Cristina De Sá Lopes
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Walter Meohas
- Specialized Care Center for Orthopedic Oncology, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Gilda Alves
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Maria Helena Faria Ornellas
- Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| |
Collapse
|
353
|
Liu S, Meng Y, Zhang Y, Qiu L, Wan X, Yang X, Zhang Y, Liu X, Wen L, Lei X, Zhang B, Han J. Integrative analysis of senescence-related genes identifies robust prognostic clusters with distinct features in hepatocellular carcinoma. J Adv Res 2025; 69:107-123. [PMID: 38614215 PMCID: PMC11954806 DOI: 10.1016/j.jare.2024.04.007] [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: 11/15/2023] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024] Open
Abstract
INTRODUCTION Senescence refers to a state of permanent cell growth arrest and is regarded as a tumor suppressive mechanism, whereas accumulative evidence demonstrate that senescent cells play an adverse role during cancer progression. The scarcity of specific and reliable markers reflecting senescence level in cancer impede our understanding of this biological basis. OBJECTIVES Senescence-related genes (SRGs) were collected for integrative analysis to reveal the role of senescence in hepatocellular carcinoma (HCC). METHODS Consensus clustering was used to subtype HCC based on SRGs. Several computational methods, including single sample gene set enrichment analysis (ssGSEA), fuzzy c-means algorithm, were performed. Data of drug sensitivities were utilized to screen potential therapeutic agents for different senescence patients. Additionally, we developed a method called signature-related gene analysis (SRGA) for identification of markers relevant to phenotype of interest. Experimental strategies consisting quantitative real-time PCR (qRT-PCR), β-galactosidase assay, western blot, and tumor-T cell co-culture system were used to validate the findings in vitro. RESULTS We identified three robust prognostic clusters of HCC patients with distinct survival outcome, mutational landscape, and immune features. We further extracted signature genes of senescence clusters to construct the senescence scoring system and profile senescence level in HCC at bulk and single-cell resolution. Senescence-induced stemness reprogramming was confirmed both in silico and in vitro. HCC patients with high senescence were immune suppressed and sensitive to Tozasertib and other drugs. We suggested that MAFG, PLIN3, and 4 other genes were pertinent to HCC senescence, and MAFG potentially mediated immune suppression, senescence, and stemness. CONCLUSION Our findings provide insights into the role of SRGs in patients stratification and precision medicine.
Collapse
Affiliation(s)
- Sicheng Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Meng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaguang Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaowen Wan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyang Yang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xueqin Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linda Wen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xue Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhong Han
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
354
|
Klg A, Priyadharshini B, Vasugi S, Dilipan E. Exploring the therapeutic potential of biosynthetic enzymes in cancer treatment: Innovations and implications. Int J Biol Macromol 2025; 292:139171. [PMID: 39732247 DOI: 10.1016/j.ijbiomac.2024.139171] [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: 09/18/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Cancer remains a major global health concern due to several factors. These include the difficulty in accessing effective drugs, the high toxicity of available treatments, and the emergence of resistance to therapy. As a result, alternative strategies, such as the use of microbial enzymes, have gained attention as potential solutions to these challenges. Microbial enzymes have shown promise in inhibiting the uncontrolled growth of tumor cells through various mechanisms. In this comprehensive review, our objective is to emphasize the importance of pivotal microbial enzymes in fighting cancer and their ability to hinder the growth of tumors or cancer cells. The review article serves as a scientific roadmap for researchers, clinicians, and industry stakeholders exploring the therapeutic potential of biosynthetic enzymes in cancer treatment. It emphasizes the quest for effective and sustainable cancer therapies, presenting the possibility of personalized treatments with fewer side effects than traditional therapies.
Collapse
Affiliation(s)
- Afeeza Klg
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Boopathy Priyadharshini
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Suresh Vasugi
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Elangovan Dilipan
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
355
|
Mei J, Yang K, Zhang X, Luo Z, Tian M, Fan H, Chu J, Zhang Y, Ding J, Xu J, Cai Y, Yin Y. Intratumoral Collagen Deposition Supports Angiogenesis Suggesting Anti-angiogenic Therapy in Armored and Cold Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409147. [PMID: 39823457 PMCID: PMC11904994 DOI: 10.1002/advs.202409147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/05/2025] [Indexed: 01/19/2025]
Abstract
A previous study classifies solid tumors based on collagen deposition and immune infiltration abundance, identifying a refractory subtype termed armored & cold tumors, characterized by elevated collagen deposition and diminished immune infiltration. Beyond its impact on immune infiltration, collagen deposition also influences tumor angiogenesis. This study systematically analyzes the association between immuno-collagenic subtypes and angiogenesis across diverse cancer types. As a result, armored & cold tumors exhibit the highest angiogenic activity in lung adenocarcinoma (LUAD). Single-cell and spatial transcriptomics reveal close interactions and spatial co-localization of fibroblasts and endothelial cells. In vitro experiments demonstrate that collagen stimulates tumor cells to express vascular endothelial growth factor A (VEGFA) and directly enhances vessel formation and endothelial cell proliferation through sex determining region Y box 18 (SOX18) upregulation. Collagen inhibition via multiple approaches effectively suppresses tumor angiogenesis in vivo. In addition, armored & cold tumors display superior responsiveness to anti-angiogenic therapy in advanced LUAD cohorts. Post-immunotherapy resistance, the transformation into armored & cold tumors emerges as a potential biomarker for selecting anti-angiogenic therapy. In summary, collagen deposition is shown to drive angiogenesis across various cancers, providing a novel and actionable framework to refine therapeutic strategies combining chemotherapy with anti-angiogenic treatments.
Collapse
Affiliation(s)
- Jie Mei
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Kai Yang
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Xinkang Zhang
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Zhiwen Luo
- Department of Sports MedicineHuashan Hospital Affiliated to Fudan UniversityShanghai200040P. R. China
| | - Min Tian
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Hanfang Fan
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Jiahui Chu
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Yan Zhang
- Departments of GynecologyThe Women's Hospital Affiliated to Jiangnan UniversityWuxi214023China
| | - Junli Ding
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Junying Xu
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Yun Cai
- Central LaboratoryChangzhou Jintan First People's HospitalThe Affiliated Jintan Hospital of Jiangsu UniversityChangzhouJiangsu213200P. R. China
| | - Yongmei Yin
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- Jiangsu Key Lab of Cancer BiomarkersPrevention and TreatmentCollaborative Innovation Center for Personalized Cancer MedicineNanjing Medical UniversityNanjingJiangsuP. R. China
| |
Collapse
|
356
|
Zhao Y, Zhu M, Ling Y, Zhao Y, Lu X, Chu B, He Y, Wang H. A DNA Nanopatch-Bacteriophage System Targeting Streptococcus Gallolyticus for Inflammatory Bowel Disease Treatment and Colorectal Cancer Prevention. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417334. [PMID: 39924920 DOI: 10.1002/adma.202417334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/28/2025] [Indexed: 02/11/2025]
Abstract
Persistent inflammation in inflammatory bowel disease (IBD) increases Streptococcus gallolyticus (Sg) colonization, increasing the risk of colorectal cancer progression via the Sg-activated cyclooxygenase-2 (COX-2) pathway and β-catenin upregulation. This study presents Sg-specific bacteriophages modified with DNA nanopatches (DNPs@P) designed to treat IBD and prevent Sg-induced malignancy. The DNPs are composed of DNA origami nanosheets and phage capture strands. The DNPs scavenge reactive oxygen species, enhancing the therapeutic efficacy of the phages while targeting and lysing pathogenic bacteria. Coating with an enteric polymer, DNPs@P ensures effective delivery in the gastrointestinal tract. These findings demonstrate significant restoration of colonic length, reduced inflammation, and improved gut microbiota diversity compared with current clinical treatments. Additionally, DNPs@P effectively prevents colonic tumourigenesis in mouse models. This approach presents a promising strategy for treating gastrointestinal diseases by remodeling the gut microenvironment, addressing a critical gap in current therapies.
Collapse
Affiliation(s)
- Yadan Zhao
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Mengna Zhu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yufan Ling
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Yingying Zhao
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xing Lu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| |
Collapse
|
357
|
Trastus LA, d'Adda di Fagagna F. The complex interplay between aging and cancer. NATURE AGING 2025; 5:350-365. [PMID: 40038418 DOI: 10.1038/s43587-025-00827-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/17/2025] [Indexed: 03/06/2025]
Abstract
Cancer is an age-related disease, but the interplay between cancer and aging is complex and their shared molecular drivers are deeply intertwined. This Review provides an overview of how different biological pathways affect cancer and aging, leveraging evidence mainly derived from animal studies. We discuss how genome maintenance and accumulation of DNA mutations affect tumorigenesis and tissue homeostasis during aging. We describe how age-related telomere dysfunction and cellular senescence intricately modulate tumor development through mechanisms involving genomic instability and inflammation. We examine how an aged immune system and chronic inflammation shape tumor immunosurveillance, fueling DNA damage and cellular senescence. Finally, as animal models are important to untangling the relative contributions of these aging-modulated pathways to cancer progression and to test interventions, we discuss some of the limitations of physiological and accelerated aging models, aiming to improve experimental designs and enhance translation.
Collapse
Affiliation(s)
| | - Fabrizio d'Adda di Fagagna
- IFOM ETS-the AIRC Institute of Molecular Oncology, Milan, Italy.
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy.
| |
Collapse
|
358
|
Li M, Huang J, Lu W, Guo Y, Xia G, Hu Q. Serum glycosylated hemoglobin and prostate cancer risk: Results from a systematic review and dose-response meta-analysis. Urol Oncol 2025; 43:195.e1-195.e9. [PMID: 39393995 DOI: 10.1016/j.urolonc.2024.09.007] [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: 07/01/2024] [Revised: 08/24/2024] [Accepted: 09/08/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES To evaluate the correlation between serum glycosylated hemoglobin (HbA1c) levels and the risk of prostate cancer incidence and mortality, providing a comprehensive analysis to inform preventative and clinical strategies. METHODS A systematic review and meta-analysis was conducted including studies based on their documentation of prostate cancer incidence and mortality related to HbA1c levels, with a minimum of 3 risk-related data levels. The Newcastle-Ottawa Quality Assessment Scale (NOQAS) was used for quality assessment and risk of bias evaluation. We employed generalized least squares (GLS) to assess the linear trend within individual studies and combined these estimates using a random effects model. Additionally, we utilized a restricted cubic spline (RCS) model to investigate potential nonlinear trends. RESULTS A total of 13 studies were included in the quantitative synthesis ultimately. The quantitative analysis did not find a significant association between HbA1c levels and prostate cancer incidence. However, a significant positive correlation was revealed between HbA1c levels and both cancer-specific mortality (CSM) and all-cause mortality (ACM), with a 1% increase in HbA1c levels associated with a 26% increase in CSM and a 21% increase in ACM. CONCLUSION The HbA1c level is significantly associated with increased prostate cancer mortality. The results highlight the importance of considering blood sugar control in the comprehensive risk assessment for prostate cancer, particularly among the nondiabetic population.
Collapse
Affiliation(s)
- Mengqi Li
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China; Department of Nursing, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingqiang Huang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Urology, Fudan University, Shanghai, China
| | - Wenwen Lu
- Department of Nursing, Huashan Hospital, Fudan University, Shanghai, China
| | - Yijun Guo
- Institute of Urology, Fudan University, Shanghai, China; Department of Urology, Jing'an District Central Hospital, Shanghai, China
| | - Guowei Xia
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Urology, Fudan University, Shanghai, China
| | - Qingfeng Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Urology, Fudan University, Shanghai, China.
| |
Collapse
|
359
|
Macias RIR, Kanzaki H, Berasain C, Avila MA, Marin JJG, Hoshida Y. The Search for Risk, Diagnostic, and Prognostic Biomarkers of Cholangiocarcinoma and Their Biological and Clinicopathologic Significance. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:422-436. [PMID: 39103092 PMCID: PMC11841489 DOI: 10.1016/j.ajpath.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/01/2024] [Accepted: 06/20/2024] [Indexed: 08/07/2024]
Abstract
Cholangiocarcinomas (CCAs) are a heterogeneous group of malignant tumors that originate from the biliary tract. They are usually diagnosed in advanced stages, leading to a poor prognosis for affected patients. As CCA often arises as a sporadic cancer in individuals lacking specific risk factors or with heterogeneous backgrounds, and there are no defined high-risk groups, the implementation of effective surveillance programs for CCA is problematic. The identification and validation of new biomarkers useful for risk stratification, diagnosis, prognosis, and prediction of treatment response remains an unmet need for patients with CCA, even though numerous studies have been conducted lately to try to discover and validate CCA biomarkers. In this review, we overview the available information about the different types of biomarkers that have been investigated in recent years using minimally invasive biospecimens (blood, serum/plasma, bile, and urine) and their potential usefulness in diagnosis, prognosis, and risk stratification. It is widely accepted that early detection of CCA will impact patients' outcomes, by improving survival rates, quality of life, and the possibility of less invasive and/or curative treatments; however, challenges to its translation and clinical application for patients with CCA need to be resolved.
Collapse
Affiliation(s)
- Rocio I R Macias
- Experimental Hepatology and Drug Targeting Group, University of Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain.
| | - Hiroaki Kanzaki
- Division of Digestive and Liver Diseases, Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Carmen Berasain
- Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, Center for Applied Medical Research, Cancer Center University of Navarra, Pamplona, Spain
| | - Matias A Avila
- Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, Center for Applied Medical Research, Cancer Center University of Navarra, Pamplona, Spain
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting Group, University of Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
360
|
Tang L, Peng S, Zhuang X, He Y, Song Y, Nie H, Zheng C, Pan Z, Lam AK, He M, Shi X, Li B, Xu WW. Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention. MEDCOMM – ONCOLOGY 2025; 4. [DOI: 10.1002/mog2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/04/2025]
Abstract
ABSTRACTMetastasis remains a leading cause of cancer‐related deaths, defined by a complex, multi‐step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post‐translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Shao‐Cong Peng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Xiao‐Wan Zhuang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Yan He
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Yu‐Xiang Song
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Hao Nie
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Can‐Can Zheng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Zhen‐Yu Pan
- Department of Radiation Oncology, The Affiliated Huizhou Hospital Guangzhou Medical University Huizhou China
| | - Alfred King‐Yin Lam
- Cancer Molecular Pathology and Griffith Medical School Griffith University Gold Coast Queensland Australia
| | - Ming‐Liang He
- Department of Biomedical Sciences City University of Hong Kong Hong Kong China
| | - Xing‐Yuan Shi
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Bin Li
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Wen Wen Xu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| |
Collapse
|
361
|
Zhang M, Wei J, Sun Y, He C, Ma S, Pan X, Zhu X. The efferocytosis process in aging: Supporting evidence, mechanisms, and therapeutic prospects for age-related diseases. J Adv Res 2025; 69:31-49. [PMID: 38499245 PMCID: PMC11954809 DOI: 10.1016/j.jare.2024.03.008] [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: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Aging is characterized by an ongoing struggle between the buildup of damage caused by a combination of external and internal factors. Aging has different effects on phagocytes, including impaired efferocytosis. A deficiency in efferocytosis can cause chronic inflammation, aging, and several other clinical disorders. AIM OF REVIEW Our review underscores the possible feasibility and extensive scope of employing dual targets in various age-related diseases to reduce the occurrence and progression of age-related diseases, ultimately fostering healthy aging and increasing lifespan. Key scientific concepts of review Hence, the concurrent implementation of strategies aimed at augmenting efferocytic mechanisms and anti-aging treatments has the potential to serve as a potent intervention for extending the duration of a healthy lifespan. In this review, we comprehensively discuss the concept and physiological effects of efferocytosis. Subsequently, we investigated the association between efferocytosis and the hallmarks of aging. Finally, we discuss growing evidence regarding therapeutic interventions for age-related disorders, focusing on the physiological processes of aging and efferocytosis.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
362
|
Horiguchi H, Kadomatsu T, Oike Y. The Two Faces of Angiopoietin-Like Protein 2 in Cancer. Cancer Sci 2025; 116:592-599. [PMID: 39686837 PMCID: PMC11875762 DOI: 10.1111/cas.16434] [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: 11/01/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/18/2024] Open
Abstract
The tumor microenvironment is composed of tumor cells and various stromal cell types, such as immune cells, fibroblasts, and vascular cells. Signaling interactions between tumor and stromal cells orchestrate the tumor microenvironment's contribution to tumor progression. Angiopoietin-like protein 2 (ANGPTL2) is a secreted glycoprotein homologous to angiopoietins. Previous studies indicate that tumor cell-derived ANGPTL2 serves as a tumor promoter. However, recent studies suggest that tumor stroma-derived ANGPTL2 shows tumor-suppressive activity by enhancing anti-tumor immune responses, supporting a dual function for ANGPTL2 in cancer pathology. Such complexity can complicate development of effective therapeutic strategies targeting ANGPTL2. In this Review, we focus on ANGPTL2 activity in the tumor microenvironment and its function in anti-cancer immunity.
Collapse
Affiliation(s)
- Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Aging and Geriatric Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Aging and Geriatric Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
363
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [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: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
364
|
Ebid N, Sharaky M, Elkhoely A, El Morsy EM, Saad SY. Cross-Talk Signaling Between Non-Small Cell Lung Cancer Cell Lines and Fibroblasts Attenuates the Cytotoxic Effect of Cisplatin. J Biochem Mol Toxicol 2025; 39:e70201. [PMID: 40025825 DOI: 10.1002/jbt.70201] [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: 11/10/2024] [Revised: 01/11/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Fibroblasts represent one of the most crucial cell types in the tumor microenvironment (TME), playing a major role in chemoresistance development. This study investigated the ability of fibroblasts to alter the response of non-small cell lung cancer (NSCLC) cell lines to cisplatin exposure. A cytotoxicity assay was performed to determine the IC50 of cisplatin using MTT. The assay was performed on NSCLC cell lines A549 and H1299 monocultures and co-cultures with fibroblasts. The co-culture was performed directly with the HSF cell line and indirectly through conditioned media. The ELISA technique was then used to determine the expression of biochemical markers at various time points of co-culture before and after cisplatin exposure. We observed a time-dependent evolution in the fibroblast-cancer cell interplay. Initially, fibroblast co-culture enhanced the cytotoxic effect of cisplatin, as reflected by decreased IC50 values after 24 h of co-culture. However, prolonged co-culture durations (48-96 h) led to the emergence of cisplatin resistance, coinciding with increased IC50 values and altered expression of key biochemical markers. The findings suggest that fibroblasts undergo a potential identity switch over time, transitioning from a tumor-restrictive to a tumor-promoting phenotype. This switch was associated with the activation of EGFR and FGF signaling pathways, increased expression of angiogenic and metastasis markers (e.g., VEGF, MMP2 & MMP9), and inhibition of apoptosis (e.g., reduced caspase expression). Our results suggest that fibroblasts may initially potentiate the effect of cisplatin on NSCLC cells; however, in a time-dependent manner, fibroblast co-culture attenuates the cytotoxic efficacy of cisplatin.
Collapse
Affiliation(s)
- Nouran Ebid
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Egypt
| | - Marwa Sharaky
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Abeer Elkhoely
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| | - Engy M El Morsy
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| | - Sherif Y Saad
- Pharmacology & Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
365
|
Zhang X, Fang H, Yang B, Qin C, Hu F, Ruan W, Chen J, Zeng D, Gai Y, Lan X. Phase I study of [ 68Ga]Ga-HX01 for targeting integrin αvβ3 and CD13 in healthy and malignancy subjects. Eur J Nucl Med Mol Imaging 2025; 52:1293-1304. [PMID: 39609274 DOI: 10.1007/s00259-024-07002-3] [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: 08/01/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE Noninvasive angiogenesis visualization is essential for evaluating tumor proliferation, progression, invasion, and metastasis. This study aimed to translate the heterodimeric PET tracer [68Ga]Ga-HX01, which targets integrin αvβ3 and CD13 in neovascularization, into phase I clinical study. METHODS This study enrolled 12 healthy volunteers (phase Ia) and 10 patients with malignant tumors (phase Ib). The subjects in phase Ia were divided into low-dose (0.05 mCi/kg) and high-dose (0.1 mCi/kg) groups. For phase Ia subjects, PET/CT images, blood and urine samples were collected to analyze the biodistribution, pharmacokinetics, radiation dosimetry, and safety of [68Ga]Ga-HX01. For phase Ib patients, PET/MR scans were performed at 30 ± 5 and 60 ± 5 min after injection. The safety and preliminary diagnostic value of [68Ga]Ga-HX01 were assessed. RESULTS In phase Ia study, [68Ga]Ga-HX01 was distributed and metabolized similarly in two dosage groups as the highest accumulations in kidneys and urine. It possessed quick renal excretion and blood clearance with an elimination half-life (T1/2) of 28.92 ± 3.97 min. The total effective dose was 2.14 × 10- 2 mSv/MBq. In phase Ib study, [68Ga]Ga-HX01 clearly detected the lesions per patient, and found a total of 59 lesions with varying uptake levels. For safety evaluation, no serious adverse events were observed during the examination. CONCLUSION [68Ga]Ga-HX01 has proved to be a translational radiopharmaceutical with reliable security, favorable pharmacokinetics, and the ability to visualize tumors. The preliminary results in malignancy patients warrant further investigation of [68Ga]Ga-HX01 in monitoring antiangiogenic therapy of patients with malignancies. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, NCT06416774. Registered 11 May, 2024.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Biao Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Fan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Weiwei Ruan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, 430022, Wuhan, Hubei, China
| | - Dexing Zeng
- Hexin (Suzhou) Pharmaceutical Technology Co., Ltd, Taicang, 215421, Jiangsu, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China.
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China.
| |
Collapse
|
366
|
Sarikaya I. Radionuclide treatments of cancer: molecular mechanisms, biological responses, histopathological changes, and role of PET imaging. Nucl Med Commun 2025; 46:193-203. [PMID: 39654504 DOI: 10.1097/mnm.0000000000001941] [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: 02/06/2025]
Abstract
Radiation treatments [radiotherapy and radionuclide treatments (RNTs)] are one of the main and effective treatment modalities of cancer. Globally, the number of cancer patients treated with radionuclides are much less as compared to number of radiotherapy cases but with the development of new radiotracers, most notably 177 Lu and 225 Ac-labeled prostate-specific membrane antigen ligands, and 223 Ra-dichloride for prostate cancer and 177 Lu-somatostatin analogs for neuroendocrine tumors, there is a significant rise in RNTs in the last decade. As therapeutic applications of nuclear medicine is on the rise, the aim of this review is to summarize biological responses to radiation treatments and molecular mechanisms of radiation-induced cell death (e.g. ionization, DNA damages such as double-strand breaks, DNA repair mechanisms, types of cell deaths such as apoptosis, necrosis, and immunogenic cell death), histopathological changes with radiation treatments, and role of PET imaging in RNTs as part of radionuclide theranostics for selecting and planning patients for RNTs, dosimetry, predicting and assessing response to RNTs, predicting toxicities, and other possible PET findings which may be seen after RNTs such as activation of immune system.
Collapse
Affiliation(s)
- Ismet Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Kirklareli University, Kirklareli, Turkey
| |
Collapse
|
367
|
Kooi EJ, Marcelis L, Wesseling P. Pathological diagnosis of central nervous system tumours in adults: what's new? Pathology 2025; 57:144-156. [PMID: 39818455 DOI: 10.1016/j.pathol.2024.11.004] [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: 10/10/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 01/18/2025]
Abstract
In the course of the last decade, the pathological diagnosis of many tumours of the central nervous system (CNS) has transitioned from a purely histological to a combined histological and molecular approach, resulting in a more precise 'histomolecular diagnosis'. Unfortunately, translation of this refinement in CNS tumour diagnostics into more effective treatment strategies is lagging behind. There is hope though that incorporating the assessment of predictive markers in the pathological evaluation of CNS tumours will help to improve this situation. The present review discusses some novel aspects with regard to the pathological diagnosis of the most common CNS tumours in adults. After a brief update on recognition of clinically meaningful subgroups in adult-type diffuse gliomas and the value of assessing predictive markers in these tumours, more detailed information is provided on predictive markers of (potential) relevance for immunotherapy especially for glioblastomas, IDH-wildtype. Furthermore, recommendations for improved grading of meningiomas by using molecular markers are briefly summarised, and an overview is given on (predictive) markers of interest in metastatic CNS tumours. In the last part of this review, some 'emerging new CNS tumour types' that may occur especially in adults are presented in a table. Hopefully, this review provides useful information on 'what's new' for practising pathologists diagnosing CNS tumours in adults.
Collapse
Affiliation(s)
- Evert-Jan Kooi
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Amsterdam, The Netherlands.
| | - Lukas Marcelis
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Amsterdam, The Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
368
|
Zhang Y, Xie S, Li W, Gu J, Zhang X, Ni B, Wang Z, Yang R, Song H, Zhong Y, Huang P, Zhou J, Cao Y, Guo J, Liu Y, Qi S, Wang H. TDP-43/ALKBH5-mediated m 6A modification of CDC25A mRNA promotes glioblastoma growth by facilitating G1/S cell cycle transition. MedComm (Beijing) 2025; 6:e70108. [PMID: 39974663 PMCID: PMC11836349 DOI: 10.1002/mco2.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 11/05/2024] [Accepted: 12/21/2024] [Indexed: 02/21/2025] Open
Abstract
Glioblastoma (GBM) exhibits significant intratumor heterogeneity (ITH), indicating the presence of tumor cells with diverse growth rates. Here, we aimed to identify fast-growing cells in GBM and elucidate the underlying mechanisms. Precisely targeting these cells could offer an improved treatment option. Our results found that targeting ALKBH5 expression impaired GBM proliferation and tumor stemness. Nuclear but not overall expression of ALKBH5 differs between monoclonal cells derived from the same patient with different proliferation rates. Mechanistically, ALKBH5 interacted with TAR DNA-binding protein 43 (TDP-43) in fast-growing cells. Furthermore, TDP-43 facilitated the nuclear localization of ALKBH5 and its binding to cell division cycle 25A (CDC25A) pre-mRNA. The TDP-43/ALKBH5 complex regulates CDC25A mRNA splicing via N6-methyladenosine (m6A) demethylation to maintain the expression of its oncogenic isoform (CDC25A-1), ultimately promoting the G1/S phase transition and growth of GBM cells. TRAD01 selectively targeted the interaction between TDP-43 and ALKBH5, leading to significant antitumor effects both in vitro and in vivo. Our study identified a novel epigenetic mechanism by which TDP-43/ALKBH5 contributes to GBM growth via m6A modification and alternative splicing. Therefore, targeting the TDP-43/ALKBH5 axis might be a promising therapeutic strategy for GBM patients.
Collapse
Affiliation(s)
- Yunxiao Zhang
- Department of Neurosurgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain DiseasesNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Sidi Xie
- Department of Neurosurgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain DiseasesNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Weizhao Li
- Department of Neurosurgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain DiseasesNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Junwei Gu
- The First People's Hospital of Xiushui CountyJiujiangChina
| | - Xi‐an Zhang
- Department of Neurosurgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain DiseasesNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Bowen Ni
- Department of Neurosurgery & Medical Research Center, Shunde HospitalSouthern Medical UniversityShundeChina
| | - Ziyu Wang
- Department of Neurosurgery & Medical Research Center, Shunde HospitalSouthern Medical UniversityShundeChina
| | - Runwei Yang
- Department of Neurosurgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain DiseasesNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Haimin Song
- Department of NeurosurgeryThe First Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| | - Yaxuan Zhong
- School of the First Clinical MedicineGannan Medical UniversityGanzhouChina
| | - Peiting Huang
- Department of Neurology, Guangdong Provincial People's HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jinyao Zhou
- Department of NeurosurgeryDongguan Tungwah HospitalDongguanChina
| | - Yongfu Cao
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jing Guo
- Epilepsy CenterGuangdong Sanjiu Brain HospitalGuangzhouChina
| | - Yawei Liu
- Department of Neurosurgery & Medical Research Center, Shunde HospitalSouthern Medical UniversityShundeChina
| | - Songtao Qi
- Department of Neurosurgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain DiseasesNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Hai Wang
- Department of Neurosurgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain DiseasesNanfang Hospital of Southern Medical UniversityGuangzhouChina
| |
Collapse
|
369
|
McAuley N, Cymer I, McAvera R, Hopkins AM, Glavey SV. Chromosome 1 Alterations in Multiple Myeloma: Considerations for Precision Therapy. Eur J Haematol 2025; 114:400-410. [PMID: 39632279 PMCID: PMC11798765 DOI: 10.1111/ejh.14352] [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: 09/25/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Multiple myeloma (MM) is an incurable blood malignancy characterized by the clonal expansion of plasma cells and the secretion of monoclonal immunoglobulins. High-risk MM, defined by specific cytogenetic abnormalities, poses significant therapeutic challenges and is associated with inferior survival outcomes compared to standard-risk disease. Although molecularly targeted therapies have shown efficacy in other hematologic malignancies, currently venetoclax is the only targeted therapy approved for MM (t(11;14)). However, chromosome 1q gains, amplifications, and 1p deletions are frequently observed in MM, and have been linked to drug resistance and poor patient prognosis. Accordingly, this review focuses on emerging MM precision therapies capable of targeting dysregulated genes within these regions. It addresses gene therapies, small molecule inhibitors and monoclonal antibodies currently under investigation to antagonize oncogenic drivers including MCL-1, BCL9, F11R, and CKS1B, all of which are implicated in cell survival, proliferation or drug resistance. In conclusion, the link between chromosome 1 abnormalities and high-risk disease in MM patients offers a compelling rationale to identify and explore therapeutic targeting of chromosome 1 gene products as a novel precision medicine approach for a poorly served patient population.
Collapse
Affiliation(s)
- Niamh McAuley
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Izabela Cymer
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Roisin McAvera
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Ann M. Hopkins
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Siobhan V. Glavey
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of HaematologyBeaumont RCSI Cancer CentreDublinIreland
| |
Collapse
|
370
|
Gamba G, Colonetti T, Uggioni MLR, Elibio LU, Balbinot EL, Heinzen R, Macedo ACL, Grande AJ, da Rosa MI. Gut microbiota and breast cancer: systematic review and meta-analysis. Breast Cancer 2025; 32:242-257. [PMID: 39652259 DOI: 10.1007/s12282-024-01658-3] [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: 09/09/2024] [Accepted: 11/27/2024] [Indexed: 02/21/2025]
Abstract
BACKGROUND The gastrointestinal microbiota can modulate systemic estrogens, potentially influencing estrogen-induced breast neoplasia development. This study aimed to assess alterations in the gut microbiota in breast cancer patients. METHODS A search strategy was developed using the terms: "Microbiota," "Gastrointestinal Microbiome," "Breast Cancer," and synonyms. Ten observational studies were included. RESULTS The total sample was 1730 women (929 cases and 801 controls). The meta-analysis of alpha diversity, assessed by the Shannon index, displayed that in the breast cancer group, the diversity of the gut microbiota was reduced compared to controls, with a standardized mean difference (SMD) of - 0.34 (95% CI - 0.59, - 0.10, I2 = 68%, p = 0.007). Regarding the premenopausal population, there was a significant reduction in the breast cancer group (SMD - 0.67, 95% CI - 1.06, - 0.28, I2 = 77%, p = 0.0009). In women with a body mass index (BMI) between overweight or obesity, no statistically significant difference was observed (SMD - 0.20; 95% CI - 0.51, 0.11; I2 52%, p = 0.20). However, in women with a BMI greater than or equal to 18.5 and less than 25.0, there was lower diversity in women with breast cancer compared to controls (SMD - 0.49, 95% CI - 0.94, - 0.04; I2 78%, p = 0.03). CONCLUSIONS The study found a significant difference in gut microbiota diversity between women with breast cancer and controls, supporting the growing evidence that the gut microbiota may play a role in mammary carcinogenesis.
Collapse
Affiliation(s)
- Guilherme Gamba
- Laboratory of Translational Biomedicine, Universidade do Extremo Sul Catarinense, Criciuma, SC, 88806-000, Brazil
| | - Tamy Colonetti
- Laboratory of Translational Biomedicine, Universidade do Extremo Sul Catarinense, Criciuma, SC, 88806-000, Brazil
| | | | - Laura Uggioni Elibio
- Laboratory of Translational Biomedicine, Universidade do Extremo Sul Catarinense, Criciuma, SC, 88806-000, Brazil
| | - Eduarda Letícia Balbinot
- Laboratory of Translational Biomedicine, Universidade do Extremo Sul Catarinense, Criciuma, SC, 88806-000, Brazil
| | - Rebeca Heinzen
- Universidade do Sul de Santa Catarina, Florianópolis, SC, Brazil
| | - Ana Cristina Lacerda Macedo
- Laboratory of Translational Biomedicine, Universidade do Extremo Sul Catarinense, Criciuma, SC, 88806-000, Brazil
| | - Antonio José Grande
- Laboratory of Evidence-Based Practice, Universidade Estadual do Mato Grosso do Sul, Dourados, MS, Brazil
| | - Maria Inês da Rosa
- Laboratory of Translational Biomedicine, Universidade do Extremo Sul Catarinense, Criciuma, SC, 88806-000, Brazil.
| |
Collapse
|
371
|
Dhanasekaran R, Suzuki H, Lemaitre L, Kubota N, Hoshida Y. Molecular and immune landscape of hepatocellular carcinoma to guide therapeutic decision-making. Hepatology 2025; 81:1038-1057. [PMID: 37300379 PMCID: PMC10713867 DOI: 10.1097/hep.0000000000000513] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
Liver cancer, primarily HCC, exhibits highly heterogeneous histological and molecular aberrations across tumors and within individual tumor nodules. Such intertumor and intratumor heterogeneities may lead to diversity in the natural history of disease progression and various clinical disparities across the patients. Recently developed multimodality, single-cell, and spatial omics profiling technologies have enabled interrogation of the intertumor/intratumor heterogeneity in the cancer cells and the tumor immune microenvironment. These features may influence the natural history and efficacy of emerging therapies targeting novel molecular and immune pathways, some of which had been deemed undruggable. Thus, comprehensive characterization of the heterogeneities at various levels may facilitate the discovery of biomarkers that enable personalized and rational treatment decisions, and optimize treatment efficacy while minimizing the risk of adverse effects. Such companion biomarkers will also refine HCC treatment algorithms across disease stages for cost-effective patient management by optimizing the allocation of limited medical resources. Despite this promise, the complexity of the intertumor/intratumor heterogeneity and ever-expanding inventory of therapeutic agents and regimens have made clinical evaluation and translation of biomarkers increasingly challenging. To address this issue, novel clinical trial designs have been proposed and incorporated into recent studies. In this review, we discuss the latest findings in the molecular and immune landscape of HCC for their potential and utility as biomarkers, the framework of evaluation and clinical application of predictive/prognostic biomarkers, and ongoing biomarker-guided therapeutic clinical trials. These new developments may revolutionize patient care and substantially impact the still dismal HCC mortality.
Collapse
Affiliation(s)
| | - Hiroyuki Suzuki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka
| | - Lea Lemaitre
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California
| | - Naoto Kubota
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
372
|
Wang F, Wei X, Zheng Y, Wang J, Ying J, Chen X, Luo S, Luo H, Yu X, Chen B, Ma L, Xu R. Safety, Pharmacokinetics, and Pharmacodynamics Evaluation of Ivonescimab, a Novel Bispecific Antibody Targeting PD-1 and VEGF, in Chinese Patients With Advanced Solid Tumors. Cancer Med 2025; 14:e70653. [PMID: 40114411 PMCID: PMC11925807 DOI: 10.1002/cam4.70653] [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: 05/06/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Ivonescimab (AK112) is a first-in-class bispecific antibody that simultaneously targets programmed death-1 (PD-1) and vascular endothelial growth factor (VEGF) with cooperative binding. We report the safety, pharmacokinetics (PK), and pharmacodynamics (PD) profiles of ivonescimab in patients suffered from advanced solid tumors. METHODS A multicenter, open-label, dose-escalation, phase I study was conducted in five hospitals in China. Ivonescimab was used as a monotherapy. The dose of ivonescimab intravenously administered was 3, 5, 10, 20, and 30 mg/kg every 2 weeks (Q2W), and 10 and 20 mg/kg every 3 weeks (Q3W). Safety, PK, and PD of ivonescimab were evaluated. RESULTS A total of 59 patients treated in the study. Only one dose-limiting toxicity (DLT) occurred in 1 out of 9 patients in the 10 mg/kg Q2W cohort, indicating that no maximum tolerated dose was reached. Among the participants, 53 patients (89.8%) experienced treatment-related adverse events (TRAEs), with the most common being proteinuria (33.9%), aspartate aminotransferase elevation (27.1%), white blood cell count decrease (22.0%), alanine aminotransferase elevation (20.3%), and anemia (20.3%). Fourteen patients (23.7%) had ≥ Grade 3 TRAEs, and 7 patients (11.9%) experienced serious TRAEs. Notably, there were no reported deaths associated with the TRAEs, and no dose-dependent increase in adverse events was observed. The half-life of ivonescimab ranged from 5.0 to 7.3 days following single-dose administration across all dose levels. The serum concentrations of ivonescimab increased with escalating doses in an approximately dose-proportional manner. Following multiple doses, the accumulation ratio ranged from 1.1 to 1.7, suggesting mild accumulation of ivonescimab. The steady state was achieved after 5 doses. Ivonescimab occupancy on PD-1 sustained over 80% across the treatment period. Serum VEGF level was rapidly down-regulated after each administration. CONCLUSIONS In patients with advanced solid tumors, ivonescimab monotherapy was well-tolerated and demonstrated a linear PK characteristics. PD profiles showed the promising potential of ivonescimab for the management of advanced solid tumors. TRIAL REGISTRATION ClinicalTrials.gov (NCT04597541).
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Neoplasms/drug therapy
- Neoplasms/pathology
- Aged
- Antibodies, Bispecific/pharmacokinetics
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/therapeutic use
- Adult
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- China
- Maximum Tolerated Dose
- Dose-Response Relationship, Drug
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/pharmacology
- Immune Checkpoint Inhibitors/pharmacokinetics
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/administration & dosage
- Immune Checkpoint Inhibitors/pharmacology
- East Asian People
Collapse
Affiliation(s)
- Fenghua Wang
- Department of Medical Oncology, Cancer Prevention Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaoli Wei
- Department of Medical Oncology, Cancer Prevention Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yulong Zheng
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Jing Wang
- Department of Gynecology, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Jieer Ying
- Department of Hepatobiliary Pancreatic Gastric Medicine, Zhejiang Cancer Hospital, Zhejiang, Hangzhou, China
| | - Xiaozhong Chen
- The Department of Head and Neck Radiation, Zhejiang Cancer Hospital, Zhejiang, Hangzhou, China
| | - Suxia Luo
- Department of Medical Oncology, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Huiyan Luo
- Department of Medical Oncology, Cancer Prevention Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xufang Yu
- Akeso Biopharma, Inc., Zhongshan, Guangdong, China
| | - Benchao Chen
- Akeso Biopharma, Inc., Zhongshan, Guangdong, China
| | - Lei Ma
- Akeso Biopharma, Inc., Zhongshan, Guangdong, China
| | - Ruihua Xu
- Department of Medical Oncology, Cancer Prevention Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
373
|
Puente-Cobacho B, Esteo C, Altea-Manzano P, Garcia-Perez JL, Quiles JL, Sanchez-Rovira P, Martín-Salvago MD, Molina-Jiménez L, Luque RJ, Fendt SM, Vera-Ramirez L. De novo lipogenesis protects dormant breast cancer cells from ferroptosis and promotes metastasis. Redox Biol 2025; 80:103480. [PMID: 39787900 PMCID: PMC11764609 DOI: 10.1016/j.redox.2024.103480] [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: 11/10/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
Dormant disseminated tumor cells (DTCs) remain viable for years to decades before establishing a clinically overt metastatic lesion. DTCs are known to be highly resilient and able to overcome the multiple biological hurdles imposed along the metastatic cascade. However, the specific metabolic adaptations of dormant DTCs remain to be elucidated. Here, we reveal that dormant DTCs upregulate de novo lipogenesis and favor the activation and incorporation of monounsaturated fatty acids (MUFAs) to their cellular membranes through the activation of acyl-coenzyme A synthetase long-chain family member 3 (ACSL3). Pharmacologic inhibition of de novo lipogenesis or genetic knockdown of ACSL3 results in lipid peroxidation and non-apoptotic cell death through ferroptosis. Clinically, ACSL3 was found to be overexpressed in quiescent DTCs in the lymph nodes of breast cancer patients and to significantly correlate with shorter disease-free and overall survival. Our work provides new insights into the molecular mechanisms enabling the survival of dormant DTCs and supports the use of de novo lipogenesis inhibitors to prevent breast cancer metastasis.
Collapse
Affiliation(s)
- Beatriz Puente-Cobacho
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Cintia Esteo
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Patricia Altea-Manzano
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jose Luis Garcia-Perez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | | | | | | | - Rafael J Luque
- Pathological Anatomy Unit, University Hospital of Jaén, Jaén, Spain
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Laura Vera-Ramirez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain; Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain.
| |
Collapse
|
374
|
Li J, Huang K, Thakur M, McBride F, Sadagopan A, Gallant DS, Khanna P, Laimon YN, Li B, Mohanna R, Ge M, Weiss CN, Achom M, Xu Q, Matar S, Lee GSM, Huang K, Gui M, Wu CL, Cornejo KM, Choueiri TK, Ryback BA, Signoretti S, Bar-Peled L, Viswanathan SR. Oncogenic TFE3 fusions drive OXPHOS and confer metabolic vulnerabilities in translocation renal cell carcinoma. Nat Metab 2025; 7:478-492. [PMID: 39915638 DOI: 10.1038/s42255-025-01218-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/09/2025] [Indexed: 02/12/2025]
Abstract
Translocation renal cell carcinoma (tRCC) is an aggressive subtype of kidney cancer driven by TFE3 gene fusions, which act via poorly characterized downstream mechanisms. Here we report that TFE3 fusions transcriptionally rewire tRCCs toward oxidative phosphorylation (OXPHOS), contrasting with the highly glycolytic nature of most other renal cancers. Reliance on this TFE3 fusion-driven OXPHOS programme renders tRCCs vulnerable to NADH reductive stress, a metabolic stress induced by an imbalance of reducing equivalents. Genome-scale CRISPR screening identifies tRCC-selective vulnerabilities linked to this metabolic state, including EGLN1, which hydroxylates HIF-1α and targets it for proteolysis. Inhibition of EGLN1 compromises tRCC cell growth by stabilizing HIF-1α and promoting metabolic reprogramming away from OXPHOS, thus representing a vulnerability for OXPHOS-dependent tRCC cells. Our study defines tRCC as being dependent on a mitochondria-centred metabolic programme driven by TFE3 fusions and nominates EGLN1 inhibition as a therapeutic strategy in this cancer.
Collapse
Affiliation(s)
- Jiao Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kaimeng Huang
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Meha Thakur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fiona McBride
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ananthan Sadagopan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Daniel S Gallant
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Prateek Khanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Bingchen Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Razan Mohanna
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Maolin Ge
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Cary N Weiss
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mingkee Achom
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Qingru Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sayed Matar
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kun Huang
- Molecular Imaging Core and Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Miao Gui
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine and Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Kristine M Cornejo
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Birgitta A Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Liron Bar-Peled
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
| | - Srinivas R Viswanathan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
375
|
Huang Y, Zhou X, Liu J, Cao Y, Fu W, Yang J. Emerging neuroimmune mechanisms in cancer neuroscience. Cancer Lett 2025; 612:217492. [PMID: 39848532 DOI: 10.1016/j.canlet.2025.217492] [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: 12/21/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
It has become increasingly recognized that neural signals can profoundly influence the prognosis of various cancer types. In the past years, we have witnessed "cancer neuroscience," which primarily focuses on the complex crosstalk between tumors and neural signals, emerging as a new, multidisciplinary direction of biomedical science. This review aims to summarize the current knowledge of this research frontier, with an emphasis on the neuroimmune mechanisms enacted through the reciprocal interactions between tumors and the central or peripheral nervous system. In addition, we wish to highlight several key questions of cancer neuroscience and its neuroimmune action that warrant future research and translational efforts, including novel strategies for manipulating neural signals for antitumor immunotherapies, as well as managing cancer-related neurological or psychiatric complications.
Collapse
Affiliation(s)
- Yingying Huang
- School of Life Sciences, Peking University Third Hospital Cancer Center, Center for Life Sciences, State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xin Zhou
- School of Life Sciences, Peking University Third Hospital Cancer Center, Center for Life Sciences, State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China; Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Jiaqi Liu
- School of Life Sciences, Peking University Third Hospital Cancer Center, Center for Life Sciences, State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Ying Cao
- School of Life Sciences, Peking University Third Hospital Cancer Center, Center for Life Sciences, State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| | - Wei Fu
- School of Life Sciences, Peking University Third Hospital Cancer Center, Center for Life Sciences, State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China; Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China.
| | - Jing Yang
- School of Life Sciences, Peking University Third Hospital Cancer Center, Center for Life Sciences, State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China; Peking Union Medical College Hospital, Beijing, 100730, China.
| |
Collapse
|
376
|
Shen Z, Zhao Y, Xu X, Yang H, He S, Ma J, Zhang S, Hou P, Sui F. Single-cell RNA sequencing integrated with bulk RNA sequencing analysis of clock circadian regulator with prognostic and immune microenvironment in thyroid cancer. Transl Oncol 2025; 53:102299. [PMID: 39892222 PMCID: PMC11833347 DOI: 10.1016/j.tranon.2025.102299] [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: 06/26/2024] [Revised: 01/05/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Disruption of circadian rhythm was found to be associated with immune infiltration and thyroid cancer. However, the role of clock circadian regulator (CLOCK) in the progression of thyroid cancer and its immune microenvironment remains largely unexplored. Therefore, our aim was to explore the role and potential mechanism of CLOCK in thyroid cancer. METHODS Single cell sequencing analysis and bulk RNA sequencing analysis was used for LASSO regression and Kaplan-Meier survival estimates. Potential mechanism analysis were gained through KEGG/GO analysis, GSEA analysis and PPI network. In vivo and in vitro experiment was used for further validation. RESULTS The result showed CLOCK protein was overexpressed in thyroid cancer compared with normal tissue in both thyroid specific mouse model and human sample. A prognostic model incorporating CLOCK and other related genes (FAT4, OR6K2, STK40, TMEM63A, HRCT1, SUPT5H, and OR2C3) was developed using LASSO regression. Functional assay and bioinformatics analysis indicated that CLOCK knockdown hindered tumor growth and the activity of MAPK signaling. Besides, analyses of gene enrichment, signaling pathways, and immune checkpoints suggested that CLOCK might inhibit immune infiltration within the tumor microenvironment. Confirmatory in vitro experiments and immunohistochemical assays in human samples further linked high CLOCK expression to reduced T cell cytotoxicity and infiltration. CONCLUSION These findings underscore the pivotal role of CLOCK in thyroid cancer prognosis and immune suppression, highlighting its potential as a target for therapeutic intervention and prognostic assessment in thyroid cancer management.
Collapse
Affiliation(s)
- Zhen Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Yuelei Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Xinxin Xu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Huini Yang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Shuting He
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Junchi Ma
- School of Information Engineering, Chang'an University, Shaanxi Province, PR China
| | - Shaoqiang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China; International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, Xi'an 710061, Shaanxi Province, PR China
| | - Fang Sui
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China.
| |
Collapse
|
377
|
Keane L, Cryan JF, Gleeson JP. Exploiting the gut microbiome for brain tumour treatment. Trends Mol Med 2025; 31:213-223. [PMID: 39256110 DOI: 10.1016/j.molmed.2024.08.008] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Increasing evidence suggests that the gut microbiome plays a key role in a host of pathological conditions, including cancer. Indeed, the bidirectional communication that occurs between the gut and the brain, known as the 'gut-brain axis,' has recently been implicated in brain tumour pathology. Here, we focus on current research that supports a gut microbiome-brain tumour link with emphasis on high-grade gliomas, the most aggressive of all brain tumours, and the impact on the glioma tumour microenvironment. We discuss the potential use of gut-brain axis signals to improve responses to current and future therapeutic approaches. We highlight that the success of novel treatment strategies may rely on patient-specific microbiome profiles, and these should be considered for personalised treatment approaches.
Collapse
Affiliation(s)
- Lily Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Jack P Gleeson
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland; CUH/UCC Cancer Centre, Cork University Hospital, Cork, Ireland.
| |
Collapse
|
378
|
Li M, Lopez‐Aguiar AG, Poultsides G, Rocha F, Weber S, Fields R, Idrees K, Cho C, Maithel SK, Zhang X, Pawlik TM. Preoperative Glycosylated Hemoglobin A1C Impacts Long-Term Outcomes Following Curative-Intent Resection of Nonfunctional Gastroenteropancreatic Neuroendocrine Tumors. J Surg Oncol 2025; 131:473-480. [PMID: 39387595 PMCID: PMC12044285 DOI: 10.1002/jso.27951] [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: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND To investigate the impact of preoperative glycosylated hemoglobin A1C (HbA1c) among patients following curative-intent resection of nonfunctional gastroentropancreatic neuroendocrine tumors (GEP-NETs). METHODS Patients who underwent curative-intent resection for GEP-NETs from 2000 to 2020 were identified from the US Neuroendocrine Tumor Study Group (US-NETSG). Preoperative blood HbA1c levels were defined as high HbA1c (≥ 6.5%) versus low HbA1c group (< 6.5%). Impact of HbA1c level on postoperative short-term and long-term overall (OS) were investigated. RESULTS A total of 130 patients with HbA1c < 6.5% and 60 patients with HbA1c ≥ 6.5% were included. Patients with HbA1c ≥ 6.5% had higher proportion of comorbidities, such as hypertension, obesity, anemia, and lower preoperative albumin levels versus patients with HbA1c < 6.5% (all p < 0.05). In addition, high level of preoperative HbA1c was associated with increased incidence of wound and infectious complications, as well as decreased long-term OS (median OS: high Hb1Ac 89.8 months vs. low Hb1Ac not reached, HR 3.487, p = 0.004) among patients with nonfunctional GEP-NETs, as well as among the subset of pancreatic NET patients (median OS: high Hb1Ac 74.3 months vs. low Hb1Ac not reached, p = 0.004), and patients with normal fasting blood glucose (< 140 mg/dL) (median OS: high Hb1Ac 75.4 months vs. low Hb1Ac not reached, p = 0.001). CONCLUSIONS Hb1Ac might have value as a screening tool to identify high-risk patients following surgical resection of nonfunctional GEP-NETs for consideration of more strict postoperative surveillance and treatment of elevated Hb1Ac level.
Collapse
Affiliation(s)
- Mu‐Xing Li
- Department of Hepatobiliary Surgery, Institute of Advanced Surgical Technology and EngineeringThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Department of General SurgeryPeking University Third HospitalBeijingChina
| | - Alexandra G. Lopez‐Aguiar
- Department of Surgery, Division of Surgical Oncology, Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | | | - Flavio Rocha
- Department of SurgeryVirginia Mason Medical CenterSeattleWashington
| | - Sharon Weber
- Department of SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Ryan Fields
- Department of SurgeryWashington University School of MedicineSt. LouisWisconsin
| | - Kamran Idrees
- Department of Surgery, Division of Surgical OncologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Cliff Cho
- Department of Surgery, Division of Hepatopancreatobiliary and Advanced Gastrointestinal SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Shishir K. Maithel
- Department of Surgery, Division of Surgical Oncology, Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Xu‐Feng Zhang
- Department of Hepatobiliary Surgery, Institute of Advanced Surgical Technology and EngineeringThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Division of Surgical OncologyThe Ohio State University Wexner Medical Center and James Comprehensive Cancer CenterColumbusOhioUSA
| | - Timothy M. Pawlik
- Division of Surgical OncologyThe Ohio State University Wexner Medical Center and James Comprehensive Cancer CenterColumbusOhioUSA
| |
Collapse
|
379
|
Zhao AR, Kouznetsova VL, Kesari S, Tsigelny IF. Machine-learning diagnostics of breast cancer using piRNA biomarkers. Biomarkers 2025; 30:167-177. [PMID: 39899375 DOI: 10.1080/1354750x.2025.2461067] [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: 12/02/2024] [Accepted: 01/25/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND AND OBJECTIVES Prior studies have shown that small non-coding RNAs (sncRNAs) are associated with cancer occurrence or development. Recently, a newly discovered class of small ncRNAs known as PIWI-interacting RNAs (piRNAs) have been found to play a vital role in physiological processes and cancer initiation. This study aims to utilize piRNAs as innovative, noninvasive diagnostic biomarkers for breast cancer. Our objective is to develop computational methods that leverage piRNA attributes for breast cancer prediction and its application in diagnostics. METHODS We created a set of piRNA sequence descriptors using information extracted from the piRNA sequences. To ensure accuracy, we found a path to convert non-standard piRNA names to standard ones to enable precise identification of these sequences. Using these descriptors, we applied machine-learning (ML) techniques in WEKA (Waikato Environment for Knowledge Analysis) to a dataset of piRNA to assess the predictive accuracy of the following classifiers: Logistic Regression model, Sequential Minimal Optimization (SMO), Random Forest classifier, and Logistic Model Tree (LMT). Furthermore, we performed Shapley additive explanations (SHAP) Analysis to understand which descriptors were the most relevant to the prediction accuracy. The ML models were then validated on an independent dataset to evaluate their effectiveness in predicting breast cancer. RESULTS The top three performing classifiers in WEKA were Logistic Regression, SMO, and LMT. The Logistic Regression model achieved an accuracy of 90.7% in predicting breast cancer, while SMO and LMT attained 89.7% and 85.65%, respectively. CONCLUSIONS Our study demonstrates the effectiveness of using ML-based piRNA classifiers in diagnosing breast cancer and contributes to the growing body of evidence supporting piRNAs as biomarkers in cancer diagnosis. However, additional research is needed to validate these findings and further assess the clinical applicability of this approach.
Collapse
Affiliation(s)
- Amy R Zhao
- Scholars Program, CureScience Institute, San Diego, CA, USA
| | - Valentina L Kouznetsova
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- BIAna Institute, San Diego, CA, USA
- CureScience Institute, San Diego, CA, USA
| | - Santosh Kesari
- Department of Neuro-oncology, Pacific Neuroscience Institute, Santa Monica, CA, USA
| | - Igor F Tsigelny
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- BIAna Institute, San Diego, CA, USA
- CureScience Institute, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
380
|
Nakatsukasa Y, Yamada Y, Yamada Y. Research of in vivo reprogramming toward clinical applications in regenerative medicine: A concise review. Regen Ther 2025; 28:12-19. [PMID: 39678397 PMCID: PMC11638634 DOI: 10.1016/j.reth.2024.11.008] [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: 11/01/2024] [Accepted: 11/17/2024] [Indexed: 12/17/2024] Open
Abstract
The successful generation of induced pluripotent stem cells (iPSCs) has significantly impacted many scientific fields. In the field of regenerative medicine, iPSC-derived somatic cells are expected to recover impaired organ functions through cell transplantation therapy. Subsequent studies using genetically engineered mouse models showed that somatic cells are also reprogrammable in vivo. Notably, cyclic expression of reprogramming factors, so-called partial reprogramming in vivo ameliorates cellular and physiological hallmarks of aging without inducing teratoma formation or premature death of animals. Subsequent studies provided evidence supporting the beneficial effects of partial reprogramming in various organs. Although in vivo reprogramming appears to be a promising strategy for tissue regeneration and rejuvenation, there remain unsolved issues that hinder its clinical application, including concerns regarding its safety, controllability, and unexpected detrimental effects. Here, we review the pathway that research of in vivo reprogramming has followed and discuss the future perspective as we look toward its clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Yoshihiko Nakatsukasa
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Yamada
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Yamada
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
381
|
Tressler CM, Sonkar K, Cheng M, Ayyappan V, Cai R, Glunde K. Molecular effects of clinically relevant chemotherapeutic agents on choline phospholipid metabolism in triple negative breast cancer cells. Transl Oncol 2025; 53:102311. [PMID: 39922048 PMCID: PMC11849126 DOI: 10.1016/j.tranon.2025.102311] [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: 08/25/2024] [Revised: 12/21/2024] [Accepted: 01/30/2025] [Indexed: 02/10/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most lethal breast cancer subtype, leading to poor patient outcomes despite aggressive treatment with surgery, radiation, and chemotherapy. There are currently no clinical tests available which measure early on whether TNBC patients respond to the selected chemotherapy treatment regimen. The magnetic resonance spectroscopy (MRS)-detected total choline (tCho) signal was shown to be a promising biomarker for assessing the response to chemotherapy treatment early on, as breast tumor tCho decreases after the first treatment cycle in patients who respond to chemotherapy cocktails. We sought to further investigate these clinical observations at the molecular level by combining metabolic and transcriptomic studies in two human TNBC cell lines treated with six different chemotherapeutic agents. Overall, our findings show that the glycerophosphocholine-to-phosphocholine ratio (GPC/PC) was a more sensitive and more broadly applicable measure of TNBC response to various chemotherapeutic agents than tCho. Specific chemotherapeutic drugs, including 5-fluorouracil and melphalan, resulted in the most significant effects on choline phospholipid metabolism, while other drugs did not significantly alter choline phospholipid metabolism. Overall, several of the six tested chemotherapeutic drugs mainly affected the expression levels of phosphatidylcholine (PtdC)-specific phospholipases and lysophospholipases, leading to the observed GPC/PC and tCho changes following treatment with the chemotherapeutic agents that altered choline phospholipid metabolism. The presented metabolic and transcriptomic findings support that the GPC/PC ratio and PtdC-phospholipases and -lysophospholipases could be further developed for assessing the response to chemotherapy treatment in TNBC patients. Statement of Significance: We show that the glycerophosphocholine-to-phosphocholine ratio and phosphatidylcholine-specific-phospholipases and -lysophospholipases are reliable markers for assessing the response to several chemotherapeutic agents, which could help with selecting correct treatments for TNBC patients.
Collapse
Affiliation(s)
- Caitlin M Tressler
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kanchan Sonkar
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Menglin Cheng
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vinay Ayyappan
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruoqing Cai
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Glunde
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
382
|
Xia RX, Zou PC, Xie JT, Tang YB, Gong MM, Fan F, Aihemaiti A, Liu YQ, Shen Y, Zhou BBS, Zhu L, Lei HM. Dependence of NPPS creates a targetable vulnerability in RAS-mutant cancers. Acta Pharmacol Sin 2025; 46:728-739. [PMID: 39506063 PMCID: PMC11845791 DOI: 10.1038/s41401-024-01409-2] [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: 07/11/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
RAS is the most frequently mutated oncoprotein for cancer driving. Understanding of RAS biology and discovery of druggable lynchpins in RAS pathway is a prerequisite for targeted therapy of RAS-mutant cancers. The recent identification of KRASG12C inhibitor breaks the "undruggable" curse on RAS and has changed the therapy paradigm of KRAS-mutant cancers. However, KRAS mutations, let alone KRASG12C mutation, account for only part of RAS-mutated cancers. Targeted therapies for cancers harboring other RAS mutations remain the urgent need. In this study we explored the pivotal regulatory molecules that allow for broad inhibition of RAS mutants. By comparing the expression levels of nucleotide pyrophosphatase (NPPS) in a panel of cell lines and the functional consequence of increased NPPS expression in RAS-mutant cells, we demonstrated that cancer cells with various kinds of RAS mutations depended on NPPS for growth and survival, and that this dependence conferred a vulnerability of RAS-mutant cancer to treatment of NPPS inhibition. RAS-mutant cells, compared with RAS-wildtype cells, bored and required an upregulation of NPPS. Transcriptomics and metabolomics analyses revealed a NPPS-dependent hyperglycolysis in RAS-mutant cells. We demonstrated that NPPS promoted glucose-derived glycolytic intermediates in RAS-mutant cells by enhancing its interaction with hexokinase 1 (HK1), the enzyme catalyzing the first committed step of glycolysis. Pharmacological inhibition of NPPS-HK1 axis using NPPS inhibitor Enpp-1-IN-1 or HK1 inhibitor 2-deoxyglucose (2-DG), or genetic interfere with NPPS suppressed RAS-mutant cancers in vitro and in vivo. In conclusion, this study reveals an unrecognized mechanism and druggable lynchpin for modulation of pan-mutant-RAS pathway, proposing a new potential therapeutic approach for treating RAS-mutant cancers.
Collapse
Affiliation(s)
- Rui-Xue Xia
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Pei-Chen Zou
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jun-Ting Xie
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ya-Bin Tang
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Miao-Miao Gong
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fu Fan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ayinazhaer Aihemaiti
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu-Qing Liu
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bin-Bing S Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui-Min Lei
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
383
|
Lehtonen E, Vertanen M, Syvälä H, Tolonen T, Auriola S, Tammela T, Siltari A, Murtola T. The Impact of Atorvastatin on Intraprostatic Biomarkers - Prognostic Value of 3LS-score - Follow-up of ESTO1-Trial. Neoplasia 2025; 61:101132. [PMID: 39919693 PMCID: PMC11851191 DOI: 10.1016/j.neo.2025.101132] [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: 06/05/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Prostate cancer (PCa) remains a global health burden, with limited reliable biomarkers beyond prostate-specific antigen (PSA). Statins have been associated with survival benefits in advanced Pca, potentially by modulating cholesterol metabolism and tumor biology. However, the causal mechanisms are not well understood. A distinct three-lipid signature (3LS) has previously been proposed as a prognostic biomarker for PCa. OBJECTIVE This study investigates the effects of atorvastatin intervention on PCa tissue markers, long-term clinical outcomes, and the prognostic value of the 3LS derived from prostate tissue lipidome. METHODS The ESTO1 trial randomized 158 statin-naïve PCa patients to receive high-dose atorvastatin (80 mg daily) or placebo before prostatectomy. Long term outcomes were assessed for 102 patients through medical records review. Prostate tissue samples were pathologically characterized, and lipidome quantified. Cox regression models were used to analyse clinical outcomes between the groups. The 3LS score was calculated by identifying the constituent lipids from the prostate lipidome. FINDINGS Higher intraprostatic atorvastatin lactone concentrations were associated with reduced Ki67 expression and PSA levels. After a median follow-up of seven years, no significant differences were observed in biochemical recurrence, overall mortality, or initiation of hormonal therapy. However, the atorvastatin arm had a lower risk of major acute cardiovascular events (HR 0.11, 95% CI 0.01-1.01). The intraprostatic 3LS correlated with higher baseline tumor aggressiveness but did not predict subsequent outcomes. CONCLUSION Higher atorvastatin lactone concentrations in the prostate tissue were linked to improved pathological variables. Pre-surgery statin intervention reduced MACE risk but no impact on other clinical outcomes was observed. The 3LS from prostate tissue does not seem to be prognostic marker in localized Pca.
Collapse
Affiliation(s)
- Eemil Lehtonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maiju Vertanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Heimo Syvälä
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Teemu Tolonen
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland; Department of Pathology, FimLab Laboratories, Tampere, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Teuvo Tammela
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland; TAYS Cancer Center, Department of Urology, Tampere, Finland
| | - Aino Siltari
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Teemu Murtola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; TAYS Cancer Center, Department of Urology, Tampere, Finland.
| |
Collapse
|
384
|
Zhan Y, Sun D, Gao J, Gao Q, Lv Y, Du T, Dong Y, Wang Y, Zhan H, Li J, Li P, Du L, Wang C. Single-cell transcriptomics reveals intratumor heterogeneity and the potential roles of cancer stem cells and myCAFs in colorectal cancer liver metastasis and recurrence. Cancer Lett 2025; 612:217452. [PMID: 39805388 DOI: 10.1016/j.canlet.2025.217452] [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: 07/05/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Metastasis and recurrence are the primary obstacles to long-term survival in colorectal cancer (CRC) patients. In this study, we employed single-cell RNA sequencing (scRNA-seq) to comprehensively delineate the transcriptomic landscape of primary and liver metastatic CRCs, and revealed novel cellular crosstalk between cancer cell subpopulation and myofibroblastic CAFs (myCAFs) at single-cell resolution. We identified a cancer cell subpopulation termed stem/transient amplifying-like (stem/TA-like) cells, which expressed genes associated with stem cell-like characteristics and metastatic potential. MyCAFs in their microenvironment showed the potential to remodel the extracellular matrix (ECM), regulate angiogenesis, and support a pro-metastatic microenvironment through paracrine signaling involving FN1, BGN, and other ECM components. Notably, we found that they may communicate through the ligand-receptor pairs FN1-CD44 and GDF15-TGFBR2, which may be linked to the liver metastatic process. Additionally, our findings suggest that both stem/TA-like cells and myCAFs could be involved in CRC recurrence following chemotherapy. A unique gene signature generated using the gene expression characteristics of stem/TA-like cells and myCAFs (SM signature) can be used to assess recurrence risk in CRC patients. Collectively, these findings highlight the intratumor heterogeneity and the potential roles of cancer stem cells and myCAFs in CRC liver metastasis and recurrence, providing new targets and insights for the prognostic assessment of CRC patients and the improved selection of effective treatment options.
Collapse
Affiliation(s)
- Yao Zhan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Dong Sun
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jie Gao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Qinglun Gao
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250031, Shandong, China
| | - Yanfeng Lv
- Department of Colorectal & Anal Surgery, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Tiantian Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Yaqi Dong
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hanxiang Zhan
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China.
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China.
| | - Lutao Du
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, 250012, Shandong, China.
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, 250033, Shandong, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, 250033, Shandong, China.
| |
Collapse
|
385
|
Tao J, Liu Y, Tang X, Nie D, Wu K, Wang K, Tang N. Hypoxia reduces SLC27A5 to promote hepatocellular carcinoma proliferation by repressing HNF4A. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119916. [PMID: 39938688 DOI: 10.1016/j.bbamcr.2025.119916] [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: 09/11/2024] [Revised: 01/10/2025] [Accepted: 02/02/2025] [Indexed: 02/14/2025]
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related mortality globally, with hypoxia recognized as a key factor in its progression. Solute carrier family 27 member 5 (SLC27A5/FATP5), a pivotal enzyme in hepatic fatty acid transport and bile acid metabolism, is frequently downregulated in hepatocellular carcinoma, resulting in poor prognosis. However, the link between hypoxia and the suppression of SLC27A5 in HCC remains to be elucidated. Here, we investigated the hypoxia-induced downregulation of SLC27A5 and its impact on HCC proliferation via the repression of hepatocyte nuclear factor 4 alpha (HNF4A). Utilizing in vitro and in vivo hepatocellular carcinoma models, we have demonstrated that hypoxic conditions significantly reduce SLC27A5 transcription, which is mediated by the suppression of HNF4A. This reduction leads to the activation of the AKT pathway and an increase in cyclin-dependent kinase 2 (CDK2) and Cyclin E1 (CCNE1) expression, promoting the transition from the G1 to S phase of the cell cycle and driving HCC proliferation. Furthermore, we show that the pharmacological activation of HNF4A using Benfluorex, in combination with the AKT inhibitor MK2206, significantly inhibits tumor growth in a subcutaneous MHCC-97H xenograft model, suggesting a synergistic therapeutic potential. Together, our study provides novel insights into the hypoxia-mediated regulatory mechanisms in HCC and highlights the HNF4A/SLC27A5/AKT axis as a promising target for combination therapy.
Collapse
Affiliation(s)
- Junji Tao
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yuanyuan Liu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xin Tang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Dan Nie
- Department of Gastroenterology, The Chongqing Hospital of Traditional Chinese Medicine, Chongqing College of Traditional Chinese Medicine, Chongqing 400011, China
| | - Kang Wu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Kai Wang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China.
| | - Ni Tang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
386
|
El-Ashmawy NE, Khedr EG, Abo-Saif MA, Hamouda SM. Cuproptosis regulation by long noncoding RNAs: Mechanistic insights and clinical implications in cancer. Arch Biochem Biophys 2025; 765:110324. [PMID: 39900259 DOI: 10.1016/j.abb.2025.110324] [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: 12/13/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Although survival rates have been improved in recent years, the prognosis of many cancer types remains inadequate, mostly owing to treatment resistance. Moreover, there is a continued need for exploring novel and reliable tumor markers to achieve accurate diagnosis. Understanding the molecular complexity of cancer allows for the development of more effective and personalized treatments and facilitates the discovery of biomarkers that surpass traditional ones and assist in cancer diagnosis and monitoring disease progression and response to treatment. Recent studies exploring the complexity of cancer biology have identified a new form of cell death, known as cuproptosis, which is driven by the accumulation of copper and subsequent stress induced by dysregulation of copper homeostasis. Increased copper level enables cancer cells to maintain their accelerated growth rates and metastatic potential, yet these cells can evade cuproptosis. Long noncoding RNAs (lncRNAs) have been recognized for their pivotal role in different hallmarks of cancer, including resistance to cell death. They have been found to be implicated in controlling copper balance and cuproptosis. Besides, lncRNAs associated with cuproptosis pathway have demonstrated their potential as diagnostic and prognostic cancer biomarkers as well as indicators of treatment response. Our review aims to summarize recent studies focusing on the intricate relationship between lncRNAs and cuproptosis and explore the mechanisms by which lncRNAs can modulate copper homeostasis and regulate cuproptosis pathway. We also highlight recent discoveries concerning the role of cuproptosis-related lncRNAs in diagnosis, prognosis, and therapy of different types of cancer. By elucidating the significance of cuproptosis-related lncRNAs, this review provides insights into how these lncRNAs can be used to develop new therapeutic strategies to improve treatment outcomes.
Collapse
Affiliation(s)
- Nahla E El-Ashmawy
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk, Cairo, Postal Code: 11837, Egypt.
| | - Eman G Khedr
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt.
| | - Mariam A Abo-Saif
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt.
| | - Sara M Hamouda
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt.
| |
Collapse
|
387
|
Chang TG, Park S, Schäffer AA, Jiang P, Ruppin E. Hallmarks of artificial intelligence contributions to precision oncology. NATURE CANCER 2025; 6:417-431. [PMID: 40055572 PMCID: PMC11957836 DOI: 10.1038/s43018-025-00917-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/21/2025] [Indexed: 03/29/2025]
Abstract
The integration of artificial intelligence (AI) into oncology promises to revolutionize cancer care. In this Review, we discuss ten AI hallmarks in precision oncology, organized into three groups: (1) cancer prevention and diagnosis, encompassing cancer screening, detection and profiling; (2) optimizing current treatments, including patient outcome prediction, treatment planning and monitoring, clinical trial design and matching, and developing response biomarkers; and (3) advancing new treatments by identifying treatment combinations, discovering cancer vulnerabilities and designing drugs. We also survey AI applications in interventional clinical trials and address key challenges to broader clinical adoption of AI: data quality and quantity, model accuracy, clinical relevance and patient benefit, proposing actionable solutions for each.
Collapse
Affiliation(s)
- Tian-Gen Chang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Seongyong Park
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro A Schäffer
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
388
|
Safeguard repressor locks hepatocyte identity and blocks liver cancer. Nat Genet 2025; 57:494-495. [PMID: 39972215 DOI: 10.1038/s41588-025-02082-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
|
389
|
Chen L, Chen M, Xie Y, Zhang Y, Mo S, He Y, Liang T, Liao Y, Huang R, Huang G, Han C, Pham TTH. 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione mediates the effect of ROS-enhanced PI3K/Akt/mTOR pathway on autophagy in breast cancer. FEBS Open Bio 2025; 15:474-489. [PMID: 39648951 PMCID: PMC11891764 DOI: 10.1002/2211-5463.13940] [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: 04/21/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024] Open
Abstract
Several studies have suggested a potential antitumor effect of 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione (DMDD). To further understand the mechanism of action of this compound, we investigated its effect on the phosphatidylinositol-3-kinase (PI3K)/serine-threonine kinase (Akt)/mammalian target of rapamycin (mTOR) signaling pathway. We show that DMDD application significantly inhibited the proliferation of breast cancer cell lines MDA-MB-231 and ER-α positive MCF-7. Furthermore, DMDD application resulted in increased intracellular reactive oxygen species (ROS) levels, apoptosis and autophagy, whereas it downregulated the expression of PI3K, Akt and mTOR mRNA and proteins, and increased the expression of LC3II/I and p62 proteins. In a mouse breast cancer xenograft model, DMDD inhibited tumor growth. Expression analyses suggest that ROS levels were higher in DMDD treated tumor tissues, whereas immunohistochemical analyses suggest that apoptotic cells were more prevalent in the DMDD treated group compared to the control group. Taken together, our results suggest that the molecular mechanism of action of DMDD may involve the enhancement of breast cancer autophagy through the PI3K/Akt/mTOR signaling pathway by mediating ROS expression.
Collapse
Affiliation(s)
- Linqian Chen
- Guangxi Medical University School of PharmacyNanningChina
| | - Meifeng Chen
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Yan Xie
- Guangxi Medical University School of PharmacyNanningChina
| | - Yuyan Zhang
- Guilin Medical College School of PharmacyGuilinChina
| | - Shutian Mo
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Yongfei He
- Department of Hepatobiliary and Pancreatic SurgeryThe Eighth Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| | - Tianyi Liang
- The Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Yuan Liao
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Renbin Huang
- Guangxi Medical University School of PharmacyNanningChina
| | - Guodong Huang
- Zhuang & Yao Medicine Research and Development CenterGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Chuangye Han
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency TumorNanningChina
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal CancerNanningChina
| | - Thi Thai Hoa Pham
- Zhuang & Yao Medicine Research and Development CenterGuangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese MedicineNanningChina
| |
Collapse
|
390
|
Huang S, Shi J, Shen J, Fan X. Metabolic reprogramming of neutrophils in the tumor microenvironment: Emerging therapeutic targets. Cancer Lett 2025; 612:217466. [PMID: 39862916 DOI: 10.1016/j.canlet.2025.217466] [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: 09/09/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Neutrophils are pivotal in the immune system and have been recognized as significant contributors to cancer development and progression. These cells undergo metabolic reprogramming in response to various stimulus, including infections, diseases, and the tumor microenvironment (TME). Under normal conditions, neutrophils primarily rely on aerobic glucose metabolism for energy production. However, within the TME featured by hypoxic and nutrient-deprived conditions, they shift to altered anaerobic glycolysis, lipid metabolism, mitochondrial metabolism and amino acid metabolism to perform their immunosuppressive functions and facilitate tumor progression. Targeting neutrophils within the TME is a promising therapeutic approach. Yet, focusing on their metabolic pathways presents a novel strategy to enhance cancer immunotherapy. This review synthesizes the current understanding of neutrophil metabolic reprogramming in the TME, with an emphasis on the underlying molecular mechanisms and signaling pathways. Studying neutrophil metabolism in the TME poses challenges, such as their short lifespan and the metabolic complexity of the environment, necessitating the development of advanced research methodologies. This review also discusses emerging solutions to these challenges. In conclusion, given their integral role in the TME, targeting the metabolic pathways of neutrophils could offer a promising avenue for cancer therapy.
Collapse
Affiliation(s)
- Shiyun Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| |
Collapse
|
391
|
Lim B, Kamal A, Gomez Ramos B, Adrian Segarra JM, Ibarra IL, Dignas L, Kindinger T, Volz K, Rahbari M, Rahbari N, Poisel E, Kafetzopoulou K, Böse L, Breinig M, Heide D, Gallage S, Barragan Avila JE, Wiethoff H, Berest I, Schnabellehner S, Schneider M, Becker J, Helm D, Grimm D, Mäkinen T, Tschaharganeh DF, Heikenwalder M, Zaugg JB, Mall M. Active repression of cell fate plasticity by PROX1 safeguards hepatocyte identity and prevents liver tumorigenesis. Nat Genet 2025; 57:668-679. [PMID: 39948437 PMCID: PMC11906372 DOI: 10.1038/s41588-025-02081-w] [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: 10/08/2023] [Accepted: 01/08/2025] [Indexed: 02/20/2025]
Abstract
Cell fate plasticity enables development, yet unlocked plasticity is a cancer hallmark. While transcription master regulators induce lineage-specific genes to restrict plasticity, it remains unclear whether plasticity is actively suppressed by lineage-specific repressors. Here we computationally predict so-called safeguard repressors for 18 cell types that block phenotypic plasticity lifelong. We validated hepatocyte-specific candidates using reprogramming, revealing that prospero homeobox protein 1 (PROX1) enhanced hepatocyte identity by direct repression of alternative fate master regulators. In mice, Prox1 was required for efficient hepatocyte regeneration after injury and was sufficient to prevent liver tumorigenesis. In line with patient data, Prox1 depletion caused hepatocyte fate loss in vivo and enabled the transition of hepatocellular carcinoma to cholangiocarcinoma. Conversely, overexpression promoted cholangiocarcinoma to hepatocellular carcinoma transdifferentiation. Our findings provide evidence for PROX1 as a hepatocyte-specific safeguard and support a model where cell-type-specific repressors actively suppress plasticity throughout life to safeguard lineage identity and thus prevent disease.
Collapse
Affiliation(s)
- Bryce Lim
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Aryan Kamal
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Borja Gomez Ramos
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juan M Adrian Segarra
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ignacio L Ibarra
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Lennart Dignas
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tim Kindinger
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kai Volz
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Mohammad Rahbari
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nuh Rahbari
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of General and Visceral Surgery, University of Ulm, Ulm, Germany
| | - Eric Poisel
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kanela Kafetzopoulou
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lio Böse
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Marco Breinig
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
| | - Suchira Gallage
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University Tuebingen, Tübingen, Germany
| | | | - Hendrik Wiethoff
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Ivan Berest
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Sarah Schnabellehner
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Jonas Becker
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Heidelberg University, Center for Integrative Infectious Diseases Research (CIID), BioQuant, Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Heidelberg University, Center for Integrative Infectious Diseases Research (CIID), BioQuant, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Darjus F Tschaharganeh
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University Tuebingen, Tübingen, Germany
| | - Judith B Zaugg
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany.
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Moritz Mall
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany.
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
392
|
Luo L, Xia R, Mao S, Liu Q, Du H, Jiang T, Yang S, Wang Y, Li W, Zhou F, Yu J, Gao G, Li X, Zhao C, Cheng L, Shi J, Chen X, Zhou C, Chen L, Ren S, Wu F. Differential expression of the MYC-Notch axis drives divergent responses to the front-line therapy in central and peripheral extensive-stage small-cell lung cancer. MedComm (Beijing) 2025; 6:e70112. [PMID: 39974662 PMCID: PMC11836348 DOI: 10.1002/mco2.70112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 02/21/2025] Open
Abstract
Central and peripheral extensive-stage small-cell lung cancer (ES-SCLC) are reported to be two distinct tumor entities, but their responses to the front-line therapies and underlying biological mechanisms remain elusive. In this study, we first compared the outcomes of central and peripheral ES-SCLC receiving front-line chemotherapy or chemo-immunotherapy with a cohort of 265 patients. Then we performed single-cell RNA sequencing (scRNA-seq) on nine treatment-naïve ES-SCLC samples to investigate potential mechanisms underlying the response differences. Under chemotherapy, the peripheral type had a lower objective response rate (44.8% vs. 71.2%, p = 0.008) and shorter progression-free survival (median 3.4 vs. 5.1 months, p = 0.001) than the central type. When comparing chemo-immunotherapy with chemotherapy, the peripheral type showed a greater potential to reduce progression (HR, 0.18 and 0.52, respectively) and death (HR, 0.44 and 0.91 respectively) risks than the central type. Concerning the scRNA-seq data, the peripheral type was associated with chemo-resistant and immune-responsive tumoral and microenvironmental features, including a higher expression level of MYC-Notch-non-neuroendocrine (MYC-Notch-non-NE) axis and a more potent antigen presentation and immune activation status. Our results revealed that central and peripheral ES-SCLC had distinct responses to front-line treatments, potentially due to differential activation statuses of the MYC-Notch-non-NE axis.
Collapse
Affiliation(s)
- Libo Luo
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Rui Xia
- Key Laboratory of Systems BiologyCenter for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shiqi Mao
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Qian Liu
- Department of OncologyXiangyang No. 1 People's HospitalHubei University of MedicineXiangyangChina
| | - He Du
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Tao Jiang
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Shuo Yang
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yan Wang
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Wei Li
- Department of RadiologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Fei Zhou
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Jia Yu
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Guanghui Gao
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Xuefei Li
- Department of Lung Cancer and ImmunologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Chao Zhao
- Department of Lung Cancer and ImmunologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Lei Cheng
- Department of Lung Cancer and ImmunologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Jingyun Shi
- Department of RadiologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Xiaoxia Chen
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Caicun Zhou
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Luonan Chen
- Key Laboratory of Systems BiologyCenter for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesChinese Academy of SciencesHangzhouChina
| | - Shengxiang Ren
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Fengying Wu
- Department of Medical OncologyShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
393
|
Prinz D, Bartsch SJ, Ehret V, Friske J, Pinker K, Helbich TH. [Multiparametric magnetic resonance imaging of the breast : What can we expect from the future?]. RADIOLOGIE (HEIDELBERG, GERMANY) 2025; 65:162-169. [PMID: 39611894 PMCID: PMC11845421 DOI: 10.1007/s00117-024-01390-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The combination of different MRI methods is described as multiparametric MRI (mpMRI) and plays a significant role in breast cancer diagnostics. Currently, mpMRI includes contrast-enhanced and diffusion-weighted MRI. For a more comprehensive characterization of the key processes involved in cancer development, additional MRI methods that capture functional processes at the cellular and molecular levels are necessary. In the context of preclinical studies, MRI methods that enable contrast-free evaluation of key processes at the metabolic and molecular levels are being developed for future clinical applications. OBJECTIVES What does multiparametric MRI in breast cancer look like in the future? METHODS Systematic literature analysis focusing on preclinical research with regard to mpMRI as well as development and modification of noninvasive MRI methods. RESULTS Some of the most promising MRI methods for the evaluation of breast cancer that can answer functional and metabolic questions are BOLD (blood oxygen level dependent), IVIM (intravoxel incoherent motion), DMI (deuterium metabolic imaging) and CEST (chemical exchange saturation transfer). A combination and, therefore, a multiparametric approach allows for a noninvasive differentiation of breast cancer subtypes and early detection of treatment response which is crucial for the future development of the disease. CONCLUSION Standardization of quantification methods as well as improvement and expansion of MRI methods enable such a multiparametric, functional, and metabolic evaluation of the tumor. Many of these are initially developed in preclinical settings before they can be translated into clinical practice.
Collapse
Affiliation(s)
- Daniela Prinz
- Division of Molecular and Structural Preclinical Imaging, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Silvester J Bartsch
- Division of Molecular and Structural Preclinical Imaging, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Viktoria Ehret
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Wien, Österreich
| | - Joachim Friske
- Division of Molecular and Structural Preclinical Imaging, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Katja Pinker
- Division of Molecular and Structural Preclinical Imaging, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Wien, Österreich
- Division of Breast Imaging, Department of Radiology, Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Thomas H Helbich
- Division of Molecular and Structural Preclinical Imaging, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Wien, Österreich.
| |
Collapse
|
394
|
Ding Y, Jing W, Kang Z, Yang Z. Exploring the role and application of mitochondria in radiation therapy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167623. [PMID: 39674289 DOI: 10.1016/j.bbadis.2024.167623] [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: 09/09/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Mitochondria are pivotal in cellular energy metabolism, the oxidative stress response and apoptosis. Recent research has focused on harnessing their functions to enhance the efficacy of radiation therapy (RT). This review focuses on the critical functions and applications of mitochondria in radiation therapy, including the targeting of mitochondrial metabolism and the modulation of mitochondria-mediated cell death and immune responses. While these strategies have demonstrated considerable potential in preclinical studies to improve radiotherapy outcomes, challenges remain, such as optimizing drug delivery systems, ensuring safety and overcoming resistance to therapy.
Collapse
Affiliation(s)
- Yi Ding
- Shandong University, Jinan 250000, China
| | - Wang Jing
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Zhichao Kang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Zhe Yang
- Shandong University, Jinan 250000, China.
| |
Collapse
|
395
|
Gu Y, Fang Y, Guo Y, Yang R, Ma J, Zhang C, Deng M, Wen Q, Gao N, Qiao H. Cytochrome P450 2E1 inhibitor Q11 is effective on hepatocellular carcinoma by promoting peritumor neutrophil chemotaxis. Int J Biol Macromol 2025; 293:139189. [PMID: 39732257 DOI: 10.1016/j.ijbiomac.2024.139189] [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: 10/24/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Current studies found that the peritumoral tissue of hepatocellular carcinoma (HCC) may be different from normal liver tissue based on proteomics, and related to progression, recurrence and metastasis of HCC. Our previous study proposed "peritumor microenvironment (PME)" to summarize the influence of peritumor tissue on occurrence and progression of HCC. Peritumor CYP2E1 activity was significantly elevated in HCC, and related to occurrence and progression of HCC. However, the effectiveness and mechanism of inhibiting CYP2E1 against HCC remain unclear. In this study, by integrating the advantages of proteomics and transcriptomics, we reanalyzed the various influencing factors in PME. Although there were large differences in the occurrence and progression, the immunity and inflammation still played crucial roles. Peritumor neutrophil were "pro-tumor" phenotype in the stage of progression, while it showed cytotoxicity for tumor cell in the occurrence stage. CYP2E1 activity is associated with peritumor neutrophil infiltration and occurrence of HCC. CYP2E1 inhibitor Q11 showed anti-tumor effects in an orthotopic HCC mouse model by promoting secretion of chemokines and infiltration of neutrophils in peritumor tissue. Overall, these findings provided a reasonable mechanism of anti-tumor effects of CYP2E1 inhibitors, which may be a new strategy for the prevention and treatment of HCC.
Collapse
Affiliation(s)
- Yuhan Gu
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Department of Clinical Pharmacy, Nanyang Central Hospital, Nanyang, China
| | - Yan Fang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Guo
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Rui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jun Ma
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Cunzhen Zhang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mengyan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Gao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hailing Qiao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
396
|
Zhao L, Zhang Y, Wang J, Li D, Hao X. UBE2I depletion regulated tumor-associated macrophage polarization into M1 type through reprogramming glycolysis and increases immunotherapy efficacy of anti-PD-L1 in ovarian cancer. Mol Immunol 2025; 179:29-41. [PMID: 39919348 DOI: 10.1016/j.molimm.2025.01.007] [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: 10/08/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/09/2025]
Abstract
There is ample evidence that ubiquitin-conjugating enzyme E2I (UBE2I) is involved in progression of diverse cancers. However, the influence of UBE2I on ovarian cancer (OC) has been poorly reported. This study tries to discover the mechanisms and functions of UBE2I in OC. Relative mRNA expression of UBE2I, CD86, iNOS, MHC II and programmed death ligand 1 (PD-L1) was detected through qRT-PCR. We identified UBE2I, Vimentin, E-Cadherin, N-Cadherin and Ki67 protein expression levels in tumor tissues through immunohistochemistry staining. Protein levels of UBE2I, cleaved caspase-3, cleaved PARP, E-cadherin, N-cadherin and Vimentin were detected through western blot. Cell viability, invasion, and migration were examined by means of cell counting kit-8 (CCK-8), transwell, and wound healing assays. Immunofluorescence was used to detect colocalization between UBE2I and CD68. We assessed expression levels of IFN-γ and TNF-α via flow cytometry and ELISA. We used the TUNEL assay to assess tumor cell apoptosis. Glycolysis was assessed through the consumption of glucose, ATP production, production of lactate, and extracellular acidification rate. For establishing a xenograft model, OC cells were subcutaneously injected into mice. UBE2I expression was boosted in OC cells and tissues, which was negatively associated with OC patients' prognosis. Silencing of UBE2I suppressed OC cell proliferation, invasion, EMT (epithelial-to-mesenchymal transition) and migration. UBE2I inhibition promoted macrophages toward the M1 phenotype and macrophage viability. After deletion of UBE2I in vivo, mice tumor growth and EMT were suppressed, and apoptosis of tumor cells was increased. Meantime, an increasing proportion of CD86+ TAMs (tumor-associated macrophages) was observed after the deletion of UBE2I. Besides, increases in consumption of glucose, lactate production, ATP production and ECAR in THP-1 cells were observed by silencing of UBE2I; however, glycolysis inhibitor reversed UBE2I-mediated polarization of M1 macrophages in a dose-dependent fashion. Importantly, UBE2I-mediated M1 macrophages promoted PD-L1 expression. Furthermore, the combinatorial therapy of UBE2I inhibitor plus anti-PD-1 repressed tumor growth, reduced Ki67 expression, and promoted apoptosis in tumor cells, exhibiting higher efficiency than UBE2I inhibitor/anti-PD-L1 alone. UBE2I inhibition regulated polarization of M1 macrophages via glycolysis and improved anti-PD-L1 immunotherapy efficacy, paving a novel avenue to prevent OC development.
Collapse
Affiliation(s)
- Lei Zhao
- College of Medical Laboratory Science and Technology, Harbin Medical University at Daqing, Daqing, Heilongjiang 163319, China.
| | - Yuxin Zhang
- College of Medical Laboratory Science and Technology, Harbin Medical University at Daqing, Daqing, Heilongjiang 163319, China
| | - Jinming Wang
- College of Medical Laboratory Science and Technology, Harbin Medical University at Daqing, Daqing, Heilongjiang 163319, China
| | - Dongliang Li
- College of Medical Laboratory Science and Technology, Harbin Medical University at Daqing, Daqing, Heilongjiang 163319, China
| | - Xuewei Hao
- College of Medical Laboratory Science and Technology, Harbin Medical University at Daqing, Daqing, Heilongjiang 163319, China
| |
Collapse
|
397
|
Moura T, Laranjeira P, Caramelo O, Gil AM, Paiva A. Breast Cancer and Tumor Microenvironment: The Crucial Role of Immune Cells. Curr Oncol 2025; 32:143. [PMID: 40136347 PMCID: PMC11941043 DOI: 10.3390/curroncol32030143] [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: 01/15/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer is the most common type of cancer in women and the second leading cause of death by cancer. Despite recent advances, the mortality rate remains high, underlining the need to develop new therapeutic approaches. The complex interaction between cancer cells and the tumor microenvironment (TME) is crucial in determining tumor progression, therapy response, and patient prognosis. Understanding the role of immune cells in carcinogenesis and tumor progression can help improve targeted therapeutic options, increasing the likelihood of a favorable prognosis. Therefore, this review aims to critically analyze the complex interaction between tumor cells and immune cells, emphasizing the clinical and therapeutic implications. Additionally, we explore advances in immunotherapies, with a focus on immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Tânia Moura
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Center of Neurosciences and Cell (CNC), University of Coimbra, 3000-504 Coimbra, Portugal
| | - Olga Caramelo
- Gynecology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-075 Coimbra, Portugal;
| | - Ana M. Gil
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Ciências Biomédicas Laboratoriais, Instituto Politécnico de Coimbra, ESTESC—Coimbra Health School, 3046-854 Coimbra, Portugal
| |
Collapse
|
398
|
Costantini S. Special Issue "Cancer Biomarker: Current Status and Future Perspectives". Int J Mol Sci 2025; 26:2164. [PMID: 40076787 PMCID: PMC11900974 DOI: 10.3390/ijms26052164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
In recent years, advancements in omics technologies have significantly accelerated the identification of a broad spectrum of biomarkers based on DNA, RNA, microRNAs (miRNAs), and long non-coding RNAs, as well as proteins and metabolic and lipid alterations (Figure 1) [...].
Collapse
Affiliation(s)
- Susan Costantini
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| |
Collapse
|
399
|
Guo Z, Xiao Y, Wu W, Zhe M, Yu P, Shakya S, Li Z, Xing F. Metal-organic framework-based smart stimuli-responsive drug delivery systems for cancer therapy: advances, challenges, and future perspectives. J Nanobiotechnology 2025; 23:157. [PMID: 40022098 PMCID: PMC11871784 DOI: 10.1186/s12951-025-03252-x] [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: 09/28/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
Cancer treatment is currently one of the most critical healthcare issues globally. A well-designed drug delivery system can precisely target tumor tissues, improve efficacy, and reduce damage to normal tissues. Stimuli-responsive drug delivery systems (SRDDSs) have shown promising application prospects. Intelligent nano drug delivery systems responsive to endogenous stimuli such as weak acidity, complex redox characteristics, hypoxia, active energy metabolism, as well as exogenous stimuli like high temperature, light, pressure, and magnetic fields are increasingly being applied in chemotherapy, radiotherapy, photothermal therapy, photodynamic therapy, and various other anticancer approaches. Metal-organic frameworks (MOFs) have become promising candidate materials for constructing SRDDSs due to their large surface area, tunable porosity and structure, ease of synthesis and modification, and good biocompatibility. This paper reviews the application of MOF-based SRDDSs in various modes of cancer therapy. It summarizes the key aspects, including the classification, synthesis, modifications, drug loading modes, stimuli-responsive mechanisms, and their roles in different cancer treatment modalities. Furthermore, we address the current challenges and summarize the potential applications of artificial intelligence in MOF synthesis. Finally, we propose strategies to enhance the efficacy and safety of MOF-based SRDDSs, ultimately aiming at facilitating their clinical translation.
Collapse
Affiliation(s)
- Ziliang Guo
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuzhen Xiao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China
| | - Wenting Wu
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiyun Yu
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Sujan Shakya
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhihui Li
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Fei Xing
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
400
|
Fuller H, Agasaro OP, Darst BF. Pre-diagnostic circulating metabolomics and prostate cancer risk: A systematic review and meta-analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.27.25321444. [PMID: 40061317 PMCID: PMC11888532 DOI: 10.1101/2025.02.27.25321444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Background Metabolomic dysregulation contributes to prostate cancer (PCa) pathogenesis, and studies suggest that circulating metabolites have strong clinical potential to act as biomarkers. However, evidence of circulating metabolite associations has not been quantitively aggregated. Methods Systematic searches were performed in PubMed and Embase (October 17th, 2024) to identify pre-diagnostic untargeted serum metabolomic studies of PCa risk. After harmonizing metabolite names across studies, restricted maximum likelihood was used to conduct meta-analyses to quantify associations between metabolites and risk of overall PCa, low- to intermediate-risk PCa, high- to very high-risk PCa and lethal PCa, as defined by the NCCN. Statistical significance was defined as FDR-adjusted P<0.05. Enrichment analyses were conducted on significant metabolites to identify biologically relevant pathways. Correlation of effect estimates between PCa outcomes was assessed via Pearson correlation. Results We identified 12 untargeted pre-diagnostic circulating metabolomic studies in a systematic review and meta-analyzed associations between up to 408 metabolites with four PCa outcomes. Three, eleven and nineteen metabolites were significantly associated with risk of overall, high/very high-risk and lethal PCa, respectively. Metabolites associated with high/very high-risk PCa were significantly enriched for lipids. Limited evidence of correlation between metabolite effects across outcomes was identified, highlighting potentially unique metabolite drivers of high-risk and lethal PCa. Follow-up analyses found that 13 of the significant metabolites were drug and/or dietary modifiable. Conclusions These findings suggest the strong potential for metabolites to inform risk of lethal PCa, which could inform risk-stratified screening strategies and facilitate the identification of targets for PCa prevention.
Collapse
Affiliation(s)
- Harriett Fuller
- Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Orietta P. Agasaro
- Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Burcu F. Darst
- Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|