151
|
Jiang M, Li J, Wei J, Yang X, Wang W. Advances in neoantigen-based immunotherapy for head and neck squamous cell carcinoma: a comprehensive review. Front Oncol 2025; 15:1593048. [PMID: 40444094 PMCID: PMC12119297 DOI: 10.3389/fonc.2025.1593048] [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: 03/13/2025] [Accepted: 04/17/2025] [Indexed: 06/02/2025] Open
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC), ranking among the six most prevalent malignancies worldwide, is characterized by significant heterogeneity. Conventional monotherapeutic approaches, including surgical intervention, radiotherapy, and chemotherapy, often fail to achieve complete tumor cell elimination, consequently leading to disease recurrence and metastatic progression. In this context, personalized immunotherapeutic strategies, particularly cancer vaccines and immune checkpoint inhibitors, have emerged as promising therapeutic modalities for patients with recurrent/metastatic (R/M) HNSCC. Neoantigens, which exhibit selective expression in tumor tissues while remaining absent in normal tissues, have garnered considerable attention as novel targets for HNSCC personalized immunotherapy. However, the marked heterogeneity of HNSCC, coupled with patient-specific HLA variations, necessitates precise technical identification and evaluation of neoantigens at the individual level-a significant contemporary challenge. This comprehensive review systematically explores the landscape of neoantigen-based immunotherapy in HNSCC, including neoantigen sources, screening strategies, identification methods, and their clinical applications. Additionally, it evaluates the therapeutic potential of combining neoantigen-based approaches with other immunotherapeutic modalities, particularly immune checkpoint inhibitors, providing valuable insights for future clinical practice and research directions in HNSCC treatment.
Collapse
Affiliation(s)
- Manzhu Jiang
- College of Life Sciences, Shandong Agricultural University, Tai’an, China
| | - Jiefu Li
- Guangzhou National Laboratory, Guangzhou, China
| | - Jianhua Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| | - Xuerong Yang
- College of Life Sciences, Shandong Agricultural University, Tai’an, China
| | - Weiqi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
152
|
Premer C, Hare JM, Yuan SY, Wilson JW. Mesenchymal stem/stromal cells as a therapeutic for sepsis: a review on where do we stand? Stem Cell Res Ther 2025; 16:245. [PMID: 40375314 PMCID: PMC12082945 DOI: 10.1186/s13287-025-04371-w] [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: 03/25/2025] [Accepted: 04/28/2025] [Indexed: 05/18/2025] Open
Abstract
Sepsis is one of the leading causes of morbidity and mortality in the United States and Worldwide despite advances in quick recognition and early antibiotics, fluids, and vasopressors. Mesenchymal stem/stromal cells (MSCs) have gained attention as a biologic therapy given their unique anti-inflammatory, immunomodulatory, and anti-bacterial characteristics. MSCs have had success in treating a range of diseases, however limited clinical trials exist specifically for MSC use in sepsis. This article reviews the properties of MSCs that make them favorable for treating sepsis, as well as the current state of clinical trials. All clinical trials presented here demonstrated MSC safety, with a mixture of efficacy results and a heterogeneity of trial methods. Ultimately, MSCs are a promising novel therapeutic for sepsis, however a consensus in cell source, dosage, preparation, and delivery needs to be further investigated for MSCs to transition from bench to bedside and become a true therapeutic for sepsis.
Collapse
Affiliation(s)
- Courtney Premer
- Department of Emergency Medicine, McGaw Medical Center of Northwestern University, 211 E Ontario Street, Suite 200, Chicago, IL, 60611, USA.
| | - Joshua M Hare
- Department of Medicine, Division of Cardiovascular Medicine, Interdisciplinary Stem Cell Institute (ISCI), University of Miami, Miami, FL, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jason W Wilson
- Department of Emergency Medicine, Morsani College of Medicine, Tampa General Hospital, University of South Florida, Tampa, FL, USA
| |
Collapse
|
153
|
Zhang S, Xu D, Li F, Wang J. CRISPR-based non-nucleic acid detection. Trends Biotechnol 2025:S0167-7799(25)00139-8. [PMID: 40368676 DOI: 10.1016/j.tibtech.2025.04.012] [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: 02/15/2025] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
Characterization of clustered regularly interspaced short palindromic repeat (CRISPR)-associated (Cas) trans-cleavage activities has initiated the era of next-generation CRISPR diagnostics. By using the trans-cleavage reaction for signal output, CRISPR systems have been engineered to detect non-nucleic acids (NNAs), including ions, inorganic small molecules, organic compounds, proteins, and bacteria. Diverse strategies are being used to specifically recognize NNAs and regulate Cas trans-cleavage activities, via generation or depletion of output signals. In this review, we introduce the principles and advantages of CRISPR-based NNA detection. We then classify CRISPR-based NNA detection strategies into three classes: the generation or depletion of free activators, synthesis of crRNAs, and reconstruction of active Cas effectors. Finally, we discuss the challenges and potential strategies to advance both clinical and nonclinical applications of CRISPR-based NNA detection.
Collapse
Affiliation(s)
- Shanshan Zhang
- School of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China
| | - Dayong Xu
- School of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China.
| | - Feng Li
- School of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China.
| | - Jin Wang
- School of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China; Tolo Biotechnology Co., Ltd, Wuxi, Jiangsu 214100, China.
| |
Collapse
|
154
|
Guan H, Wang H, Cai X, Wang J, Chai Z, Wang J, Wang H, Zhang M, Wu Z, Zhu J, Zhong J, Yue B. Liquid-liquid phase separation of membrane-less condensates: from biogenesis to function. Front Cell Dev Biol 2025; 13:1600430. [PMID: 40438142 PMCID: PMC12116561 DOI: 10.3389/fcell.2025.1600430] [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: 03/26/2025] [Accepted: 04/30/2025] [Indexed: 06/01/2025] Open
Abstract
Membrane-less condensates (MLCs) are highly concentrated non-membrane-bounded structures in mammalian cells, comprising heterogeneous mixtures of proteins and/or nucleic acids. As dynamic compartments, MLCs can rapidly exchange components with the cellular environment, and their properties are easily altered in response to environmental signals, thus implicating that they can mediate numerous critical biological functions. A basic understanding of these condensates' formation, function, and underlying biomolecular driving forces has been obtained in recent years. For example, MLCs form through a liquid-liquid phase separation (LLPS) phenomenon similar to polymer condensation, which is primarily maintained via multivalent interactions of multi-domain proteins or proteins harboring intrinsically disordered regions (IDRs) as well as RNAs with binding sites. Moreover, an accumulating body of research indicates that MLCs are pathophysiologically relevant and involved in gene expression regulation and cellular stress responses. Here, we review the emerging field and explore what is currently known about the varied progress in LLPS of MLCs and how their features affect various cellular process, focusing on RNAs, including in skeletal myogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Binglin Yue
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
| |
Collapse
|
155
|
Song L, Sun L, Ren Y, Wang X, Xian L. Sex disparities in hepatocellular carcinoma immunotherapy: hormonal and genetic influences on treatment efficacy. Front Immunol 2025; 16:1607374. [PMID: 40438106 PMCID: PMC12116488 DOI: 10.3389/fimmu.2025.1607374] [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: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive liver cancer with a rising incidence globally. Immunotherapy, particularly immune checkpoint inhibitors (ICIs), has revolutionized HCC treatment, yet response rates remain variable. Sex-based disparities in immunotherapy efficacy have become increasingly recognized as important factors influencing treatment outcomes in HCC. This review examines the role of biological sex in HCC progression and immunotherapy responses. It discusses the epidemiology of sex differences in HCC incidence, prognosis, and therapeutic outcomes, highlighting the impact of sex hormones, such as estrogen and testosterone, on immune system function and tumor biology. Estrogen's protective effects, including enhanced T cell activation and improved immune surveillance, contribute to better treatment responses in females, while testosterone's immunosuppressive effects lead to poorer outcomes in males. The review also explores the influence of the tumor microenvironment, including immune cell composition and macrophage polarization, on treatment efficacy. Emerging evidence suggests that sex-specific factors, including hormonal status, should be considered in clinical trials and personalized treatment strategies. By addressing these disparities, tailored immunotherapeutic approaches could optimize efficacy and minimize toxicity in both male and female HCC patients, ultimately improving overall outcomes.
Collapse
Affiliation(s)
- Lei Song
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, China
| | - Liyan Sun
- Pediatric Outpatient Department, The First Hospital of Jilin University, Changchun, China
| | - Yuning Ren
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, The First Hospital of Jilin University, Changchun, China
| | - Xiaodan Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, The First Hospital of Jilin University, Changchun, China
| | - Lei Xian
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
156
|
Hosseini SS, Mansouri Majd S, Salimi A. Polarity-Switchable Dual-Mode Photoelectrochemical Cancer Marker Immunoassay Based on a Metal-Organic Framework@Nitrogen-Doped Graphdiyne Heterojunction. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27759-27771. [PMID: 40304230 DOI: 10.1021/acsami.5c00533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
A photocurrent polarity-switching photoelectrochemical (PEC) assay has been used for its anti-interference ability and superior accuracy compared to a conventional PEC measuring system. In this work, an ultrasensitive photocurrent polarity-switchable assay was established for sensitive prostate-specific antigen (PSA) detection based on a novel metal-organic framework (MOF) and graphdiyne@polyaniline (GDY@PANI)-sensitized structure as a photoactive material. The nitrogen-doped carbon nanolayers wrapped around graphdiyne and a zinc-based MOF were synthesized via a hydrothermal method. As an excellent photoactive material, the type II heterostructure (MOF/GDY@PANI) not only reduced the recombination of generated electron-hole pairs but also resulted in a significant increase in photoelectric conversion efficiency. Furthermore, its photocurrent was 4.6-fold higher than that of GDY@PANI and 37-fold higher than the proposed MOF. The integrated MOF/GDY@PANI/antibody (Ab) glassy carbon photoelectrode (GCE) was used as a PEC immunosensor for PSA detection (signal-off mode) and exhibited a wide linear dynamic range from 0.1 fg/mL to 10 pg/mL and a limit of detection of 0.05 fg/mL. The GCE modified with MOF and primary antibody (Ab1) (GCE/MOF/Ab1) produced a cathodic photocurrent, and in the presence of PSA, after the introduction of GDY@PANI-labeled-secondary antibody (Ab2) onto the surface of GCE/MOF/Ab1 and formation of an immunocomplex, the photocurrent amplified and switched to an anodic current. Due to high photoelectric conversion efficiency and good polarity-switching ability of GDY@PANI, the proposed immunosensor presented a turn-on photoelectrochemical performance for PSA detection at a wide linear range from 0.1 to 10 pg/mL and ultralow detection limit of 0.03 fg/mL. Compared to signal-off mode, the sensitivity increased 2-fold and the effect of interferences produces more reliable results due to photocurrent switching, and its effectiveness was evaluated against an enzyme-linked immunosorbent assay (ELISA) using spiked real human serum samples. The positive and promising outcomes achieved by the proposed immunosensor imply that the developed platform has the potential to serve as an excellent enzyme-free photoanode immunosensor for early cancer diagnosis and therapeutic monitoring.
Collapse
Affiliation(s)
| | - Samira Mansouri Majd
- Department of Chemistry, University of Kurdistan, Sanandaj 66177-15175, Iran
- Department of Semiconductors, Materials and Energy Research Center (MERC), Post Office Box 31787-316, Karaj 31779-83634, Iran
- Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran 19916-33361, Iran
| | - Abdollah Salimi
- Department of Chemistry, University of Kurdistan, Sanandaj 66177-15175, Iran
- Research Center for Nanotechnology, University of Kurdistan, Sanandaj 66177-15175, Iran
| |
Collapse
|
157
|
Azad MG, Russell T, Gu X, Zhao X, Richardson V, Wijesinghe TP, Babu G, Guo X, Kaya B, Dharmasivam M, Deng Z, Richardson DR. NDRG1 and its Family Members: More than Just Metastasis Suppressor Proteins and Targets of Thiosemicarbazones. J Biol Chem 2025:110230. [PMID: 40378957 DOI: 10.1016/j.jbc.2025.110230] [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: 03/15/2025] [Revised: 04/19/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
N-Myc downstream regulated gene-1 (NDRG1) and the other three members of this family (NDRG2, 3, and 4) play various functional roles in the cellular stress response, differentiation, migration, and development. These proteins are involved in regulating key signaling proteins and pathways that are often dysregulated in cancer, such as EGFR, PI3K/AKT, c-Met, and the Wnt pathway. NDRG1 is the primary, well-examined member of the NDRG family, and is generally characterized as a metastasis suppressor that inhibits the first step in metastasis, the epithelial-mesenchymal transition. While NDRG1 is well-studied, emerging evidence suggests NDRG2, NDRG3, and NDRG4 also play significant roles in modulating oncogenic signaling and cellular homeostasis. NDRG family members are regulated by multiple mechanisms, including transcriptional control by hypoxia-inducible factors, p53, and Myc, as well as post-translational modifications such as phosphorylation, ubiquitination, and acetylation. Pharmacological targeting of the NDRG family is a therapeutic strategy against cancer. For instance, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) have been extensively shown to up-regulate NDRG1 expression, leading to metastasis suppression and inhibition of tumor growth in multiple cancer models. Similarly, targeting NDRG2 demonstrates its pro-apoptotic and anti-proliferative effects, particularly in glioblastoma and colorectal cancer. This review provides a comprehensive analysis of the structural features, regulatory mechanisms, and biological functions of the NDRG family and their roles in cancer and neurodegenerative diseases. Additionally, NDRG1-4 are explored as therapeutic targets in oncology, focusing on recent advances in anti-cancer agents that induce the expression of these proteins. Implications for future research and clinical applications are also discussed.
Collapse
Affiliation(s)
- Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Tiffany Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xuanling Gu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Golap Babu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xinnong Guo
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Zhao Deng
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
158
|
Shu Z, Ye T, Wu W, Su M, Wang J, Zhang M, Qian Z, Huang H, Zheng S, Xia Q. Preoperative plasma cell-free DNA chromosomal instability predicts microvascular invasion in hepatocellular carcinoma: a prospective study. BMC Cancer 2025; 25:867. [PMID: 40361115 PMCID: PMC12076900 DOI: 10.1186/s12885-025-14268-9] [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: 11/22/2024] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Microvascular invasion (MVI) has been recognized as a risk factor for early recurrence after hepatectomy in patients with hepatocellular carcinoma (HCC). This study aimed to estimate the performance of an ultrasensitive chromosomal aneuploidy detector (UCAD) model for preoperative MVI prediction in operable HCC patients based on plasma cell-free DNA (cfDNA). METHODS A prospective study included HCC patients who underwent surgery in 2021. Preoperative peripheral plasma samples of eligible patients were collected to extract cfDNA, which was then subject to next generation sequencing. Low-coverage whole-genome sequencing data were analyzed for chromosomal instability using different parameters, including Z-score, chromosomal instability score (CIN score), tumor fraction (TFx) and a UCAD model (UCAD = CIN score + TFx + Z-score of all chromosomes). Receiver operating characteristic (ROC) curve was used to evaluate the performance of these parameters in preoperative MVI prediction. RESULTS Finally, a total of 74 patients with HCC who undergone hepatectomy were prospectively enrolled. Chromosomal instability was evaluated by copy number alterations and oncogenes MCL1 (located at 1q), MYC (located at 8q), TERT (located at 5p), EGFR (located at 7p), and VEGFA (located at 6p) were identified in plasma cfDNA. The UCAD model was a superior parameter in predicting preoperative MVI, with an area under curve (AUC) value 0.749 with a sensitivity of 0.938 specificity of 0.466. Univariate analysis revealed that tumor size (≥ 5 cm) and UCAD (> 0.199) significantly increased the risk of MVI, which were further demonstrated by multivariate analysis, with odd ratio of 1.338 (95%CI, 1.060-1.689) and 2.028 (95%CI, 1.053-3.908) (P < 0.05). CONCLUSIONS Our cfDNA-based UCAD model has shown a promising performance for preoperative MVI prediction in operable HCC patients. TRIAL REGISTRATION This study was registered at https://clinicaltrials.gov/ on 16 May 2022, retrospectively registered, Registration number: NCT05371873.
Collapse
Affiliation(s)
- Zheyue Shu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China
- Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ting Ye
- Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Menghan Su
- Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jingcheng Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, 310003, China
| | - Min Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, 310003, China
| | - Ziliang Qian
- The First People's Hospital of Chenzhou, Chenzhou, 424512, China
| | - Haifen Huang
- The First People's Hospital of Chenzhou, Chenzhou, 424512, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences, Hangzhou, 310003, China.
| | - Qi Xia
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
159
|
Qiu Y, Zhu Q, Cui K, Wang H, Zhang W, Li X, Yu J, Li Y, Luo Y, Wang Y, Xie W, Xia Q, Xiao Z. Polymer-Based Raman/PET Dual-Modal Probe for Preoperative Tumor Diagnosis and Intraoperative Image-Guided Surgery and Phototherapy. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40359337 DOI: 10.1021/acsami.5c03752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Integration of Raman imaging and positron emission tomography (PET) holds great promise for providing complementary information for precise cancer diagnosis and imaging-guided therapy. However, current existing Raman/PET dual-modal probes primarily rely on surface-enhanced Raman scattering, raising clinical concerns about the biosafety of the substrates. Herein, we developed a novel substrate-free Raman/PET probe-[18F]-AS-DPPT-TT NPs, which integrates a biocompatible polymer DPPT-TT that possesses strong Raman signals and photothermal effects, along with radiolabeling by short-lived 18F radionuclide and functionalization with tumor-targeting AS1411 aptamer. The [18F]-AS-DPPT-TT NPs generate ultrasensitive resonance Raman signals under 785 nm excitation, due to their absorption peak closely matching the excitation light, and exhibit significant photothermal effects for tumor cell ablation under 808 nm excitation. In the orthotopic colon cancer mouse models, [18F]-AS-DPPT-TT NPs enabled preoperative PET imaging for high-contrast whole-body tumor localization and simultaneously provided intraoperative Raman imaging for accurate tumor boundary delineation and micrometastasis detection (as small as 0.58 mm × 0.32 mm). Moreover, Raman imaging-guided surgery combined with photothermal therapy achieved complete elimination of primary and metastatic tumors, significantly decreasing the recurrence rates. This Raman/PET dual-modal probe effectively combines the strengths of Raman and PET imaging, providing a robust platform for comprehensive tumor management from preoperative planning to intraoperative intervention.
Collapse
Affiliation(s)
- Yuanyuan Qiu
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qi Zhu
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes,Department of Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Kai Cui
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haoze Wang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wenxian Zhang
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinyi Li
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiapei Yu
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yumeng Li
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yating Luo
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yiyuan Wang
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenhui Xie
- Department of Nuclear Medicine, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qian Xia
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes,Department of Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zeyu Xiao
- Department of Pharmacology and Chemical Biology, Institute of Molecular Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
160
|
Liao R, Mao J, Wang S, Wang S, Deng W, Tan Y, Xie Q. Coordination-Induced Photocurrent Polarity Switching of Black Titanium Dioxide for Tyrosinase Activity Assay. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:11776-11784. [PMID: 40319501 DOI: 10.1021/acs.langmuir.5c01300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Polarity-switchable photoelectrochemical (PEC) analysis has garnered significant attention owing to its enhanced accuracy and superior anti-interference capability. Developing a straightforward and efficient approach for polarity-switchable PEC analysis is critically essential. We present here a novel approach for polarity-switchable PEC assay by leveraging the coordination interactions between black titanium dioxide (b-TiO2) and o-diphenol-functionalized BiOI. Specifically, 4-hydroxyphenylacetic acid was covalently functionalized onto hydrolyzed 3-aminopropyl triethoxysilane-modified BiOI nanosheets, and the monophenol groups on BiOI are further converted into o-diphenol groups through tyrosinase (TYR) catalysis. Consequently, the o-diphenol-functionalized BiOI nanosheets are attached to a b-TiO2-based photoelectrode via coordination interactions between b-TiO2 and the o-diphenol groups, enabling photocurrent polarity switching of the b-TiO2-based electrode and polarity-switchable PEC assay of TYR activity. The developed PEC assay of TYR activity exhibits a broad linear range from 0.010 to 10 U mL-1 and a low detection limit of 0.002 U mL-1. The present work not only reports an innovative photocurrent polarity switchable strategy but also establishes an effective method for TYR activity assay.
Collapse
Affiliation(s)
- Rong Liao
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| | - Jiaxin Mao
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| | - Siyu Wang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| | - Shihua Wang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| | - Wenfang Deng
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| | - Yueming Tan
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| | - Qingji Xie
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
161
|
Sun L, Zhang T, Che Y, Shi J, Ren Y, Yan Y, Ji H, Song W, Gong Y, Zang L, Che Y, Zhao J. Photoinduced Proton-Transfer-Mediated Molecular Recognition in Molecular Crystals. Anal Chem 2025; 97:10010-10018. [PMID: 40301006 DOI: 10.1021/acs.analchem.5c00876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Molecular recognition has traditionally been focused on ground-state interactions. However, leveraging photoenergy to access excited-state molecular recognition, which may enable enhanced sensitivity or selectivity that is unattainable in the ground state, remains underexplored. In this study, we demonstrate a novel photoinduced molecular recognition mechanism using self-assembled crystalline microribbons composed of a donor-acceptor (D-A) molecule with a twisted molecular backbone for ultratrace phenol vapor detection. We confirm that photoinduced proton transfer occurs from phenol to the C═N group in the pyridine moiety of the D-A system, generating a protonated D-A molecule and a phenoxide ion that triggers fluorescence quenching. This proton-transfer-mediated recognition mechanism endows the microribbons with exceptional sensitivity and selectivity toward phenol vapor, achieving a limit of detection (LOD) of 0.6 parts per trillion (ppt). Our findings reveal that harnessing light energy to drive molecular recognition opens new avenues for advancing fluorescence sensing technologies.
Collapse
Affiliation(s)
- Lishan Sun
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianyan Zhang
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanxue Che
- HT-NOVA Company Limited, Zhuyuan Road, Shunyi District, Beijing 101312, China
| | - Jiangfan Shi
- Department of Materials Science and Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Yangyang Ren
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Yan
- Institute of Green Chemistry and Chemical Technology, School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Hongwei Ji
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjing Song
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanjun Gong
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - Ling Zang
- Department of Materials Science and Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Yanke Che
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jincai Zhao
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
162
|
Wu Q, Liu S, Zhao M, Wang Y, Lv K, Zhu J, Liu J. Pyroptosis-preconditioned mesenchymal stromal cell-derived extracellular vesicles as advanced nanomedicines for treating inflammatory diseases. Biomater Sci 2025; 13:2690-2703. [PMID: 40165652 DOI: 10.1039/d4bm01505c] [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: 04/02/2025]
Abstract
Uncontrolled inflammation is one of the major causes of various forms of tissue injury, and nanomedicines with immunoregulatory effects are needed. Mesenchymal stromal cell-derived extracellular vesicles (e.g., MSC-EVs) have been proposed as promising therapies, but the highly efficient generation of EVs with desirable properties is still a considerable challenge in this field. Here, we report that preconditioning MSCs with a critical immune process (pyroptosis) is a robust method for improving both the yield and anti-inflammatory potency of MSC-EVs. In brief, pyroptosis-preconditioned MSCs using a combined lipopolysaccharide (LPS) and adenosine triphosphate (ATP) stimulation showed elevated EV yields compared with those of MSCs cultured under normal conditions. Pyroptosis preconditioning upregulated multiple pathways (e.g., cell proliferation, DNA repair, and the immune response) in MSCs, leading to the enrichment of immunoregulatory cargos (e.g., PD-L2 and STC2) in MSC-EVs. In vitro, pyroptosis-preconditioned MSC-EVs (P-EVs) treatment has greater potential to suppress cytokine expression and cell death in pyroptotic macrophages than treatment with normal MSC-EVs (N-EVs). Compared with N-EV treatment, P-EV treatment showed superior potency in attenuating proinflammatory cell infiltration, cytokine/chemokine expression, resident tissue cell death, and the severity of pathological injury in different models of inflammatory diseases (acute lung or kidney injury), and these effects are likely the joint result of diverse functional cargos delivered by such EVs. This study highlights that pyroptosis preconditioning is a promising strategy for the highly efficient production of MSC-EVs with advanced therapeutic potential for treating diverse inflammatory diseases.
Collapse
Affiliation(s)
- Qianyi Wu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shuyun Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| | - Meng Zhao
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yizhuo Wang
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| | - Ke Lv
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| | - Jiaying Zhu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jingping Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| |
Collapse
|
163
|
Yang F, Song H, Wu W, Guo J. Targets and promising adjuvants for improving breast tumor response to radiotherapy. Bioorg Chem 2025; 162:108582. [PMID: 40393355 DOI: 10.1016/j.bioorg.2025.108582] [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: 02/23/2025] [Revised: 05/08/2025] [Accepted: 05/10/2025] [Indexed: 05/22/2025]
Abstract
Breast cancer ranks among the most common cancers globally, with significant mortality rates in advanced stages. Despite progress in treatment, therapy resistance, particularly to radiotherapy, remains a major challenge. Radiosensitization offers a promising solution to enhance radiotherapy effectiveness. This approach specifically increases tumor cells' vulnerability to IR. Recent research has explored molecular targets and strategies to improve radiosensitivity in breast cancer. Examples include inhibiting DNA repair pathways, altering the TME, targeting signaling pathways, and using immunomodulators. These strategies not only amplify destructive effects of IR but may also reduce required radiation doses, thereby minimizing normal tissue injury. This review examines promising molecular targets and combination therapies to boost radiosensitivity in breast cancer. It also highlights recent advances in immune modulation, TME remodeling, targeted molecular therapy, and metabolic pathway targeting. These advancements offer insights into the future of radiosensitization research. By systematically analyzing these strategies, the article aims to provide a comprehensive understanding of radiosensitization's current state and future potential in breast cancer treatment.
Collapse
Affiliation(s)
- Fusen Yang
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Hui Song
- Department of Traditional Chinese Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Weihong Wu
- Chinese Medicine Teaching and Research Group, Medical Advanced Vocational School of Shandong, Jinan, Shandong 250002, China
| | - Junmei Guo
- Department of Traditional Chinese Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China.
| |
Collapse
|
164
|
Feng W, Yang K, Zou Y, Xiao Z, Qian R, Qian R. Progress of ursolic acid on the regulation of macrophage: summary and prospect. Front Immunol 2025; 16:1576771. [PMID: 40421013 PMCID: PMC12104263 DOI: 10.3389/fimmu.2025.1576771] [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: 04/15/2025] [Indexed: 05/28/2025] Open
Abstract
Ursolic acid (UA), a prevalent pentacyclic triterpenoid found in numerous fruits and herbs, has garnered significant attention for its vital role in anti-inflammatory processes and immune regulation. The study of immune cells has consistently been a focal point, particularly regarding macrophages, which play crucial roles in antigen presentation, immunomodulation, the inflammatory response, and pathogen phagocytosis. This paper reveals the underlying regulatory effects of UA on the function of macrophages and the specific therapeutic effects of UA on a variety of diseases. Owing to the superior effect of UA on macrophages, different types of macrophages in different tissues have been described. Through the multifaceted regulation of macrophage function, UA may provide new ideas for the development of novel anti-inflammatory and immunomodulatory drugs. However, to facilitate its translation into actual medical means, the specific mechanism of UA in macrophages and its clinical application still need to be further studied.
Collapse
Affiliation(s)
- Wenjing Feng
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kehong Yang
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Zou
- Department of Anatomy, Anatomy Teaching Center of Hunan University of Chinese Medicine, Changsha, China
| | - Zhaohua Xiao
- Xiangya Hospital, Central South University, Changsha, China
| | - Rongkang Qian
- Department of Integrated Traditional Chinese and Western Medicine, Qian Rongkang Clinic, Loudi, China
| | - Ronghua Qian
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
165
|
Fu T, Duan B, Sun P, Ma W, Wang T, Wang T, Tong Z, Wang Y. Innovative applications of silicon dioxide nanoparticles for targeted liver cancer treatment. Front Bioeng Biotechnol 2025; 13:1595772. [PMID: 40421114 PMCID: PMC12104587 DOI: 10.3389/fbioe.2025.1595772] [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: 03/25/2025] [Accepted: 04/24/2025] [Indexed: 05/28/2025] Open
Abstract
Liver cancer remains a major global health challenge, characterized by high mortality and limited treatment efficacy. Conventional therapies, including chemotherapy, immunotherapy, and viral vectors, are hindered by systemic toxicity, drug resistance, and high costs. Silica nanoparticles (SiO2NPs) have emerged as promising platforms for liver cancer therapy, offering precise drug delivery, stimuli-responsive release, and integrated diagnostic-therapeutic capabilities. This review critically examines the potential of SiO2NPs to overcome these therapeutic limitations. Notable advances include their high drug-loading capacity, customizable surface modifications, and dual-responsive systems (pH/redox/NIR-II) that enable >90% tumor-specific drug release. Preclinical studies have demonstrated synergistic efficacy in combination therapies. Additionally, theranostic SiO2NPs enable MRI-guided tumor delineation and real-time treatment monitoring. Despite promising results, challenges remain in long-term biosafety, scalable synthesis, and regulatory compliance. Early-phase clinical trials, including those using NIR-II-responsive platforms, highlight their translational potential but underscore the need for further validation of toxicity profiles and manufacturing standards. Future research should focus on optimizing combinatory treatment strategies, scaling up production, and aligning with evolving regulatory frameworks. By bridging nanomaterial innovation with clinical applications, SiO2NPs offer unparalleled potential for advancing precision oncology in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Tiantian Fu
- Department of Thoracic Radiation Oncology Ward 1, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Boshi Duan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Peng Sun
- Department of Hand Surgery 4 Ward, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Wei Ma
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | | | | | - Zhuang Tong
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yue Wang
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
166
|
Zeng M, Hu C, Chen T, Zhao T, Dai X. Advancements in Cell Membrane-Derived Biomimetic Nanotherapeutics for Breast Cancer. Int J Nanomedicine 2025; 20:6059-6083. [PMID: 40385497 PMCID: PMC12083498 DOI: 10.2147/ijn.s502144] [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: 11/13/2024] [Accepted: 04/11/2025] [Indexed: 05/20/2025] Open
Abstract
Breast cancer remains the leading cause of female mortality worldwide, necessitating innovative and multifaceted approaches to address its various subtypes. Nanotechnology has attracted considerable attention due to its nanoscale dimensions, diverse carrier types, suitability for hydrophobic drug delivery, and capacity for controlled and targeted administration. Nano-sized particles have become prevalent carriers for therapeutic agents targeting breast cancer, thanks to their reproducible synthesis and adjustable properties, including size, shape, and surface characteristics. In addition, certain nanoparticles can enhance therapeutic effects synergistically. However, the immune system often detects and removes these nanoparticles, limiting their efficacy. As a promising alternative, cell membrane-based delivery systems have gained attention due to their biocompatibility and targeting specificity. These membrane-coated drug delivery systems are derived from various cell sources, including blood cells, cancer cells, and stem cells. Leveraging the unique properties of these cell membranes enables precise targeting of breast cancer tumors and associated biomarkers. Inspired by natural structures, cell membranes disguise nanoparticles in the bloodstream, enhancing their retention time in vivo and improving tumor targeting. Consequently, cell membrane-derived nanoparticles (CMDNPs) have been investigated for their potential applications in breast cancer diagnostics, photothermal therapy (PTT), and vaccine development. This review comprehensively explores the potential and limitations of cell membrane-derived drug delivery systems in clinical applications against breast cancer.
Collapse
Affiliation(s)
- Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Chenji Hu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Tao Chen
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400014, People’s Republic of China
| | - Tingrui Zhao
- Department of Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, People’s Republic of China
| | - Xinhua Dai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
167
|
Li Z, Shen M, Meng F, Zhang Y, Duan W, Hou C, Zhang M. Engineering Oxidase-Based Cascade Nanoreactors Design, Catalytic Efficiency, and Applications in Disease Monitoring. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501976. [PMID: 40351055 DOI: 10.1002/smll.202501976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/14/2025] [Indexed: 05/14/2025]
Abstract
Inspired by the advantages of biological cascade catalytic systems, it has been devoted to the discovery of novel oxidase-based cascade catalytic systems for disease monitoring. However, the low stability, easy inactivation, and poor reproducibility of oxidase significantly limit their practical applications. Immobilization of the oxidase can be enabled to protect them from external mediators and improve catalytic efficiency and reproducibility. Notably, the substrate channels and spatial confinement play an essential role in the construction of immobilized cascade nanoreactors to enhance the overall activity. Moreover, nanozymes, a class of enzyme mimics, have not only enzyme-like activity but also high stability and tunable catalytic properties, which bolster the development of cascade nanoreactors. Herein, recent advances in the assembly of cascade reactors involving enzymes/nanozymes are described. The importance of substrate channeling and spatial distribution in regulating the catalytic efficiency of the nanoreactor is highlighted. Then, along with an in-depth discussion of the cascade biosensors for disease monitoring, the design and application of innovative devices based on these sensing principles are also summarized, including microfluidic systems, hydrogel-based platforms, and test paper technologies. Finally, challenges and prospects for cascade nanoreactors are briefly discussed and prospected.
Collapse
Affiliation(s)
- Zongda Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi, 830046, P. R. China
- College of Life Science & Technology, Xinjiang University, Urumqi, 830046, P. R. China
| | - Mingping Shen
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi, 830046, P. R. China
- College of Life Science & Technology, Xinjiang University, Urumqi, 830046, P. R. China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Fanxing Meng
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi, 830046, P. R. China
- College of Life Science & Technology, Xinjiang University, Urumqi, 830046, P. R. China
| | - Youning Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi, 830046, P. R. China
- School of Intelligent Agriculture and Technology, Xinjiang University, Urumqi, 830046, P. R. China
| | - Weiwei Duan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi, 830046, P. R. China
- College of Life Science & Technology, Xinjiang University, Urumqi, 830046, P. R. China
| | - Chengyi Hou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Minwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Urumqi, 830046, P. R. China
- School of Intelligent Agriculture and Technology, Xinjiang University, Urumqi, 830046, P. R. China
| |
Collapse
|
168
|
Li L, Liu X, Xu S, Zhang S, Yang Z, Xiao D. Electrochromic platform for the visual detection of the neuroblastoma biomarkers vanillylmandelic acid and homovanillic acid. Analyst 2025; 150:2153-2159. [PMID: 40223690 DOI: 10.1039/d5an00228a] [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: 04/15/2025]
Abstract
Vanillylmandelic acid (VMA) and homovanillic acid (HVA) are biomarkers for the diagnosis and course-of-disease monitoring of malignant tumor neuroblastomas, which endanger infants and children. Herein, we demonstrated a proof-of-concept visual detection of VMA and HVA on an electrochromic basis, in which the viologen 1,1'-dibenzyl-4,4'-bipyridinium dichloride was used as a coloration chromophore. It was found that VMA and HVA can be used as effective electron mediators to improve the electrochromic performance of devices. It is interesting to note that VMA and HVA reduce the driving voltage of electrochromic devices (ECDs) down to -1.0 V, which is lower than that (-2.1 V) achieved without these additives, and the coloration of ECDs is undoubtedly dependent on the concentration of VMA and HVA from 0.8 to 10-6 mol L-1. Thus, this study presents an ECD platform as a breakthrough strategy for the facile, routine and portable visual detection of the neuroblastoma biomarkers VMA and HVA with obvious advantages over other detection techniques such as HPLC/MS used in clinical diagnosis.
Collapse
Affiliation(s)
- Lu Li
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing 211816, China.
| | - Xiaodi Liu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing 211816, China.
| | - Shijie Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing 211816, China.
| | - Shiming Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing 211816, China.
| | - Zhuangzhuang Yang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing 211816, China.
| | - Debao Xiao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Flexible Electronics (Future Technologies), Nanjing Tech University (NanjingTech), Nanjing 211816, China.
| |
Collapse
|
169
|
Teng W, Li H, Yang H, Chen Y, Xi L, Xin F, Zhang A, Yu L, Zheng L, Wang M, Bai J, Ke F, Wang Y, Sun F, Zhang H, Wu L, Liu J. Discovery and validation of a novel dual-target blood test for the detection of hepatocellular carcinoma across stages from cirrhosis. BMC Med 2025; 23:278. [PMID: 40350453 PMCID: PMC12067762 DOI: 10.1186/s12916-025-04115-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common cancers. Early detection of HCC helps improve the patients' 5-year survival rate. Our goal was to identify superior methylation biomarkers to develop a methylation-specific quantitative PCR (MS‒qPCR) assay. METHODS A five-phase case-control study identified HCC methylation biomarkers via capture sequencing, TCGA/RNA-seq filtering, technical (MS-qPCR/Sanger) and biological (quadruplex MS-qPCR) validation. Methylated biomarkers were selected based on differential methylation expression using a tissue discovery cohort (43 HCC, 32 normal) and validated in plasma validation cohorts (Phase 1: 53 HCC, 52 cirrhosis, 20 benign, 50 healthy; Phase 2: 67 HCC, 81 cirrhosis). Then, the final assay's HCC detection performance was compared with existing blood-based surveillance methods. RESULTS Two methylated genes, OSR2 and TSPYL5, and a novel internal reference gene, SDF4, were identified and developed into an MS‒qPCR assay named Qliver. Qliver had an AUC of 0.955 (95% CI: 0.924-0.987) for distinguishing HCC patients from non-HCC patients in the Phase 1 plasma cohort, with a sensitivity of 88.68% (95% CI: 76.97%-95.73%) and a specificity of 89.34% (95% CI: 82.47%-94.20%), and 0.958 (95% CI: 0.927-0.989) for distinguishing HCC patients from cirrhosis patients in the Phase 2 plasma cohort, with a sensitivity of 88.06% (95% CI: 77.82%-94.70%) and a specificity of 92.59% (95% CI: 84.57%-97.23%). For the Phase 1 plus Plasma 2 cohort, Qliver had an AUC of at least 0.958 for detecting HCC in healthy individuals, cirrhosis patients and patients with benign liver diseases, which was superior to that of the GALAD score (AUC: 0.777 to 0.849). For BCLC stage 0 and A HCC patients, the sensitivity of Qliver ranged from 62.50% (95% CI: 24.49%-91.48%) to 72.73% (39.03%-93.98%), with a specificity of 90%. Overall, Qliver was superior to the AFP, AFP-L3, DCP and the GALAD score in terms of cirrhosis history, tumor stage, tumor size and tumor count. CONCLUSIONS Qliver demonstrated superior performance in detecting HCC compared with currently widely used blood biomarkers, suggesting its potential clinical benefit in HCC surveillance in high-risk populations.
Collapse
Grants
- 2023J05234, 2023J011297 Natural Science Foundation of Fujian Province
- 2023J05234, 2023J011297 Natural Science Foundation of Fujian Province
- 2021Y9232, 2021Y9227, 2024Y9620 Joint Funds for the Innovation of Science and Technology, Fujian province
- 2021Y9232, 2021Y9227, 2024Y9620 Joint Funds for the Innovation of Science and Technology, Fujian province
- 2021Y9232, 2021Y9227, 2024Y9620 Joint Funds for the Innovation of Science and Technology, Fujian province
- 2022ZQNZD009 Young and Middle-aged Scientific Research Major Project of Fujian Provincial Health Commission
- 2022YNG01 high level talents training project of Fujian Cancer Hospital
Collapse
Affiliation(s)
- Wenhao Teng
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hui Li
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Hao Yang
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
- Genetrix Biotech Corporation, Beijing, China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Liying Xi
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Fuli Xin
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Aiyuan Zhang
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Lihui Yu
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lu Zheng
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Ming Wang
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jian Bai
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Fayong Ke
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yin Wang
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Fuming Sun
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
- Genetrix Biotech Corporation, Beijing, China
| | - Hui Zhang
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Lin Wu
- Berry Oncology Corporation, Beijing, China.
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China.
| | - Jingfeng Liu
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China.
| |
Collapse
|
170
|
Xiao H, Raza F, Li K, Song J, Zafar H, Yang S, Su J, Qiu M. Cell membrane derived biomimetic nanomedicine for precision delivery of traditional Chinese medicine in cancer therapy. J Control Release 2025; 383:113829. [PMID: 40355044 DOI: 10.1016/j.jconrel.2025.113829] [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: 02/19/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
The rapidly developing modern nanotechnology has brought new vitality to the application of traditional Chinese medicine (TCM), improving the pharmacokinetics and bioavailability of unmodified natural drugs. However, synthetic materials inevitably introduce incompatibilities. This has led to focusing on biomimetic drug delivery systems (DDS) based on biologically derived cell membranes. This "top-down" approach to nanomedicine preparation is simple and effective, as the inherited cell membranes and cell surface substances can mimic nature when delivering drugs back into the body, interacting similarly to the source cells at the biological interface. The concept of biologically derived TCM and biomimetic membranes aligns well with nature, the human body, and medicine, thereby enhancing the in vivo compatibility of TCM. This review focused on the recent progress using biomimetic membranes for TCM in cancer therapy, emphasizing the effective integration of biomimetic nanomedicine and TCM in applications such as cancer diagnosis, imaging, precision treatment, and immunotherapy. The review also provided potential suggestions on the challenges and prospects in this field.
Collapse
Affiliation(s)
- Hang Xiao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kunwei Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinpu Song
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiqi Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
171
|
Shen Q, Liu J, Zeng L, Ren Y, Liao J, Chen S, Tang Y, Zhang Z, Jiang M, Liao H, Wang L, Xu X, Chen J. Pancreas-targeted lipid nanoparticles for relatively non-invasive interleukin-12 mRNA therapy in orthotopic pancreatic ductal adenocarcinoma. J Control Release 2025; 381:113588. [PMID: 40032009 DOI: 10.1016/j.jconrel.2025.113588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents 90 % of pancreatic cancers and shows limited response to immune therapy owing to the highly immunosuppressive tumor microenvironment (TME). Cytokine-encoded mRNA therapy demonstrates a great promise in converting "cold" tumors into "hot" ones, while it is typically administered through intratumoral injection and applicable only to superficial tumors, which limites their application in PDAC. In this study, we design and develop a lipid nanoparticle (LNP) delivery system capable of targeting pancreatic tissue via intraperitoneal (I.P.) injection. This system not only efficiently delivers mRNA to pancreatic tissues but also selectively targets immune cells in PDAC. A single I.P. injection of LNP encapsulating interleukin-12 (IL-12) mRNA (LNP/mIL-12) activates both myeloid and lymphoid cells in PDAC, reprogramming the immunosuppressive TME. Remarkably, I.P. injection of LNP/mIL-12 induces eradication of orthotopic PDAC in some cases. Our work represents the first relatively non-invasive method to deliver IL-12 mRNA for targeted treatment of orthotopic PDAC, offering a novel approach for PDAC immunotherapy.
Collapse
Affiliation(s)
- Qian Shen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Jia Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China; Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Ling Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Yupeng Ren
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Hunan Provincial Key Laboratory of Advanced Materials for New Energy Storage and Conversion, School of Materials Science and Engineering 2 Taoyuan Street, Xiangtan 411201, PR China
| | - Jing Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Sijie Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Yingsen Tang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Zixi Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Meng Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China
| | - Hangping Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Hunan Provincial Key Laboratory of Advanced Materials for New Energy Storage and Conversion, School of Materials Science and Engineering 2 Taoyuan Street, Xiangtan 411201, PR China
| | - Lingyun Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China.
| | - Jinjin Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, PR China.
| |
Collapse
|
172
|
Ma Y, He Y, Hao C, Li X, Li L, Zhao Y, Zhang Y, Zhang W. Versatile core-shell MnO 2@PANI nanocomposites: Bridging photo-assisted zinc-ion batteries and wearable strain sensors for self-powered systems. J Colloid Interface Sci 2025; 696:137860. [PMID: 40367873 DOI: 10.1016/j.jcis.2025.137860] [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: 02/20/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
With the continuous development of self-powered sensing systems, achieving efficient energy self-supply and high-performance signal acquisition simultaneously has emerged as a main trend for next-generation sensors. However, developing functional materials for such sensing systems is still facing crucial challenges. Herein, a multifunctional MnO2@PANI (MP) composite material was designed to realize a synchronized "integration" strategy. Specifically, the MP serves as a cathode in photo-assisted zinc-ion batteries (PA-ZIBs), enabling solar energy conversion and storage. The p-n heterojunction formed between MnO2 and PANI promotes effective separation of photogenerated electron-hole pairs and enhances Zn2+ kinetics. Therefore, the PA-ZIBs deliver a high self-charging voltage of 1.08 V and 33 % enhancement in specific capacity (reaching 249.5 mAh g-1 at 0.1 A g-1) under illumination. Meanwhile, it also functions as the active layer of a flexible strain sensor, achieving efficient mechanical signal detection. The core-shell structure and composite component improve both structural stability and conductivity. As a result, the sensor exhibits high sensitivity, a fast response time of 0.4 s, and excellent durability over 2000 cycles. Furthermore, proof-of-concept demonstrations validate the device's capability to simultaneously harvest solar energy and detect physiological signals. This work offers a new solution for designing self-powered sensing systems and intelligent healthcare applications by merging energy-autonomous operation with multifunctional sensing capabilities.
Collapse
Affiliation(s)
- Yueyue Ma
- Province-Ministry Co-construction Collaborative Innovation Center of Hebei Photovoltaic Technology, College of Physics Science and Technology, Hebei University, Baoding 071002, China
| | - Yadong He
- Province-Ministry Co-construction Collaborative Innovation Center of Hebei Photovoltaic Technology, College of Physics Science and Technology, Hebei University, Baoding 071002, China
| | - Chenlong Hao
- Province-Ministry Co-construction Collaborative Innovation Center of Hebei Photovoltaic Technology, College of Physics Science and Technology, Hebei University, Baoding 071002, China
| | - Xiang Li
- Province-Ministry Co-construction Collaborative Innovation Center of Hebei Photovoltaic Technology, College of Physics Science and Technology, Hebei University, Baoding 071002, China
| | - Ling Li
- Province-Ministry Co-construction Collaborative Innovation Center of Hebei Photovoltaic Technology, College of Physics Science and Technology, Hebei University, Baoding 071002, China.
| | - Youwei Zhao
- Province-Ministry Co-construction Collaborative Innovation Center of Hebei Photovoltaic Technology, College of Physics Science and Technology, Hebei University, Baoding 071002, China; National & Local Joint Engineering Research Center of Metrology Instrument and System, Hebei Key Laboratory of Energy Metering and Safety Testing Technology, College of Quality and Technical Supervision, Hebei University, Baoding 071002, China.
| | - Yucang Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian Province 361021, China.
| | - Wenming Zhang
- Province-Ministry Co-construction Collaborative Innovation Center of Hebei Photovoltaic Technology, College of Physics Science and Technology, Hebei University, Baoding 071002, China.
| |
Collapse
|
173
|
Song K, Ming J, Tao B, Zhao F, Huang S, Wu W, Jiang C, Li X. Emerging glucose oxidase-delivering nanomedicines for enhanced tumor therapy. J Control Release 2025; 381:113580. [PMID: 40024341 DOI: 10.1016/j.jconrel.2025.02.076] [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: 08/30/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Abnormalities in glucose metabolism have been shown to characterize malignant tumors. Glucose depletion by glucose oxidase (GOD) has shown great potential in tumor therapy by causing tumor starvation. Since 2017, nanomedicines have been designed and utilized to deliver GOD for more precise and effective glucose modulation, which can overcome intrinsic limitations of different cancer therapeutic modalities by remodeling the tumor microenvironment to enhance antitumor therapy. To date, the topic of GOD-delivering nanomedicines for enhancing tumor therapy has not been comprehensively summarized. Herein, this review aims to provide an overview and discuss in detail recent advances in GOD delivery and directly involved starvation therapy strategies, GOD-sensitized various tumor therapy strategies, and GOD-mediated multimodal antitumor strategies. Finally, the challenges and outlooks for the future progress of the emerging tumor therapeutic nanomedicines are discussed. This review provides intuitive and specific insights to a broad audience in the fields of nanomedicines, biomaterials, and cancer therapy.
Collapse
Affiliation(s)
- Kaiyue Song
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jiang Ming
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Feng Zhao
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China.
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Cong Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200092, China.
| | - Xianglong Li
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
174
|
Ling S, Jin Y, He C, Fu L, Deng Y, Qi X, Wang Z, Wu Y. Determination of tacrolimus in whole blood of pediatric liver transplant patients by UPLC-MS/MS: Informed its individualized dose. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1261:124645. [PMID: 40359669 DOI: 10.1016/j.jchromb.2025.124645] [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: 02/05/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
Tacrolimus is a potent macrolide immunosuppressant widely used in pediatric liver transplant patients. However, its narrow therapeutic window and significant inter-individual pharmacokinetic differences result in a poor correlation between blood concentrations and administered doses. To support its individualized dose, a sensitive, fast and robust ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed to measure tacrolimus concentrations in whole blood of pediatric liver transplant patients. The method employed acetonitrile for protein precipitation, and sample separation was achieved using an Acquity UPLC CSH C18 column (2.1 × 50 mm, 1.7 μm) with gradient elution. The mobile phase consisted of ammonium solution-water (0.5:1000, v/v) and methanol. The method demonstrated good linearity within a concentration range of 0.20-50.00 ng/mL. The intra- and inter-day precisions of tacrolimus in whole blood were less than 13.5 %, with the accuracy ranged from -7.2 % to 13.0 %. Selectivity, carryover, matrix effects, recovery, dilution reliability, and stability all met the standards set by relevant guidelines. The established UPLC-MS/MS method was successfully applied to measure the concentration of tacrolimus in the whole blood of pediatric liver transplant patients and support its individualized dose. Under the rapid UPLC-MS/MS method, 25 (65.8 %) patients required a dose increase, 3 (7.9 %) patients needed a dose reduction, and 10 (26.3 %) patients maintained their original dosage without adjustment.
Collapse
Affiliation(s)
- Saisai Ling
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China; Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, West China Hospital, Sichuan University, Chengdu 610041, China; Clinical Trial Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Jin
- Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, West China Hospital, Sichuan University, Chengdu 610041, China; Clinical Trial Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cuiyao He
- Department of Pharmacy Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Lisha Fu
- Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, West China Hospital, Sichuan University, Chengdu 610041, China; Clinical Trial Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuhua Deng
- Department of Hepatobiliary Surgery Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Xiaohui Qi
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China; Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, West China Hospital, Sichuan University, Chengdu 610041, China; Clinical Trial Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhenglei Wang
- Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, West China Hospital, Sichuan University, Chengdu 610041, China; Clinical Trial Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yiying Wu
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China.
| |
Collapse
|
175
|
Wu J, Ge Y, Huang W, Zhang L, Huang J, Huang N, Luo Y. Natural bioactive compounds modified with mesenchymal stem cells: new hope for regenerative medicine. Front Bioeng Biotechnol 2025; 13:1446537. [PMID: 40416310 PMCID: PMC12098461 DOI: 10.3389/fbioe.2025.1446537] [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: 06/10/2024] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have the potential to differentiate into various cell types, providing important sources of cells for the development of regenerative medicine. Although MSCs have various advantages, there are also various problems, such as the low survival rate of transplanted cells and poor migration and homing; therefore, determining how to reform MSCs to improve their utilization is particularly important. Although many natural bioactive compounds have shown great potential for improving MSCs, many mechanisms and pathways are involved; however, in the final analysis, natural bioactive compounds promoted MSC proliferation, migration and homing and promoted differentiation and antiaging. This article reviews the regulatory effects of natural bioactive compounds on MSCs to provide new ideas for the therapeutic effects of modified MSCs on diseases.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ying Ge
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Wendi Huang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Li Zhang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Juan Huang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yong Luo
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
176
|
Markin NS, Gordeev IS, Fu HE, Ivannikov SI, Kim YB, Samardak AY, Samardak AS, Kim YK, Ognev AV. Secondary electron dynamics in core-shell-satellite nanoparticles: a computational strategy for targeted cancer treatment. NANOSCALE 2025; 17:11691-11702. [PMID: 40260843 DOI: 10.1039/d5nr00270b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
As the global incidence of cancer escalates, there exists an urgent necessity for innovative therapeutic modalities. While radiation therapy is indispensable in oncology, it faces significant challenges in achieving an optimal equilibrium between tumour ablation and the preservation of surrounding healthy tissues. Noteworthy advancements such as intensity-modulated radiation therapy (IMRT) and three-dimensional conformal radiation therapy (3D-CRT) have enhanced the precision of treatment; however, their efficacy is still constrained by the accuracy of tumour delineation. The utilization of radiosensitizers, with a particular emphasis on metal nanoparticles, presents a promising avenue for augmenting the susceptibility of neoplastic cells to ionizing radiation. This research examines the potential of core-shell-satellite Fe3O4-SiO2-Au nanoparticles as effective radiosensitizers. By investigating the interaction of individual nanoparticles situated within a water phantom of 20 micrometers in diameter with monochromatic photon beams at energies of 50, 100, and 150 keV, we analyse how variations in the structural composition of Au nanoparticles and their concentrations within these multifaceted nanoparticles influence the efficacy of radiation therapy, employing Monte Carlo simulations corroborated by the general-purpose radiation transport code PHITS. Our investigation aspires to refine nanoparticle-based methodologies to enhance cancer treatment outcomes, potentially facilitating the development of more targeted therapeutic interventions that minimize adverse effects while improving patient survival rates.
Collapse
Affiliation(s)
- Nikita Sergeevich Markin
- Laboratory of Thin Film Technologies, ITAM, Far Eastern Federal University, Vladivostok 690922, Russia
- Institute of Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690022, Russia
| | - Ivan Sergeevich Gordeev
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna 141980, Russia
- Dubna State University, Dubna 141982, Russia
| | - Hong En Fu
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea.
| | | | - Yeon Beom Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea.
| | - Alexey Yurievich Samardak
- Laboratory of Thin Film Technologies, ITAM, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Alexander Sergeevich Samardak
- Laboratory of Thin Film Technologies, ITAM, Far Eastern Federal University, Vladivostok 690922, Russia
- Sakhalin State University, Yuzhno-Sakhalinsk 693000, Russia
| | - Young Keun Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea.
| | - Alexey Vyacheslavovich Ognev
- Laboratory of Thin Film Technologies, ITAM, Far Eastern Federal University, Vladivostok 690922, Russia
- Sakhalin State University, Yuzhno-Sakhalinsk 693000, Russia
| |
Collapse
|
177
|
Wei LH, Chen X, Shen AL, Fang Y, Xie QR, Guo Z, Sferra TJ, Chen YQ, Peng J. Pien Tze Huang Attenuates Cell Proliferation and Stemness Promoted by miR-483-5p in Hepatocellular Carcinoma Cells. Chin J Integr Med 2025:10.1007/s11655-025-4126-0. [PMID: 40342037 DOI: 10.1007/s11655-025-4126-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 05/11/2025]
Abstract
OBJECTIVE To investigate the effect of miR-483-5p on hepatocellular carcinoma (HCC) cells proliferation and stemness, as well as the attenuating effect of Pien Tze Huang (PZH). METHODS Differentially expressed miRNA between HepG2 cells and hepatic cancer stem-like cells (HCSCs) were identified by a miRNA microarray assay. miR-483-5p mimics were transfected into HepG2 cells to explore the effects of miR-483-5p on cell proliferation and stemness. HepG2 cells and HCSCs were treated with PZH (0, 0.25, 0.50 and 0.75 mg/mL) to explore the effects of PZH on the proliferation and stemness, both in non-induced state and the state induced by miR-483-5p mimics. RESULTS miR-483-5p was significantly up-regulated in HCSCs and its overexpression increased cell proliferation and stemness in HepG2 cells (P<0.05). PZH not only significantly inhibited proliferation in HepG2 cells, but also significantly suppressed the cell proliferation and self-renewal of HCSCs (P<0.05). The effects of miR-483-5p mimics on proliferation and stemness of HepG2 cells were partially abolished by PZH. CONCLUSIONS miR-483-5p promotes proliferation and enhances stemness of HepG2 cells, which were attenuated by PZH, demonstrating that miR-483-5p is a potential molecular target for the treatment of HCC and provide experimental evidence to support clinical use of PZH for patients with HCC.
Collapse
Affiliation(s)
- Li-Hui Wei
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Xi Chen
- Department of Oncology, the 900th Hospital of the Chinese People's Liberation Army Joint Service Support Force, Fuzhou, 350003, China
| | - A-Ling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Yi Fang
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Qiu-Rong Xie
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Zhi Guo
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Thomas J Sferra
- Department of Pediatrics, Rainbow Babies and Children's Hospital and Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - You-Qin Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Department of Pediatrics, Rainbow Babies and Children's Hospital and Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
178
|
Chen Y, Huang X, Hu R, Lu E, Luo K, Yan X, Zhang Z, Ma Y, Zhang M, Sha X. Inhalable biomimetic polyunsaturated fatty acid-based nanoreactors for peroxynitrite-augmented ferroptosis potentiate radiotherapy in lung cancer. J Nanobiotechnology 2025; 23:338. [PMID: 40340938 PMCID: PMC12060495 DOI: 10.1186/s12951-025-03409-8] [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: 01/26/2025] [Accepted: 04/19/2025] [Indexed: 05/10/2025] Open
Abstract
The limited efficacy and poor tumor accumulation remain crucial challenges for radiotherapy against lung cancer. To address these limitations, we rationally developed a polyunsaturated fatty acid (PUFA)-based nanoreactor (DHA-N@M) camouflaged with macrophage cell membrane to improve tumoral distribution and achieve peroxynitrite-augment ferroptosis for enhanced radiotherapy against lung cancer. After nebulization, the nanoreactors exhibited superior pulmonary accumulation in orthotopic lung cancer-bearing mice, with 70-fold higher than intravenously injected nanoreactors at 12 h post-administration, and distributed deeply in the tumors. DHA-N@M selectively released nitric oxide (NO) in glutathione (GSH)-enriched tumor cells, with consumption of GSH and subsequent inactivation of glutathione peroxidase 4 (GPX4). Under radiation, NO reacted with radiotherapy-induced reactive oxygen species (ROS) to generate peroxynitrite (ONOO-), resulting in redox homeostasis disruption. Combined with docosahexaenoic acid (DHA)-induced lipid metabolism disruption, overwhelming ferroptosis was induced both in vitro and in vivo. Notably, DHA-N@M mediated ferroptosis-radiotherapy significantly suppressed tumor growth with a 93.91% inhibition in orthotopic lung cancer models. Therefore, this design provides a nebulized ferroptosis-radiotherapy strategy for lung cancer.
Collapse
Affiliation(s)
- Yiting Chen
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xueli Huang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Ruining Hu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Enhao Lu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Kuankuan Luo
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xin Yan
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Zhiwen Zhang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Yan Ma
- Department of Pharmacy, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| | - Minghe Zhang
- Naval Medical Center, Naval Medical University, Shanghai, Shanghai, 200052, China.
| | - Xianyi Sha
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China.
- Quzhou Fudan Institute, 108 Minjiang Avenue, Kecheng District, Quzhou, Zhejiang Province, 324002, China.
| |
Collapse
|
179
|
Lin J, He R, Qu Z, Dong J, Krabill AD, Wu L, Bai Y, Conroy LR, Bruntz RC, Miao Y, Jassim BA, Babalola B, Nguele Meke FGB, Sun R, Gentry MS, Zhang ZY. Discovery and Evaluation of Active Site-Directed, Potent, and Selective Sulfophenyl Acetic Amide-Based Inhibitors for the Laforin Phosphatase. J Med Chem 2025; 68:9220-9240. [PMID: 40238926 DOI: 10.1021/acs.jmedchem.4c02580] [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: 04/18/2025]
Abstract
Lafora disease is a rare and fatal progressive myoclonus epilepsy characterized by the accumulation of insoluble glycogen deposits in the brain and peripheral tissues. Mutations in the gene encoding the glycogen phosphatase laforin result in Lafora disease. Currently, there are no laforin-specific chemical probes, limiting our understanding of the roles of laforin in glycogen metabolism and other cellular processes. Here, we identified sulfophenyl acetic amide (SPAA), as a novel nonhydrolyzable phosphotyrosine mimetic for laforin inhibition. Using fragment-based and scaffold-hopping strategies, we discovered several highly potent and selective active site-directed laforin inhibitors. Among them, compound 9c displayed a Ki value of 1.9 ± 0.2 nM and more than 8300-fold preference for laforin. Moreover, these inhibitors efficiently block laforin-mediated glucan dephosphorylation inside the cell and possess favorable pharmacokinetic properties in mice. These chemical probes will enable further investigation of the roles of laforin in normal physiological processes and in diseases.
Collapse
Affiliation(s)
- Jianping Lin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Rongjun He
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Zihan Qu
- The James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jiajun Dong
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Aaron D Krabill
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Li Wu
- Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Yunpeng Bai
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lindsey R Conroy
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Ronald C Bruntz
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Yiming Miao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Brenson A Jassim
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Benjamin Babalola
- The James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | | | - Ramon Sun
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida 32610, United States
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida 32610, United States
- McKnight Brain Institute, University of Florida, Gainesville, Florida 32610, United States
| | - Matthew S Gentry
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida 32610, United States
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida 32610, United States
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
- The James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
180
|
Kim Y, Mondal S, Shin H, Tak S, Doan VHM, Oh J, Kang HW. Advanced Precision Dual Photothermal and Photodynamic Therapy for Prostate Cancer Using PSMA-ICG-Conjugated Gold Nanorods. ACS Biomater Sci Eng 2025. [PMID: 40340375 DOI: 10.1021/acsbiomaterials.5c00209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Prostate cancer is the second most common cancer among men globally. In this study, we developed a prostate-cancer-targeted gold nanoparticle-based photothermal and photodynamic complex (GNR-ICG-FA@PSMA) to enhance the targeting efficiency of prostate cancer cells and simultaneously deliver photothermal therapy (PTT) and photodynamic therapy (PDT). For the in vitro tests, ROS assays, annexin V/PI staining, and MTT assays were conducted. In the in vivo tests, fluorescence and photoacoustic imaging systems were used to track the distribution of nanoparticles in animal models. Tumor tissues were analyzed post-treatment using Triphenyl tetrazolium chloride (TTC) staining, Hematoxylin and Eosin (HE) staining, and Immunohistochemistry (IHC) staining. The in vitro results showed that GNR-ICG with laser irradiation produced high levels of ROS, the highest rate of apoptosis, and the lowest cell viability. In the in vivo tests, tail-injected GNR-ICG-FA@PSMA reached the tumor within 9 h. During laser irradiation, GNRs increased the temperature (<50 °C), inducing necrosis, while ICGs generated ROS, leading to apoptosis. The results demonstrated that folic acid (FA) and PSMA antibodies improved prostate cancer-specific targeting. GNRs and ICGs contributed to the photothermal and photodynamic effects, respectively. This study confirms the potential of GNR-ICG-FA@PSMA for targeted photothermal and photodynamic therapy of prostate cancer.
Collapse
Affiliation(s)
- Yeongeun Kim
- Biomedical Engineering, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Sudip Mondal
- Digital Healthcare Research Center, Pukyong National University, Busan 48513, Korea
| | - Hwarang Shin
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
- Marine-integrated Biomedical Technology Center, Pukyong National University, Busan 48513, Korea
| | - Soonhyuk Tak
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Vu Hoang Minh Doan
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Korea
| | - Junghwan Oh
- Digital Healthcare Research Center, Pukyong National University, Busan 48513, Korea
- Marine-integrated Biomedical Technology Center, Pukyong National University, Busan 48513, Korea
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Korea
| | - Hyun Wook Kang
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
- Department of Biomedical Engineering, Pukyong National University, Busan 48513, Korea
- Marine-integrated Biomedical Technology Center, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
181
|
Wang B, Wang Y, Zhu Y, Guo L, Zeng H, Wu S, Wang L, Mu J, Shao X, Cheng N, Ying J, Tian Y, Xue L. Predictive factors for neoadjuvant combined immunotherapy in gastric adenocarcinoma: Focusing on the primitive enterocyte phenotype and PVR. Br J Cancer 2025:10.1038/s41416-025-03031-3. [PMID: 40341250 DOI: 10.1038/s41416-025-03031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Neoadjuvant combined immunotherapy has provided more treatment options for patients with gastric adenocarcinoma (GA). However, some GA patients, especially those with primitive enterocyte phenotype (GAPEP) show a poor response to immunotherapy, even with positive PD-L1 expression. METHOD We enrolled multiple cohorts from our center and utilized public data to identify the predictive factors and explore the immunosuppressive features of GAPEP by multi-omics methods. RESULTS Forty-seven patients with neoadjuvant combined immunotherapy were enrolled. After testing, we found PD-L1 combined positive score (CPS) ≥ 50 in biopsy tissues was significantly associated with major pathological response (MPR) (P = 0.04). RNA testing and immunohistochemical staining highlighted the cytotoxicity-associated markers (GZMA and PRF1) as the predictors to better response. Notably, GAPEP patients demonstrated resistance to therapy and exhibited worse survival outcomes. Our own and public bulk/single-cell transcriptomic analyses identified PVR as a predictor of treatment resistance and as an important immune suppressor, especially in GAPEP. Cell interaction analyses, multiple staining, and cell experiments verified the association between GAPEP and PVR. CONCLUSION Cytotoxic markers, especially GZMA and PRF1, could predict the benefit of neoadjuvant combined immunotherapy in GA than PD-L1 CPS, while PVR is a negative predictor, particularly for GAPEP patients.
Collapse
Affiliation(s)
- Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yinong Wang
- 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, China
| | - Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hua Zeng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuai Wu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Long Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiali Mu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinxin Shao
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Na Cheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yantao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
182
|
Ma N, Gao J, Pang X, Wu K, Yang S, Wei H, Hao Y. Formulation-optimized oncolytic viruses: Advancing systemic delivery and immune amplification. J Control Release 2025; 383:113822. [PMID: 40348130 DOI: 10.1016/j.jconrel.2025.113822] [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: 02/25/2025] [Revised: 04/06/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Cancer is a major global public health challenge. Traditional treatments such as surgery, radiotherapy, and chemotherapy often show limited efficacy, minimal improvements in survival rates, and high recurrence risks. With limited therapeutic options for solid tumors, tumor immunotherapy, which harness the body's immune system, has gained significant attention. Oncolytic viruses (OVs) selectively infect and destroy tumor cells, induce immunogenic cell death (ICD) and stimulate antitumor immune responses. However, current OVs therapies, which are predominantly administered via intratumoral injection, have numerous limitations, including the need for guidance, suboptimal viral spread, extracellular matrix barriers, and immune clearance. These challenges hinder repeated dosing effectiveness and restrict its clinical applicability. Although genetic engineering has improved the tumor selectivity and immune activation of OVs, significant delivery challenges remain. Recently, optimizing pharmaceutical formulations to enhance tumor targeting and viral accumulation has emerged as a key approach to improving OV therapy and expanding clinical applicability. This review highlights the critical role of pharmaceutical formulations in biologics and outlines recent advances in OVs formulations. Specifically, we discuss strategies aimed at enhancing tumor targeting, reducing adverse effects, and promoting antitumor immunity. These strategies significantly enhance OV therapeutic potential and inform novel delivery systems for clinical translation.
Collapse
Affiliation(s)
- Ningye Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jian Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Xiaoao Pang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Kexin Wu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Shihua Yang
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning 110042, China; Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang 110001, China.
| | - Heng Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Yingying Hao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
183
|
Zhao X, Peng S, Hui T, Yu X, Li H, Ni M, Liu X, Chen Y, Zhang J, Zhang H. Optimization on the extraction conditions of flavonoids from Suaeda glauca and the research of its hepatoprotection in mice. Fitoterapia 2025; 184:106606. [PMID: 40348032 DOI: 10.1016/j.fitote.2025.106606] [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: 10/20/2024] [Revised: 05/03/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Liver disease is a serious threat to health worldwide. Flavonoids from Suaeda glauca (SGF) is able to alleviate liver lipid peroxidation. However, it is unclear whether SGF could protect against liver glycogen accumulation, inflammation and fibrosis. In this study, the extraction conditions of SGF were optimized with response surface methodology. The qualitative analysis of components in SGF was carried out by a LC-MS/MS method. Moreover, SGF was administered orally to male mice given 10 % carbon tetrachloride (CCl4) for 4 weeks at doses of 25 and 50 mg/kg once daily for 4 weeks. The optimal extraction conditions of SGF were as follows: the ratio of material to liquid 1:35, the temperature 67 °C, the time 3 h, and the ethanol concentration 89 %. Thirty-five compounds were preliminarily identified in SGF. Furthermore, SGF could significantly improve liver dysfunction, regulate the hepatic protein levels of glucose-6-phosphatase, glycogen synthase, glycogen phosphorylase L, Laforin, interleukin-1β, gasdermin-D, NOD-like receptor (NLR) family, pyrin domain containing 3 inflammasome, α-smooth muscle actin, the hepatic mRNA levels and enzyme activities of matrix metallopeptidase 9, tissue inhibitor of metalloproteinases 1, and collagen 1α1, reduce the liver glycogen accumulation, inflammation and fibrosis in mice induced by CCl4. These results indicated that SGF may be a promising drug for the treatment of liver injury.
Collapse
Affiliation(s)
- Xiaojuan Zhao
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China.
| | - Shengjie Peng
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Ting Hui
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Xinrong Yu
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Haorong Li
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Min Ni
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Xue Liu
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Yu Chen
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Jiankang Zhang
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China
| | - Hua Zhang
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, PR China.
| |
Collapse
|
184
|
Huang Y, Li Q, He F, Yang T, Zhou Q, Zheng Y, Li Y. Cationic Azobenzene Tag to Enhance Liposomal Prodrug Retention and Tumor-Targeting Prodrug Activation for Improved Antitumor Efficacy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26323-26337. [PMID: 40285708 DOI: 10.1021/acsami.5c01634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
In this study, we reported a cationic azobenzene (Azo) tag to increase the retention of camptothecin (CPT) prodrugs in liposomes driven by π-π stacking interaction between Azo. Compared with a cationic CPT prodrug without Azo, the liposome-encapsulating Azo-linked CPT prodrugs (AzoCPT-Lips) exhibited slower prodrug leakage in plasma and a longer blood circulation time in mice. The AzoCPT-Lips had a high encapsulation efficiency (95%), loading capacity (20%, by weight), and good storage stability. The AzoCPT was efficiently taken up by 4T1 tumor cells (100-fold higher than CPT) and readily converted into active CPT in the cytoplasm to exert 10-fold higher cytotoxicity than free CPT. More importantly, AzoCPT-Lips resulted in 5-20 times higher tumor distribution of active CPT than that of CPT solution or those in other tissues, which further led to more potent antitumor activity and lower toxicities in the 4T1 breast cancer xenograft. Such a cationic Azo tag represents an effective strategy for developing liposomal antitumor drugs with improved antitumor efficacy.
Collapse
Affiliation(s)
- Yuanyuan Huang
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qiunan Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Fei He
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tao Yang
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu 610051, China
| | - Yaxin Zheng
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yang Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
185
|
Hu Y, Zhou J, Gao Y, Fan Y, Chen B, Su J, Li H. Multifunctional nanocomposite hydrogels: an effective approach to promote diabetic wound healing. Biomed Mater 2025; 20:032006. [PMID: 40273939 DOI: 10.1088/1748-605x/add06f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/24/2025] [Indexed: 04/26/2025]
Abstract
Diabetes, a metabolic disease that is becoming increasingly severe globally, presents a significant challenge in the medical field. Diabetic wounds are characterized by their chronicity, difficulty healing, and complex microenvironment that harbors multiple adverse factors, including elevated hyperglycemia, persistent inflammation, susceptibility to infections, and oxidative stress, all of which contribute to the impaired healing process. Nanocomposite hydrogels, as materials with unique physicochemical properties and biocompatibility, have gained growing attention in recent years for their potential applications in diabetic wound healing. These hydrogels provide a moist healing environment for wounds and regulate cellular behavior and signaling pathways, promoting wound repair and healing. By introducing specific functional groups and nanoparticles, nanocomposite hydrogels can respond to pathological features of wounds, enabling adaptive drug release. Owing to their diverse bioactive functions, nanocomposite hydrogels are powerful tools for the treatment of diabetic wounds. Thus, this article provides an overview of recent progress in the use of nanocomposite hydrogels for diabetic wound healing.
Collapse
Affiliation(s)
- Yuchen Hu
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Junchao Zhou
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Yuhang Gao
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Ying Fan
- Chongqing University Jiangjin Hospital, Chongqing 402260, People's Republic of China
| | - Ban Chen
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Jiangtao Su
- National '111' Centre for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Hong Li
- School of Pharmacy, Guangxi Medical University, Nanning 530021, People's Republic of China
| |
Collapse
|
186
|
Zhang D, Chen Q, Zhang J, Xing X, Zhou Y, Ou X, Dai S, Chen Q, Liu X, Chen X, Zeng Y. Amplifying X-ray-Induced Charge Transfer Facilitates Direct Sensitization of Photosensitizers in Radiotherapy. ACS NANO 2025; 19:16775-16793. [PMID: 40277128 DOI: 10.1021/acsnano.5c01506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
X-ray-induced photodynamic therapy offers substantial promise for treating deep-seated tumors, but it is still limited by highly inefficient energy transfer processes and the stringent requirements for scintillators with high luminescence quantum yield and significant singlet-triplet intersystem crossing ratios. Herein, we describe X-ray-induced electron-dynamic therapy (X-eDT), which obviates the need for intersystem crossing by exposing nonluminescent hafnium-silica nanoparticles to X-rays, to generate high-energy electrons that can sensitize lower-lying triplet states of various photosensitizers. Our approach strongly induced the production of singlet oxygen (6.18-fold) in vitro even at lower X-ray doses, and in mice it strongly inhibited the growth of xenografts derived from liver, breast, or colon cancer cell lines (CDX), and growth of patient-derived xenografts (PDX) of hepatocellular carcinoma. In these CDX preclinical systems, X-eDT was not only effective against the irradiated xenograft but also against untreated xenografts in the same animal, and these abscopal effects involved enhanced tumor infiltration by CD4+T cells, CD8+T cells, and IFN-γ-polarized M1 macrophages within the tumor microenvironment. X-eDT even stimulated the production of memory T cells that inhibited rechallenges after treatment. These findings suggest that X-eDT can be effective against primary and metastatic tumors as well as tumor recurrence, which makes it much more powerful than conventional X-PDT.
Collapse
Affiliation(s)
- Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Qingjing Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Junrong Zhang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, P. R. China
| | - Xiaohua Xing
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fuzhou University, Fuzhou 350108, P. R. China
| | - Yang Zhou
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xiangyu Ou
- Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping SE-58183, Sweden
| | - Shuheng Dai
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fuzhou University, Fuzhou 350108, P. R. China
| | - Qiushui Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fuzhou University, Fuzhou 350108, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 138667, Singapore
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, P. R. China
| |
Collapse
|
187
|
Miller J, Perrier Q, Rengaraj A, Bowlby J, Byers L, Peveri E, Jeong W, Ritchey T, Gambelli AM, Rossi A, Calafiore R, Tomei A, Orlando G, Asthana A. State of the Art of Bioengineering Approaches in Beta-Cell Replacement. CURRENT TRANSPLANTATION REPORTS 2025; 12:17. [PMID: 40342868 PMCID: PMC12055624 DOI: 10.1007/s40472-025-00470-y] [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] [Accepted: 04/09/2025] [Indexed: 05/11/2025]
Abstract
Purpose of the Review Despite recent advancements in technology for the treatment of type 1 diabetes (T1D), exogenous insulin delivery through automated devices remains the gold standard for treatment. This review will explore progress made in pancreatic islet bioengineering within the field of beta-cell replacement for T1D treatment. Recent Findings First, we will focus on the use of decellularized extracellular matrices (dECM) as a platform for pancreatic organoid development. These matrices preserve microarchitecture and essential biochemical signals for cell differentiation, offering a promising alternative to synthetic matrices. Second, advancements in 3D bioprinting for creating complex organ structures like pancreatic islets will be discussed. This technology allows for increased precision and customization of cellular models, crucial for replicating native pancreatic islet functionality. Finally, this review will explore the use of stem cell-derived organoids to generate insulin-producing islet-like cells. While these organoids face challenges such as functional immaturity and poor vascularization, they represent a significant advancement for disease modeling, drug screening, and autologous islet transplantation. Summary These innovative approaches promise to revolutionize T1D treatment by overcoming the limitations of traditional therapies based on human pancreatic islets.
Collapse
Affiliation(s)
- Jake Miller
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Quentin Perrier
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
- Univ. Grenoble Alpes, Department of Pharmacy, Grenoble Alpes University Hospital, Grenoble, France
| | - Arunkumar Rengaraj
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Joshua Bowlby
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Lori Byers
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Emma Peveri
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Wonwoo Jeong
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Thomas Ritchey
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | | | - Arianna Rossi
- Department of Engineering, University of Perugia, Perugia, Italy
| | | | - Alice Tomei
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Giuseppe Orlando
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Amish Asthana
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| |
Collapse
|
188
|
Zhang Y, Chen H, You S, Chi X, Chen Y, Wu Z, Zheng J, Cheng X, Liu J, Li J. Preliminary investigation of nicotinamide N-methyltransferase as an HBV-specific biomarker for hepatocellular carcinoma diagnosis. Int J Biol Markers 2025:3936155251330664. [PMID: 40329575 DOI: 10.1177/03936155251330664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
BackgroundNicotinamide N-methyltransferase (NNMT), a metabolic enzyme in the liver, has been implicated in various biological processes, and its high expression in hepatocellular carcinoma has been linked to tumor metastasis and poor prognosis. However, its potential as a serum biomarker for hepatocellular carcinoma diagnosis remains unexplored.MethodsA total of 172 subjects were included in this study, consisting of 71 hepatocellular carcinoma patients (64 with hepatitis B virus (HBV)-associated hepatocellular carcinoma and 7 with non-HBV-associated hepatocellular carcinoma), as well as 70 healthy controls and 31 HBV-infected individuals. Serum NNMT levels were measured, and clinical-pathological correlations were analyzed. The diagnostic efficacy of serum NNMT for HBV-related hepatocellular carcinoma was evaluated using receiver operating characteristic (ROC) curve analysis.ResultsSerum NNMT levels were significantly elevated in HBV-infected individuals and correlated with poorer prognosis, including reduced overall survival and shorter disease-free survival. Kaplan-Meier analysis revealed that low NNMT expression was associated with longer overall survival (75 vs. 12 months, P < 0.0001) and disease-free survival (21.5 vs. 5 months, P < 0.01). In HBV-related hepatocellular carcinoma patients, NNMT levels correlated with biochemical markers including alfa-fetoprotein, aspartate transaminase, triglycerides, total cholesterol, low-density lipoprotein, apolipoprotein B, TB, and albumin, with decreased albumin, and high-density lipoprotein levels promoting NNMT expression. ROC analysis showed that NNMT outperformed alfa-fetoprotein (area under the curve (AUC) 0.869 vs. 0.775), with a sensitivity of 95.2%, specificity of 87.9%, and a combined AUC of 0.947, demonstrating its superior diagnostic value for HBV-related hepatocellular carcinoma.ConclusionsSerum NNMT is a promising biomarker for predicting the risk of hepatocellular carcinoma in HBV-infected individuals and may serve as an indicator for the prognosis of hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Yi Zhang
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Huayang Chen
- Department of Colorectal Surgery, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Song You
- Department of Hepatobiliary Surgery, Meng Chao Hepatobiliary Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xiaoqin Chi
- Department of Hepatobiliary Surgery, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen Translational Medical Key Laboratory of Digestive System Tumor, Zhongshan Hospital of Xiamen University, Fujian Province, China
| | - Yongxi Chen
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Zhiyong Wu
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Jiepeng Zheng
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Xin Cheng
- Vascular Surgery Department, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Jianming Liu
- Department of Hepatobiliary Surgery, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen Translational Medical Key Laboratory of Digestive System Tumor, Zhongshan Hospital of Xiamen University, Fujian Province, China
| | - Jie Li
- Department of Hepatobiliary Surgery, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen Translational Medical Key Laboratory of Digestive System Tumor, Zhongshan Hospital of Xiamen University, Fujian Province, China
| |
Collapse
|
189
|
Saadh MJ, Hussein A, Bayani A, Dastafkan S, Amiri M, Akbari A, Shahsavan S, Soleimani Samarkhazan H, Shirani Asl V. Mesenchymal stem cell-derived exosomes: a novel therapeutic frontier in hematological disorders. Med Oncol 2025; 42:199. [PMID: 40327167 DOI: 10.1007/s12032-025-02742-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells valued for their immunomodulatory and regenerative properties, positioning them as a cornerstone of regenerative medicine. Their derived exosomes small extracellular vesicles laden with bioactive molecules such as proteins, lipids, and nucleic acids have emerged as critical mediators of MSC therapeutic effects. This review systematically explores the biology of MSC-derived exosomes, detailing their biogenesis, molecular composition, and pivotal roles in hematopoiesis, inflammation, and immune regulation. In hematological disorders, including leukemia, lymphoma, and myelodysplastic syndromes, these exosomes exhibit significant therapeutic potential by modulating the tumor microenvironment, enhancing hematopoietic recovery, and suppressing malignant cell proliferation. Notable findings include their ability to induce cell cycle arrest in leukemia cells via the p53 pathway and to reduce chemoresistance through targeted signaling mechanisms, such as the IRF2/INPP4B axis. However, clinical translation is hindered by several challenges, including the standardization of isolation techniques such as ultracentrifugation which are costly and susceptible to contamination as well as difficulties in optimizing large-scale production and ensuring long-term safety and efficacy. Despite these obstacles, MSC-derived exosomes offer a promising, cell-free therapeutic alternative that minimizes risks such as immune rejection and tumorigenicity associated with whole-cell therapies. Future research must prioritize the refinement of isolation and production protocols, the development of precise delivery strategies, and the execution of comprehensive safety evaluations to unlock their full clinical potential in treating hematological disorders and beyond. This review integrates recent advancements to provide a clearer understanding of their multifaceted contributions and highlights the critical gaps that remain.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Ahmed Hussein
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Alireza Bayani
- Division of Hematology and Blood Bank, Department of Laboratory Science, School of Paramedical Science, Shiraz University of Med1ical Sciences, Shiraz, Iran
| | - Shayan Dastafkan
- Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahdie Amiri
- Department of Laboratory Sciences, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Atie Akbari
- Department of Family Medicine, School of Medicine, Ziaeian Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shaghayegh Shahsavan
- HSCT Research Center, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Soleimani Samarkhazan
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Vida Shirani Asl
- Division of Hematology and Blood Bank, Department of Laboratory Science, School of Paramedical Science, Shiraz University of Med1ical Sciences, Shiraz, Iran.
| |
Collapse
|
190
|
Ferrari S, Ferulli F, Galla R, Vicini R, Cattaneo V, Mulè S, Uberti F. Effective Restoration of Gastric and Esophageal Tissues in an In Vitro Model of GERD: Mucoadhesive and Protective Properties of Xyloglucan, Pea Proteins, and Polyacrylic Acid. Int J Mol Sci 2025; 26:4409. [PMID: 40362645 PMCID: PMC12073054 DOI: 10.3390/ijms26094409] [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: 02/17/2025] [Revised: 05/02/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025] Open
Abstract
Esophageal barrier dysfunction is a crucial pathophysiological mechanism of gastroesophageal reflux disease (GERD). However, treatments mainly aim to reduce gastric acidity rather than improve tissue integrity. This study evaluated the protective and mucoadhesive properties of a formulation containing xyloglucan, pea proteins, and polyacrylic acid (XPPA) in gastric and esophageal cells. Cells were exposed to hydrochloric acid (HCl) and subsequently treated with the test compound. Trans-epithelial electrical resistance (TEER), tight junction (TJ) expression, and mucoadhesion of XPPA on gastric and esophageal cells were evaluated. To further confirm the protective ability of XPPA, a Lucifer Yellow assay was performed on a human reconstructed esophageal epithelium to assess the ability of XPPA to prevent HCl-induced hyperpermeability. XPPA possesses noteworthy mucoadhesive properties, ensuring an extended contact time between the product and the damaged mucosa to allow sustained mucosal protection. Furthermore, XPPA effectively restored gastroesophageal barrier integrity after HCl-induced damage, as assessed with TEER, after 1 h (p < 0.05). Finally, XPPA helped to restore TJ expression (p < 0.05) and protected the tissues from hyperpermeability for at least 2 h (p < 0.05). These results pave the way for using XPPA as a promising treatment to ameliorate gastroesophageal barrier properties in GERD patients.
Collapse
Affiliation(s)
- Sara Ferrari
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | | | - Rebecca Galla
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
- Noivita Srls, Spin Off, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | | | | | - Simone Mulè
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | - Francesca Uberti
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| |
Collapse
|
191
|
Ren Q, Wang F, Du F, He C, Wang X, Wang J, Zhang Z, Sun Y. Asiaticoside enhances the anti-tumor effect of anti-PDL1 by regulating T cell activity through increasing LCK activity. Pathol Res Pract 2025; 271:155995. [PMID: 40373489 DOI: 10.1016/j.prp.2025.155995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/12/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025]
Abstract
Anti-PD-L1 antibody confers anti-tumor effects, but its long-term use can provoke resistance and adverse effects. Asiaticoside, a bioactive triterpene glycoside from Centella asiatica L., regulates immune function and induces apoptosis of hepatocellular carcinoma (HCC) cells. T cells play a vital role in killing tumor cells and require lymphocyte-specific protein tyrosine kinase (LCK) for activation. Here, we examined whether a combined asiaticoside and anti-PD-L1 treatment regulates T cells via LCK activation to enhance the anti-tumor effect in vivo. We established a subcutaneous mouse HCC model using Hepa1-6 cells and measured spleen and tumor weight. Morphological changes of tumor tissues were assessed by hematoxylin-eosin staining. Tumor cell apoptosis and proliferation were determined by TUNEL staining and KI67 immunohistochemistry. The proportion of activated T cells in the spleen was detected by flow cytometry, and the levels of phosphorylated p-LCK and p-AKT in the spleen were determined by Western blotting. Changes in the levels of serum inflammatory factors were detected with ELISA. Our results revealed that the combined asiaticoside and anti-PD-L1 treatment inhibited tumor growth by enhancing apoptosis and reducing tumor cell proliferation. The treatment activated T cells to increase the proportion of effector T cells in the spleen, evidenced by upregulated p-LCK and p-AKT levels. It also increased the level of TNF-α in the serum and decreased IL-6, implying an enhanced immune response. In conclusion, the combined asiaticoside and anti-PD-L1 treatment enhances the anti-HCC effect in vivo by promoting LCK activation to regulate T cells.
Collapse
Affiliation(s)
- Qingyi Ren
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Fang Wang
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China; Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China; Central Nervous System Drug Key Laboratory of Sichuan Province
| | - Fei Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chenxi He
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jun Wang
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China; Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| | - Yuhong Sun
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| |
Collapse
|
192
|
Zaccariotto GDC, Bistaffa MJ, Zapata AMM, Rodero C, Coelho F, Quitiba JV, Lima L, Sterman R, Cardoso VDO, Zucolotto V. Cancer Nanovaccines: Mechanisms, Design Principles, and Clinical Translation. ACS NANO 2025; 19:16204-16223. [PMID: 40202241 PMCID: PMC12060653 DOI: 10.1021/acsnano.4c15765] [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: 11/05/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Cancer immunotherapy has transformed the landscape of oncological treatment by employing various strategies to teach the immune system to eliminate tumors. Among these, cancer nanovaccines are an emerging strategy that utilizes nanotechnology to enhance immune activation in response to tumor antigens. This review addresses the principles behind the different technologies in this field aimed at generating a robust and effective immune response. The diversity of strategies adopted for the design of nanovaccines is discussed, including the types of active agents, nanocarriers, their functionalizations, and the incorporation of adjuvants. Furthermore, strategies to optimize nanoparticle formulations to enhance the antigen presentation, target immune cells, and organs and promote strong and durable antitumor responses are explored. Finally, we analyze the current state of clinical application, highlighting ongoing clinical trials and the future potential of cancer nanovaccines. The insights presented in this review aim to guide future research and development efforts in the field, contributing to the advancement of more effective and targeted nanovaccines in the fight against cancer.
Collapse
Affiliation(s)
- Gabriel de Camargo Zaccariotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Maria Julia Bistaffa
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Angelica Maria Mazuera Zapata
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Camila Rodero
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Fernanda Coelho
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - João Victor
Brandão Quitiba
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Lorena Lima
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Raquel Sterman
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | | | - Valtencir Zucolotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| |
Collapse
|
193
|
Tian S, Yu Z, Wang Y, Chen S, Li M, Tang D. Crystal Facet Engineering Modulated Electron Transfer Mechanisms: A Self-Powered Photoelectrochemical Sensing Platform for Noninvasive Detection of Uric Acid. Anal Chem 2025; 97:9518-9526. [PMID: 40289317 DOI: 10.1021/acs.analchem.5c01345] [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: 04/30/2025]
Abstract
Crystal facet engineering is a pivotal strategy to design high-performance photoelectrodes and suppress electron and hole complexation, thus enhancing photoelectrochemical (PEC) activity through carrier enrichment at specific crystal facets. However, there is still a lack of systematic resolution on the intrinsic principles of crystal facet tuning energy band structure and the specific adsorption of signaling molecules. In this work, a multidimensional synergistic optimization strategy was proposed to achieve precise prediction and targeted crystal facet design of photoelectrodes by establishing a quantitative structure-activity relationship (QSAR) model of "crystal configuration-molecular recognition-carrier transport". A three-dimensional hierarchical TiO2 nanoflower (3D HTNF) photoelectrode dominated by the {110} facet exhibited a significant positive photocurrent toward uric acid (UA). Integrated with a microelectromechanical system (MEMS), a miniaturized self-powered PEC biosensor provided an innovative solution for high-throughput, noninvasive UA monitoring in saliva and displayed a linear range of 0.01-50 μM with a detection limit of 8.76 nM. In addition, the advantages of photoelectrodes in light harvesting, charge separation and migration, molecular adsorption, and surface reactions were verified by density functional theory (DFT) calculations to reveal the path selectivity and carrier transport mechanisms of the photo-oxidation reactions on specific crystal surfaces. This study elucidates the interplay mechanism of the crystal surface tuning energy band structure and the interfacial kinetics of response. The program can be extended to precisely detect biomarkers in complex biological matrices, promoting the leapfrog development of noninvasive health monitoring technology.
Collapse
Affiliation(s)
- Shuo Tian
- Key Laboratory of Analytical Science for Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Zhichao Yu
- Key Laboratory of Analytical Science for Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Yunsen Wang
- Key Laboratory of Analytical Science for Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Shuyun Chen
- Key Laboratory of Analytical Science for Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Meijin Li
- Key Laboratory of Analytical Science for Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Dianping Tang
- Key Laboratory of Analytical Science for Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| |
Collapse
|
194
|
Cheng Z, Ye Q, Lao J, Liu X, Wu P. Conjugated Polymer-Photosensitizers for Cancer Photodynamic Therapy and Their Multimodal Treatment Strategies. Polymers (Basel) 2025; 17:1258. [PMID: 40363042 PMCID: PMC12074309 DOI: 10.3390/polym17091258] [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: 04/14/2025] [Revised: 04/29/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Conjugated polymers (CPs) have emerged as promising candidates for photodynamic therapy (PDT) in cancer treatment due to their high fluorescence quantum yield, excellent photostability, and remarkable reactive oxygen species (ROS) generation capability. This review systematically summarizes molecular design strategies to augment CP photosensitivity efficiency, including: (1) constructing donor-acceptor (D-A) alternating structures, (2) incorporating aggregation-induced emission (AIE) moieties, (3) employing heavy-atom effects, and (4) designing hyperbranched architectures. In addition, considering the limitations of monotherapy like tumor heterogeneity, we will further discuss the synergistic treatment strategies of CP-mediated PDT in combination with other therapeutic modalities, including photothermal therapy (PTT)-PDT, immunotherapy-PDT, chemotherapy-PDT, Chemiluminescence (CL)-PDT, diagnostic technology-PDT, and chemodynamic therapy (CDT)-PDT. These multimodal approaches leverage complementary mechanisms to achieve enhanced tumor eradication efficacy.
Collapse
Affiliation(s)
- Zhengqing Cheng
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (Z.C.); (Q.Y.); (J.L.)
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Qiuting Ye
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (Z.C.); (Q.Y.); (J.L.)
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Jieling Lao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (Z.C.); (Q.Y.); (J.L.)
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (Z.C.); (Q.Y.); (J.L.)
| | - Pan Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; (Z.C.); (Q.Y.); (J.L.)
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
195
|
Ho NHJG, Talvard-Balland N, Köhler N, Zeiser R. Immune Escape of Acute Myeloid Leukemia after Transplantation. Blood Cancer Discov 2025; 6:168-181. [PMID: 40168448 PMCID: PMC12050969 DOI: 10.1158/2643-3230.bcd-24-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/13/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025] Open
Abstract
SIGNIFICANCE We discuss the mechanisms of AML immune evasion including loss or downregulation of MHC class I and II, reduced TRAIL receptor expression, inhibitory metabolite production, inhibitory ligand expression, impaired proinflammatory cytokine production, and AML niche alterations.
Collapse
Affiliation(s)
- Nguyen Huong Jenny Giang Ho
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
| | - Nana Talvard-Balland
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
| | - Natalie Köhler
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
- CIBSS – Centre for Integrative Biological Signaling Studies, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
| |
Collapse
|
196
|
Xu M, Yu Z, Wei Q, Tang D. Inflammatory microenvironment-responsive electrochemical biosensing for cancer cell discrimination using PtZnCd-anchored multi-walled carbon nanotubes. Talanta 2025; 294:128277. [PMID: 40334508 DOI: 10.1016/j.talanta.2025.128277] [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: 02/13/2025] [Revised: 04/15/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
The quantification of intracellular hydrogen peroxide (H2O2) serves as a critical biomarker for characterizing cellular physiological states, providing essential insights into metabolic regulation and signaling pathways. This analytical paradigm not only advances our understanding of pathological mechanisms but also contributes to the development of novel diagnostic approaches and precision therapeutic interventions. Herein, we established an innovative electrochemical microsensing platform capable of discriminating between malignant and normal cells through their distinct inflammatory responses under external stimulation. This innovative methodology integrates three critical technical advancements: (i) optimization of a one-pot microwave synthesis protocol for fabricating high-performance PtZnCd nanoparticles anchored on multi-walled carbon nanotubes (MWCNTs), which serve as the core sensing element; (ii) systematic electrochemical characterization coupled with density functional theory (DFT) calculations demonstrating that this hybrid architecture significantly reduces interfacial charge-transfer resistance while enhancing heterogeneous electron transfer kinetics; (iii) comprehensive biocompatibility evaluations confirming the composite material's favorable cytotoxicity profile and biological safety, supporting its potential for cellular classification applications. Through real-time monitoring of dynamic metabolic fluctuations and intracellular inflammatory microenvironment changes in response to ascorbic acid (AA) and dehydroascorbic acid (DHA) stimulation, we established distinct response signatures that effectively differentiate neoplastic cells from their healthy counterparts. This study introduces an innovative electrochemical sensing paradigm that synergistically combines biocompatible nanocatalysts with inflammatory microenvironment dynamics, establishing a robust platform for dual discrimination between cancerous and normal cells, with significant implications for biomedical research and clinical diagnostics.
Collapse
Affiliation(s)
- Man Xu
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Zhichao Yu
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Qiaohua Wei
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China.
| | - Dianping Tang
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China.
| |
Collapse
|
197
|
Li B, Liu XJ, Zhu HW, Guan HP, Guo RT. Recent Progress of Round-the-Clock Photocatalytic System in Environmental and Energy Applications: A Review. Chemphyschem 2025; 26:e202401144. [PMID: 39934953 DOI: 10.1002/cphc.202401144] [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: 12/23/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
The prevailing overdependence on fossil fuels contributes to energy supply instability and pronounced price volatility. The combustion of these fuels emits considerable greenhouse gases and pollutants, further deteriorating the climate and environment. In response, Round-the-Clock Photocatalytic Systems (RTCPs) have emerged as a viable technological solution, attracting significant research interest due to their convenience, sustainability, and environmental benefits etc. which act as "photo-batteries" facilitate catalytic processes in the absence of light, offering continuous operation. Given the considerable potential of RTCPs, a timely examination of recent advancements is essential to optimize efforts. This review delineates the fundamental mechanisms of RTCPs, explores innovative strategies and current developments, and addresses the challenges of scaling up production. It aims to provide new insights and serve as a foundational reference for future research on RTCPs.
Collapse
Affiliation(s)
- Bo Li
- College of Energy Source and Mechanical Engineering, Shanghai University of Electric Power, Shanghai, 200090, People's Republic of China
| | - Xiao-Jing Liu
- College of Energy Source and Mechanical Engineering, Shanghai University of Electric Power, Shanghai, 200090, People's Republic of China
| | - Hao-Wen Zhu
- College of Energy Source and Mechanical Engineering, Shanghai University of Electric Power, Shanghai, 200090, People's Republic of China
| | - Hua-Peng Guan
- College of Energy Source and Mechanical Engineering, Shanghai University of Electric Power, Shanghai, 200090, People's Republic of China
| | - Rui-Tang Guo
- College of Energy Source and Mechanical Engineering, Shanghai University of Electric Power, Shanghai, 200090, People's Republic of China
- Shanghai Non-Carbon Energy Conversion and Utilization Institute, Shanghai, 200090, People's Republic of China
| |
Collapse
|
198
|
Ultimo A, Jain A, Gomez-Gonzalez E, Alex TS, Moreno-Borrallo A, Jana S, Ghosh S, Ruiz-Hernandez E. Nanotherapeutic Formulations for the Delivery of Cancer Antiangiogenics. Mol Pharm 2025; 22:2322-2349. [PMID: 40184281 PMCID: PMC12056699 DOI: 10.1021/acs.molpharmaceut.4c00822] [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: 07/24/2024] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/06/2025]
Abstract
Antiangiogenic medications for cancer treatment have generally failed in showing substantial benefits in terms of prolonging life on their own; their effects are noticeable only when combined with chemotherapy. Moreover, treatments based on prolonged antiangiogenics administration have demonstrated to be ineffective in stopping tumor progression. In this scenario, nanotherapeutics can address certain issues linked to existing antiangiogenic treatments. More specifically, they can provide the ability to target the tumor's blood vessels to enhance drug accumulation and manage release, ultimately decreasing undesired side effects. Additionally, they enable the administration of multiple angiogenesis inhibitors at the same time as chemotherapy. Key reports in this field include the design of polymeric nanoparticles, inorganic nanoparticles, vesicles, and hydrogels for loading antiangiogenic substances like endostatin and interleukin-12. Furthermore, nanoformulations have been proposed to efficiently control relevant pro-angiogenic pathways such as VEGF, Tie2/Angiopoietin-1, HIF-1α/HIF-2α, and TGF-β, providing powerful approaches to block tumor growth and metastasis. In this article, we outline a selection of nanoformulations for antiangiogenic treatments for cancer that have been developed in the past ten years.
Collapse
Affiliation(s)
- Amelia Ultimo
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Ayushi Jain
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Elisabet Gomez-Gonzalez
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Thomson Santosh Alex
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Almudena Moreno-Borrallo
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Sukanya Jana
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Shubhrima Ghosh
- Trinity
Translational Medicine Institute, Trinity College Dublin, the University
of Dublin, St. James’s
Hospital, Dublin 8 D08 NHY1, Ireland
- School
of Biological, Health and Sports Sciences, Technological University Dublin, Grangegorman Lower, Dublin 7 D07 ADY7, Ireland
| | - Eduardo Ruiz-Hernandez
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| |
Collapse
|
199
|
Chen D, Zhao Z, Hong R, Yang D, Gong Y, Wu Q, Wang Y, Cao Y, Chen J, Tai Y, Liu H, Li J, Fan J, Zhang W, Song Y, Zhan Q. Harnessing the FGFR2/NF2/YAP signaling-dependent necroptosis to develop an FGFR2/IL-8 dual blockade therapeutic strategy. Nat Commun 2025; 16:4128. [PMID: 40319089 PMCID: PMC12049493 DOI: 10.1038/s41467-025-59318-9] [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] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
The multifaceted roles and mechanisms of necroptosis in cancer cells remain incompletely understood. Here, we demonstrate that FGFR2 inhibition potently inhibits esophageal squamous cell carcinoma (ESCC) by inducing necroptosis in a RIP1/MLKL-dependent manner and show RIP3 is dispensable in this pathway. Notably, MST1 is identified as a necroptotic pathway component that interacts with RIP1 and MLKL to promote necroptosis by phosphorylating MLKL at Thr216. Additionally, FGFR2 inhibition induces Ser518 phosphorylation and triggers ubiquitin-mediated degradation of NF2, culminating in Hippo pathway suppression. Subsequently, YAP activation promotes RIP1 and MLKL transcriptional upregulation, further amplifying necroptosis. Intriguingly, IL-8 derived from necrotic cells stimulates peripheral surviving tumor cells to increase PD-L1 expression. Dual blockade of FGFR2/PD-L1 or FGFR2/IL-8-CXCR1/2 robustly impedes tumor growth in humanized mouse xenografts. Collectively, our findings delineate an alternative FGFR2-NF2-YAP signaling-dependent necroptotic pathway and shed light on the immunoregulatory role of FGFR2, offering promising avenues for combinatorial therapeutic strategies in clinical cancer management.
Collapse
Affiliation(s)
- Dongshao Chen
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Zitong Zhao
- 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, China
| | - Ruoxi Hong
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Di Yang
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Gong
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingnan Wu
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Wang
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiren Cao
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Chen
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Yidi Tai
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Haoyu Liu
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Jinting Li
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiawen Fan
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Weimin Zhang
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China.
- Department of Oncology, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, China.
| | - Yongmei Song
- 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, China.
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China.
- Department of Oncology, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, China.
- International Cancer Institute, Peking University Health Science Center, Beijing, China.
- Soochow University Cancer institute, Suzhou, Jiangsu, China.
| |
Collapse
|
200
|
Shu Y, Tao Q, Xu Q, Chen Y, Xu Y, Ma T, Zhu Z, Wei X, Liu F, Wu Z, Zeng Y, Chen M, Shao M, Cao X, Zhou Y, Peng W, Li C, Shi Y. Loss of NUMB promotes hepatomegaly and hepatocellular carcinoma through the AKT/glycogen/hippo signaling. Oncogene 2025:10.1038/s41388-025-03430-z. [PMID: 40319143 DOI: 10.1038/s41388-025-03430-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Excessive glycogen deposition is a common feature of liver enlargement, liver adenoma, and liver cancer, yet the underlying mechanisms remain poorly understood. In this study, we found that NUMB, a well-known cell fate determinant, is downregulated in glycogen-rich adenomas and hepatocellular carcinoma (HCC). NUMB-deficient livers developed excessive glycogen accumulation and adenoma formation particularly in aged mice. Surprisingly, the Alb-Cre:Trp53loxP/loxP liver displayed no similar defective morphology and function, although p53 is considered an important downstream target of NUMB and closely related to glucose metabolism. Instead, we observed a synergistic interaction between NUMB and p53 in regulating glycogen metabolism in HCC tissues and cell lines. Combined knockout of NUMB and p53 in mice significantly enhances glycogen accumulation and hepatomegaly, particularly when mice are subjected to a high sugar diet (HSD), leading to higher cancer incidence. Mechanistically, NUMB deficiency disrupts the PTEN-PI3K/AKT signaling pathway, promoting glycogen accumulation. Subsequently, successive glycogen deposition triggers hepatomegaly and tumorigenesis via the Hippo signaling pathway. Our results suggest that NUMB plays a crucial role in maintaining the homeostasis of glucose metabolism and suppressing the development of liver tumors associated with glycogen deposition.
Collapse
Affiliation(s)
- Yuke Shu
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Tao
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Xu
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuwei Chen
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yahong Xu
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingting Ma
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiqi Zhu
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Wei
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Liu
- Department of Biliary Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenru Wu
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuting Zeng
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Menglin Chen
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingyang Shao
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoyue Cao
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongjie Zhou
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Peng
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Li
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yujun Shi
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Institute of Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|