1
|
Chen DTH, Huang SY, Liu TC, Chen YK. Novel compounds of Taiwanese green propolis induce apoptosis of human glioblastoma cells by daylight photodynamic action. Future Sci OA 2025; 11:2464491. [PMID: 39925210 PMCID: PMC11812390 DOI: 10.1080/20565623.2025.2464491] [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: 10/08/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Glioblastoma, an aggressive brain cancer, has limited treatment options and poor prognosis. Taiwanese green propolis, known for its tumor-inhibitory properties, shows promise when combined with photodynamic therapy (PDT), a targeted, low-toxicity treatment. This study investigated a novel Taiwanese green propolis-based compound for inducing apoptosis in glioblastoma cells and its synergistic potential with daylight PDT. METHODS Ethanol extracts of green propolis, wheatgrass, and mulberry leaves were combined and analyzed using High-Performance Liquid Chromatography (HPLC). Apoptosis induction in U87 glioblastoma cells was assessed via the MTT assay following treatment with the compound alone and in combination with daylight PDT at 570 nm. RESULTS We identified Artepillin C as the main active component in the compound by HPLC, which significantly induced apoptosis in glioblastoma cells. Combined with daylight PDT, it demonstrated enhanced efficacy, with cell viability reduced from 95.2% at 0.25 µL to 11.3% at 8 µL of the compound extract. The EC50 decreased, indicating greater apoptotic activity compared to the extract alone. CONCLUSION This study provides the first in vitro evidence of synergistic anti-tumor effects of a Taiwanese green propolis-based compound daylight PDT (GPDT), highlighting a promising novel therapeutic approach that warrants further clinical investigation.
Collapse
Affiliation(s)
- Daniel Tzu-Hsuan Chen
- Nuffield Department of Primary Care Health Science, University of Oxford, Oxford, UK
| | | | - Ta-Chun Liu
- Department of Oncology, University of Oxford, Oxford, UK
| | - Yao-Kuan Chen
- Dr. Oxford Biotech Factory Co., Ltd, Taichung, Taiwan
| |
Collapse
|
2
|
Lin D, Li Y, Fang J, Xie X, Zhang B, Ye X, Huang Y, Zhang X, Xue A. Global, regional and national burden of pancreatitis in children and adolescents, 1990-2021: a systematic analysis for the global burden of disease study 2021. Ann Med 2025; 57:2499699. [PMID: 40326502 PMCID: PMC12057782 DOI: 10.1080/07853890.2025.2499699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Pancreatitis poses a significant global health burden, disproportionately affecting children and adolescents. This study uses the global burden of disease (GBD) 2021 dataset to evaluate pancreatitis epidemiology in this demographic, focusing on disparities by age, sex, and region. OBJECTIVE To assess global trends in pediatric pancreatitis, identify risk factors, and forecast disease burden to 2035. METHODS We analysed GBD 2021 data on deaths and disability-adjusted life years (DALYs) for pancreatitis in individuals under 20. The socio-demographic index (SDI) assessed the link between societal development and health outcomes. Bayesian age-period-cohort (BAPC) modelling and Poisson's linear models were applied to project future burdens and estimate annual percentage changes (EAPCs) in age-standardized rates. RESULTS In 2021, pancreatitis caused 1120.09 deaths in children and adolescents, comprising 2% of all pancreatitis-related deaths. Age-standardized death rate (ASDR) and DALYs rate declined from 1990 to 2021 (EAPC -0.92 and -0.86, respectively). Low-middle SDI regions, notably Andean and Central Latin America and Eastern Europe, faced the highest burden. Alcohol was a leading risk factor, accounting for 3.51% of related deaths, and males had higher death and DALYs rate. CONCLUSIONS Despite declining pancreatitis-related mortality and DALYs, the disease remains a challenge, particularly in low-middle SDI regions. Alcohol consumption is a key risk factor, underscoring the need for targeted public health interventions. Gender-, age-, and region-specific strategies are essential to mitigate pancreatitis impact in children and adolescents.
Collapse
Affiliation(s)
- Derong Lin
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Yue Li
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Jingya Fang
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Xiaohua Xie
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Bin Zhang
- The Fourth Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiaolin Ye
- The Seventh Affiliated Hospital of Guangzhou University of Chinese Medicine, Bao’an District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yiheng Huang
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Xiaowen Zhang
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Aiguo Xue
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| |
Collapse
|
3
|
Rezaei Z, Wang N, Yang Y, Govindaraj K, Velasco JJ, Martinez Blanco AD, Bae NH, Lee H, Shin SR. Enhancing organoid technology with carbon-based nanomaterial biosensors: Advancements, challenges, and future directions. Adv Drug Deliv Rev 2025; 222:115592. [PMID: 40324529 DOI: 10.1016/j.addr.2025.115592] [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: 01/03/2025] [Revised: 03/26/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Various carbon-based nanomaterials (CBNs) have been utilized to develop nano- and microscale biosensors that enable real-time and continuous monitoring of biochemical and biophysical changes in living biological systems. The integration of CBN-based biosensors into organoids has recently provided valuable insights into organoid development, disease modeling, and drug responses, enhancing their functionality and expanding their applications in diverse biomedical fields. These biosensors have been particularly transformative in studying neurological disorders, cardiovascular diseases, cancer progression, and liver toxicity, where precise, non-invasive monitoring is crucial for understanding pathophysiological mechanisms and assessing therapeutic efficacy. This review introduces intra- and extracellular biosensors incorporating CBNs such as graphene, carbon nanotubes (CNTs), graphene oxide (GO), reduced graphene oxide (rGO), carbon dots (CDs), and fullerenes. Additionally, it discusses strategies for improving the biocompatibility of CBN-based biosensors and minimizing their potential toxicity to ensure long-term organoid viability. Key challenges such as biosensor integration, data accuracy, and functional compatibility with specific organoid models are also addressed. Furthermore, this review highlights how CBN-based biosensors enhance the precision and relevance of organoid models in biomedical research, particularly in organ-specific applications such as brain-on-a-chip systems for neurodegenerative disease studies, liver-on-a-chip platforms for hepatotoxicity screening, and cardiac organoids for assessing cardiotoxicity in drug development. Finally, it explores how biosensing technologies could revolutionize personalized medicine by enabling high throughput drug screening, patient-specific disease modeling, and integrated sensing platforms for early diagnostics. By capturing current advancements and future directions, this review underscores the transformative potential of carbon-based nanotechnology in organoid research and its broader impact on medical science.
Collapse
Affiliation(s)
- Zahra Rezaei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Niyou Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Yipei Yang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Department of Orthopedic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China
| | - Kannan Govindaraj
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Department of Developmental Bioengineering, TechMed Centre, University of Twente, Drienerlolaan 5, Enschede 7522NB, the Netherlands
| | - Jose Joaquin Velasco
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Monterrey Institute of Technology, School of Science and Engineering, Eugenio Garza Sada Avenue 2501 South, Monterrey, Nuevo Leon 64849, Mexico
| | - Alvaro Dario Martinez Blanco
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; Monterrey Institute of Technology, School of Science and Engineering, Epigmenio González 500, Fraccionamiento San Pablo, Santiago de Querétaro, Querétaro 76130, Mexico
| | - Nam Ho Bae
- Center for Nano-Bio Developement, National NanoFab Center (NNFC), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - HeaYeon Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA; MARA Nanotech, INC. 4th floor, Hanmir Hall, Yongdang Campus, Pukyung National University, 365 Sinseon-ro, Nam-gu, Busan 48547, Republic of Korea; MARA Nanotech New York, INC. 1 Pennsylvania Plaza, Suite 1423, New York, NY 10119, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA.
| |
Collapse
|
4
|
Yu M, Chen X, Zheng Z. Comprehensive conditional survival analysis of pancreatic signet ring cell carcinoma: chemotherapy's role and predictive model development using the SEER database. Discov Oncol 2025; 16:1074. [PMID: 40504325 PMCID: PMC12162426 DOI: 10.1007/s12672-025-02946-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Pancreatic signet ring cell carcinoma (PSRCC) is a rare and aggressive subtype of pancreatic cancer, with a poor prognosis and limited evidence on the survival benefit of chemotherapy. From the perspective of conditional survival (CS) prognosis, this study sought to assess the effect of chemotherapy on PSRCC survival and to construct a predictive model integrating CS analysis. METHODS Using the SEER database, 708 PSRCC patients diagnosed between 2000 and 2019 were analyzed. Propensity score matching (PSM) and Kaplan-Meier curves were employed to assess chemotherapy's impact on survival. The CS analysis was performed to evaluate dynamic survival probabilities. A nomogram was developed based on key prognostic factors identified through random survival forests (RSF), least absolute shrinkage and selection operator (LASSO) regression, and multivariate Cox analysis with a stepwise backward elimination procedure. And multiple evaluation methods were employed to assess the performance of the nomogram. RESULTS The CS analysis for all cohort showed a rapid decline in survival probability within the first few years, dropping to 18% by year 1, 5% by year 3 and 3% by year 5. Chemotherapy improved short-term survival, with a 30% one-year survival rate compared to 8% in the non-chemotherapy group. However, long-term survival probabilities converged after the first year. Key prognostic factors included age, tumor size, stage, site, surgery, and chemotherapy were identified to develop a CS-integrated nomogram. And the nomogram was found to have strong predictive accuracy and clinical utility, validated by calibration, ROC, and decision curve analyses. CONCLUSION Chemotherapy offered significant early survival benefits in PSRCC, although its long-term impact is limited. The developed nomogram provided a reliable tool for personalized survival prediction, with further validation needed in prospective studies.
Collapse
Affiliation(s)
- Mingxu Yu
- Department of Hepatobiliary and pancreatic surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou city, 325000, China
| | - Xiaodong Chen
- Department of Emergency, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou city, China
| | - Zihao Zheng
- Department of Hepatobiliary and pancreatic surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou city, 325000, China.
| |
Collapse
|
5
|
Xin H, Chang Z, Niu M. Multifaceted Applications of Nanomaterials in Colorectal Cancer Management: Screening, Diagnostics, and Therapeutics. Int J Nanomedicine 2025; 20:7271-7294. [PMID: 40520059 PMCID: PMC12166954 DOI: 10.2147/ijn.s520616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/29/2025] [Indexed: 06/18/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor worldwide. Early detection and treatment of CRC can significantly improve patient survival and quality of life, while advanced-stage patients still face numerous challenges, such as drug resistance and adverse effects. Consequently, researchers are developing more efficient early screening and diagnostic strategies for CRC. Consequently, researchers are actively developing more efficient strategies for diagnosis and refined treatments. This review comprehensively examines the diverse applications of various nanomaterials in CRC management, including screening, diagnostic imaging, surgical guidance, drug delivery, radiotherapy, and modulation of the tumor microenvironment. Firstly, we explored how nanomaterials are revolutionizing CRC screening by enhancing the detection of early-stage tumors. In the realm of diagnostic imaging, nanomaterials are employed to improve the clarity and specificity of imaging modalities, thereby facilitating more accurate diagnoses. The review also examines the use of nanomaterials in surgical guidance, where they aid in the precise identification and removal of tumors, potentially improving surgical outcomes. Furthermore, the review underscores the significance of nanomaterials in drug delivery systems, which enable targeted therapy and reduce systemic side effects. We also discussed the role of nanomaterials in radio-sensitization, where they enhance the efficacy of radiotherapy by increasing the sensitivity of tumor cells to radiation. Additionally, the modulation of the tumor immune microenvironment using nanomaterials is highlighted as a promising strategy to induce immune response against cancer cells. Throughout the review, the mechanisms of action of these nanomaterials are meticulously examined, providing insights into how they interact with biological systems to achieve their therapeutic effects. The efficacy of these nanomaterials in overcoming drug resistance is also a focal point, as this is a critical factor in improving the long-term outcomes for CRC patients. In conclusion, while nanomaterials hold great promise for the management of CRC, addressing their biocompatibility and clinical translation challenges is crucial for their safe and effective application in clinical settings.
Collapse
Affiliation(s)
- He Xin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Zhihui Chang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Meng Niu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
6
|
Xu X, Cao J, Mu Y, Zhang H, Wang YL, Chen M, Li Y, Hua Q. Ultrasound-Induced Nitric Oxide-Propelled Nanomotor for Multimodal Theranostics of Cancer with Deep Penetration and Extended Lifetime. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e16709. [PMID: 40492501 DOI: 10.1002/advs.202416709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/21/2025] [Indexed: 06/12/2025]
Abstract
Cancer treatment is often ineffective due to poor bioimaging and resistance to standard therapies. This issue is exacerbated by multiple low-penetrable bio-barriers that limit the theranostic agents' effectiveness in tumors. Here, a hollow nanomotor PM-HMSN/Arg is fabricated by a sequential process involving: electrostatic adsorption of Mn2+, loading of l-Arg, and coating of platelet membrane (PM), respectively. This nanomotor uses l-Arg as an NO donor and ultrasound (US) as a trigger for NO release. After administration, it improves tumor penetration via a "tethering-relaxing-drilling" mechanism, overcoming bio-barriers during delivery from blood vessels to tumor cells. NO regulates the metabolism of tumor vascular endothelial cells, facilitating relaxation, and enhances cytotoxicity by participating in reactive oxygen species metabolism. More importantly, the nanomotor's active motion enhances tissue penetration and retention in cancer, increasing therapeutic effects. In addition, continuous in situ NO generation extends US imaging signal lifetime. This innovative nanomotor shows promise for multimodal theranostics in low-penetrable tumors.
Collapse
Affiliation(s)
- Xue Xu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Jinxu Cao
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Yan Mu
- School of Life Sciences, School of Chinese Medicine, Beijing University of Chinese medicine, Beijing, 100029, China
| | - Hao Zhang
- School of Life Sciences, School of Chinese Medicine, Beijing University of Chinese medicine, Beijing, 100029, China
| | - Ya-Lei Wang
- School of Life Sciences, School of Chinese Medicine, Beijing University of Chinese medicine, Beijing, 100029, China
| | - Mengzhen Chen
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Yuce Li
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Qian Hua
- School of Life Sciences, School of Chinese Medicine, Beijing University of Chinese medicine, Beijing, 100029, China
| |
Collapse
|
7
|
Ding Q, Rha H, Yoon C, Kim Y, Hong SJ, Kim HJ, Li Y, Lee MH, Kim JS. Regulated cell death mechanisms in mitochondria-targeted phototherapy. J Control Release 2025; 382:113720. [PMID: 40228665 DOI: 10.1016/j.jconrel.2025.113720] [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: 01/25/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Phototherapy, comprising photodynamic therapy (PDT) and photothermal therapy (PTT), was first introduced over a century ago and has since evolved into a versatile cancer treatment modality. While numerous studies have explored regulated cell death (RCD) mechanisms induced by phototherapy, a comprehensive synthesis centered on mitochondria-targeted phototherapeutic strategies and agents as mediators of RCD is still lacking. This review provides a systematic and in-depth analysis of recent advances in mitochondria-centered mechanisms driving phototherapy-induced death pathways, including apoptosis, autophagy, pyroptosis, immunogenic cell death, ferroptosis, and cuproptosis. We highlight the critical role of mitochondria as central regulators of these death pathways in response to phototherapeutic interventions. Moreover, we discuss fundamental design strategies for developing precision-targeted phototherapeutic materials to enhance efficacy and minimize off-target effects. Finally, we identify prevailing challenges and propose future research directions to address these hurdles, paving the way for next-generation mitochondria-targeted phototherapy as a highly effective strategy for cancer management.
Collapse
Affiliation(s)
- Qihang Ding
- School of Chemical Engineering & Pharmacy, Pharmaceutical Research Institute, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Wuhan Institute of Technology, Wuhan 430205, China; Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Hyeonji Rha
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Changyu Yoon
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Yujin Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - So Jin Hong
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hui Ju Kim
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yang Li
- School of Chemical Engineering & Pharmacy, Pharmaceutical Research Institute, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Min Hee Lee
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
8
|
Chen Y, Xu X, Hu X. Design and fabrication of polymer-coated vincristine-loaded core - shell iron nanoparticle system with mitochondrial-targeted ultrasound improved colorectal cancer therapy. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-21. [PMID: 40489545 DOI: 10.1080/09205063.2025.2501095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/25/2025] [Indexed: 06/11/2025]
Abstract
Vincristine (VCR)-based anticancer drugs show significant potential in improving treatment results for aggressive malignancies, including colorectal cancer (CRC). This study presents a core - shell iron nanoparticle system (IR780@VCR@GA-Fe-BSA@PLGA, termed as PVIBs) that combines VCR chemotherapy, sonodynamic treatment (SDT) utilizing IR780, and Fenton reaction-improved oxidative stress. The technology exhibited pH-response release of therapeutic compounds, synergic anticancer effects, and mitochondrial targeting when coupled with ultrasonic (US) irradiation. PVIBs markedly elevated ROS generation, exacerbated mitochondrial impairment, and augmented apoptosis rates in colorectal cancer therapies. These findings underscore the promise of this US-enhanced nanoplatform in tackling problems related to colorectal cancer treatment, providing a highly successful and least intrusive therapeutic approach.
Collapse
Affiliation(s)
- Yanbin Chen
- General Surgery, Cancer Center, Department of Colorectal Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaodong Xu
- General Surgery, Cancer Center, Department of Colorectal Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xinye Hu
- General Surgery, Cancer Center, Department of Colorectal Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Dogan C, Acikbas I, Er Urganci B, Azizi Z. The role of the NEAT1/miR410-3p axis in the invasion of breast cancer cells. Gene 2025; 951:149379. [PMID: 40037420 DOI: 10.1016/j.gene.2025.149379] [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: 06/24/2024] [Revised: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer, which is the most common cancer among women in Türkiye and throughout the world, is also one of the leading causes of cancer-related deaths. A significant factor in these deaths is metastatic breast cancer, which spreads to distant organs. The metastasis of the breast tumor follows a series of steps. Many proteins and signal molecules are in charge of these processes. In addition, NEAT1, a long noncoding RNA (lncRNA), was reported to play a key role in breast cancer cell proliferation and survival. Numerous cancer kinds were also shown to have extraordinary miR-410-3p expression levels. NEAT1 and miR-410-3p expression patterns in MCF-7 and MCF-10A cell lines were investigated using quantitative real-time polymerase chain reaction (qRT-PCR) in this study. The results demonstrated that NEAT1 was elevated by 2.30-fold in cancer cells in comparison to normal cells, whereas miR-410-3p was diminished by -2.85-fold. Furthermore, the transwell invasion experiment demonstrated the invasive potential of the MCF-7 cell line, whereas the MCF-10A cells could not invade. The target analysis revealed that functions of the targets were associated with biological adhesion and cel growth. In conclusion, a correlation was found between overexpression of NEAT1 and increased invasiveness of target cells, as well as inhibition of miR-410-3p, which is a regulatory target of NEAT1.
Collapse
Affiliation(s)
- Cihangir Dogan
- Pamukkale University, Faculty of Medicine, Medical Biology Department, Denizli, Turkey.
| | - Ibrahim Acikbas
- Pamukkale University, Faculty of Medicine, Medical Biology Department, Denizli, Turkey.
| | - Buket Er Urganci
- Pamukkale University, Faculty of Medicine, Medical Biology Department, Denizli, Turkey.
| | - Zahra Azizi
- Pamukkale University, Faculty of Medicine, Medical Biology Department, Denizli, Turkey.
| |
Collapse
|
10
|
Delmas D, Mialhe A, Cotte AK, Connat JL, Bouyer F, Hermetet F, Aires V. Lipid metabolism in cancer: Exploring phospholipids as potential biomarkers. Biomed Pharmacother 2025; 187:118095. [PMID: 40311223 DOI: 10.1016/j.biopha.2025.118095] [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: 02/12/2025] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Aberrant lipid metabolism is increasingly recognized as a hallmark of cancer, contributing to tumor growth, metastatic dissemination, and resistance to therapy. Cancer cells reprogram key metabolic pathways-including de novo lipogenesis, lipid uptake, and phospholipid remodeling-to sustain malignant progression and adapt to microenvironmental demands. This review summarizes current insights into the role of lipid metabolic reprogramming in oncogenesis and highlights recent advances in lipidomics that have revealed cancer type- and stage-specific lipid signatures with diagnostic and prognostic relevance. We emphasize the dual potential of lipid metabolic pathways-particularly those involving phospholipids-as sources of clinically relevant biomarkers and therapeutic targets. Enzymes and transporters involved in these pathways have emerged as promising candidates for both diagnostic applications and pharmacological intervention. We also examine persistent challenges hindering the clinical translation of lipid-based approaches, including analytical variability, insufficient biological validation, and the lack of standardized integration into clinical workflows. Furthermore, the review explores strategies to overcome these barriers, highlighting the importance of incorporating lipidomics into multi-omics frameworks, supported by advanced computational tools and AI-driven analytics, to decipher the complexity of tumor-associated metabolic networks. We discuss how such integrative approaches can facilitate the identification of actionable metabolic targets, improve the specificity and robustness of lipid-based biomarkers, and enhance patient stratification in the context of precision oncology.
Collapse
Affiliation(s)
- Dominique Delmas
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France; Centre de Lutte Contre le Cancer Georges François Leclerc Center, Dijon F-21000, France; Inserm UMS58 - Biologie Santé Dijon (BioSanD), Dijon F-21000, France.
| | - Aurélie Mialhe
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Alexia K Cotte
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Jean-Louis Connat
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Florence Bouyer
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - François Hermetet
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Virginie Aires
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| |
Collapse
|
11
|
Han L, Wang R, He M, Chen Z, Wang F. METTL3/YTDHF1 Stabilizes CSRP1 mRNA to Regulate Glycolysis and Promote Acute Myeloid Leukemia Progression. Cell Biochem Biophys 2025; 83:1993-2007. [PMID: 39565517 DOI: 10.1007/s12013-024-01610-4] [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] [Accepted: 10/29/2024] [Indexed: 11/21/2024]
Abstract
CSRP1 (Cysteine and Glycine-Rich Protein 1) is a protein often overactivated in various cancers, promoting cell proliferation and survival, making it a key factor in cancer development. However, it is worth noting that the effect of this protein on the glycolysis process in Acute Myeloid Leukemia (AML) has not yet been studied. This study aims to investigate the role of the METTL3/YTHDF1 axis in regulating Glycolysis and its impact on AML progression by stabilizing CSRP1 mRNA. We analyzed CSRP1 expression in AML tissues and cell lines using quantitative real-time PCR (qRT-PCR) and Western blotting. Functional assays, including cell viability, colony formation, glycolysis related indicators, were performed to assess the impact of CSRP1 knockdown or overexpression on AML cells. RNA immunoprecipitation (RIP) and RNA stability assays were conducted to elucidate the mechanism of METTL3/YTHDF1-mediated regulation of CSRP1 mRNA. CSRP1 was significantly upregulated in AML tissues and cell lines. Knockdown of CSRP1 inhibited AML cell proliferation and glycolysis. Overexpression of CSRP1 promoted AML cell survival. Mechanistically, METTL3 enhanced CSRP1 mRNA stability via m6A modification, recognized and bound by YTHDF1, preventing mRNA degradation. The METTL3/YTHDF1/ CSRP1 axis plays a critical role in AML progression by regulating glycolysis. Targeting this pathway may provide a novel therapeutic strategy for AML treatment.
Collapse
Affiliation(s)
- Lili Han
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ruiyan Wang
- Nursing College, Bengbu Medical College, Bengbu, China
| | - Mengyu He
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhenyue Chen
- Nursing College, Bengbu Medical College, Bengbu, China
| | - Feng Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
12
|
Bakrania A, Mo Y, Zheng G, Bhat M. RNA nanomedicine in liver diseases. Hepatology 2025; 81:1847-1877. [PMID: 37725757 PMCID: PMC12077345 DOI: 10.1097/hep.0000000000000606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023]
Abstract
The remarkable impact of RNA nanomedicine during the COVID-19 pandemic has demonstrated the expansive therapeutic potential of this field in diverse disease contexts. In recent years, RNA nanomedicine targeting the liver has been paradigm-shifting in the management of metabolic diseases such as hyperoxaluria and amyloidosis. RNA nanomedicine has significant potential in the management of liver diseases, where optimal management would benefit from targeted delivery, doses titrated to liver metabolism, and personalized therapy based on the specific site of interest. In this review, we discuss in-depth the different types of RNA and nanocarriers used for liver targeting along with their specific applications in metabolic dysfunction-associated steatotic liver disease, liver fibrosis, and liver cancers. We further highlight the strategies for cell-specific delivery and future perspectives in this field of research with the emergence of small activating RNA, circular RNA, and RNA base editing approaches.
Collapse
Affiliation(s)
- Anita Bakrania
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yulin Mo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Mamatha Bhat
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Gastroenterology, University Health Network and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Qu B, Wang Q, Zhou Y, Ning X, Wang Q, Zhou Z, Bai P, Zhang R. pH-driven butterfly effect for cascade-amplified tumor therapy based on thalidomide coordinated Fe-HMME nanoplatform. Mater Today Bio 2025; 32:101691. [PMID: 40225132 PMCID: PMC11986554 DOI: 10.1016/j.mtbio.2025.101691] [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: 01/13/2025] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
A promising approach for treating intractable cancers has been presented by photodynamic therapy (PDT). However, the limited penetration depth of PDT and suboptimal monotherapy efficacy of PDT significantly restrict its clinical applications. In this study, we constructed an acidic tumor microenvironment (TME)-activated carrier-free nanoplatform (HMME-Fe-Thal, abbreviated as HFT) through self-assembly of iron ions, photosensitizer hematoporphyrinmonomethyl ether (HMME) and anti-angiogenesis drug thalidomide (Thal). Near infrared (NIR) triggers PDT behavior before the degradation of the HFT nanoplatform. Subsequently, the HFT nanoplatform degrades, releasing Thal for chemotherapy, iron ions for chemodynamic therapy (CDT), which reinforce the therapeutic benefits of PDT synergistically. Moreover, the iron ions released by HFT degradation turn on the MRI signal, which can suggest the most appropriate time for PDT, divide the treatment into two stages (First-stage: PDT, Second-stage: CDT/chemotherapy), and gradually achieve cascade-amplified tumor therapy. In this sense, HFT modulates TME and leads to a "butterfly effect" of CDT/chemotherapy/glutathione (GSH) depletion for enhanced PDT efficacy. This strategy compensates the deficient shadow penetration and poor treatment efficacy from PDT monotherapy. This work presents the selection and rational design of HFT constructed by endogenous components for tumor regression, and greatly push nanomaterials towards the development of PDT application.
Collapse
Affiliation(s)
- Botao Qu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Qian Wang
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Yuxin Zhou
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaogang Ning
- School of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Qian Wang
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Ziyi Zhou
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Peirong Bai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
14
|
Wongkrasant P, Wallace LE, MacNaughton WK, Sharkey KA. Fructooligosaccharides slow colonic motility and activate myenteric neurons via calcium sensing and 5-HT 3 receptors in the proximal colon. Am J Physiol Gastrointest Liver Physiol 2025; 328:G734-G745. [PMID: 40279204 DOI: 10.1152/ajpgi.00039.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/05/2025] [Accepted: 04/10/2025] [Indexed: 04/27/2025]
Abstract
Calcium-sensing receptors (CaSR) regulate a variety of functions in the gastrointestinal tract. Recently, prebiotic-independent effects of fructooligosaccharides (FOS) on epithelial barrier function were found to be mediated by CaSR. Here, we tested the hypothesis that FOS acts via the CaSR to regulate colonic motility and neuronal activity in the enteric nervous system. Using immunohistochemistry, we determined that CaSR were localized on the colonic epithelium of the mouse proximal colon and that a small proportion of enterochromaffin cells coexpress CaSR. We demonstrated that intraluminal administration of FOS slows colonic motility in vivo in male and female mice, an effect that is mediated by both CaSR and 5-HT3 receptors. We assessed neuronal activity in response to luminally perfused FOS in intact segments of the proximal colon from male and female mice expressing a genetically encoded fluorescent calcium reporter in intrinsic primary afferent neurons (Calb1-GCaMP6 mice) or in all enteric neurons (Wnt1-GCaMP6 mice) using live cell confocal imaging. In both Calb1-GCaMP6 mice and Wnt1-GCaMP6 mice, intraluminal FOS perfusion induced a sustained elevation of intracellular Ca2+ in neurons of the myenteric plexus. This effect was sensitive to tetrodotoxin and mediated by CaSR and 5-HT3 receptors. Serosal application of FOS was without effect. Our results demonstrate that FOS acts acutely to slow colonic motility in vivo and activates the enteric nervous system via CaSR and 5-HT3 receptors.NEW & NOTEWORTHY Calcium-sensing receptors regulate a variety of functions in the gastrointestinal tract. Here, we demonstrate a novel action of fructooligosaccharides to regulate colonic motility in vivo and activate the enteric nervous system. These effects are mediated by calcium-sensing and 5-HT3 receptors.
Collapse
Affiliation(s)
- Preedajit Wongkrasant
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Laurie E Wallace
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
15
|
Yang Z, Wang L, Kang F. Immunogenic cell death: A promising mechanism involving different therapeutic strategies for liver cancer. Int J Cancer 2025. [PMID: 40448266 DOI: 10.1002/ijc.35496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/30/2025] [Accepted: 05/21/2025] [Indexed: 06/02/2025]
Abstract
Hepatocellular carcinoma is a malignant tumor with a high mortality rate that seriously endangers human health. Although there are various treatments for hepatocellular carcinoma, the 5-year survival rate and prognosis of patients are still poor, depending on the stage. The proposal of immunogenic cell death provides a new idea and direction for the treatment of HCC. A variety of drugs act as effective inducers of ICD to induce the immunogenicity of tumor cells, significantly kill tumor cells, activate the body's inherent and adaptive immunity while producing and releasing damage-related molecular patterns, and significantly improve the treatment effect and side effects. This article briefly classifies the existing ICD inducers and describes how DAMPs change in this process. By summarizing the existing ICD-related studies applied to HCC treatment and proposing improvement methods for existing problems, this paper provides a theoretical summary for the future exploration of new therapies for HCC.
Collapse
Affiliation(s)
- Ziye Yang
- Department of Hepatology, Bethune International Peace Hospital, Shijiazhuang, China
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ling Wang
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fubiao Kang
- Department of Hepatology, Bethune International Peace Hospital, Shijiazhuang, China
| |
Collapse
|
16
|
Wei X, Liu R, Li W, Yu Q, Yang QT, Li T. Advances in research regarding epithelial-mesenchymal transition and prostate cancer. Front Cell Dev Biol 2025; 13:1583255. [PMID: 40519259 PMCID: PMC12162976 DOI: 10.3389/fcell.2025.1583255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/09/2025] [Indexed: 06/18/2025] Open
Abstract
Prostate cancer (PCa) is the most prevalent cancer in men and the fifth leading cause of cancer-related mortality among men globally. Despite substantial advancements in patient prognosis attributable to improvements in PCa treatment, individuals with metastatic castration-resistant prostate cancer continue to experience poor outcomes. Epithelial-mesenchymal transition (EMT) is characterized as a cellular event in which epithelial cells adopt a mesenchymal phenotype while simultaneously losing their epithelial characteristics. EMT has been demonstrated to be associated with the progression of PCa, encompassing tumor metastasis, recurrence, drug resistance, and the development of an immunosuppressive microenvironment. Consequently, this review synthesizes recent studies on EMT in PCa, consolidating the events mediated by EMT in the progression of PCa and the molecular mechanisms linked to EMT activation in this context.
Collapse
Affiliation(s)
| | | | | | | | | | - Tao Li
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
17
|
Gu D, Liang X, Shao Z, He H, Zhu L, Qiu S, Liu Z, Wang S, Chen H, Ci S, Wu H, Wang Y. A cascaded amplification carrier-free nanoplatform for synergistic photothermal/ferroptosis therapy via dual antioxidant pathway disruption in cervical cancer. J Mater Chem B 2025; 13:6128-6143. [PMID: 40337781 DOI: 10.1039/d5tb00627a] [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: 05/09/2025]
Abstract
Cellular defense mechanisms against ferroptosis are primarily mediated by antiferroptotic regulators, particularly glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1). Notably, singlet oxygen (1O2) generated through photoactivation of organic small-molecule photosensitizers (PSs) has been demonstrated to deplete both glutathione (GSH) and nicotinamide adenine dinucleotide phosphate (NADPH). This dual depletion mechanism effectively disrupts the GSH/GPX4 redox axis and the NADPH/FSP1/ubiquinone (CoQ) antioxidant system, thereby potentiating ferroptosis. In this study, we engineered a tumor-targeting amphiphilic iridium-based photosensitizer nanoplatform (Ir-TCF3P-FA NPs) for synergistic photothermal-ferroptosis therapy. Specifically, GSH depletion and NADPH oxidation by 1O2 produced via Ir-TCF3P-FA NPs at 450 nm can suppress the expression of GPX4 and FSP1, amplifying ferroptosis. Additionally, TCF3P exhibited high photothermal conversion efficiency at 808 nm, which not only can enhance photothermal therapy (PTT) efficacy but also facilitated 1O2 generation. The Ir-TCF3P-FA NPs enable effective tumor-targeted delivery and fluorescence/photoacoustic imaging for in vivo distribution tracking. In vivo studies revealed that dual-laser irradiation of Ir-TCF3P-FA NPs provided potent therapeutic efficacy, significantly inhibiting human cervical cancer progression in murine models. This cascaded amplification carrier-free nanoplatform holds promise for clinical multimodal treatment of cervical cancer.
Collapse
Affiliation(s)
- Dihai Gu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| | - Xiao Liang
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Zitong Shao
- The First Clinical Medical College, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Haonan He
- The First Clinical Medical College, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Lin Zhu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Shali Qiu
- Department of Pathology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Zhen Liu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Senlin Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| | - Haijiao Chen
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Shusheng Ci
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Hongshuai Wu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Yihong Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| |
Collapse
|
18
|
Zhang H, Yuan F, Zhao N, Tang W, Zhao P, Liu C, Chen S, Hou X, Xia C, Chu J. Nanoparticle-mediated SIRT1 inhibition suppresses M2 macrophage polarization and hepatocarcinogenesis in chronic hepatitis B. J Nanobiotechnology 2025; 23:385. [PMID: 40426198 PMCID: PMC12117705 DOI: 10.1186/s12951-025-03447-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major complication of chronic hepatitis B (CHB), with macrophage M2 polarization playing a critical role in shaping the tumor-promoting hepatic immune microenvironment. Sirtuin 1 (SIRT1) has been implicated in immune modulation and liver carcinogenesis. This study investigates the potential of Mimetic Nanoparticles (MNPs) for delivering SIRT1 inhibitors to regulate macrophage polarization and remodel the hepatic immune microenvironment, aiming to prevent HCC development post-CHB. A transgenic mouse model of CHB was established, and RNA sequencing (RNA-seq) and proteomics analyses revealed significant dysregulation of genes associated with M2 macrophage polarization, particularly SIRT1. Functional enrichment analysis highlighted key pathways, including PI3K-Akt and NF-κB, that contribute to CHB-driven immune alterations. Synthesized and characterized MNPs successfully delivered SIRT1 inhibitors, effectively inhibiting M2 macrophage polarization in vitro. In vivo administration of MNPs-SIRT1-FN significantly reduced M2 macrophage infiltration and suppressed tumor growth. These findings suggest that nanoparticle-mediated SIRT1 inhibition is a promising strategy for immunomodulation and HCC prevention in CHB patients. This study provides novel insights into nanoparticle-based immunotherapy for CHB-related HCC and highlights a potential therapeutic avenue for liver cancer prevention.
Collapse
Affiliation(s)
- He Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Feng Yuan
- Laboratory of Inflammation and Vaccines, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Nan Zhao
- Institute of Clinical Medicine, National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Wenqiang Tang
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, 850009, China
| | - Pengwei Zhao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Chunfa Liu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
| | - Shan Chen
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
| | - Xiaolin Hou
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China.
| | - Changyou Xia
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China.
| | - Jun Chu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China.
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, 850009, China.
| |
Collapse
|
19
|
Huang L, Wei M, Li H, Yu M, Wan L, Zhao R, Gao Q, Sun L, Hou X, Mo Y, Huang Q, Zhen L, Yang X, Li J, Wang N, Zhang C, Jin H, Zhou L, Xu Y, Lin H, Zhang X, Li B, Han Y, Yuan J, Zhang R, Wu F, Zhong H, Wei C. GP73-dependent regulation of exosome biogenesis promotes colorectal cancer liver metastasis. Mol Cancer 2025; 24:151. [PMID: 40414849 DOI: 10.1186/s12943-025-02350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
Colorectal cancer (CRC) liver metastasis is the main cause of cancer-related mortality. How liver influences intercellular communication to support CRC liver metastasis remains unknown. Herein, we link GP73, whose chronic upregulation in hepatocytes triggers non-obese metabolic-dysfunction associated steatotic liver disease (MASLD) in mice, with exosome biogenesis and CRC liver metastasis. Mice with high liver GP73 expression exhibited increased CRC liver metastasis in an exosome-dependent manner. GP73 modulated the cholesterol contents in endosomal compartments to promote exosome production. Quantitative proteomics revealed GP73 reshaped hepatocyte exosomal proteome and produced NAV2-rich exosomes. Clinically, serum GP73 levels positively correlated with exosomal NAV2 levels in CRC patients with liver metastasis. Knockdown of liver NAV2 suppressed enhanced CRC liver metastasis in GP73-induced non-obese mice, and GP73 blockade mitigated the increased CRC liver metastasis in obese mice fed by high-fat diet or high-fructose diet. Our findings suggest GP73 blockade as a potential therapeutic strategy for mitigating CRC liver metastasis.
Collapse
Affiliation(s)
- Linfei Huang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Meng Wei
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, 530021, China
| | - Huilong Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Mingxin Yu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, State Key Laboratory of Digestive Health, Beijing Key Laboratory of Early Gastrointestinal Cancer Medicine and Medical Devices, No. 95 Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Luming Wan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Ruzhou Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Qi Gao
- Beijing Youngen Technology Co. Ltd, No. 55 Qingfeng West Road, Daxing District, Beijing, 102629, China
| | - Lijuan Sun
- Beijing Youngen Technology Co. Ltd, No. 55 Qingfeng West Road, Daxing District, Beijing, 102629, China
| | - Xufeng Hou
- Beijing Youngen Technology Co. Ltd, No. 55 Qingfeng West Road, Daxing District, Beijing, 102629, China
| | - Yunhai Mo
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, 530021, China
| | - Qing Huang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, 530021, China
| | - Lan Zhen
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, 530021, China
| | - Xiaopan Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Jingfei Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Nan Wang
- Department of Radiotherapy, Changzhi People's Hospital, No. 502, Changxing Middle Road, Luzhou District, Changzhi, Shanxi, 046000, China
| | - Chundong Zhang
- Department of Surgical Oncology and Central Laboratory, the Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang, Liaoning, 110032, China
| | - Haoran Jin
- Department of Colorectal Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong Distrct, Shenyang, Liaoning, 110042, China
| | - Li Zhou
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Yixin Xu
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, 530021, China
| | - Haotian Lin
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China
| | - Xuhui Zhang
- Beijing Youngen Technology Co. Ltd, No. 55 Qingfeng West Road, Daxing District, Beijing, 102629, China
| | - Boan Li
- Clinical Laboratory, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, No. 100 Xisihuan Middle Road, Beijing, 100039, China.
| | - Yue Han
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan South Road, Chaoyang District, Beijing, 100021, China.
| | - Jing Yuan
- Capital Institute of Pediatrics, Capital Center for Children's Health, Capital Medical University, No. 2 Yabao Road, Chaoyang District, Beijing, 100020, China.
| | - Rui Zhang
- Department of Colorectal Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong Distrct, Shenyang, Liaoning, 110042, China.
| | - Feixiang Wu
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, 530021, China.
| | - Hui Zhong
- Beijing Youngen Technology Co. Ltd, No. 55 Qingfeng West Road, Daxing District, Beijing, 102629, China.
| | - Congwen Wei
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, No. 20 Dongdajie, Fengtai District, Beijing, 100071, China.
| |
Collapse
|
20
|
Deng Y, Yang B, Yang Z, Xiao H, Zou Y, Zou C, Yang S, Sun X, Wang Y, Bai J, Fang L, Wang Z. Engineered E. coli OMVs Carrying the Membrane-Binding hGC33 Fragment Precisely Target Liver Cancer and Effectively Treat Tumor. Int J Nanomedicine 2025; 20:6573-6590. [PMID: 40433120 PMCID: PMC12106912 DOI: 10.2147/ijn.s513508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background Glypican-3 (GPC3), which is a membrane-associated antigen that is overexpressed in hepatocellular carcinoma (HCC). hGC33, a humanized anti-GPC3 antibody, has been validated as a potential antibody drug with good antitumor activity by preclinical studies and the Phase II clinical trial. However, free drug usually lack good tumor penetration. Outer membrane vesicles (OMVs) that are secreted by Escherichia coli function as natural vectors for molecule delivery and mediators of biological signals across tissues. Our study aimed to engineer E. coli for use as a platform to precisely deliver the hGC33 single-chain variable fragment (hGC33-scFv) for the targeted treatment of HCC. Methods In this study, we utilized E. coli BL21(DE3) to express Hbp-hGC33-scFv fusion protein and generated E. coli hGC33-OMVs. After isolation and characterization, we assessed their chemotaxis toward HepG2 cells by Transwell, coimmunoprecipitation (co-IP) to confirm hGC33-GPC3 binding, and immunofluorescence (IF) to evaluate the localization of hGC33 on OMV membranes. The in vivo efficacy was assessed in BALB/c nude mice harboring HepG2 cell-derived xenografts, and tumor targeting was analyzed with Cy7-labeled OMVs and live imaging. Proliferation assays, cell cycle analysis, and Wnt pathway expression analysis were performed to elucidate the underlying mechanisms. Results hGC33-OMVs exhibited spherical bilayered nanostructures and displayed hGC33-scFv on their surface. hGC33-OMVs preferentially accumulated in tumors, significantly reducing tumor volume compared with controls and downregulating the proliferation markers Ki67 and PCNA. Transwell assays revealed increased tropism of hGC33-OMVs toward HepG2 cells, while Co-IP confirmed the direct interaction between hGC33 and GPC3. Meanwhile, hGC33-OMVs suppressed HepG2 cell proliferation, induced G1-phase arrest, and reduced Wnt3a, β-catenin, Cyclin D1, and C-myc expression. Conclusion Engineered E. coli hGC33-OMVs effectively target HCC via the hGC33-GPC3 interaction, inhibit tumor growth by suppressing Wnt signaling, and demonstrate potential for use as a versatile platform for antibody delivery.
Collapse
Affiliation(s)
- Yufei Deng
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Bangya Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zelan Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Hanyu Xiao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yan Zou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Cheng Zou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Song Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Xi Sun
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yiting Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jin Bai
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Liaoqiong Fang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- National Engineering Research Center of Ultrasound Medicine, Chongqing, 401121, People’s Republic of China
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- National Engineering Research Center of Ultrasound Medicine, Chongqing, 401121, People’s Republic of China
| |
Collapse
|
21
|
Muteeb G, El-Morsy MT, Abo-Taleb MA, Mohamed SK, Khafaga DSR. Herbal Medicine: Enhancing the Anticancer Potential of Natural Products in Hepatocellular Carcinoma Therapy Through Advanced Drug Delivery Systems. Pharmaceutics 2025; 17:673. [PMID: 40430962 PMCID: PMC12114929 DOI: 10.3390/pharmaceutics17050673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive and prevalent liver cancer with a poor prognosis. Nanotechnology combined with natural products has emerged as a promising strategy to enhance HCC treatment efficacy. This review assesses the current literature on the application of nanotechnology in delivering natural products for HCC therapy. A comprehensive search was conducted in PubMed, Science Direct, Web of Science, and Google Scholar to identify relevant studies published up to the present articles focusing on nanotechnology-based drug delivery systems using natural products for HCC therapy, including different nanoparticle (NP) formulations and therapeutic interventions, were included. Natural products with anticancer properties have been encapsulated using various nanocarriers such as liposomes, polymeric nanoparticles, and quantum dots, which have improved drug stability, prolonged circulation time, and enhanced targeted delivery to HCC cells. These advancements have led to increased therapeutic efficacy and reduced side effects. Additionally, combining multiple natural products or integrating them with conventional therapies via nanocarriers enables personalized treatment approaches based on patient characteristics and molecular profiles. The integration of nanotechnology with natural products shows great potential for improving HCC treatment outcomes, representing a significant advancement in precision medicine for liver cancer and paving the way for more effective and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Ghazala Muteeb
- Department of Nursing, College of Applied Medical Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Manar T. El-Morsy
- Bio-Nanotechnology Department, Faculty of Nanotechnology, Cairo University, Giza 12613, Egypt;
| | | | | | | |
Collapse
|
22
|
Cao L, Abudureheman W, Shen G, Ouyang Y, Yang W, Zhao Q, Lu T, Lin C. LncRNA NEAT1 regulation of the miR-101-3p/RAC1 axis affects cervical cancer aerobic glycolysis and progression. Sci Rep 2025; 15:17436. [PMID: 40394096 PMCID: PMC12092591 DOI: 10.1038/s41598-025-01698-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
Cervical cancer is a prevalent malignancy among women worldwide. Long-chain non-coding rna (lncRNAs) play a key role in the development of several cancers. Here, we found that the expression of lncRNA NEAT1 was significantly increased in cervical cancer cells and tissues and was closely associated with poor patient prognosis. Subsequently, we found that down-regulation of NEAT1 inhibited the proliferation, migration and invasion of cervical cancer cells. Subsequent studies showed that NEAT1, a competitive endogenous RNA, effectively enhanced RAC1 expression by adsorbing miR-101-3p. Glycolysis-related genes were predicted to be enriched in cervical cancers with high NEAT1 expression by bioinformatics analysis and confirmed by in vivo experiments. Our results suggest that NEAT1 enhances the Warburg effect through the miR-101-3p/RAC1 axis and promotes the proliferation, migration and invasion of cervical cancer cells. Therefore, elucidating this potential mechanism and targeting the NEAT1/miR-101-3p/RAC1 pathway may provide valuable insights.
Collapse
Affiliation(s)
- Lingling Cao
- Xinjiang Key Laboratory of Molecular Biology of Endemic Diseases, School of Basic Medical Science, Xinjiang Medical University, Xinjiang Medical University Graduate School, Urumqi, 830017, Xinjiang, China
| | - Wumidan Abudureheman
- Xinjiang Key Laboratory of Molecular Biology of Endemic Diseases, School of Basic Medical Science, Xinjiang Medical University, Xinjiang Medical University Graduate School, Urumqi, 830017, Xinjiang, China
| | - Guqun Shen
- Department of Gynecological Surgery, The Third Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Yunshan Ouyang
- Xinjiang Key Laboratory of Molecular Biology of Endemic Diseases, School of Basic Medical Science, Xinjiang Medical University, Xinjiang Medical University Graduate School, Urumqi, 830017, Xinjiang, China
| | - Wang Yang
- Xinjiang Key Laboratory of Molecular Biology of Endemic Diseases, School of Basic Medical Science, Xinjiang Medical University, Xinjiang Medical University Graduate School, Urumqi, 830017, Xinjiang, China
| | - Qian Zhao
- Xinjiang Key Laboratory of Molecular Biology of Endemic Diseases, School of Basic Medical Science, Xinjiang Medical University, Xinjiang Medical University Graduate School, Urumqi, 830017, Xinjiang, China
| | - Tianze Lu
- Xinjiang Medical University Tumor Hospital, Urumqi, 830011, Xinjiang, China
| | - Chen Lin
- Xinjiang Key Laboratory of Molecular Biology of Endemic Diseases, School of Basic Medical Science, Xinjiang Medical University, Xinjiang Medical University Graduate School, Urumqi, 830017, Xinjiang, China.
| |
Collapse
|
23
|
Lin Z, Rasinski P, Nilsson T, Holstensson M, Song Y, Blomgren A, Jutidamrongphan W, Pandya K, Hong J, Rominger A, Shi K, Axelsson R, Lan X, Seifert R. FAPI PET Versus FDG PET/CT in Gastrointestinal Cancers: An Overview. Semin Nucl Med 2025:S0001-2998(25)00056-X. [PMID: 40399164 DOI: 10.1053/j.semnuclmed.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/23/2025]
Abstract
Fibroblast activation protein (FAP) is a type II transmembrane serine protease that is highly expressed in cancer-associated fibroblasts (CAFs) but absent in quiescent fibroblasts. Its overexpression is associated with poor prognosis in various cancers and contributes to treatment resistance. In recent years, radiolabeled FAP inhibitors (FAPI) for PET imaging have shown promising clinical value across a range of cancers. Gastrointestinal (GI) malignancies, which often exhibit a desmoplastic reaction with a high density of FAP-expressing CAFs, are particularly well-suited for FAPI PET. Given the limitations of [18F]FDG PET in GI cancers, such as low sensitivity in certain histological subtypes and high physiological background uptake, FAPI PET is expected to serve as a complementary method, potentially enhancing both diagnostic accuracy and treatment guidance. This review provides a comprehensive comparison of the clinical applications of FAPI PET and [18F]FDG PET in various GI cancers, including their value in diagnosis, staging, and treatment guidance. Additionally, this review summarizes studies on the expanding role of FAPI PET, including its use in assessing treatment response and predicting prognosis, aiming to provide insights into its potential contribution to the improved management of GI malignancies.
Collapse
Affiliation(s)
- Zhaoguo Lin
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Pawel Rasinski
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ted Nilsson
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Holstensson
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan, China; Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - August Blomgren
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Warissara Jutidamrongphan
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Kalyani Pandya
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jimin Hong
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Rimma Axelsson
- Department of Nuclear Medicine and Medical Physics, Karolinska University Hospital, Huddinge, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China; Hubei Key Laboratory of Molecular Imaging, Wuhan, China; Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Robert Seifert
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
24
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
25
|
Wu Y, Chen W, Deng J, Cao X, Yang Z, Chen J, Tan Q, Zhou E, Li M, Liu J, Guo M, Jin Y. Tumour-derived microparticles obtained through microwave irradiation induce immunogenic cell death in lung adenocarcinoma. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01922-3. [PMID: 40389640 DOI: 10.1038/s41565-025-01922-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/31/2025] [Indexed: 05/21/2025]
Abstract
Tumour-derived microparticles (TMPs), extracellular vesicles traditionally obtained upon ultraviolet (UV) radiation of tumour cells, hold promise in tumour immunotherapies and vaccines and have demonstrated potential as drug delivery systems for tumour treatment. However, concerns remain regarding the limited efficacy and safety of UV-derived TMPs. Here we introduce a microwave (MW)-assisted method for preparing TMPs, termed MW-TMPs. Brief exposure of tumour cells to short-wavelength MW radiation promotes the release of TMPs showing superior in vivo antitumour activity and safety compared with UV-TMPs. MW-TMPs induce immunogenic cell death and reprogramme suppressive tumour immune microenvironments in different lung tumour models, enabling dual targeting of tumour cells by natural killer and T cells. We show that they can efficiently deliver methotrexate to tumours, synergistically boosting the efficacy of PD-L1 blockade. This MW-TMP development strategy is simpler, more efficient and safer than traditional UV-TMP methods.
Collapse
Affiliation(s)
- Yali Wu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjuan Chen
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Deng
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinghui Cao
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zimo Yang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangbin Chen
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - E Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minglei Li
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiatong Liu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
26
|
Restrepo-Acevedo A, Murillo MI, Orvain C, Thibaudeau C, Recberlik S, Verget L, Gómez Vidales V, Gaiddon C, Mellitzer G, Le Lagadec R. Protoporphyrin IX-Derived Ruthenium(II) Complexes for Photodynamic Therapy in Gastric Cancer Cells. Inorg Chem 2025; 64:9684-9702. [PMID: 40315445 DOI: 10.1021/acs.inorgchem.5c00896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
In recent years, photodynamic therapy (PDT) has emerged as a promising alternative to classical chemotherapy for treating cancer. PDT is based on a nontoxic prodrug called photosensitizer (PS) activated by light at the desired location. Upon irradiation, the PS reacts with the oxygen present in the tumor, producing cytotoxic reactive oxygen species (ROS). Compounds with highly conjugated π-bond systems, such as porphyrins and chlorins, have proven to be excellent light scavengers, and introducing a metal atom in their structure improved the generation of ROS. In this work, a series of tetrapyrrole-ruthenium(II) complexes derived from protoporphyrin IX and the commercial drug verteporfin were designed as photosensitizers for PDT. The complexes were almost nontoxic on human gastric cancer cells under dark conditions, revealing remarkable cytotoxicity upon irradiation with light. The ruthenium atom in the central cavity of the chlorin ligand allowed combined mechanisms in photodynamic therapy, as both singlet oxygen and superoxide radicals were detected. Additionally, one complex produced large amounts of singlet oxygen under hypoxic conditions. Biological assays demonstrated that the ruthenium derivatives caused cell death through a caspase 3 mediated apoptotic pathway and via CHOP, an endoplasmic reticulum stress-inducible transcription factor involved in apoptosis and growth arrest.
Collapse
Affiliation(s)
- Andrés Restrepo-Acevedo
- Universidad Nacional Autonoma de México, Instituto de Química UNAM, Circuito Exterior s/n Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | - María Isabel Murillo
- Universidad Nacional Autonoma de México, Instituto de Química UNAM, Circuito Exterior s/n Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | - Christophe Orvain
- Inserm UMR_S U1113; IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Chloé Thibaudeau
- Inserm UMR_S U1113; IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Sevda Recberlik
- Inserm UMR_S U1113; IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Lucas Verget
- Universidad Nacional Autonoma de México, Instituto de Química UNAM, Circuito Exterior s/n Ciudad Universitaria, 04510 Ciudad de México, Mexico
- Faculté de Chimie, Sorbonne Université, 4 place Jussieu, 75005 Paris, France
| | - Virginia Gómez Vidales
- Universidad Nacional Autonoma de México, Instituto de Química UNAM, Circuito Exterior s/n Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | - Christian Gaiddon
- Inserm UMR_S U1113; IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Georg Mellitzer
- Inserm UMR_S U1113; IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Ronan Le Lagadec
- Universidad Nacional Autonoma de México, Instituto de Química UNAM, Circuito Exterior s/n Ciudad Universitaria, 04510 Ciudad de México, Mexico
| |
Collapse
|
27
|
Fu Y, Huang R, Qin G, Huang S, Li J, Zhan W, Bai F, Xie X, Ling J, Cai Y, Xie Y, Wu Y, Cai R, Huang X, Deng Y. TRIM29 promotes liver metastasis via enhancing hepatic colonization by stabilizing FAM83H to regulate keratin network in colorectal cancer. Cell Signal 2025:111871. [PMID: 40389046 DOI: 10.1016/j.cellsig.2025.111871] [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: 04/01/2025] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
Liver metastasis is a frequent and severe event of colorectal cancer (CRC), and patients with liver metastases typically exhibit poor prognosis, high recurrence rates and low responsiveness to treatment. However, the precise molecular mechanisms underlying the liver metastasis in CRC remain poorly understood. In this study, through a comprehensive multi-omics approach, we here identify CRC cells with high tripartite motif-containing protein 29 (TRIM29) expression as the critical subset responsible for liver metastasis. Omics-sequencing pathway analyses combined with in vitro functional assays revealed that CRC cells expressing high TRIM29 expression displayed enhanced cell adhesion, proliferation and liver metastasis capabilities. Mechanistically, TRIM29 interacts with FAM83H and stabilizes it by reducing its ubiquitination and degradation, thereby redistributing cellular keratins, which activates the NF-κB pathway and upregulates PLXNB2 expression to enhance cell adhesion and proliferation to promote hepatic colonization and drive CRC liver metastasis. Interestingly, TRIM29 upregulates the expression of PLXNB2 that can bind to the hepatocyte-specific ligand SEMA4G. Importantly, targeting TRIM29-FAM83H-elicited keratin redistribution and PLXNB2 elevation effectively abrogated CRC liver metastasis. Our findings position TRIM29 as a central driver of liver metastasis in CRC and highlight its potential as a therapeutic target for reducing the risk of liver metastasis in patients.
Collapse
Affiliation(s)
- Yang Fu
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Runqing Huang
- School of Life Science, Huizhou University, Huizhou 516007, China
| | - Ge Qin
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Shishi Huang
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jianxia Li
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Weixiang Zhan
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Fan Bai
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xiaoyu Xie
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jiayu Ling
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yue Cai
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yuqian Xie
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - You Wu
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Runkai Cai
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xinrun Huang
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yanhong Deng
- Department of Medical Oncology, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| |
Collapse
|
28
|
Liang T, He Y, Mo S, Liao Y, Huang K, Gao Q, Shen X, Yang C, Liao X, Qin W, Zhu G, Su H, Ye X, Han C, Peng T. Predictive value of intra-hepatectomy ICGR15 of the remnant liver for post-hepatectomy liver failure in hemi-hepatectomy: a prospective study. BMC Cancer 2025; 25:881. [PMID: 40380127 PMCID: PMC12082978 DOI: 10.1186/s12885-025-14296-5] [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: 02/14/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Post-hepatectomy liver failure (PHLF) is one of the major complications following hepatectomy for hepatocellular carcinoma (HCC). Early identification and precise prediction of PHLF are essential for effective management. This study aimed to evaluate the predictive value of intra-hepatectomy indocyanine green retention rate at 15 min (ICGR15) for the remnant liver for grade B/C PHLF in HCC patients undergoing hemi-hepatectomy. METHODS This prospective study recruited 31 HCC patients who underwent hemi-hepatectomy. ICGR15 was measured at three time points: pre-hepatectomy, intra-hepatectomy (for the remnant liver), and post-hepatectomy. The primary endpoint was the occurrence of grade B/C PHLF according to ISGLS criteria. Logistic regression analysis was employed to evaluate the predictive performance of each parameter and to conduct risk assessment. The XGBoost algorithm was utilized to compare the predictive values of various parameters by calculating the mean Shap values. RESULTS Among the study participants, 25.8% (8 patients) developed grade B/C PHLF. The intra-hepatectomy ICGR15 for remnant liver exhibited the highest predictive accuracy for grade B/C PHLF, with a ROC-AUC of 0.864 and a PR-AUC of 0.791. The optimal threshold for ICGR15-intra was established at 19.8%. Patients with ICGR15-intra value of 19.8% or higher were found at significantly increased risk of grade B/C PHLF (OR[95% CI] = 3.602[1.437-6.750], P value = 0.004), and experienced a higher incidence of severe post-hepatectomy complications. CONCLUSION Intra-hepatectomy ICGR15 for the remnant liver was an important predictor of grade B/C PHLF in patients undergoing hemi-hepatectomy for HCC. An intra-hepatectomy ICGR15 threshold of 19.8% might effectively identify patients at high risk of developing grade B/C PHLF and severe post-hepatectomy complications, helping surgeons' final decision-making on the table.
Collapse
Affiliation(s)
- Tianyi Liang
- Department of Hepatobiliary, Pancreatic and Vascular Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongfei He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Shutian Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Yuan Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Qiang Gao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xiaoqiang Shen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Wei Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China.
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China.
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China.
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China.
| |
Collapse
|
29
|
Wu W, Mao H, Song J, Yang F. Bibliometric analysis of hepatocellular carcinoma and tyrosine kinase inhibitors. Medicine (Baltimore) 2025; 104:e42015. [PMID: 40388796 PMCID: PMC12091622 DOI: 10.1097/md.0000000000042015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 03/13/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignant tumor globally and in China, and its incidence and mortality rate are increasing year by year, and it faces many challenges and difficulties in treatment. Tyrosine kinase inhibitors (TKIs) have important roles in cell growth, proliferation, and differentiation, and have now become important drugs for cancer treatment. There are no bibliometric studies on liver cancer and TKIs to date. METHODS We retrieved 2848 records from the Web of Science™ Core Collection (WoSCC) database and analyzed them scientifically and metrically using CiteSpace and VOSviewer in terms of temporal and spatial distributions, author distributions, journal distributions, references, and keywords. RESULTS From January 1, 2004, to December 31, 2023, the WoSCC database documented 2848 publications related to tyrosinase inhibitors in HCC, comprising 2151 articles and 697 reviews. This literature involved 80 countries and regions, 3265 institutions, and 16,653 authors. Analysis shows a steady increase in publications annually since 2004, divided into 3 phases: 2004 to 2010 with fewer than 100 papers annually, suggesting minimal research attention; 2011 to 2019 with gradual growth, indicating increasing research interest; and a rapid surge post-2020, peaking in 2023, signaling heightened global interest in this field. CONCLUSION Our bibliometric analysis on TKIs and HCC spans years, countries, institutions, authors, disciplines, and journals. Since 2004, this field has gained attention, with current research focusing on inflammatory and immune mechanisms, associated diseases, cytokines, and TKIs' applications in liver cancer treatment, including combination therapies. These areas signify emerging research directions.
Collapse
Affiliation(s)
- Wurihan Wu
- Department of Neurology Department, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hejun Mao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Jian Song
- Emergency Intensive Care Unit, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, China
| | - Fan Yang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
30
|
Di Benedetto G, Varvarà P, Drago SE, Cantone AF, Mauro N, Gaudio G, Burgaletto C, Bellanca CM, Broggi G, Caltabiano R, Pitarresi G, Cantarella G, Giammona G, Bernardini R. Targeted delivery of sorafenib via biotin decorated polyaminoaspartamide-based nanoparticles for the hepatocarcinoma treatment. Int J Pharm 2025; 678:125729. [PMID: 40379225 DOI: 10.1016/j.ijpharm.2025.125729] [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: 04/07/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver cancer, faces treatment challenges due to drug resistance and poor bioavailability, with sorafenib, a key therapy, characterized by rapid clearance and significant side effects. This paper describes the development of amphiphilic graft copolymers for efficient loading and delivery of sorafenib through controlled Atom Transfer Radical Polymerization (ATRP). The amphiphilic graft copolymer PHEA-g-IB-(pButMA)-g-PEG-Bt was synthesized to enhance tumor specificity via biotin-mediated targeting. The synthesis involved a three-step process, with successful functionalization confirmed through NMR and Size Exclusion Chromatography (SEC) analyses. Sorafenib-loaded nanoparticles, prepared via dialysis-based nanoprecipitation, exhibited a mean size of ∼ 300 nm, suitable for oral and parenteral administration, while drug release studies confirmed a sustained release profile, minimizing premature systemic loss and reducing the need for frequent administration. Evaluation of cytocompatibility and anticancer efficacy tested in vitro on HepG2 and HuH-7 cell lines revealed that biotinylated sorafenib-loaded nanoparticles had the highest ability to reduce cell viability. The enhanced anticancer effect of biotinylated NPs was validated in vivo using a murine tumor xenograft model, as evidenced by reduced tumor growth, lower Ki-67 proliferation index, and diminished CD31-positive vasculature. Protein expression analysis demonstrated that PBB-Bt@SOR elicited the strongest activation of p-p38 MAPK and caspase-8-mediated apoptosis, while enhancing the expression of the pro-survival AKT pathway. Overall, the study confirms that biotinylated sorafenib-loaded nanoparticles improve tumor suppression in HCC models, demonstrating their effectiveness in targeted drug delivery. These findings suggest biotin decorated polyamino aspartamide-based nanoparticles as a promising strategy to optimize chemotherapy regimens, minimizing systemic toxicity in HCC treatment.
Collapse
Affiliation(s)
- Giulia Di Benedetto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Paola Varvarà
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Salvatore Emanuele Drago
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Flavia Cantone
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Nicolò Mauro
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Gabriella Gaudio
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Chiara Burgaletto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Carlo Maria Bellanca
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Giuseppe Broggi
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Via Santa Sofia 87, 95123 Catania, Italy
| | - Rosario Caltabiano
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Via Santa Sofia 87, 95123 Catania, Italy
| | - Giovanna Pitarresi
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy.
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.
| | - Gaetano Giammona
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| |
Collapse
|
31
|
Ijaz M, Hasan I, Aslam B, Yan Y, Zeng W, Gu J, Jin J, Zhang Y, Wang S, Xing L, Guo B. Diagnostics of brain tumor in the early stage: current status and future perspectives. Biomater Sci 2025; 13:2580-2605. [PMID: 40200902 DOI: 10.1039/d4bm01503g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Early diagnosis of brain tumors is challenging due to their complexity and delicate structure. Conventional imaging techniques like MRI, CT, and PET are unable to provide detailed visualization of early-stage brain tumors. Early-stage detection of brain tumors is vital for enhancing patient outcomes and survival rates. So far, several scientists have dedicated their efforts to innovating advanced diagnostic probes to efficiently cross the BBB and selectively target brain tumors for optimal imaging. The integration of these techniques presents a viable pathway for non-invasive, accurate, and early-stage tumor identification. Herein, we provide a timely update on the various imaging probes and potential challenges for the diagnosis of early-stage brain tumors. Furthermore, this review highlights the significance of integrating advanced imaging probes for improving the early detection of brain tumors, ultimately enhancing treatment outcomes. Hopefully, this review will stimulate the interest of researchers to accelerate the development of new imaging probes and even their clinical translation for improving the early diagnosis of brain tumors.
Collapse
Affiliation(s)
- Muhammad Ijaz
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Bilal Aslam
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Yuqian Yan
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Wenjun Zeng
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China
| | - Jian Jin
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yinghe Zhang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| | - Shaohua Wang
- Diagnostic Center of Infectious Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China.
| | - Lu Xing
- Department of Sleep Medicine, Shenzhen Kangning Hospital, No. 1080 Cuizhu Road, Guangdong 518020, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of, Technology, Shenzhen-518055, China.
| |
Collapse
|
32
|
Feng S, Zhou M, Huang Z, Xiao X, Zhong B. A machine learning-based prediction model for colorectal liver metastasis. Clin Exp Med 2025; 25:156. [PMID: 40353895 PMCID: PMC12069471 DOI: 10.1007/s10238-025-01699-8] [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: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 05/14/2025]
Abstract
Colorectal liver metastasis (CRLM) is a primary factor contributing to poor prognosis and metastasis in colorectal cancer (CRC) patients. This study aims to develop and validate a machine learning (ML)-based risk prediction model using conventional clinical data to forecast the occurrence of CRLM. This retrospective study analyzed the clinical data of 865 CRC patients between January 2018 and September 2024. Patients were categorized into non-CRLM and CRLM groups. The least absolute shrinkage and selection operator regression was employed to identify key clinical variables, and five ML algorithms were utilized to develop prediction models. The optimal model was selected based on performance metrics including the receiver operating characteristic curve, precision-recall curve, decision curve analysis, and calibration curve, which collectively evaluated both the predictive accuracy and clinical utility of the model. Among the five ML algorithms evaluated, Random forest demonstrated the best performance. Leveraging the Random forest algorithm, we developed the CRLM-Lab6 prediction model, which incorporates six features: LDH, CA199, ALT, CEA, TBIL, and AGR. This model exhibits robust predictive performance, achieving an area under the curve of 0.94, a sensitivity of 0.88, and a specificity of 0.93. To enhance its practical utility, the model has been integrated into an accessible web application. This study developed a novel risk prediction model by integrating ML algorithms with conventional laboratory test data to evaluate the likelihood of CRLM occurrence. The model demonstrates excellent predictive performance and has significant clinical application potential.
Collapse
Affiliation(s)
- Sisi Feng
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Manli Zhou
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zixin Huang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaomin Xiao
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Baiyun Zhong
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
33
|
Wang L, Zhao L, Liu J, Cheng P, Han M, Zheng Z. lncSLERT Promotes Liver Metastasis in Colorectal Cancer by Down-Regulating HUNK Expression via RBM15-Mediated m6A Modification. Onco Targets Ther 2025; 18:631-646. [PMID: 40371234 PMCID: PMC12075449 DOI: 10.2147/ott.s514001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025] Open
Abstract
Background Metastasis is a hallmark of cancer and the leading cause of cancer-related mortality. However, the mechanism underlying liver metastasis in colorectal cancer (CRC) remains incompletely understood. This study explores the role of long non-coding RNA (lncRNA) SLERT in promoting CRC liver metastasis by downregulating HUNK expression. Methods SLERT expression levels in CRC tissues were analyzed and correlated with patient survival outcomes. Functional assays, including migration and invasion assays, were performed to assess the impact of SLERT knockdown and overexpression on metastatic behavior. Mechanistic studies examined SLERT's interaction with the RNA-binding protein RBM15 and its effect on HUNK mRNA stability. The subcellular localization of SLERT was also determined. Results SLERT was significantly upregulated in CRC tissues and associated with poor survival outcomes. Silencing SLERT inhibited CRC cell migration and invasion, whereas its overexpression enhanced these metastatic properties. Mechanistically, SLERT interacted with RBM15, impairing its ability to stabilize HUNK mRNA, leading to decreased HUNK expression and increased metastatic potential. SLERT was primarily localized in the cytoplasm, indicating its active role in gene regulation within the tumor microenvironment. Conclusion LERT promotes liver metastasis in CRC by downregulating HUNK expression through RBM15-mediated mRNA destabilization. These findings suggest that SLERT could serve as a diagnostic biomarker and therapeutic target. Targeting SLERT or restoring HUNK expression may provide novel strategies to combat CRC liver metastasis and improve patient prognosis.
Collapse
Affiliation(s)
- Lin Wang
- The State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Liming Zhao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Jialiang Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Pu Cheng
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Mingyu Han
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Zhaoxu Zheng
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
34
|
Liu X, Haugh W, Zhang Z, Huang J. Emerging Role of Long, Non-Coding RNA Nuclear-Enriched Abundant Transcript 1 in Stress- and Immune-Related Diseases. Int J Mol Sci 2025; 26:4413. [PMID: 40362651 PMCID: PMC12072541 DOI: 10.3390/ijms26094413] [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: 04/08/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025] Open
Abstract
Long, non-coding RNAs (lncRNAs) are a class of RNAs exceeding 200 nucleotides in length, lacking the ability to be translated into proteins. Over the past few decades, an increasing number of publications have established lncRNAs as potent regulators in a broad spectrum of diseases. They modulate the expression of critical genes by affecting transcription, post-transcription, translation, and protein modification. This regulation frequently involves the interaction of lncRNAs with various molecules, such as proteins, RNA, and DNA. lncRNAs are involved in diseases where stress is a significant factor. In recent years, lncRNAs have been identified as regulators of both innate and adaptive immune responses, playing significant roles in the onset and progression of diseases. Additionally, lncRNAs hold potential as biomarkers or therapeutic targets for numerous stress- and immune-related diseases. lncRNA nuclear-enriched abundant transcript 1 (NEAT1) is a notable example. This review consolidates the latest findings about the role of lncRNA NEAT1 in stress response and immune cell function in non-cancer diseases. It summarizes studies on NEAT1 regulating stress response, both innate and adaptive immunity, and its potential as a biomarker and therapeutic target for stress- and immune-related diseases.
Collapse
Affiliation(s)
- Xingliang Liu
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR 97213, USA; (X.L.); (W.H.)
| | - William Haugh
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR 97213, USA; (X.L.); (W.H.)
| | - Ziqiang Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jianguo Huang
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR 97213, USA; (X.L.); (W.H.)
| |
Collapse
|
35
|
Pawar P, Prabhu A. Smart SPIONs for Multimodal Cancer Theranostics: A Review. Mol Pharm 2025; 22:2372-2391. [PMID: 40223773 DOI: 10.1021/acs.molpharmaceut.5c00411] [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] [Indexed: 04/15/2025]
Abstract
Despite significant advancements in anticancer research, the performance statistics of current therapeutic regimens yield unsatisfactory outcomes. Issues such as high metastasis rates, drug resistance, limited efficacy, and severe side effects underscore the urgent need for safer and more effective strategies for tumor mitigation. One promising approach lies in the use of superparamagnetic iron oxide nanoparticles (SPIONs) for hybridized cancer therapy, leveraging their unique properties and functional versatility to enhance treatment efficacy and safety. They can serve as platforms for various therapeutic as well as diagnostic applications, enhancing imaging techniques such as magnetic resonance imaging. This paper presents an in-depth compilation of the application of nanoparticulate SPIONs amalgamates for multimodal cancer therapeutics. Physical phenomena such as light, heat, sound, and magnetism can be coupled to nanoparticulate delivery systems for developing targeted, precision medicine against cancer. Integration of noninvasive and effective platforms technologies such as photodynamic therapy, photothermal therapy, magnetic hyperthermia, and sonodynamic therapy hold great promise in counteracting the daunting challenges within cancer therapeutics.
Collapse
Affiliation(s)
- Pradip Pawar
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, 400056 Mumbai, India
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, 400056 Mumbai, India
| |
Collapse
|
36
|
Tang T, Guo T, Zhu B, Tian Q, Wu Y, Liu Y. Interpretable machine learning model for predicting post-hepatectomy liver failure in hepatocellular carcinoma. Sci Rep 2025; 15:15469. [PMID: 40316613 PMCID: PMC12048636 DOI: 10.1038/s41598-025-97878-4] [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: 10/20/2024] [Accepted: 04/08/2025] [Indexed: 05/04/2025] Open
Abstract
Post-hepatectomy liver failure (PHLF) is a severe complication following liver surgery. We aimed to develop a novel, interpretable machine learning (ML) model to predict PHLF. We enrolled 312 hepatocellular carcinoma (HCC) patients who underwent hepatectomy, and 30% of the samples were utilized for internal validation. Variable selection was performed using the least absolute shrinkage and selection operator regression in conjunction with random forest and recursive feature elimination (RF-RFE) algorithms. Subsequently, 12 distinct ML algorithms were employed to identify the optimal prediction model. The area under the receiver operating characteristic curve, calibration curves, and decision curve analysis (DCA) were utilized to assess the model's predictive accuracy. Additionally, an independent prospective validation was conducted with 62 patients. The SHapley Additive exPlanations (SHAP) analysis further explained the extreme gradient boosting (XGBoost) model. The XGBoost model exhibited the highest accuracy with AUCs of 0.983 and 0.981 in the training and validation cohorts among 12 ML models. Calibration curves and DCA confirmed the model's accuracy and clinical applicability. Compared with traditional models, the XGBoost model had a higher AUC. The prospective cohort (AUC = 0.942) further confirmed the generalization ability of the XGBoost model. SHAP identified the top three critical variables: total bilirubin (TBIL), MELD score, and ICG-R15. Moreover, the SHAP summary plot was used to illustrate the positive or negative effects of the features as influenced by XGBoost. The XGBoost model provides a good preoperative prediction of PHLF in patients with resectable HCC.
Collapse
Affiliation(s)
- Tianzhi Tang
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Tianyu Guo
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Bo Zhu
- Department of Cancer Prevention and Treatment, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Qihui Tian
- Department of Cancer Prevention and Treatment, Cancer Hospital of China Medical University/Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Yang Wu
- Medical Oncology Department of Thoracic Cancer (2), Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning Province, People's Republic of China.
| | - Yefu Liu
- Department of Hepatobiliary and Pancreatic Surgery, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, People's Republic of China.
| |
Collapse
|
37
|
Lu D, Xia B, Feng T, Qi G, Ma Z. The Role of Cancer Organoids in Ferroptosis, Pyroptosis, and Necroptosis: Functions and Clinical Implications. Biomolecules 2025; 15:659. [PMID: 40427552 PMCID: PMC12108908 DOI: 10.3390/biom15050659] [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: 04/04/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
The enduring prevalence of cancer worldwide constitutes a significant public health challenge, thereby emphasizing the imperative for the development of therapeutic models capable of accounting for the heterogeneity inherent in tumors. In this context, cancer organoids have emerged as powerful tools for studying tumor biology, providing valuable insights into the complex interactions within the tumor microenvironment. Concurrently, research is increasingly focused on non-apoptotic forms of regulated cell death (RCD)-including ferroptosis, pyroptosis, and necroptosis-which exert pivotal influences on cancer development and progression. Cancer organoids not only recapitulate the genetic and phenotypic heterogeneity of the original tumors but also enable more precise investigations into the roles of non-apoptotic RCDs within oncology. This review explores the utility of cancer organoids in delineating the molecular mechanisms underlying RCDs and their implications for cancer biology and treatment responses. By synthesizing recent research findings, it highlights the essential role of organoid models in uncovering the intricate details of non-apoptotic RCDs. Furthermore, it emphasizes promising directions for future research that aim to deepen our understanding of these pathways and their therapeutic potential. The integration of organoid models into investigations of ferroptosis, pyroptosis, and necroptosis provides novel insights into oncogenic mechanisms and facilitates the development of targeted therapeutic strategies. By bridging cancer organoids with human pathophysiology, this approach not only provides a transformative framework for dissecting oncogenic pathways but also enables the design of precision therapeutics that selectively target the molecular machinery underlying non-apoptotic RCDs.
Collapse
Affiliation(s)
- Dingci Lu
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
- Department of Clinical Laboratory, The Second Hospital of Jingzhou, Jingzhou 434000, China
| | - Bingqian Xia
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Tianquan Feng
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Gui Qi
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Zhaowu Ma
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| |
Collapse
|
38
|
Zhang B, Wang K, Xu T, Zhu H, Wang K, Wang J, Xiang Y, He X, Zhu S, An C, Yan D. Deep learning model for predicting the RAS oncogene status in colorectal cancer liver metastases. J Cancer Res Ther 2025; 21:362-370. [PMID: 40317140 DOI: 10.4103/jcrt.jcrt_1910_24] [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: 09/30/2023] [Accepted: 12/09/2024] [Indexed: 05/07/2025]
Abstract
BACKGROUND To develop a deep learning radiomics (DLR) model based on contrast-enhanced computed tomography (CECT) to assess the rat sarcoma (RAS) oncogene status and predict targeted therapy response in colorectal cancer liver metastases (CRLM). METHODS This multicenter retrospective study comprised 185 CRLM patients who were categorized into three cohorts: training (n = 88), internal test (n = 39), and external test (n = 58). A total of 1126 radiomic features and 2589 DL signatures were extracted from each region of interest in the CECT. Fourteen significant radiomic features associated with RAS mutation were selected. Subsequently, various models (DL-arterial phase (AP), DL-venous phase (VP), AP+VP-DL, radiomics, and DL-R) were developed and validated. The model performance was compared using the area under the receiver operating characteristic (AUROC) curves and the DeLong test. The predictive usefulness of the DL score for progression-free survival and overall survival (OS) was determined. RESULTS The AP+VP-DL model achieved the highest AUC (0.98), outperforming the radiomics (0.90), DL-AP (0.93), DL-VP (0.87), and DL-R (0.97) models. Significant associations were observed between OS and the carcinoembryonic antigen (CEA), disease control rate (DCR), and DL scores, leading to the development of a DL nomogram. A high-risk RAS mutation status correlated with significantly lower 1-year (88% vs. 96%), 3-year (12% vs. 35%), and 5-year (0% vs. 15%) cumulative survival rates compared to a low-risk status (P = 0.03). CONCLUSIONS The DL model demonstrated satisfactory predictive performance, aiding clinicians in noninvasively predicting the RAS gene status for informed treatment decisions.
Collapse
Affiliation(s)
- Baogen Zhang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| | - Kai Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| | - Ting Xu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Hai-Dian District, Beijing, China
| | - Haibin Zhu
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Kangjie Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| | - Jing Wang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| | - Yaoxian Xiang
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| | - Xuelei He
- School of Information Science and Technology, Northwest University, No. 1 Xuefu Avenue, Chang'an District, Xi'an, Shaanxi Province, P.R. China
| | - Siyu Zhu
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| | - Chao An
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| | - Dong Yan
- Department of Oncology, Beijing Luhe Hospital Affiliated to Capital Medical University. Xinhua South Road, Tongzhou District, Beijing, China
| |
Collapse
|
39
|
Li H, Zeng J, You Q, Zhang M, Shi Y, Yang X, Gu W, Liu Y, Hu N, Wang Y, Chen X, Mu J. X-ray-activated nanoscintillators integrated with tumor-associated neutrophils polarization for improved radiotherapy in metastatic colorectal cancer. Biomaterials 2025; 316:123031. [PMID: 39709848 DOI: 10.1016/j.biomaterials.2024.123031] [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: 08/15/2024] [Revised: 11/26/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
Radiotherapy, employing high-energy rays to precisely target and eradicate tumor cells, plays a pivotal role in the treatment of various malignancies. Despite its therapeutic potential, the effectiveness of radiotherapy is hindered by the tumor's inherent low radiosensitivity and the immunosuppressive microenvironment. Here we present an innovative approach that integrates peroxynitrite (ONOO-)-mediated radiosensitization with the tumor-associated neutrophils (TANs) polarization for the reversal of immunosuppressive tumor microenvironment (TME), greatly amplifying the potency of radiotherapy. Our design employs X-ray-activated lanthanide-doped scintillators (LNS) in tandem with photosensitive NO precursor to achieve in-situ ONOO- generation. Concurrently, the co-loaded TGF-β inhibitor SB525334, released from the LNS-RS nanoplatform in response to the overexpressed GSH in tumor site, promotes the reprogramming of TANs from N2 phenotype toward N1 phenotype, effectively transforming the tumor-promoting microenvironment into a tumor-inhibiting state. This 'one-two punch' therapy efficiently trigger a robust anti-tumor immune response and exert potent therapeutic effects in orthotopic colorectal cancer and melanoma mouse model. Meanwhile, it also significantly prevents liver metastasis and recurrence in metastatic colorectal cancer. The development of X-ray-controlled platforms capable of activating multiple therapeutic modalities may accelerate the clinical application of radiotherapy-based collaborative therapy.
Collapse
Affiliation(s)
- Hui Li
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China; Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Junyi Zeng
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
| | - Miaomiao Zhang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Yuanchao Shi
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Xiaodong Yang
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Wenxing Gu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China; Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Yajie Liu
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Ning Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Yu Wang
- Institute of Microscale Optoelectronics, Shenzhen University, 518060, Shenzhen, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
| | - Jing Mu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China.
| |
Collapse
|
40
|
Jeong B, Heo S, Lee SS, Kim SO, Shin YM, Kim KM, Ha TY, Jung DH. Predicting post-hepatectomy liver failure in patients with hepatocellular carcinoma: nomograms based on deep learning analysis of gadoxetic acid-enhanced MRI. Eur Radiol 2025; 35:2769-2782. [PMID: 39528755 DOI: 10.1007/s00330-024-11173-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: 04/22/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES This study aimed to develop nomograms for predicting post-hepatectomy liver failure (PHLF) in patients with hepatocellular carcinoma (HCC), using deep learning analysis of Gadoxetic acid-enhanced hepatobiliary (HBP) MRI. METHODS This retrospective study analyzed patients who underwent gadoxetic acid-enhanced MRI and hepatectomy for HCC between 2016 and 2020 at two referral centers. Using a deep learning algorithm, volumes and signal intensities of whole non-tumor liver, expected remnant liver, and spleen were measured on HBP images. Two multivariable logistic regression models were formulated to predict PHLF, defined and graded by the International Study Group of Liver Surgery: one based on whole non-tumor liver measurements (whole liver model) and the other on expected remnant liver measurements (remnant liver model). The models were presented as nomograms and a web-based calculator. Discrimination performance was evaluated using the area under the receiver operating curve (AUC), with internal validation through 1000-fold bootstrapping. RESULTS The study included 1760 patients (1395 male; mean age ± standard deviation, 60 ± 10 years), with 137 (7.8%) developing PHLF. Nomogram predictors included sex, gamma-glutamyl transpeptidase, prothrombin time international normalized ratio, platelets, extent of liver resection, and MRI variables derived from the liver volume, liver-to-spleen signal intensity ratio, and spleen volume. The whole liver and the remnant liver nomograms demonstrated strong predictive performance for PHLF (optimism-corrected AUC of 0.78 and 0.81, respectively) and symptomatic (grades B and C) PHLF (optimism-corrected AUC of 0.81 and 0.84, respectively). CONCLUSION Nomograms based on deep learning analysis of gadoxetic acid-enhanced HBP images accurately stratify the risk of PHLF. KEY POINTS Question Can PHLF be predicted by integrating clinical and MRI-derived volume and functional variables through deep learning analysis of gadoxetic acid-enhanced MRI? Findings Whole liver and remnant liver nomograms demonstrated strong predictive performance for PHLF with the optimism-corrected area under the curve of 0.78 and 0.81, respectively. Clinical relevance These nomograms can effectively stratify the risk of PHLF, providing a valuable tool for treatment decisions regarding hepatectomy for HCC.
Collapse
Affiliation(s)
- Boryeong Jeong
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Subin Heo
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Radiology, Ajou University School of Medicine, Suwon, Korea
| | - Seung Soo Lee
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Seon-Ok Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong Moon Shin
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kang Mo Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae-Yong Ha
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong-Hwan Jung
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
Wang Y, Li T, Dong Z, Zhang Q, Mi J, Wang Q, Lin G, Ma Q, Jia R, Huang S. Extracellular Vesicles From Lactobacillus fermentum Enhance Intestinal Barrier Integrity and Restore Gut Microbial Homeostasis in Experimental Murine Colitis. J Nutr 2025; 155:1311-1323. [PMID: 40058701 DOI: 10.1016/j.tjnut.2025.03.001] [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: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Lactobacillus fermentum has been shown to improve intestinal health and treat colitis; however, its precise efficacy and mechanisms in inflammatory bowel disease remain unclear. OBJECTIVES This study aimed to evaluate whether L fermentum and its metabolites, extracellular vesicles, and other components could modulate intestinal barrier function and gut microbiota to alleviate dextran sulfate sodium (DSS)-induced colitis in mice. METHODS Forty-eight mice were randomly assigned to 6 groups: control, DSS, L fermentum+DSS group (LF+DSS), heat-inactivated L fermentum+DSS group (LHF+DSS), L fermentum supernatant solution+DSS group (LSF+DSS), and L fermentum extracellular vesicles+DSS group (LEV+DSS). After a 1-wk acclimation, mice were gavaged daily for 3 wk. Fresh cultures, including live (LF+DSS), heat-inactivated (LHF+DSS), supernatant (LSF+DSS), and extracellular vesicles (LEV+DSS), were prepared daily. During the final 7 d, the control group received normal water, and the other groups received 3% DSS. Data were collected daily, followed by sample collection from the mice. RESULTS In this study, significant reductions (P < 0.05) in body weight changes, disease activity index, intestinal damage, and histology scores were observed in the treatment groups, especially LEV+DSS and LF+DSS. Additionally, compared with the DSS group, colonic mucus secretion, as well as claudin-1 and occludin expression, increased significantly (P < 0.05) in the LEV+DSS and LF+DSS groups, whereas proinflammatory cytokines IL-1β and TNF-α decreased (P < 0.05) and IL-10 increased (P < 0.05) in the LEV+DSS group. L fermentum and its components significantly regulated gut microbiota α-diversity and β-diversity, affecting overall composition. Linear discriminant analysis effect size analysis revealed an enrichment of beneficial bacteria including Prevotellaceae_UCG-001, Romboutsia, and Ruminococcus species in the LF+DSS group and Akkermansia, Odoribacter, and Marvinbryantia species in the LEV+DSS group. Both L fermentum and its extracellular vesicles significantly downregulated the gene expression of TNF-α and IL-1β, whereas the expression of IL-10 was upregulated, thereby contributing to the alleviation of colitis symptoms. CONCLUSIONS This study reveals that L fermentum alleviates colitis through modulation of the gut microbiota and reinforcement of the intestinal mucosal barrier, with its extracellular vesicles potentially playing a key role in this regulatory process.
Collapse
Affiliation(s)
- Yanwei Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; School of Life Science, Shanxi University, Taiyuan, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Zhuo Dong
- Hubei International Travel Healthcare Center, Hubei, China
| | - Qiyue Zhang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jingqiu Mi
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Qingfeng Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Gang Lin
- Institute of Quality Standards and Testing Technology for Agricultural Products, Chinese Academy of Agricultural Science, Beijing, China
| | - Qiugang Ma
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Ru Jia
- School of Life Science, Shanxi University, Taiyuan, China.
| | - Shimeng Huang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China.
| |
Collapse
|
42
|
Santol J, Kim S, Gregory LA, Baumgartner R, Murtha-Lemekhova A, Birgin E, Gloor S, Braunwarth E, Ammann M, Starlinger J, Pereyra D, Ammon D, Ninkovic M, Kern AE, Rumpf B, Ortmayr G, Herrmann Y, Dong Y, Huber FX, Weninger J, Thiels CA, Warner SG, Smoot RL, Truty MJ, Kendrick ML, Nagorney DM, Cleary SP, Beldi G, Rahbari NN, Hoffmann K, Gilg S, Assinger A, Gruenberger T, Hackl H, Starlinger P. An APRI+ALBI-Based Multivariable Model as a Preoperative Predictor for Posthepatectomy Liver Failure. Ann Surg 2025; 281:861-871. [PMID: 37860868 PMCID: PMC11974630 DOI: 10.1097/sla.0000000000006127] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
OBJECTIVE AND BACKGROUND Clinically significant posthepatectomy liver failure (PHLF B+C) remains the main cause of mortality after major hepatic resection. This study aimed to establish an aspartate aminotransferase to platelet ratio combined with an albumin-bilirubin grade (APRI+ALBI), based multivariable model (MVM) to predict PHLF and compare its performance to indocyanine green clearance (ICG-R15 or ICG-PDR) and albumin-ICG evaluation (ALICE). METHODS A total of 12,056 patients from the National Surgical Quality Improvement Program database were used to generate a MVM to predict PHLF B+C. The model was determined using stepwise backwards elimination. The performance of the model was tested using receiver operating characteristic curve analysis and validated in an international cohort of 2525 patients. In 620 patients, the APRI+ALBI MVM, trained in the National Surgical Quality Improvement Program cohort, was compared with the MVM's based on other liver function tests (ICG clearance, ALICE) by comparing the areas under the curve (AUC). RESULTS A MVM including APRI+ALBI, age, sex, tumor type, and extent of resection was found to predict PHLF B+C with an AUC of 0.77, with comparable performance in the validation cohort (AUC: 0.74). In direct comparison with other MVM's based on more expensive and time-consuming liver function tests (ICG clearance, ALICE), the APRI+ALBI MVM demonstrated equal predictive potential for PHLF B+C. A smartphone application for the calculation of the APRI+ALBI MVM was designed. CONCLUSION Risk assessment through the APRI+ALBI MVM for PHLF B+C increases preoperative predictive accuracy and represents a universally available and cost-effective risk assessment before hepatectomy, facilitated by a freely available smartphone app.
Collapse
Affiliation(s)
- Jonas Santol
- Department of Surgery, HPB Center, Vienna Health Network, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
- Institute of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sarang Kim
- Medical University of Vienna, Vienna, Austria
| | - Lindsey A. Gregory
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ruth Baumgartner
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Anastasia Murtha-Lemekhova
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Emrullah Birgin
- Department of Surgery, University Medicine Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Severin Gloor
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Eva Braunwarth
- Department of Visceral, Transplantation and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Ammann
- Department of Surgery, State Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | | | - David Pereyra
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Daphni Ammon
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Marijana Ninkovic
- Department of Visceral, Transplantation and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna E. Kern
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Benedikt Rumpf
- Department of Surgery, Hospital Barmherzige Schwestern, Vienna, Austria
| | - Gregor Ortmayr
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | | | - Yawen Dong
- Department of Surgery, HPB Center, Vienna Health Network, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
| | | | | | - Cornelius A. Thiels
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Susanne G. Warner
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Rory L. Smoot
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Mark J. Truty
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Michael L. Kendrick
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - David M. Nagorney
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sean P. Cleary
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
- Division of General Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Guido Beldi
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nuh N. Rahbari
- Department of Surgery, University Medicine Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Katrin Hoffmann
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Gilg
- Department of HPB surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Gruenberger
- Department of Surgery, HPB Center, Vienna Health Network, Clinic Favoriten and Sigmund Freud Private University, Vienna, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrick Starlinger
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
Sun Z, Xu H, Lu G, Yang C, Gao X, Zhang J, Liu X, Chen Y, Wang K, Guo J, Li J. AKT1 Phosphorylates FDX1 to Promote Cuproptosis Resistance in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408106. [PMID: 39976173 PMCID: PMC12061301 DOI: 10.1002/advs.202408106] [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] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/23/2025] [Indexed: 02/21/2025]
Abstract
Cuproptosis, a recently defined copper-dependent cell death pathway, remains largely unexplored in tumor therapies, particularly in breast cancer. This study demonstrates that triple-negative breast cancer (TNBC) bears a relatively elevated copper levels and exhibits resistance to cuproptosis. Mechanistically, copper activates the AKT signaling pathway, which inhibits ferredoxin-1 (FDX1), a key regulator of cuproptosis. AKT1-mediated FDX1 phosphorylation not only abrogates FDX1-induced cuproptosis and aerobic respiration but also promotes glycolysis. Consequently, the combination of AKT1 inhibitors and the copper ionophores synergistically alleviate TNBC tumorigenesis both in vitro and in vivo. In summary, the findings reveal a crucial mechanism underlying TNBC resistance to cuproptosis and suggest a potential therapeutic approach for TNBC.
Collapse
Affiliation(s)
- Zicheng Sun
- Department of Breast and Thyroid SurgeryGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Huazhen Xu
- Department of Breast and Thyroid SurgeryGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Guanming Lu
- Department of Breast and Thyroid SurgeryAffiliated Hospital of Youjiang Medical University for Nationalities and Key Laboratory of Molecular Pathology in Tumors of GuangxiGuangxi533000China
| | - Ciqiu Yang
- Department of Breast CancerGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdong510000China
| | - Xinya Gao
- Department of Breast and Thyroid SurgeryGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Jing Zhang
- Department of Breast and Thyroid SurgeryGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Xin Liu
- Department of Breast and Thyroid SurgeryGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Yongcheng Chen
- Department of Breast and Thyroid SurgeryAffiliated Hospital of Youjiang Medical University for Nationalities and Key Laboratory of Molecular Pathology in Tumors of GuangxiGuangxi533000China
| | - Kun Wang
- Department of Breast CancerGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdong510000China
| | - Jianping Guo
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000China
| | - Jie Li
- Department of Breast and Thyroid SurgeryGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510000China
| |
Collapse
|
44
|
Saravanan N, Sundaramurthy A, Park S. MoS 2 nanoflowers surface decorated with CuS nanorods and carbon dots for fluorescent and ultrasound imaging in cancer therapy. Colloids Surf B Biointerfaces 2025; 249:114503. [PMID: 39823948 DOI: 10.1016/j.colsurfb.2025.114503] [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: 09/24/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025]
Abstract
In recent years, the design of various ultrasound responsive echogenic nanomaterials offers many advantages such as deep tissue penetration, high signal intensity, colloidal stability, biocompatibility and less expensive for ultrasound-based cancer cell imaging while providing the option to monitor the progress of tumor volume during the treatment. Further, the ability of nanomaterials to combine photo-thermal therapy (PTT) and chemotherapy has opened a new avenue in the development of cancer theranostics for synergistic cancer therapy. Herein, we report MoS2 nanoflowers (NFs) surface decorated with CuS nanorods (NRs) and folic acid-derived carbon dots (FACDs) using cystine-polyethyleneimine (PEI) linker for PTT-chemotherapy. The size of NFs was found to be 350 ± 50 nm which increased to 500 ± 50 nm after surface decoration. The morphology of MoS2 NFs before and after surface decoration was investigated using scanning electron microscopy, X-ray diffraction, X-ray photoelectron spectroscopy, Fourier transform infrared spectroscopy, and UV-Vis-NIR spectroscopy. The photo-thermal heat generation was found to be increasing as a function of the concentration of NFs. The encapsulation efficiency of doxorubicin (DOX) and photo-thermal conversion efficiency (PCE) for surface-decorated MoS2 NFs (MoS2@CuS/FACDs NFs) was estimated to be 42 and 44 %, respectively. The surface decoration of CuS NRs and FACDs on MoS2 NFs not only improved the anticancer activity but also increased the signal intensity in ultrasound and fluorescence imaging of cancer cells. The MoS2@CuS/FACDs NFs exhibited excellent cytotoxicity against MDA-MB-231 cancer cells. Hence, the hybrid system demonstrated here showed high potential for use as a combined probe for non-invasive ultrasound imaging and fluorescence imaging for PTT-chemotherapy.
Collapse
Affiliation(s)
- Nishakavya Saravanan
- Department of Physics and Nanotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603203, India
| | - Anandhakumar Sundaramurthy
- Biomaterials Research Laboratory (BMRL), Department of Chemical Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603203, India; Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea.
| | - Sukho Park
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea.
| |
Collapse
|
45
|
Kim SJ, Jeong WK, Han HJ, Choi GS, Kim KH, Kim J. Comparison of initial treatments for resectable hepatocellular carcinoma within Milan criteria: an observational study based on a nationwide survey. Ann Surg Treat Res 2025; 108:279-294. [PMID: 40352802 PMCID: PMC12059244 DOI: 10.4174/astr.2025.108.5.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 05/14/2025] Open
Abstract
Purpose Treatment options for hepatocellular carcinoma (HCC) vary according to known guidelines among liver resection (LR), liver transplantation (LT), radiofrequency ablation (RFA), and transarterial chemoembolization (TACE). This study aimed to compare the outcomes of initial treatment for patients with resectable HCC within Milan criteria (MC) via nationwide data. Methods Patients with resectable HCC (Child-Pugh class A; platelet count, ≥100,000/µL) within MC from the Korean Liver Cancer Association databank were analyzed, retrospectively. Outcomes according to initial treatment and subgroups according to tumor size and number were analyzed. Overall survival (OS) rates after initial treatment were compared. Results A total of 3,241 patients who underwent LR (n = 1,371), LT (n = 12), RFA (n = 679), or TACE (n = 1,179) were included. The 5-year OS rates differed significantly between the groups (P < 0.05), except for LT (LR, 84.9%; LT, 82.5%; RFA, 76.2%; and TACE, 59.9%). For patients with a single tumor of any size, the 5-year OS rates of the LR group were significantly higher than RFA and TACE groups. For patients with multiple tumors, the 5-year OS rates were 78.2%, 100%, 74.3%, and 53.0% for the LR, LT, RFA, and TACE groups, respectively, but without significant difference between LR and RFA (P = 0.86). Conclusion For resectable HCC within MC, the LR had the highest OS rate for a single tumor of any size. LR and RFA showed no significant differences in OS rate for multiple tumors. LR has a much more optimistic outlook for HCC within MC.
Collapse
Affiliation(s)
- Sang Jin Kim
- Division of Hepatobiliary-Pancreas and Transplant Surgery, Department of Surgery, Korea University Ansan Hospital, Ansan, Korea
| | - Woo Kyoung Jeong
- Department of Radiology and Center for Imaging Sciences, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyung-Joon Han
- Division of Hepatobiliary-Pancreas and Transplant Surgery, Department of Surgery, Korea University Ansan Hospital, Ansan, Korea
| | - Gyu-Seong Choi
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyun-Hwan Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Jongman Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Li ZF, Zhang JN, Tian S, Sun C, Ma Y, Ye ZX. Dual-Time-Point Radiomics for Prognosis Prediction in Colorectal Liver Metastasis Treated with Neoadjuvant Therapy Before Radical Resection: A Two-Center Study. Ann Surg Oncol 2025; 32:3516-3525. [PMID: 39907877 DOI: 10.1245/s10434-025-16941-6] [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: 11/19/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Optimal prognostic stratification for colorectal liver metastases (CRLM) patients undergoing surgery with neoadjuvant therapy (NAT) remains elusive. This study aimed to develop and validate dual-time-point radiomic models for CRLM prognosis prediction using pre- and post-NAT imaging features. METHODS Radiomic features were extracted from four MRI sequences in 100 cases of CRLM patients who underwent NAT and radical resection. RAD scores were generated, and clinical/pathologic variables were incorporated into uni- and multivariate Cox regression analyses to construct prognosis models. Time-ROC, time-C index, decision curve analysis (DCA), and calibration curves assessed the predictive performance of Fong score and pre- and post-NAT models for overall survival (OS) and disease-free survival (DFS) in a testing set. RESULTS The final models included four variables for OS and three variables for DFS. The post-NAT models outperformed the pre-NAT models in time-ROC, time-C index, calibration, and DCA analysis, except for the 1-year DFS area under the curve (AUC). The Fong score models underperformed. The post-NAT OS RAD score effectively stratified patients into prognostic subgroups. CONCLUSIONS The radiomic models incorporating pre- and post-NAT MRI features and clinical/pathologic variables effectively stratified CRLM patients prognositically. The post-NAT models demonstrated superior performance.
Collapse
Affiliation(s)
- Zhuo-Fu Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | - Jia-Ning Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | | | - Chao Sun
- Department of Radiology, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Ying Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhao-Xiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China.
| |
Collapse
|
47
|
Xuan Z, Wu Z, Cheng L, Jiang J, Zhang Y, Xia Y. SCGB3A1-Epi and KLK10-Epi Crosstalk With Fibroblasts Promotes Liver Metastasis of Breast Cancer and Pancreatic Ductal Adenocarcinoma. Cancer Med 2025; 14:e70904. [PMID: 40357856 PMCID: PMC12070254 DOI: 10.1002/cam4.70904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/21/2025] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The liver often serves as the principal site for metastatic spread from a variety of solid tumors, and metastasis to the liver markedly diminishes patient survival. Single-cell RNA sequencing (scRNA-seq) has helped uncover the complexity of liver tumor metastasis. However, the key cellular subtypes of breast cancer and pancreatic ductal adenocarcinoma (PDAC) with liver metastasis and their mechanisms of action are unclear, making treatment difficult. METHODS We used integrated scRNA-seq data to dissect liver metastasis-specific epithelial cell subtypes in breast cancer and PDAC, and elucidated their mechanisms through functional analyses and intercellular interactions with fibroblasts. RESULTS Interestingly, our results show that SCGB3A1-Epi and KLK10-Epi are key drivers of liver metastasis in breast cancer and PDAC, respectively. These subtypes are associated with high malignancy rates and involved in oxidative phosphorylation and other critical pathways. Specific ligand-receptor interactions were observed between these epithelial subtypes and fibroblasts, with significant interactions between CD74-APP receptors in SCGB3A1-Epi and Fib-11 in breast cancer and between SPP1-CD44 receptors in KLK10-Epi and Fib-11 in PDAC. High expression levels of Fib-11 and CD74 were correlated with improved survival in breast cancer, whereas high SPP1 and CD44 expression predicted worse PDAC outcomes. Fib-11 is implicated in signaling pathways associated with tumor metastasis, particularly those involving cell adhesion molecules. CONCLUSIONS We revealed the cellular heterogeneity of liver metastasis and provided a crucial research foundation for developing novel therapeutic strategies to specifically target metastatic cell subtypes, thereby enhancing patient prognosis.
Collapse
Affiliation(s)
- Zixue Xuan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
- Department of PharmacyZhejiang Provincial People's Hospital Bijie HospitalBijieGuizhouChina
| | - Zhongxiu Wu
- Department of PharmacyZhejiang Provincial People's Hospital Bijie HospitalBijieGuizhouChina
| | - Lei Cheng
- Department of PharmacyZhejiang Provincial People's Hospital Bijie HospitalBijieGuizhouChina
| | - Jinying Jiang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
- Department of PharmacyZhejiang Provincial People's Hospital Bijie HospitalBijieGuizhouChina
| | - Yuan Zhang
- Department of PharmacyZhejiang Provincial People's Hospital Bijie HospitalBijieGuizhouChina
| | - Yuxuan Xia
- Outpatient Department, Zhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhouZhejiangChina
| |
Collapse
|
48
|
Shi P, Xu J, Cui H. Targeting oxygenases could be a viable anti-metastatic approach in cancer therapy. Int J Biol Macromol 2025; 310:143375. [PMID: 40268020 DOI: 10.1016/j.ijbiomac.2025.143375] [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: 02/28/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
Malignant tumors are characterized by irregular boundaries, rapid and uncontrolled cell growth, the ability to invade surrounding tissues, and the potential to spread and metastasize to other parts of the body through the bloodstream or lymphatic system. More than 90 % of cancer-related deaths are attributed to the metastasis of cancer cells. When malignant tumors metastasize, the metabolic processes within the cells undergo significant changes, with enzymes playing a crucial role in regulating metabolism and serving as key mediators in both synthesis and degradation. Oxygenases are a group of oxidative enzymes that catalyze the incorporation of oxygen atoms into various substrates. Advances in our understanding of the genome and proteome of malignant tumors have revealed that oxygenases are highly expressed in many metastatic tumor cells, where they can enhance the activity of specific proteins that regulate tumor metastasis. Furthermore, there is a growing recognition that certain drugs can specifically target oxygenases to inhibit tumor metastasis, with several of these agents are currently undergoing clinical evaluation. In this context, we summarize the mechanisms by which oxygenases influence cancer cell behavior, along with the preclinical and clinical studies related to targeted therapies involving oxygenases.
Collapse
Affiliation(s)
- Pengfei Shi
- Jinfeng Laboratory, 401329 Chongqing, China; Cancer Center, Medical Research Institute, Southwest University, 400716 Chongqing, China
| | - Jie Xu
- Jinfeng Laboratory, 401329 Chongqing, China; Cancer Center, Medical Research Institute, Southwest University, 400716 Chongqing, China
| | - Hongjuan Cui
- Jinfeng Laboratory, 401329 Chongqing, China; Cancer Center, Medical Research Institute, Southwest University, 400716 Chongqing, China.
| |
Collapse
|
49
|
Bartholomä WC, Gilg S, Lundberg P, Larsen PN, Sallinen V, Sternby Eilard M, Urdzik J, Lindell G, Brismar TB, Fallentin E, Ovissi A, Socratous A, Bjerner T, Kollbeck S, Tellman J, Holmquist F, Dahlström N, Woisetschläger M, Björnsson B, Sparrelid E, Sandström P. Magnetic resonance-derived hepatic uptake index improves the identification of patients at risk of severe post-hepatectomy liver failure. Br J Surg 2025; 112:znaf103. [PMID: 40439010 PMCID: PMC12120443 DOI: 10.1093/bjs/znaf103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 06/02/2025]
Abstract
BACKGROUND Post-hepatectomy liver failure (PHLF) is a leading cause of mortality after major liver resection. Accurate preoperative risk assessment is essential, yet current methods have limitations. Gadoxetic acid-enhanced MRI (Gd-EOB MRI) enables both morphological and functional evaluation of the liver. The aim of this study was to evaluate the efficacy of the hepatic uptake index (HUI) obtained from routine preoperative Gd-EOB MRI for identifying patients at risk of severe PHLF. METHODS This observational retrospective multicentre study included 292 patients who underwent major hepatectomy between 2010 and 2020 in Sweden, Denmark, and Finland. Preoperative Gd-EOB MRI was performed for each patient and the HUI, hepatic uptake index of the standardized future liver remnant (sFLR-HUI), and Model for End-Stage Liver Disease Version 3 (MELD 3) score were evaluated. Statistical analyses included logistic regression and receiver operating characteristic (ROC) curve assessment to determine cut-off values and discriminative accuracies for severe PHLF (International Study Group of Liver Surgery grades B and C). RESULTS Among the 292 patients, 25 (8.6%) developed severe PHLF. Patients with severe PHLF had significantly lower HUI and sFLR-HUI values (P < 0.001). The HUI demonstrated superior discriminative performance for severe PHLF (area under the curve (AUC) 0.758) compared with volume-only assessments, such as the standardized future liver remnant (sFLR) (AUC 0.628). Combining the HUI with the MELD 3 score improved performance further (AUC 0.803). CONCLUSION The HUI obtained from routine Gd-EOB MRI outperforms volume-based biomarkers (sFLR) for identification of patients at risk of severe PHLF. Incorporating image-derived functional assessments, such as the HUI, with independent biomarkers, such as the MELD 3 score, may optimize preoperative risk stratification for severe PHLF and improve outcomes after major hepatectomy.
Collapse
Affiliation(s)
- Wolf C Bartholomä
- Department of Radiology and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Stefan Gilg
- Division of Surgery and Oncology, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Lundberg
- Department of Radiology and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Radiation Physics and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Peter N Larsen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ville Sallinen
- Department of Gastroenterological Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Malin Sternby Eilard
- Transplant Institute, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jozef Urdzik
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Gert Lindell
- Department of Surgery, Skåne University Hospital Comprehensive Cancer Centre, Lund University, Lund, Sweden
| | - Torkel B Brismar
- Department of Radiology, Karolinska University Hospital in Huddinge, Stockholm, Sweden
| | - Eva Fallentin
- Department of Radiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ali Ovissi
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Andreas Socratous
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tomas Bjerner
- Department of Radiology and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Sophie Kollbeck
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jens Tellman
- Department of Radiation Physics and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Fredrik Holmquist
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Nils Dahlström
- Department of Radiology and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Mischa Woisetschläger
- Department of Radiology and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Bergthor Björnsson
- Department of Surgery and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ernesto Sparrelid
- Division of Surgery and Oncology, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per Sandström
- Department of Surgery and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
50
|
Sabir S, Thani ASB, Abbas Q. Nanotechnology in cancer treatment: revolutionizing strategies against drug resistance. Front Bioeng Biotechnol 2025; 13:1548588. [PMID: 40370595 PMCID: PMC12075138 DOI: 10.3389/fbioe.2025.1548588] [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: 12/19/2024] [Accepted: 04/15/2025] [Indexed: 05/16/2025] Open
Abstract
A notable increase in cancer-related fatalities and morbidity worldwide is attributed to drug resistance. The factors contributing to drug resistance include drug efflux via ABC transporters, apoptosis evasion, epigenetic alterations, DNA repair mechanisms, and the tumor microenvironment, among others. Systemic toxicities and resistance associated with conventional cancer diagnostics and therapies have led to the development of alternative approaches, such as nanotechnology, to enhance diagnostic precision and improve therapeutic outcomes. Nanomaterial, including carbon nanotubes, dendrimers, polymeric micelles, and liposomes, have shown significant benefits in cancer diagnosis and treatment due to their unique physicochemical properties, such as biocompatibility, stability, enhanced permeability, retention characteristics, and targeted delivery. Building on these advantages, this review is conducted through comprehensive analysis of recent literature to explore the principal mechanisms of drug resistance, the potential of nanomaterials to revolutionize selective drug delivery and cancer treatment. Additionally, the strategies employed by nanomaterials to overcome drug resistance in tumors, such as efflux pump inhibition, multidrug loading, targeted delivery to the tumor microenvironment, and gene silencing therapies are discussed in detail. Furthermore, we examine the challenges associated with nanomaterials that limit their application and impede their transition to clinical use.
Collapse
Affiliation(s)
- Shazia Sabir
- Department of Biology, College of Science, Sakhir Campus, University of Bahrain, Sakhir, Bahrain
| | | | - Qamar Abbas
- Department of Biology, College of Science, Sakhir Campus, University of Bahrain, Sakhir, Bahrain
| |
Collapse
|