1
|
Gong G, Huang H, Tong Z, Zheng Y, Bian D, Zhang Y. Implant derived high local concentration of magnesium inhibits tumorigenicity of osteosarcoma. Biomaterials 2025; 320:123263. [PMID: 40132359 DOI: 10.1016/j.biomaterials.2025.123263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/25/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
Osteosarcoma (OS) is a fatal malignant tumor that occurs in bone, whose main treatment is surgical resection. With anti-tumor and osteogenic effects, Magnesium (Mg) is a promising biodegradable metal for postoperative treatment in OS, however, its anti-OS effect and mechanism still need to be explored. Here, while holding the ability to promote osteogenesis, Mg metal at the same time significantly reduces the proliferation, migration and invasion of various OS cells (UMR106, 143B, K7M2) in vitro. Similarly, it inhibits the growth and lung metastasis of UMR106 induced tumors in xenograft models in vivo. The mRNA-seq analysis shows that Mg significantly inhibits Wnt-pathway (increased APC, Axin2 and GSK3β to induce degradation of β-catenin) in typical OS, which is further verified by western blotting and immunofluorescence analyses. A Mg2+ concentration of 240 mg/L, either from Mg metal extract or Mg salt (MgCl2), equivalently exhibits significantly increased APC, Axin2, GSK3β and decreased β-catenin, and then inhibits tumorigenicity of typical OS cells. This work reveals that a local high concentration of Mg can inhibit OS by down-regulating Wnt-pathway, and meanwhile favors for normal health bone, which demonstrates a new approach and mechanism in the treatment of OS with Mg-based biodegradable metals.
Collapse
Affiliation(s)
- Gencheng Gong
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Orthopedics, Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - He Huang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450003, China
| | - Zhipei Tong
- Medical Research Institute, Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yufeng Zheng
- Department of Orthopedics, Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Dong Bian
- Medical Research Institute, Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Yu Zhang
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Orthopedics, Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
2
|
Xie Y, Guo J, Hu J, Li Y, Zhang Z, Zhu Y, Deng F, Qi J, Zhou Y, Chen W. A factorial design-optimized microfluidic LNP vaccine elicits potent magnesium-adjuvating cancer immunotherapy. Mater Today Bio 2025; 32:101703. [PMID: 40230646 PMCID: PMC11994397 DOI: 10.1016/j.mtbio.2025.101703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025] Open
Abstract
Human papillomavirus (HPV)-associated cancers remain a critical health challenge, prompting the development of effective therapeutic vaccines. This study presents a lipid nanoparticle (LNP)-based vaccine co-loading E7 antigen peptide and magnesium ions as the adjuvant. Microfluidic technology was employed to optimize LNP preparation and formulation, ensuring efficient co-delivery of antigen and adjuvant. Magnesium ions were chosen over conventional aluminum-based adjuvants, which often suffer from limited efficacy and adverse effects, particularly for cancer immunotherapy. Compared to aluminum, magnesium ions exhibited superior capabilities in enhancing T-cell activation and promoting cellular immune response. Mechanistic insights suggest that magnesium ions facilitate dendritic cell maturation and antigen presentation via a collagen-CD36 axis, contributing to the adjuvant activity of magnesium. Through design of experiments (DoE) optimization, the LNP formulation was tailored for enhanced encapsulation and stability, positioning it as a targeted system for immune activation. These findings support the promise of magnesium ions as effective and safer adjuvants in LNP-based vaccines, marking a potential advancement for therapeutic cancer vaccination.
Collapse
Affiliation(s)
- Yongyi Xie
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiaxin Guo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jialin Hu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zhongqian Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongcheng Zhu
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW, 2052, Australia
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, PR China
| | - You Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenjie Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| |
Collapse
|
3
|
Hao H, Sun S, Fu Y, Wen S, Wen Y, Yi Y, Peng Z, Fang Y, Tang J, Wang T, Wu M. Magnesium peroxide-based biomimetic nanoigniter degrades extracellular matrix to awake T cell-mediated cancer immunotherapy. Biomaterials 2025; 317:123043. [PMID: 39754969 DOI: 10.1016/j.biomaterials.2024.123043] [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: 08/20/2024] [Revised: 11/25/2024] [Accepted: 12/21/2024] [Indexed: 01/06/2025]
Abstract
As the elite force of our immune system, T cells play a determining role in the effectiveness of cancer immunotherapy. However, the clever tumor cells construct a strong immunosuppressive tumor microenvironment (TME) fortress to resist the attack of T cells. Herein, a magnesium peroxide (MP)-based biomimetic nanoigniter loaded with doxorubicin (DOX) and metformin (MET) is rationally designed (D/M-MP@LM) to awake T cell-mediated cancer immunotherapy via comprehensively destroying the strong TME fortress. The nanoigniter not only effectively initiate CD8+ T cell-mediated immune response by promoting the presentation of tumor antigens, but also greatly facilitate the infiltration of T cells by degrading rigid extracellular matrix (ECM). More importantly, the nanoigniter significantly augment the effector functions of infiltrated CD8+ T cells by Mg2+-mediated metalloimmunotherapy and avoid the exhaustion of CD8+ T cells by improving the acidic TME. Thus, the nanoigniter comprehensively awakes T cells and achieves remarkable tumor inhibition efficacy.
Collapse
Affiliation(s)
- Huisong Hao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yanan Fu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Simin Wen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yingfei Wen
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yunfei Yi
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhangwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yixuan Fang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jia Tang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Tianqi Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
4
|
Zhang Y, Zhao L, Wang X, Zhang C, Zuo H, Gao D. Impact of Geographical Origin on the Contents of Inorganic Elements and Bioactive Compounds in Polygonum perfoliatum L. Molecules 2025; 30:2231. [PMID: 40430408 PMCID: PMC12113674 DOI: 10.3390/molecules30102231] [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: 04/14/2025] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
This study investigated the correlation between thirteen inorganic elements, five key bioactive compounds, and environmental factors in Polygonum perfoliatum L. from fifteen different origins. Analyses were conducted using techniques such as ultrasound-assisted extraction, HPLC, ICP-AES, PCA, and HCA. The results indicate that the geographical origin significantly influences the contents of inorganic elements and bioactive compounds in Polygonum perfoliatum L., and a certain correlation exists among elements, compounds, and environmental factors. This research provides a theoretical foundation for the development and utilization of Polygonum perfoliatum L.
Collapse
Affiliation(s)
- Yanping Zhang
- Chemistry and Chemical Engineering School, Henan University of Science and Technology, Luoyang 471023, China
| | | | | | | | | | | |
Collapse
|
5
|
Li L, Sun Z, Sun W, Zhai Y, Ding N, Wang W. Associations between ionomic profile and metabolic abnormalities in a murine model of sodium sulfide induced alopecia areata. Front Pharmacol 2025; 16:1507348. [PMID: 40438604 PMCID: PMC12117264 DOI: 10.3389/fphar.2025.1507348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 04/28/2025] [Indexed: 06/01/2025] Open
Abstract
Background Alopecia areata (AA) is a common autoimmune disorder marked by non-scarring hair loss, which imposes significant psychosocial stress on patients. To investigate key metabolites and ions involved in AA's pathogenesis, we utilized gas chromatography-mass spectrometry (GC-MS) for non-targeted metabolomics and inductively coupled plasma mass spectrometry (ICP-MS) for ionomics. Methods A total of 36 six-week-old Kunming mice were divided into control (n = 12), an AA model (n = 12), and tofacitinib-treated groups (n = 12). A mouse model of AA was established by sodium sulfide (Na2S) induction in both the model and treatment groups, while the treatment group (n = 12) received tofacitinib treatment at a dose of 1 mg/kg. GC-MS was used to determine the metabolic profiling in serum samples, and ICP-MS was applied to assess ionomic changes in the serum samples. Potential metabolites and ions were identified using orthogonal partial least squares-discriminant analysis (OPLS-DA). Subsequently, MetaboAnalyst 5.0 and the Kyoto Encyclopedia of Genes and Genomes database (KEGG) were used to map the metabolic pathways. Spearman correlation analysis was conducted to identify relationships and potential regulatory interactions between differential metabolites and individual ions. Results Metabolomics analysis revealed that D-lactic acid, glycolic acid, linoleic acid, petroselinic acid, and stearic acid are key differential metabolites between the control, AA model, and tofacitinib groups. Pathway analysis highlighted that the biosynthesis of unsaturated fatty acids and linoleic acid metabolism are pivotal pathways implicated in the onset and progression of AA. Furthermore, ionomics analysis identified magnesium, aluminum, titanium, and nickel as differential ions among the three groups. The integrated metabolomics and ionomics analysis indicated that linoleic acid, a key differential metabolite according to the KEGG database, shows a positive correlation with phosphorus, vanadium, magnesium, and zinc. Among these, Mg2+ (Mg2+) play a crucial role in modulating CD8+ T cell infiltration, thereby influencing the disease progression in AA. Conclusion Tofacitinib inhibits CD8+ T cell infiltration in hair follicles affected by sodium sulfide-induced AA by modulating the linoleic acid metabolism-Mg2+ pathway. Our findings offer new insights and potential avenues for the clinical diagnosis and treatment of AA, suggesting that targeting metabolic and ionic pathways could enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Luning Li
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining, Shandong, China
| | - Zhen Sun
- Department of Clinical Pharmacy, Jining First People’s Hospital, Jining, Shandong, China
| | - Wenxue Sun
- Department of Clinical & Translational Medicine, Jining First People’s Hospital, Jining, Shandong, China
| | - Yujuan Zhai
- Department of Dermatology, Jining First People’s Hospital, Jining, Shandong, China
| | - Na Ding
- Department of Dermatology, Jining First People’s Hospital, Jining, Shandong, China
| | - Wei Wang
- Department of Dermatology, Jining First People’s Hospital, Jining, Shandong, China
| |
Collapse
|
6
|
Wang Z, Zhang F, Zhou B, Sun L, Liu B, Liu M, Wang S, Xu L, Liu H, Dong B. Gradient-driven deep penetration of self-electrophoretic nanoparticles in acidic tumor microenvironments for enhanced antitumor therapy. Biomaterials 2025; 322:123398. [PMID: 40344880 DOI: 10.1016/j.biomaterials.2025.123398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Difficulty of nanomedicines to effectively penetrate the tumor core and achieve effective killing of tumor stem cells is an important factor leading to recurrence, metastasis and drug resistance of tumors. Strategies based on the tumor microenvironment offer new perspectives and approaches to address the challenges associated with deep tumor treatment. Here, we designed novel MgF2@L-Arg nanoparticles (ML NPs) by integrating basic L-arginine into MgF2. Under the endogenous acid gradient within the tumor, ML NPs selectively protonate their proximal amines, leading to spatial charge asymmetry. This promotes the sustained diffusion and permeation of ML NPs deep into the tumor, achieving a penetration distance of up to 197 μm. Moreover, aside from enabling synergistic effects in sonodynamic therapy (SDT) and gas therapy, ML NPs can reduce the expression of hypoxia-inducible factor 1-alpha (HIF-1α) and heat shock protein 70 (HSP 70) within tumor cells, induce immunogenic cell death, and bind to the co-stimulatory molecule LFA-1 on the surface of tumor cells, thereby enhancing the specific cytotoxicity of CD8+ T cells. This mechanism significantly improves the immune response against cancer cells and effectively suppresses tumor metastasis. Our research proposes a viable new strategy for the deep penetration of nanoparticles into tumors and for effective deep tumor treatment, demonstrating the tremendous potential of such materials in enhancing anti-tumor efficacy.
Collapse
Affiliation(s)
- Zhifang Wang
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Fanrou Zhang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Jilin University, Changchun, 130021, China
| | - Bingshuai Zhou
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Liheng Sun
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Bailong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
| | - Min Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China.
| | - Shimeng Wang
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, 130021, China
| | - Lin Xu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Haipeng Liu
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, 130021, China.
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China.
| |
Collapse
|
7
|
Tin E, Rutella S, Khatri I, Na Y, Yan Y, MacLean N, Vadakekolathu J, Minden MD, Schimmer AD, Lee J, Zhang L. SOCS1 Protects Acute Myeloid Leukemia against Allogeneic T Cell-Mediated Cytotoxicity. Blood Cancer Discov 2025; 6:217-232. [PMID: 39928733 PMCID: PMC12050964 DOI: 10.1158/2643-3230.bcd-24-0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/14/2024] [Accepted: 01/28/2025] [Indexed: 02/12/2025] Open
Abstract
SIGNIFICANCE Our investigation of the SOCS1 pathway in AML and T-cell interactions provides insights into potential mechanisms of resistance of AML to allogeneic hematopoietic stem cell transplantation and demonstrates the potential of targeting SOCS1 and its downstream mediators to enhance antileukemic T-cell activity. See related commentary by Fry, p. 157.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Suppressor of Cytokine Signaling 1 Protein/metabolism
- Suppressor of Cytokine Signaling 1 Protein/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Hematopoietic Stem Cell Transplantation
- Transplantation, Homologous
- Cytotoxicity, Immunologic
- Animals
Collapse
Affiliation(s)
- Enoch Tin
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Sergio Rutella
- John van Geest Cancer Research Center, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Ismat Khatri
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Yoosu Na
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Yongran Yan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Jayakumar Vadakekolathu
- John van Geest Cancer Research Center, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Mark D. Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Aaron D. Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - JongBok Lee
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Li Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
8
|
Barik P, Mondal S. Immunomodulatory effects of metal nanoparticles: current trends and future prospects. NANOSCALE 2025; 17:10433-10461. [PMID: 40202489 DOI: 10.1039/d5nr01030f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The advent of nanotechnology has steered into a new era of medical advancements, with metal nanoparticles (MNPs) emerging as potent agents for precise regulation of the immune system. This review provides a comprehensive overview of the immunomodulatory roles of MNPs, including gold, silver, and metal oxide nanoparticles, in regulating innate and adaptive immunity. Additionally, we discuss the immunological effects of metal ions and metal complexes, offering a comparative analysis with nanoparticulate systems. We analyse cutting-edge strategies utilising MNPs to optimise vaccine efficacy, achieve targeted delivery to immune cells, and orchestrate inflammatory responses. Additionally, we discuss the therapeutic potential of MNPs in combating autoimmune diseases, cancers, and infectious agents, which is evaluated within the framework of precision medicine. Furthermore, we critically assess challenges such as biocompatibility, potential toxicity, and regulatory hurdles. Finally, we propose future directions for integrating MNPs with advanced delivery systems and other nanomaterials to propel the frontiers of immunotherapy. This review aims to provide a foundational understanding of MNP-mediated immunomodulation, inspiring further research and development in this burgeoning field.
Collapse
Affiliation(s)
- Puspendu Barik
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
- Department of Physics, College of Arts and Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
| | - Samiran Mondal
- Department of Chemistry, Rammohan College (University of Calcutta), 102/1-Raja Rammohan Sarani, Kolkata 700009, West Bengal, India.
| |
Collapse
|
9
|
Salem Y, Yacov N, Kafri P, Propheta-Meiran O, Karni A, Maharshak N, Furer V, Elkayam O, Mendel I. MOSPD2 regulates the activation state of αLβ2 integrin to control monocyte migration: applicability for treatment of chronic inflammatory diseases. Immunol Res 2025; 73:78. [PMID: 40312574 PMCID: PMC12045827 DOI: 10.1007/s12026-025-09633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
Monocytes are innate immune cells that drive the chronicity of various inflammatory diseases. Monocyte migration to inflamed tissues involves multiple steps of interaction with the vascular endothelium and the extracellular matrix (ECM), a process mediated through conformational transitions in cell surface integrins. We previously described motile sperm domain-containing protein 2 (MOSPD2) as a surface protein expressed on myeloid cells that is essential for the migration of monocytes and a key regulator of inflammation. Investigating MOSPD2's mechanism of action, we assessed whether it plays a role in regulating integrin activation and monocyte adhesion. Data show that silencing of MOSPD2 expression in the THP-1 monocytic cell line significantly increased cell adhesion to various ECM molecules. Employing IW-601, a humanized anti-human MOSDP2 monoclonal antibody, on primary human monocytes increased adhesion to ECM molecules as well as to adhesion molecules. At the molecular level, silencing of MOSPD2 or blocking MOSPD2 using IW-601 led to a transition in integrin αLβ2 (CD11a/CD18, LFA-1) conformation into an active high-affinity binding form and to the induction of adhesion-associated signaling pathways. Co-immunoprecipitation experiments showed that MOSPD2 binds integrin-β2 (CD18), but not integrin-β1 (CD29). Our results reveal a novel mechanism controlling monocyte migration, in which MOSPD2 acts as an adhesion checkpoint that governs the balance between monocyte adhesion and release. By demonstrating the inhibitory effect of IW-601 on the migration of primary monocytes isolated from patients with chronic inflammatory diseases, we provide proof of concept for translating MOSPD2's mechanism into a potential treatment for inflammatory diseases, further supported by in vivo data in models of RA and IBD.
Collapse
Affiliation(s)
- Yaniv Salem
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
- , Current Address: 77, Shtern Yair St., 5560706, Kiryat-Ono, Israel
| | - Niva Yacov
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
| | - Pinhas Kafri
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
- Current Address: Teva Pharmaceuticals, 12 Hatrufa St, 4250483, Netanya, Israel
| | - Oshrat Propheta-Meiran
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA
| | - Arnon Karni
- Neuroimmunology and Multiple Sclerosis Unit of the Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Neurology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nitsan Maharshak
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Victoria Furer
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ori Elkayam
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Itzhak Mendel
- ImmuneWalk Therapeutics, SVP Research, 20, Hamagshimim St, 4934829, Petach-Tikva, Israel.
- ImmuneWalk Therapeutics, 1 Blue Hill Plaza, Pearl River, NY, 10965, USA.
| |
Collapse
|
10
|
Tan J, Ding B, Chen H, Meng Q, Li J, Zhang W, Yang Z, Ma X, Han D, Yang M, Zheng P, Ma P, Lin J. Gallium-Magnesium Layered Double Hydroxide for Elevated Tumor Immunotherapy Through Multi-Network Synergistic Regulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501256. [PMID: 40190140 DOI: 10.1002/adma.202501256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/12/2025] [Indexed: 05/28/2025]
Abstract
Immunotherapeutic efficacy is often limited by poor immunogenicity, immunosuppressive tumor microenvironment (TME), and cytoprotective mechanisms, leading to low immune activation. To this end, here, L-amino acid oxidase (LAAO) loaded gallium-magnesium layered double hydroxide (MG-LAAO) is prepared for significantly enhanced tumor immunotherapy through multi-network synergistic regulation. First, MG-LAAO induces tumor cell pyroptosis by initiating caspase-1/GSDMD and caspase-3/GSDME pathways, further triggering immunogenic cell death (ICD). Then the released Ga3+ induces mitochondrial iron overload, resulting in ferroptosis. In addition, MG-LAAO also hinders autophagy of tumor cells, and reshapes the immunosuppressive tumor microenvironment (TME) by neutralizing H+ and inhibiting lactic acid accumulation, thus destroying the cytoprotective mechanism and avoiding immune escape. Furthermore, this multi-network synergy further activates the cGAS-STING signaling pathway, generating powerful antitumor immunotherapy. This work highlights the critical role of synergies between autophagy block, pyroptosis, ferroptosis, and ICD in tumor immunotherapy, demonstrating the important role of this multi-network synergy in effectively overcoming immunosuppressive TME and enhancing immunogenicity. In particular, the mechanism of gallium-induced pyroptosis is revealed for the first time, providing theoretical support for the design of new materials for tumor immunotherapy in the future.
Collapse
Affiliation(s)
- Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Wenying Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Zhuang Yang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Xinyu Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Di Han
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Mingkai Yang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Key Laboratory of Superlight Materials and Surface Technology of Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
11
|
Gile JJ, Mondello P, Wang Z, Li Y, Bansal R, Gandhi S, Zhang H, Babadi E, Martinez K, McCoy G, Shao Z, Regan K, Hathcock MA, Wang P, Wang J, Al Saleh AS, Ruan G, Ansell SM, Bennani NN, Johnston PB, Paludo J, Villasboas-Bisneto JC, Khurana A, Durani U, Wang Y, Hampel PJ, Rosenthal A, Munoz J, Moreno E, Castro JE, Murthy HS, Kharfan-Dabaja M, Kenderian SS, Kim JJ, Shen R, Mattie M, Lin Y, Witzig TE. Hypomagnesemia in lymphoma patients receiving CAR T therapy correlates with immune dysfunction and decreased survival. Exp Hematol Oncol 2025; 14:63. [PMID: 40307941 PMCID: PMC12044716 DOI: 10.1186/s40164-025-00623-w] [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: 12/27/2024] [Accepted: 02/24/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Hypomagnesemia has been correlated with inferior outcomes in patients with large B cell lymphoma (LBCL) undergoing stem cell transplants. As T-cell and myeloid cell dysfunction have been associated with low magnesium conditions, we investigated whether serum magnesium (Mg) levels could predict clinical outcomes in LBCL patients who received chimeric antigen receptor T-cell therapy. METHODS Patients with LBCL who received axi-cel under the ZUMA-1 trial or as FDA approved therapy at Mayo Clinic were examined. Serum samples were obtained at specified time points and cytokine analysis was performed. Single cell RNA sequencing was performed on peripheral blood mononuclear cells. The Student T-test, Kruskal Wallis, or Fisher's Exact Tests were used to compare differences in demographics across Mg levels. Survival curves were plotted using the Kaplan-Meier methodology and compared using the Wilcoxon test. RESULTS We found that hypomagnesemia before lymphodepletion chemotherapy predicted inferior progression-free and overall survival in the pivotal study ZUMA-1 (NCT02348216). These results were validated in an independent cohort of LBCL patients receiving axicabtagene ciloleucel (axi-cel) at Mayo Clinic. Hypomagnesemia correlated with increased inflammatory serum markers and cytokine levels including ferritin, IL-6, IL1Ra, IL-8, and MIP1a. scRNAseq analysis unveiled altered immune interactions between monocytes and T cells with a concordant immune suppressive transcriptome. CONCLUSIONS Hypomagnesemia at the time of CAR-T infusion is associated with an unfavorable inflammatory profile and decreased response and survival in LBCL patients receiving axi-cel. These findings suggest a potentially actionable prognostic factor for patients with large cell lymphoma undergoing CAR-T.
Collapse
Affiliation(s)
- Jennifer J Gile
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Patrizia Mondello
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Zixing Wang
- KITE, a Gilead Company, Santa Monica, CA, 90404, USA
| | - Ying Li
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Radhika Bansal
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Sangeetha Gandhi
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Henan Zhang
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Elham Babadi
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Kodi Martinez
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Gabrielle McCoy
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Zuoyi Shao
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Kevin Regan
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Matthew A Hathcock
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Panwen Wang
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Junwen Wang
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Abdullah S Al Saleh
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Gordon Ruan
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Stephen M Ansell
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - N Nora Bennani
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Patrick B Johnston
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Jonas Paludo
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | | | - Arushi Khurana
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Urshila Durani
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Yucai Wang
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Paul J Hampel
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Allison Rosenthal
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Javier Munoz
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Eider Moreno
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Januario E Castro
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Hemant S Murthy
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Saad S Kenderian
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA
| | - Jenny J Kim
- KITE, a Gilead Company, Santa Monica, CA, 90404, USA
| | - Rhine Shen
- KITE, a Gilead Company, Santa Monica, CA, 90404, USA
| | - Mike Mattie
- KITE, a Gilead Company, Santa Monica, CA, 90404, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA.
- Division of Experimental Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Thomas E Witzig
- Division of Hematology, Mayo Clinic, 200 SW First Street, Rochester, MN, 55905, USA.
| |
Collapse
|
12
|
Liao P, Zhou Y, Qiu Y, Hu R, Li H, Sun H, Li Y. Metal-modulated T cell antitumor immunity and emerging metalloimmunotherapy. Cancer Metastasis Rev 2025; 44:49. [PMID: 40301229 DOI: 10.1007/s10555-025-10266-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/19/2025] [Indexed: 05/01/2025]
Abstract
In recent years, increasing evidence has shown that metals play important roles in both innate and adaptive immunity. An emerging concept of metalloimmunotherapy has been proposed, which may accelerate the development of immunotherapy for cancers. Here, we discuss how metals affect T cell function through different signaling pathways. Metals impact the fate of T cells, including their activation, proliferation, cytotoxicity, and differentiation. Most importantly, metals also participate in mitochondrial operation by regulating energy production and reactive oxygen species homeostasis in T cells. We also identified the metal-based mutual effects between tumor cells and T cells in the tumor microenvironment. Overall, the antitumor effect of T cells can be improved by targeting metal metabolism and metalloimmunotherapy, which will be a step forward in the treatment of cancers.
Collapse
Affiliation(s)
- Peiyun Liao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Zhou
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Rong Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyan Li
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Hongzhe Sun
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.
- Guangdong Engineering Research Center of Precision Immune Cell Therapy Technology, Zhujiang Hospital, No. 253, Gongye Road, Guangzhou, China.
| |
Collapse
|
13
|
Vargas MH, Chávez J, Del-Razo-Rodríguez R, Muñoz-Perea C, Romo-Domínguez KJ, Báez-Saldaña R, Rumbo-Nava U, Guerrero-Zúñiga S. Lower Serum Magnesium Is Associated with Mortality in Severe COVID-19: A Secondary Analysis of a Randomized Trial. Biol Trace Elem Res 2025:10.1007/s12011-025-04619-9. [PMID: 40234280 DOI: 10.1007/s12011-025-04619-9] [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: 01/03/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Many abnormalities in laboratory tests have been described in severe coronavirus disease (COVID-19), but most of them probably just reflect the degree of organ dysfunction and are not true risk factors for death. The present study is a secondary analysis of a clinical trial carried out in patients hospitalized due to severe COVID-19 (ClinicalTrials.gov identifier No. NCT04443673). We explored the association of clinical laboratory tests and serum cytokines with death in COVID-19 patients, either considering only the initial measurement obtained shortly after the patient's arrival at the emergency room, or by means of the weighted average of all measurements during the entire hospitalization. The study included 56 patients with a mean age of 58.6 years (range from 31.8 to 86.2 years), with a fatality rate of 58.9% (33 patients). Among initial laboratory tests, only mean corpuscular volume (MCV), erythrocyte count, serum magnesium, and age showed a trend (p < 0.10, univariable logistic regression) for an association with a fatal outcome. However, in the multivariable logistic regression, only MCV and magnesium remained associated with death, with adjusted odds ratios (95% confidence intervals) of 1.253 (1.047-1.501, p = 0.014) and 0.091 (0.010-0.798, p = 0.03), respectively. Serum magnesium tended to decrease during the hospital stay in both groups, survivors and non-survivors. Compared with survivors, patients who died had a higher weighted average of urea, blood urea nitrogen (BUN), procalcitonin, MCV, neutrophils, neutrophil/lymphocyte ratio, fibrinogen/albumin ratio, C-reactive protein/albumin ratio, BUN/albumin ratio, IL-6, and IL-10, as well as decreased weighted average of albumin, lymphocytes, and monocytes, among others. In conclusion, patients with severe COVID-19 who had lower serum magnesium on their arrival at the emergency room were more prone to die. On the other hand, serum magnesium tended to decrease during the patients' hospital stay, independently of the outcome.Trial Registration: ClinicalTrials.gov NCT04443673.
Collapse
Affiliation(s)
- Mario H Vargas
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico.
| | - Jaime Chávez
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, CP 14080, Mexico City, Mexico
| | - Rosangela Del-Razo-Rodríguez
- Servicio Clínico de Neumología Pediátrica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Carolina Muñoz-Perea
- Servicio Clínico de Neumología Pediátrica, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Karina Julieta Romo-Domínguez
- Servicio de Urgencias, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Servicio de Neumología, Hospital Infantil del Estado de Sonora, Hermosillo, Sonora, Mexico
| | - Renata Báez-Saldaña
- Servicio Clínico 3, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Uriel Rumbo-Nava
- Servicio Clínico 3, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Selene Guerrero-Zúñiga
- Unidad de Medicina del Sueño, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| |
Collapse
|
14
|
Zhang Z, Rana I, Nam J. Metal coordination polymer nanoparticles for cancer therapy. Essays Biochem 2025; 69:EBC20253012. [PMID: 40209056 DOI: 10.1042/ebc20253012] [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: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025]
Abstract
Metal ions are essential elements in biological processes and immune homeostasis. They can regulate cancer cell death through multiple distinct molecular pathways and stimulate immune cells implicated in antitumor immune responses, suggesting opportunities to design novel metal ion-based cancer therapies. However, their small size and high charge density result in poor target cell uptake, uncontrolled biodistribution, and rapid clearance from the body, reducing therapeutic efficacy and increasing potential off-target toxicity. Metal coordination polymer nanoparticles (MCP NPs) are nanoscale polymer networks composed of metal ions and organic ligands linked via noncovalent coordination interactions. MCP NPs offer a promising nanoplatform for reshaping metal ions into more drug-like formulations, improving their in vivo pharmacological performance and therapeutic index for cancer therapy applications. This review provides a comprehensive overview of the inherent biological functions of metal ions in cancer therapy, showcasing examples of MCP NP systems designed for preclinical cancer therapy applications where drug delivery principles play a critical role in enhancing therapeutic outcomes. MCP NPs offer versatile metal ion engineering approaches using selected metal ions, various organic ligands, and functional payloads, enabling on-demand nano-drug designs that can significantly improve therapeutic efficacy and reduce side effects for effective cancer therapy.
Collapse
Affiliation(s)
- Zhengzheng Zhang
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| | - Isra Rana
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwanju 61186, South Korea
| |
Collapse
|
15
|
Wang S, Qin L, Liu F, Zhang Z. Unveiling the crossroads of STING signaling pathway and metabolic reprogramming: the multifaceted role of the STING in the TME and new prospects in cancer therapies. Cell Commun Signal 2025; 23:171. [PMID: 40197235 PMCID: PMC11977922 DOI: 10.1186/s12964-025-02169-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025] Open
Abstract
The cGAS-STING signaling pathway serves as a critical link between DNA sensing and innate immunity, and has tremendous potential to improve anti-tumor immunity by generating type I interferons. However, STING agonists have shown decreasing biotherapeutic efficacy in clinical trials. Tumor metabolism, characterized by aberrant nutrient utilization and energy production, is a fundamental hallmark of tumorigenesis. And modulating metabolic pathways in tumor cells has been discovered as a therapeutic strategy for tumors. As research concerning STING progressed, emerging evidence highlights its role in metabolic reprogramming, independent its immune function, indicating metabolic targets as a strategy for STING activation in cancers. In this review, we delve into the interplay between STING and multiple metabolic pathways. We also synthesize current knowledge on the antitumor functions of STING, and the metabolic targets within the tumor microenvironment (TME) that could be exploited for STING activation. This review highlights the necessity for future research to dissect the complex metabolic interactions with STING in various cancer types, emphasizing the potential for personalized therapeutic strategies based on metabolic profiling.
Collapse
Affiliation(s)
- Siwei Wang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lu Qin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology), Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhanguo Zhang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
16
|
Wang X, Liao Y, Liu D, Zheng J, Shi M. Presetting CAR-T cells during ex vivo biomanufacturing. Mol Ther 2025; 33:1380-1406. [PMID: 39988874 PMCID: PMC11997485 DOI: 10.1016/j.ymthe.2025.02.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the treatment of hematologic malignancies. However, it continues to encounter significant obstacles, including treatment relapse and limited efficacy in solid tumors. While effector T cells exhibit robust cytotoxicity, central memory T cells and stem cell-like T cells are essential for in vivo expansion, long-term survival, and persistence. Strategies such as genetic engineering to enhance CAR-T cell efficacy and durability are often accompanied by increased safety risks, which not only raise regulatory approval thresholds but also escalate CAR-T production costs. In contrast, optimizing ex vivo manufacturing conditions represents a more straightforward and practical approach, offering the potential for rapid application to commercially approved CAR-T products and enhancement of their clinical outcomes. This review examines several factors that have been shown to improve T cell memory phenotype and in vivo cytotoxic activity, including cytokines, electrolytes, signaling pathway inhibitors, metabolic modulators, and epigenetic agents. The insights provided will guide the optimization of CAR-T cell industrial production. Furthermore, considerations for selecting appropriate conditions are discussed, balancing effectiveness, cost-efficiency, safety, and regulatory compliance while addressing current challenges in the field.
Collapse
Affiliation(s)
- Xu Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ying Liao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Dan Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
17
|
Li YH, Huang XJ, Zhao XY. Translational study of the regulatory mechanism by which immune synapses enhance immune cell function. Cancer Lett 2025; 614:217542. [PMID: 39924076 DOI: 10.1016/j.canlet.2025.217542] [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: 12/20/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
Immune synapses, which were initially discovered at the interface between antigen-presenting cells (APCs) and T cells, are special structures formed at the contact site between antigen-presenting cells and immune cells and constitute the structural basis for immune cells to kill tumours and synthesise antibodies. Their structures are very similar to those of neural synapses in the nervous system, and they contain different functional structural regions. With the development of cell visualization research, scientists have increasingly conducted in-depth research on immune synapses. At present, it is known that T cells, B cells, and NK cells can form different immune synapses with target cells. Immune synapses formed by different cell subsets as well as CAR-T cells have their own characteristics, mainly in terms of their structure, formation process and regulatory mechanism. Therefore, how to enhance immune cell killing function by enhancing immune synaptic function has long been a research hotspot. At present, the killing function of immune cells can be enhanced by influencing the signalling molecules of immune synapses and the cell microenvironment and modifying the structure of immune synapses. Through a review of the factors affecting immune synapses, we can better explore the target for enhancing immune system function.
Collapse
Affiliation(s)
- Ya-Hui Li
- Peking University People's Hospital, Peking University Institute of Haematology, National Clinical Research Center for Haematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Haematology, National Clinical Research Center for Haematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Haematology, National Clinical Research Center for Haematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing, China.
| |
Collapse
|
18
|
Yang P, Yao X, Tian X, Wang Y, Gong L, Yang Y, Jie J. Supramolecular peptide hydrogel epitope vaccine functionalized with CAR-T cells for the treatment of solid tumors. Mater Today Bio 2025; 31:101517. [PMID: 39925713 PMCID: PMC11804731 DOI: 10.1016/j.mtbio.2025.101517] [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: 08/02/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy, which benefits from the perfect combination of gene editing techniques and antibody engineering, has shown outstanding clinical efficacy in hematological malignancies. Solid tumors present the next challenge due to their extremely complicated microenvironment and structural characteristics. Targeting efficiency and persistence are currently bottleneck issues in the clinical treatment of CAR-T. Beyond drugs and cytokines, biomaterials can modulate the immune response, assisting adoptive CAR-T cells in exerting their function. In this study, a supramolecular peptide hydrogel epitope vaccine was designed to serve as both a preparation medium and a reservoir for CAR-T cells. The self-assembling peptide formed a nanofiber scaffold through non-covalent interactions of amphiphilic amino acids and ion stabilizers. Firstly, the complementary peptide conjugated vaccine epitopes and CAR-T target sites were derived from different extracellular domains of the HER2 protein, and the combination treatment improved tumor antigen spreading and targeting efficiency. The epitope hydrogel promoted CAR-T cell proliferation, cytotoxic activity, and lymphocyte subpopulation transformation. Furthermore, the supramolecular peptide epitope vaccine encapsulated CAR-T (SPEV-CAR-T) induced endogenous humoral and cellular immune responses through a sustained release of the hydrogel and CAR-T cells, demonstrating superior anti-tumor effects in an in vivo mouse model. Most importantly, SPEV-CAR-T induced central memory cells in systemic immune tissues, addressing the poor persistence of single CAR-T therapy. The integration and complementation of active and passive immune responses in this all-in-one hydrogel epitope vaccine and CAR-T system facilitated a sequential succession of endogenous and exogenous immune responses, promoting persistent and specific tumor attack. SPEV-CAR-T showed superior therapeutic effects in solid tumors.
Collapse
Affiliation(s)
- Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, PR China
- Engineering Research Center of Integration and Application of Digital Learning Technology, Ministry of Education, 100034, Beijing, PR China
| | - Xiaomin Yao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, PR China
| | - Xue Tian
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, 226001, Nantong, PR China
| | - Yuehan Wang
- Medical School of Nantong University, Nantong University, 226001, Nantong, PR China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, PR China
- Engineering Research Center of Integration and Application of Digital Learning Technology, Ministry of Education, 100034, Beijing, PR China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, PR China
- Engineering Research Center of Integration and Application of Digital Learning Technology, Ministry of Education, 100034, Beijing, PR China
| | - Jing Jie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, 226001, Nantong, PR China
| |
Collapse
|
19
|
Enbar T, Tang L. Mimicking the TCR immune synapse for improved CAR-T cell function. Cell Res 2025:10.1038/s41422-025-01086-8. [PMID: 40000772 DOI: 10.1038/s41422-025-01086-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Affiliation(s)
- Tom Enbar
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland.
| |
Collapse
|
20
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
21
|
Gong H, Lin X, Huang S. Association between magnesium depletion score and prostate cancer. Sci Rep 2025; 15:4801. [PMID: 39922926 PMCID: PMC11807168 DOI: 10.1038/s41598-025-89506-y] [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: 11/11/2024] [Accepted: 02/05/2025] [Indexed: 02/10/2025] Open
Abstract
The Magnesium Depletion Score (MDS) is a practical tool used to assess magnesium deficiency. Studies have indicated that MDS is associated with various urological conditions, such as kidney stones and the prognosis of chronic kidney disease. However, the relationship between MDS and prostate cancer (PCa) remains unclear. Therefore, this study aims to investigate the association between MDS and PCa. This study conducted a cross-sectional analysis of 16,043 participants from the 2005-2018 NHANES database. Subgroup analysis, restricted cubic splines (RCS), and multivariable logistic regression were employed to examine the association between MDS and the prevalence of PCa. A total of 16,043 participants were included in this study, of whom 511 had PCa. After adjusting for all variables using multivariable logistic regression, each 1-unit increase in MDS was associated with a 26% higher prevalence of PCa (OR = 1.26, 95% CI: 1.05, 1.50). Additionally, compared to an MDS of 0, an MDS of 3 or higher was associated with a 3.04-fold increase in PCa prevalence (OR = 3.04, 95% CI: 1.53, 6.01). RCS analysis demonstrated a significant linear positive correlation between MDS and PCa prevalence. Subgroup analysis indicated that the positive association between MDS and PCa was generally consistent across different population groups. This study indicates a significant association between MDS and the risk of PCa, with higher MDS linked to an increased prevalence of PCa. These findings highlight the potential role of MDS in PCa. Further research is needed to determine whether a causal relationship exists between MDS and PCa, which would help assess the appropriateness of potential interventions.
Collapse
Affiliation(s)
- Hongyang Gong
- Department of Oncology Surgery, Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, No.102 Gudong Road, Gulou District, Fuzhou City, 350001, Fujian Province, China
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Xiaomei Lin
- Department of Orthopedics, Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou City, Fujian Province, China
| | - Shaoqun Huang
- Department of Oncology Surgery, Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, No.102 Gudong Road, Gulou District, Fuzhou City, 350001, Fujian Province, China.
| |
Collapse
|
22
|
Wang F, Mao Y, Sun J, Yang J, Xiao L, Huang Q, Wei C, Gou Z, Zhang K. Models based on dietary nutrients predicting all-cause and cardiovascular mortality in people with diabetes. Sci Rep 2025; 15:4600. [PMID: 39920222 PMCID: PMC11805981 DOI: 10.1038/s41598-025-88480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 01/28/2025] [Indexed: 02/09/2025] Open
Abstract
Dietary intervention plays a vital role in improving the prognosis of people with diabetes mellitus (DM). Currently, there is a lack of systematic analysis of the relation between dietary nutrients and long-term mortality risk in people with DM. The study aims to establish models predicting long-term mortality and explore dietary nutrients associated with reduced long-term events to guide daily dietary decisions in people with DM. The retrospective cohort study collected 5060 participants with DM from the National Health and Nutrition Examination Survey (NHANES) 1999-2018. The least absolute shrinkage and selection operator (LASSO) regression and random forest (RF) algorithm were applied to identify key mortality-related dietary factors, which were subsequently incorporated into risk prediction nomogram models. The receiver operating characteristic (ROC) curve, calibration plot and decision curve analysis (DCA) were utilized to evaluate the performance of the models. The association of key dietary nutrients with all-cause and cardiovascular mortality were visualized by restricted cubic spline (RCS) models both in the whole and subgroups by sex, age, drinking and smoking status. The overall median age of the cohort was 62.0 years (interquartile range (IQR) 52.0-70.0), 2564 (50.67%) being male. During a median follow-up period of 56.0 months, 997 (19.70%) all-cause deaths were recorded, with 219 (21.97%) of which being ascribed to cardiovascular disease. The nomogram models based on key dietary nutrients identified by LASSO and RF demonstrated a significant predicative value for all-cause and cardiovascular mortality. Dietary fiber and magnesium were the common predictive nutrients in the two nomogram models. The RCS curve revealed that dietary fiber and magnesium were negatively associated with long-term mortality in the whole and subgroups of people with DM after adjustment of potential confounders. The diet of people with DM is closely associated with mortality. The nomogram models based on dietary nutrients can predict long-term mortality of people with DM, and the higher intake of dietary fiber and magnesium was associated with reduced risks of both long-term all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Fang Wang
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China
| | - Yukang Mao
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China
| | - Jinyu Sun
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China
| | - Jiaming Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Li Xiao
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China
| | - Qingxia Huang
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China
| | - Chenchen Wei
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China
| | - Zhongshan Gou
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China.
| | - Kerui Zhang
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242# Guangji Road, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
23
|
Feng Y, Zhang H, Wang X. Response to Letter Entitled "Elevated serum magnesium levels prompt favourable outcomes in cancer patients treated with immune checkpoint blockers". Eur J Cancer 2025; 216:115250. [PMID: 39843338 DOI: 10.1016/j.ejca.2025.115250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Affiliation(s)
- Yingfang Feng
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Huilai Zhang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| | - Xianhuo Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| |
Collapse
|
24
|
Feng Y, Zhang H, Wang X. Response to letter re: "Elevated serum magnesium levels prompt favourable outcomes in cancer patients treated with immune checkpoint blockers". Eur J Cancer 2025; 216:115249. [PMID: 39843337 DOI: 10.1016/j.ejca.2025.115249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Affiliation(s)
- Yingfang Feng
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Huilai Zhang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| | - Xianhuo Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| |
Collapse
|
25
|
Tintelnot J, Paschold L, Goekkurt E, Schultheiss C, Matschl U, Santos Cruz M, Bauer M, Wickenhauser C, Thuss-Patience P, Lorenzen S, Ettrich TJ, Riera-Knorrenschild J, Jacobasch L, Kretzschmar A, Kubicka S, Al-Batran SE, Reinacher-Schick A, Pink D, Bokemeyer C, Sinn M, Lindig U, Hinke A, Hegewisch-Becker S, Stein A, Binder M. Inflammatory Stress Determines the Need for Chemotherapy in Patients with HER2-Positive Esophagogastric Adenocarcinoma Receiving Targeted Therapy and Immunotherapy. Cancer Immunol Res 2025; 13:200-209. [PMID: 39527097 PMCID: PMC11788649 DOI: 10.1158/2326-6066.cir-24-0561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/10/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Anti-PD-1, trastuzumab, and chemotherapy are used in the treatment of patients with advanced HER2-positive esophagogastric adenocarcinoma, but long-term survival remains limited. In this study, we report extended follow-up data from the INTEGA trial (NCT03409848), which investigated the efficacy of the anti-PD-1 nivolumab, trastuzumab, and FOLFOX chemotherapy (FOLFOX arm) in comparison with a chemotherapy-free regimen involving nivolumab, trastuzumab, and the anti-CTLA-4 ipilimumab (Ipi arm) in the first-line setting for advanced disease. The 12-month overall survival (OS) showed no statistical difference between the arms, with 57% OS (95% confidence interval, 41%-71%) in the Ipi arm and 70% OS (95% confidence interval, 54%-82%) in the FOLFOX arm. Crossing of the survival curves indicated a potential long-term benefit for some patients within the Ipi arm, but early progressors in the Ipi arm underlined the need for biomarker-guided strategies to optimize treatment selection. To this end, metabolomic and cytokine analyses demonstrated elevated levels of normetanephrine, cortisol, and IL6 in immunotherapy-unresponsive patients in the Ipi arm, suggesting a role for systemic inflammatory stress in modulating antitumor immune responses. Patients with this signature also showed an increased neutrophil to lymphocyte ratio that persisted in the Ipi arm, but not in the FOLFOX arm, and strongly correlated with survival. Furthermore, a low neutrophil to lymphocyte ratio characterized patients benefiting from immunotherapy and targeted therapy without the need for additional chemotherapy. These data suggest that patient selection based on inflammatory stress-driven immune changes could help customize first-line treatment in patients with advanced HER2-positive esophagogastric adenocarcinoma to potentially improve long-term survival.
Collapse
Affiliation(s)
- Joseph Tintelnot
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Paschold
- Department of Internal Medicine IV - Oncology/Hematology, University Hospital, Martin-Luther University, Halle, Germany
| | - Eray Goekkurt
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hematology-Oncology Practice Eppendorf (HOPE), Hamburg, Germany
| | - Christoph Schultheiss
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Urte Matschl
- Department of Virus Immunology, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Mariana Santos Cruz
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Bauer
- Institute of Pathology, University Hospital, Martin-Luther University, Halle, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, University Hospital, Martin-Luther University, Halle, Germany
| | | | - Sylvie Lorenzen
- Rechts der Isar Hospital, Technical University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | - Daniel Pink
- Klinik und Poliklinik für Innere Medizin C, University Greifswald, Greifswald, Germany
- Klinik für Hämatologie, Onkologie und Palliativmedizin, Sarkomzentrum Berlin-Brandenburg, HELIOS Klinikum Bad-Saarow, Bad Saarow, Germany
| | - Carsten Bokemeyer
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Sinn
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | - Alexander Stein
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
26
|
Wang Y, Yang Q, Dong Y, Wang L, Zhang Z, Niu R, Wang Y, Bi Y, Liu G. Piezo1-directed neutrophil extracellular traps regulate macrophage differentiation during influenza virus infection. Cell Death Dis 2025; 16:60. [PMID: 39890818 PMCID: PMC11785962 DOI: 10.1038/s41419-025-07395-5] [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: 08/16/2024] [Revised: 12/20/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Neutrophils and macrophages are critical for antiviral immunity, but their reciprocal regulatory roles and mechanisms in the response to viral infection remain unclear. Herein, we found that the ion channel Piezo1 directs neutrophil extracellular trap (NET) formation and regulates macrophage functional differentiation in anti-influenza virus immunity. Genetic deletion of Piezo1 in neutrophils inhibited the generation of NETs and M1 macrophage differentiation while driving the development of M2 macrophages during viral infection. Piezo1-directed neutrophil NET DNA directly regulates macrophage differentiation in vitro and in vivo. Mechanistically, neutrophil Piezo1 deficiency inhibited NET DNA production, leading to decreased TLR9 and cGAS-STING signalling activity while inducing reciprocal differentiation from M1 to M2 macrophages. In addition, Piezo1 integrates magnesium signalling and the SIRT2-hypoxia-inducible factor-1 alpha (HIF1α)-dependent pathway to orchestrate reciprocal M1 and M2 macrophage lineage commitment through neutrophil-derived NET DNA. Our studies provide critical insight into the role of neutrophil-based mechanical regulation of immunopathology in directing macrophage lineage commitment during the response to influenza virus infection.
Collapse
Affiliation(s)
- Yuexin Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, 100080, Beijing, China
| | - Zhiyuan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Ruiying Niu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, 100080, Beijing, China.
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China.
| |
Collapse
|
27
|
Nascimento Da Conceicao V, Sun Y, Venkatesan M, De La Chapa J, Ramachandran K, Jasrotia RS, Drel V, Chai X, Mishra BB, Madesh M, Singh BB. Naltriben promotes tumor growth by activating the TRPM7-mediated development of the anti-inflammatory M2 phenotype. NPJ Precis Oncol 2025; 9:29. [PMID: 39875485 PMCID: PMC11775176 DOI: 10.1038/s41698-025-00815-x] [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: 04/10/2024] [Accepted: 01/19/2025] [Indexed: 01/30/2025] Open
Abstract
Macrophage plasticity is critical for maintaining immune function and developing solid tumors; however, the macrophage polarization mechanism remains incompletely understood. Our findings reveal that Mg2+ entry through distinct plasma membrane channels is critical to macrophage plasticity. Naïve macrophages displayed a previously unidentified Mg2+ dependent current, and TRPM7-like activity, which modulates its survival. Significantly, in M1 macrophages, Mg2+ entry is facilitated by a novel Mg²-dependent current that relies on extracellular Mg2+, which was crucial for activating iNOS/NFκB pathways and cellular bioenergetics, which drives pro-inflammatory cytokines. Conversely, in M2 macrophages, Mg2+ entry occurs primarily through TRPM7 channels, pivotal for IL-4 and IL-10-mediated anti-inflammatory cytokine secretion. Notably, the Mg2+ deficient diet or addition of TRPM7 agonist Naltriben suppresses the M1 phenotype while promoting angiogenic factors and fostering tumor growth. These findings suggest that Mg2+ flux via specific channels is indispensable for macrophage polarization, with its dysregulation playing a pivotal role in tumor progression.
Collapse
Affiliation(s)
| | - Yuyang Sun
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Manigandan Venkatesan
- Center for Mitochondrial Medicine, Department of Medicine/Cardiology Division, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Jorge De La Chapa
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Karthik Ramachandran
- Center for Mitochondrial Medicine, Department of Medicine/Cardiology Division, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Rahul S Jasrotia
- Department of Developmental Dentistry, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Victor Drel
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Xiufang Chai
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Bibhuti B Mishra
- Department of Developmental Dentistry, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Muniswamy Madesh
- Center for Mitochondrial Medicine, Department of Medicine/Cardiology Division, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| | - Brij B Singh
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
28
|
Upadhyay S, Murugu L, Svensson L. Tumor cells escape immunosurveillance by hampering LFA-1. Front Immunol 2025; 16:1519841. [PMID: 39911389 PMCID: PMC11794523 DOI: 10.3389/fimmu.2025.1519841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
During tumor immunosurveillance, leukocytes play a crucial role in the cellular defense system, working collaboratively with other immune components to recognize and eliminate aberrant cells. Integral to this process is the integrin Lymphocyte Function-Associated Antigen 1 (LFA-1). LFA-1 facilitates adhesion during leukocyte migration and helps establish stable cell-to-cell contacts between leukocytes and their targets. Additionally, as a receptor, LFA-1 signaling activates leukocytes, promoting their differentiation and effector functions against cancer. However, tumors can develop mechanisms to evade immune clearance by disrupting LFA-1 functions or hijacking its pathways. In this review, we first detail how leukocytes utilize LFA-1 during immunosurveillance and then explore how tumors counteract this process in the tumor microenvironment (TME) by either altering LFA-1 functions or exploiting it to drive tumorigenesis. Moreover, we discuss therapeutic strategies targeting LFA-1, including inhibitors tested in laboratory studies and animal models, highlighting their potential as anticancer interventions and the need for further research to evaluate their clinical utility.
Collapse
Affiliation(s)
| | - Lewis Murugu
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Lena Svensson
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|
29
|
Rodriguez R, Müller S, Colombeau L, Solier S, Sindikubwabo F, Cañeque T. Metal Ion Signaling in Biomedicine. Chem Rev 2025; 125:660-744. [PMID: 39746035 PMCID: PMC11758815 DOI: 10.1021/acs.chemrev.4c00577] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/10/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Complex multicellular organisms are composed of distinct tissues involving specialized cells that can perform specific functions, making such life forms possible. Species are defined by their genomes, and differences between individuals within a given species directly result from variations in their genetic codes. While genetic alterations can give rise to disease-causing acquisitions of distinct cell identities, it is now well-established that biochemical imbalances within a cell can also lead to cellular dysfunction and diseases. Specifically, nongenetic chemical events orchestrate cell metabolism and transcriptional programs that govern functional cell identity. Thus, imbalances in cell signaling, which broadly defines the conversion of extracellular signals into intracellular biochemical changes, can also contribute to the acquisition of diseased cell states. Metal ions exhibit unique chemical properties that can be exploited by the cell. For instance, metal ions maintain the ionic balance within the cell, coordinate amino acid residues or nucleobases altering folding and function of biomolecules, or directly catalyze specific chemical reactions. Thus, metals are essential cell signaling effectors in normal physiology and disease. Deciphering metal ion signaling is a challenging endeavor that can illuminate pathways to be targeted for therapeutic intervention. Here, we review key cellular processes where metal ions play essential roles and describe how targeting metal ion signaling pathways has been instrumental to dissecting the biochemistry of the cell and how this has led to the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Raphaël Rodriguez
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Sebastian Müller
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Ludovic Colombeau
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Stéphanie Solier
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
- Université
Paris-Saclay, UVSQ, 78180 Montigny-le-Bretonneux, France
| | | | - Tatiana Cañeque
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| |
Collapse
|
30
|
Zhao L, Gui Y, Cai J, Deng X. Biometallic ions and derivatives: a new direction for cancer immunotherapy. Mol Cancer 2025; 24:17. [PMID: 39815289 PMCID: PMC11734411 DOI: 10.1186/s12943-025-02225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/01/2025] [Indexed: 01/18/2025] Open
Abstract
Biometallic ions play a crucial role in regulating the immune system. In recent years, cancer immunotherapy has become a breakthrough in cancer treatment, achieving good efficacy in a wide range of cancers with its specificity and durability advantages. However, existing therapies still face challenges, such as immune tolerance and immune escape. Biometallic ions (e.g. zinc, copper, magnesium, manganese, etc.) can assist in enhancing the efficacy of immunotherapy through the activation of immune cells, enhancement of tumor antigen presentation, and improvement of the tumor microenvironment. In addition, biometallic ions and derivatives can directly inhibit tumor cell progression and offer the possibility of effectively overcoming the limitations of current cancer immunotherapy by promoting immune responses and reducing immunosuppressive signals. This review explores the role and potential application prospects of biometallic ions in cancer immunotherapy, providing new ideas for future clinical application of metal ions as part of cancer immunotherapy and helping to guide the development of more effective and safe therapeutic regimens.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Jing Cai
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Xiangying Deng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China.
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China.
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
31
|
Li W, Tian H, Yan Z, Yu X, Li B, Dai Y. Magnesium-Phenolic Nanoeditor Refining Gliomatous T Cells for Metalloimmunotherapy. ACS NANO 2025; 19:1222-1237. [PMID: 39701951 PMCID: PMC11752515 DOI: 10.1021/acsnano.4c13388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
More than the sparse infiltration in glioblastoma, cytotoxic T lymphocytes (CTLs) also function inefficiently and overexpress the inhibitory markers, especially the identified NK cell receptor (NK1.1). However, most studies solely focus on how to augment tumor-infiltrating CTLs and overlook their killing maintenance. Metalloimmunotherapy has been proven to improve the functionalities of CTLs, but it has barely adapted to glioblastoma due to the severe limitations of safe delivery and the brain's physiological properties. Herein, we synthesized an amphipathic polyethylene glycol (PEG) polymer (designated as MPP) modified with the choline analogue 2-methacryloyloxyethyl phosphorylcholine (MPC) and polyphenol moieties to customize a nanoeditor (Mg2+@MK-8931@MPP) by coordinating Mg2+ and entrapping the hydrophobic BACE1 inhibitor MK-8931, then precisely redressing the gliomatous CTL sparsity and cytotoxic dysfunction. Upon MPC-assisted local accumulation in glioblastoma, Mg2+@MK-8931@MPP nanoeditors release MK-8931 to repolarize M2-like macrophages, facilitating CTL infiltration quantitatively. The cenogenetic immune adjuvant Mg2+ ulteriorly fortifies the T-cell receptor downstream signals to enhance the functionality of the ingoing CTLs in quality, leading to the secretion of high-level antitumor cytokines and cytotoxic proteins. Further blocking the inhibitory NK1.1 on CTLs by anti-NK1.1 antibodies can extend their cytolytic endgame. Studies on T-cell-deficient and wild-type mouse models support the immunomodulating feasibility of Mg2+@MK-8931@MPP. This gliomatous CTL-tailored strategy concurrently broadens metalloimmunotherapy to glioblastoma treatment and highlights the necessity of enforcing gliomatous CTLs' functionality.
Collapse
Affiliation(s)
- Wenxi Li
- Cancer
Centre and Institute of Translational Medicine, Faculty of Health
Sciences, University of Macau, Macau SAR 999078, China
- MoE
Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| | - Hao Tian
- Cancer
Centre and Institute of Translational Medicine, Faculty of Health
Sciences, University of Macau, Macau SAR 999078, China
- MoE
Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| | - Ziliang Yan
- Cancer
Centre and Institute of Translational Medicine, Faculty of Health
Sciences, University of Macau, Macau SAR 999078, China
- MoE
Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| | - Xinying Yu
- Cancer
Centre and Institute of Translational Medicine, Faculty of Health
Sciences, University of Macau, Macau SAR 999078, China
- MoE
Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| | - Bei Li
- Cancer
Centre and Institute of Translational Medicine, Faculty of Health
Sciences, University of Macau, Macau SAR 999078, China
- MoE
Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| | - Yunlu Dai
- Cancer
Centre and Institute of Translational Medicine, Faculty of Health
Sciences, University of Macau, Macau SAR 999078, China
- MoE
Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR 999078, China
| |
Collapse
|
32
|
Zou Q, Liao K, Li G, Huang X, Zheng Y, Yang G, Luo M, Xue EY, Lan C, Wang S, Shen Y, Luo D, Ng DKP, Liu Q. Photo-metallo-immunotherapy: Fabricating Chromium-Based Nanocomposites to Enhance CAR-T Cell Infiltration and Cytotoxicity against Solid Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407425. [PMID: 38899741 PMCID: PMC11733712 DOI: 10.1002/adma.202407425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The infiltration and cytotoxicity of chimeric antigen receptor (CAR)-T cells are crucial for effective elimination of solid tumors. While metallo-immunotherapy is a promising strategy that can activate the antitumor immunity, its role in promoting CAR-T cell therapy remains elusive. The first single-element nanomaterial based on chromium nanoparticles (Cr NPs) for cancer photo-metallo-immunotherapy has been reported previously. Herein, an extended study using biodegradable polydopamine as a versatile carrier for these nanoparticles, enabling synergistic CAR-T cell therapy, is reported. The results show that these nanocomposites with or without further encapsulation of the anticancer drug alpelisib can promote the CAR-T cell migration and antitumor effect. Upon irradiation with near-infrared light, they caused mild hyperthermia that can "warm" the "cold" tumor microenvironment (TME). The administration of B7-H3 CAR-T cells to NOD severe combined immunodeficiency gamma mice bearing a human hepatoma or PIK3CA-mutated breast tumor can significantly inhibit the tumor growth after the induction of tumor hyperthermia by the nanocomposites and promote the secretion of serum cytokines, including IL-2, IFN-γ, and TNF-α. The trivalent Cr3+ ions, which are the major degradation product of these nanocomposites, can increase the CXCL13 and CCL3 chemokine expressions to generate tertiary lymphoid structures (TLSs) in the tumor tissues, facilitating the CAR-T cell infiltration.
Collapse
Affiliation(s)
- Qingshuang Zou
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Ke Liao
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
- Institute of Pharmacy and PharmacologySchool of Pharmaceutical ScienceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001China
| | - Guangchao Li
- Department of HematologyThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Xin Huang
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Yongwei Zheng
- Research and Development DepartmentGuangzhou Bio‐Gene Technology Co. Ltd.Guangzhou510530China
| | - Gun Yang
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Min Luo
- Research and Development DepartmentGuangzhou Bio‐Gene Technology Co. Ltd.Guangzhou510530China
| | - Evelyn Y. Xue
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Chuanqing Lan
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Shuai Wang
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Yao Shen
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Dixian Luo
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Dennis K. P. Ng
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Quan Liu
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| |
Collapse
|
33
|
Qi X, Chen G, Cao YJ. Optimal Structural Designs of Trispecific Antibodies to Enhance Therapeutic Efficacy in Solid Tumors and Hematological Malignancies. Methods Mol Biol 2025; 2930:277-294. [PMID: 40402462 DOI: 10.1007/978-1-0716-4558-1_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Engineered antibody therapies have significantly propelled the advancement of tumor immunotherapy. However, a considerable proportion of patients fail to respond to treatment or experience relapses following an initial positive response. Moreover, recurrence rates have increased due to antigen escape or downregulation. To address this challenge, novel strategies to engineer trispecific antibodies (tsAbs) have been developed in recent years. Here, we describe the molecular procedures of construction, expression, and purification of optimal tsAbs [specifically, Her2/VEGFR2/CD3 (SO) and CD19/CD22/CD3 (CC) tsAbs]. Also, we detail the experimental methods of functional validation of optimal tsAbs, including immunofluorescence analysis, cytokine release detection, and in vitro cytotoxicity assay and in vivo efficacy studies.
Collapse
Affiliation(s)
- Xuexiu Qi
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Guang Chen
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
34
|
Chu X, Mi B, Xiong Y, Wang R, Liu T, Hu L, Yan C, Zeng R, Lin J, Fu H, Liu G, Zhang K, Bian L. Bioactive nanocomposite hydrogel enhances postoperative immunotherapy and bone reconstruction for osteosarcoma treatment. Biomaterials 2025; 312:122714. [PMID: 39079462 DOI: 10.1016/j.biomaterials.2024.122714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Osteosarcoma, a malignant bone tumor often characterized by high hedgehog signaling activity, residual tumor cells, and substantial bone defects, poses significant challenges to both treatment response and postsurgical recovery. Here, we developed a nanocomposite hydrogel for the sustained co-delivery of bioactive magnesium ions, anti-PD-L1 antibody (αPD-L1), and hedgehog pathway antagonist vismodegib, to eradicate residual tumor cells while promoting bone regeneration post-surgery. In a mouse model of tibia osteosarcoma, this hydrogel-mediated combination therapy led to remarkable tumor growth inhibition and hence increased animal survival by enhancing the activity of tumor-suppressed CD8+ T cells. Meanwhile, the implanted hydrogel improved the microenvironment of osteogenesis through long-term sustained release of Mg2+, facilitating bone defect repair by upregulating the expression of osteogenic genes. After 21 days, the expression levels of ALP, COL1, RUNX2, and BGLAP in the Vis-αPD-L1-Gel group were approximately 4.1, 5.1, 5.5, and 3.4 times higher than those of the control, respectively. We believe that this hydrogel-based combination therapy offers a potentially valuable strategy for treating osteosarcoma and addressing the tumor-related complex bone diseases.
Collapse
Affiliation(s)
- Xiangyu Chu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, PR China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, PR China
| | - Yuan Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ruinan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Tuozhou Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Liangcong Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, PR China
| | - Chenchen Yan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, PR China
| | - Ruiyin Zeng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, PR China
| | - Jiali Lin
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Hao Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, PR China.
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China.
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
35
|
Gile JJ, Maurer M, Ruan GJ, Abeykoon JP, Heimgartner JR, Baumann NA, McMahon M, Lin Y, Witzig TE. Low magnesium levels and prognosis in newly diagnosed diffuse large B-cell lymphoma. Oncologist 2024; 29:e1779-e1782. [PMID: 39418117 PMCID: PMC11630732 DOI: 10.1093/oncolo/oyae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/17/2024] [Indexed: 10/19/2024] Open
Abstract
Magnesium (Mg) is an essential element involved in cellular metabolism. We demonstrated that in patients with diffuse large B-cell lymphoma (DLBCL) undergoing autologous stem cell transplant (SCT), those with a serum Mg < 2.0 mg/dL at the time of transplant had worse outcomes. In this study, we aimed to learn the prognostic value of low serum Mg in patients with untreated DLBCL. We analyzed serum from 408 patients and tested 2 Mg cutpoints-low (<1.7 mg/dL) and low normal (<2.0 mg/dL), a range we found associated with lower survival in the SCT group. We found 3% of patients with low levels and 23% with low normal levels. Low normal serum Mg levels were associated with a higher stage at diagnosis, more extranodal involvement, higher international prognostic index score, lower overall survival (OS), and event-free survival. These data warrant testing Mg replacement to a target of >2.0 mg/dL to learn if survival can be improved.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Female
- Prognosis
- Male
- Middle Aged
- Magnesium/blood
- Aged
- Adult
- Aged, 80 and over
Collapse
Affiliation(s)
- Jennifer J Gile
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Matthew Maurer
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, United States
| | - Gordon J Ruan
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Jithma P Abeykoon
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Joy R Heimgartner
- Division of Endocrinology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Nikola A Baumann
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Molly McMahon
- Division of Endocrinology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Yi Lin
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Thomas E Witzig
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
36
|
Feng Y, Gao M, Xu X, Liu H, Lu K, Song Z, Yu J, Liu X, Han X, Li L, Qiu L, Qian Z, Zhou S, Zhang H, Wang X. Elevated serum magnesium levels prompt favourable outcomes in cancer patients treated with immune checkpoint blockers. Eur J Cancer 2024; 213:115069. [PMID: 39489925 DOI: 10.1016/j.ejca.2024.115069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Magnesium deficiency influences the activation and cytotoxicity of immune cells. Nevertheless, whether serum magnesium levels influence the clinical outcomes of immune checkpoint blockers (ICBs) treatment still remains ambiguous. There is an urgent need for clinical research to elucidate the relationship between serum magnesium levels and the outcomes of ICB therapy. Such insights could offer new perspectives on immunotherapy for cancer. METHODS A multi-center retrospective study involving in pan-cancer patients treated with ICBs at three large cancer centers from August 2012 to May 2023 was conducted. The primary objective was to assess the correlation between serum magnesium levels and therapeutic response in patients receiving ICBs, and further evaluate the associations between serum magnesium levels and progression-free survival (PFS) and overall survival (OS). RESULTS A total of 1441 patients treated with ICBs, including 1042 with lung cancer, 270 with esophageal cancer, and 129 with Hodgkin lymphoma, were enrolled in this study. We found that patients with elevated serum magnesium levels exhibited a favourable response to ICBs treatment. The optimal cut-off point for serum magnesium level (0.79 mmol/L) was applied for stratifying patients into distinct groups. In the three tumor cohorts, patients in high magnesium level group (Mg2+ ≥ 0.79 mmol/L) had longer PFS and OS than those in low magnesium level group (Mg2+ < 0.79 mmol/L). Univariate and multivariate analyses confirmed that the serum Mg2+ level serves as an independent prognostic factor for cancer patients receiving ICBs therapy. CONCLUSION Our multi-center study demonstrated that among patients receiving ICBs therapy, those with elevated serum magnesium levels exhibit significantly better clinical outcomes than those with low serum magnesium levels. Further prospective validation studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Yingfang Feng
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Meng Gao
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China; The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010030, China
| | - Xiyue Xu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Hengqi Liu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Ke Lu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China; Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Zheng Song
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Jingwei Yu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Xia Liu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Xue Han
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Lanfang Li
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Lihua Qiu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Zhengzi Qian
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Shiyong Zhou
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Huilai Zhang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| | - Xianhuo Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| |
Collapse
|
37
|
Zhang FQ, Chen J, Fan H. Eating for immunity: how diet shapes our defenses. Curr Opin Immunol 2024; 91:102486. [PMID: 39353254 PMCID: PMC11609002 DOI: 10.1016/j.coi.2024.102486] [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: 06/26/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
Emerging studies on the diet-immune axis have uncovered novel dietary immune regulators and identified crucial targets and pathways mediating the crosstalk between specific dietary components and diverse immune cell populations. Here, we discuss the recent discovery and mechanisms by which diet-derived components, such as vitamins, amino acids, fatty acids, and antioxidants, could impact immune cell metabolism, alter signaling pathways, and reprogram the overall cellular responses. We also note crucial considerations that need to be tackled to make these findings clinically relevant, acknowledging that our current understanding often relies on simplified models that may not adequately represent the intricate network of factors influencing the diet-immune axis at the whole organism level. Overall, our growing understanding of how diet shapes our defenses underscores the importance of lifestyle choices and illuminates the potential to fine-tune immune responses through targeted nutritional strategies, thereby fortifying the immune system and bolstering our defenses against diseases.
Collapse
Affiliation(s)
- Freya Q Zhang
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Jing Chen
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA.
| | - Hao Fan
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
38
|
Piemonte E, Bruzzaniti S, Galgani M. Type 2 Diabetes: Fighting Inflammation Fire of T Lymphocytes With Magnesium. J Clin Endocrinol Metab 2024; 109:e2348-e2349. [PMID: 38593086 DOI: 10.1210/clinem/dgae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Affiliation(s)
- Erica Piemonte
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131 Naples, Italy
| | - Sara Bruzzaniti
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131 Naples, Italy
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Mario Galgani
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131 Naples, Italy
| |
Collapse
|
39
|
Drenthen LCA, Ajie M, de Baaij JHF, Tack CJ, de Galan BE, Stienstra R. Magnesium Supplementation Modulates T-cell Function in People with Type 2 Diabetes and Low Serum Magnesium Levels. J Clin Endocrinol Metab 2024; 109:e2240-e2245. [PMID: 38412117 PMCID: PMC11570359 DOI: 10.1210/clinem/dgae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/31/2024] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
CONTEXT Low magnesium levels, which are common in people with type 2 diabetes, are associated with increased levels of proinflammatory molecules. It is unknown whether magnesium supplementation decreases this low-grade inflammation in people with type 2 diabetes. OBJECTIVE We performed multidimensional immunophenotyping to better understand the effect of magnesium supplementation on the immune system of people with type 2 diabetes and low magnesium levels. METHODS Using a randomized, double-blind, placebo-controlled, 2-period, crossover study, we compared the effect of magnesium supplementation (15 mmol/day) with placebo on the immunophenotype, including whole blood immune cell counts, T-cell and CD14+ monocyte function after ex vivo stimulation, and the circulating inflammatory proteome. RESULTS We included 12 adults with insulin-treated type 2 diabetes (7 males, mean ± SD age 67 ± 7 years, body mass index 31 ± 5 kg/m2, HbA1c 7.5 ± 0.9%) and low magnesium levels (0.73 ± 0.05 mmol/L). Magnesium treatment significantly increased serum magnesium and urinary magnesium excretion compared with placebo. Interferon-γ production from phorbol myristate acetate/ionomycin stimulated CD8+ T-cells and T-helper 1 cells, as well as interleukin (IL) 4/IL5/IL13 production from T-helper 2 cells was lower after treatment with magnesium compared with placebo. Magnesium supplementation did not affect immune cell numbers, ex vivo monocyte function, and circulating inflammatory proteins, although we found a tendency for lower high sensitivity C-reactive protein levels after magnesium supplementation compared with placebo. CONCLUSION In conclusion, magnesium supplementation modulates the function of CD4+ and CD8+ T-cells in people with type 2 diabetes and low serum magnesium levels.
Collapse
Affiliation(s)
- Linda C A Drenthen
- Department of Internal Medicine, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Mandala Ajie
- Department of Internal Medicine, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Medical Biosciences, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Bastiaan E de Galan
- Department of Internal Medicine, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Department of Internal Medicine, Maastricht UMC+, 6229 HX Maastricht, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine, Radboudumc, 6500 HB Nijmegen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
40
|
Lin F, Yang H, Huang Z, Li Y, Ding Q, Ye Y, Qiu S. Magnesium-related gene ITGAL: a key immunotherapy predictor and prognostic biomarker in pan-cancer. Front Pharmacol 2024; 15:1464830. [PMID: 39605903 PMCID: PMC11598444 DOI: 10.3389/fphar.2024.1464830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background Integrin subunit alpha L (ITGAL) is crucial for activating CD8+ T cells through magnesium-mediated immune synapse formation and specific cytotoxicity. ITGAL might exert an important function in the growth and transformation of cancer. Methods Our study comprehensively analyzed ITGAL expression across various cancers, validated by Immunochemistry (IHC) in the laboratory. ITGAL showed prognostic significance in pan-cancer patients, correlated with clinical features, and associated with specific signaling pathways. We also observed a relationship between ITGAL and immune cell infiltration. In HNSCC, ITGAL demonstrated prognostic value and potential implications for immunotherapy response and novel drug targets. Results ITGAL expression linked to tumor prognosis across 27 cancers. Elevated ITGAL correlated with good prognosis in CESC, LUAD, SARC, HNSCC, and SKCM. ITGAL involved in immune regulation pathways and showed positive correlation with immune cell infiltration. ITGAL associated with CD8+ T cell infiltration. And high ITGAL expression in CD8+ T cells and NK cells. In HNSCC, ITGAL linked to favorable prognosis and sensitivity to immunotherapy. Predicted potential drugs for HNSCC. Conclusion ITGAL is remarkably associated with CD8+T cells and crucial in the tumor immune microenvironment of pan-cancer. Furthermore, our findings may provide a targeted anti-tumor strategy for ITGAL by influencing the tumor immune microenvironment.
Collapse
Affiliation(s)
- Fengjie Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, China
| | - Hanxuan Yang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, China
| | - Zongwei Huang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, China
| | - Ying Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, China
| | - Qin Ding
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, China
| | - Yunbin Ye
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer, Hospital, Fuzhou, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, China
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, China
| |
Collapse
|
41
|
Yuan K, Zhang C, Pan X, Hu B, Zhang J, Yang G. Immunomodulatory metal-based biomaterials for cancer immunotherapy. J Control Release 2024; 375:249-268. [PMID: 39260573 DOI: 10.1016/j.jconrel.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Cancer immunotherapy, as an emerging cancer treatment approach, harnesses the patient's own immune system to effectively prevent tumor recurrence or metastasis. However, its clinical application has been significantly hindered by relatively low immune response rates. In recent years, metal-based biomaterials have been extensively studied as effective immunomodulators and potential tools for enhancing anti-tumor immune responses, enabling the reversal of immune suppression without inducing toxic side effects. This review introduces the classification of bioactive metal elements and summarizes their immune regulatory mechanisms. In addition, we discuss the immunomodulatory roles of biomaterials constructed from various metals, including aluminum, manganese, gold, calcium, zinc, iron, magnesium, and copper. More importantly, a systematic overview of their applications in enhancing immunotherapy is provided. Finally, the prospects and challenges of metal-based biomaterials with immunomodulatory functions in cancer immunotherapy are outlined.
Collapse
Affiliation(s)
- Kangzhi Yuan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xinlu Pan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, Jiangsu 215004, China.
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
42
|
Su Z, Boucetta H, Shao J, Huang J, Wang R, Shen A, He W, Xu ZP, Zhang L. Next-generation aluminum adjuvants: Immunomodulatory layered double hydroxide NanoAlum reengineered from first-line drugs. Acta Pharm Sin B 2024; 14:4665-4682. [PMID: 39664431 PMCID: PMC11628803 DOI: 10.1016/j.apsb.2024.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 12/13/2024] Open
Abstract
Aluminum adjuvants (Alum), approved by the US Food and Drug Administration, have been extensively used in vaccines containing recombinant antigens, subunits of pathogens, or toxins for almost a century. While Alums typically elicit strong humoral immune responses, their ability to induce cellular and mucosal immunity is limited. As an alternative, layered double hydroxide (LDH), a widely used antacid, has emerged as a novel class of potent nano-aluminum adjuvants (NanoAlum), demonstrating advantageous physicochemical properties, biocompatibility and adjuvanticity in both humoral and cellular immune responses. In this review, we summarize and compare the advantages and disadvantages of Alum and NanoAlum in these properties and their performance as adjuvants. Moreover, we propose the key features for ideal adjuvants and demonstrate that LDH NanoAlum is a promising candidate by summarizing its current progress in immunotherapeutic cancer treatments. Finally, we conclude the review by offering our integrated perspectives about the remaining challenges and future directions for NanoAlum's application in preclinical/clinical settings.
Collapse
Affiliation(s)
- Zhenwei Su
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Hamza Boucetta
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jiahui Shao
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Jinling Huang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ran Wang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Aining Shen
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhi Ping Xu
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Lingxiao Zhang
- Interdisciplinary Nanoscience Center (INANO), Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
43
|
Xu Z, Wu Y, Hu J, Mei Z, Zhao Y, Yang K, Shi Y, Xu X. Recent advances in nanoadjuvant-triggered STING activation for enhanced cancer immunotherapy. Heliyon 2024; 10:e38900. [PMID: 39640775 PMCID: PMC11620084 DOI: 10.1016/j.heliyon.2024.e38900] [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: 04/30/2024] [Revised: 07/22/2024] [Accepted: 10/01/2024] [Indexed: 12/07/2024] Open
Abstract
The development of effective cancer treatments is a popular in contemporary medical research. Immunotherapy, the fourth most common cancer treatment method, relies on activating autoimmune function to eradicate tumors and exhibits advantages such as a good curative effect and few side effects. In recent years, tumor vaccines that activate the stimulator of interferon genes (STING) pathway are being actively researched in the field of immunotherapy; however, their application is still limited because of the rapid clearance rate of tumor-related lymph nodes and low efficiency of antigen presentation. The rise of nanomedicine has provided new opportunities for solving these problems. By preparing materials with adjuvant effects nanoparticles, the small size of nanoparticles can be exploited to enable the entry of vaccines into tumor-related lymph nodes to accurately deliver STING agonists and activate the immune response. Based on this, this paper reviews various types of nano-adjuvants based on metals, platinum chemotherapy drugs, camptothecin derivatives, deoxyribonucleic acid, etc. and highlights the transformation prospects of these nano-adjuvants in tumor vaccines to provide a reference for promoting the development of nano-medicine and tumor vaccinology.
Collapse
Affiliation(s)
- Zicong Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yihong Wu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Junjie Hu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Zhaozhao Mei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yutong Zhao
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, PR China
| | - Keda Yang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, PR China
| | - Xiaoling Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| |
Collapse
|
44
|
Zhang R, Hu M, Liu Y, Li W, Xu Z, He S, Lu Y, Gong Y, Wang X, Hai S, Li S, Qi S, Li Y, Shu Y, Du D, Zhang H, Xu H, Zhou Z, Lei P, Chen HN, Dai L. Integrative Omics Uncovers Low Tumorous Magnesium Content as A Driver Factor of Colorectal Cancer. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae053. [PMID: 39052867 PMCID: PMC11514849 DOI: 10.1093/gpbjnl/qzae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 03/04/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Magnesium (Mg) deficiency is associated with increased risk and malignancy in colorectal cancer (CRC), yet the underlying mechanisms remain elusive. Here, we used genomic, proteomic, and phosphoproteomic data to elucidate the impact of Mg deficiency on CRC. Genomic analysis identified 160 genes with higher mutation frequencies in Low-Mg tumors, including key driver genes such as KMT2C and ERBB3. Unexpectedly, initiation driver genes of CRC, such as TP53 and APC, displayed higher mutation frequencies in High-Mg tumors. Additionally, proteomic and phosphoproteomic data indicated that low Mg content in tumors may activate epithelial-mesenchymal transition (EMT) by modulating inflammation or remodeling the phosphoproteome of cancer cells. Notably, we observed a negative correlation between the phosphorylation of DBN1 at S142 (DBN1S142p) and Mg content. A mutation in S142 to D (DBN1S142D) mimicking DBN1S142p up-regulated MMP2 and enhanced cell migration, while treatment with MgCl2 reduced DBN1S142p, thereby reversing this phenotype. Mechanistically, Mg2+ attenuated the DBN1-ACTN4 interaction by decreasing DBN1S142p, which in turn enhanced the binding of ACTN4 to F-actin and promoted F-actin polymerization, ultimately reducing MMP2 expression. These findings shed new light on the crucial role of Mg deficiency in CRC progression and suggest that Mg supplementation may be a promising preventive and therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Rou Zhang
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Hu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Liu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanmeng Li
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqiang Xu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Siyu He
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Lu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiuxuan Wang
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan Hai
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuangqing Li
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiqian Qi
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Li
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Shu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Du
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huiyuan Zhang
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Heng Xu
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zongguang Zhou
- Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Lei
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Strazza M, Song R, Hiner S, Mor A. Changing the location of proteins on the cell surface is a promising strategy for modulating T cell functions. Immunology 2024; 173:248-257. [PMID: 38952142 PMCID: PMC11987702 DOI: 10.1111/imm.13828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
Targeting immune receptors on T cells is a common strategy to treat cancer and autoimmunity. Frequently, this is accomplished through monoclonal antibodies targeting the ligand binding sites of stimulatory or inhibitory co-receptors. Blocking ligand binding prevents downstream signalling and modulates specific T cell functions. Since 1985, the FDA has approved over 100 monoclonal antibodies against immune receptors. This therapeutic approach significantly improved the care of patients with numerous immune-related conditions; however, many patients are unresponsive, and some develop immune-related adverse events. One reason for that is the lack of consideration for the localization of these receptors on the cell surface of the immune cells in the context of the immune synapse. In addition to blocking ligand binding, changing the location of these receptors on the cell surface within the different compartments of the immunological synapse could serve as an alternative, efficient, and safer approach to treating these patients. This review discusses the potential therapeutic advantages of altering proteins' localization within the immune synapse and summarizes published work in this field. It also discusses the novel use of bispecific antibodies to induce the clustering of receptors on the cell surface. It presents the rationale for developing novel antibodies, targeting the organization of signalling receptor complexes on the cell surface. This approach offers an innovative and emerging technology to treat cancer patients resistant to current immunotherapies.
Collapse
Affiliation(s)
- Marianne Strazza
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
| | - Ruijiang Song
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
| | - Shannon Hiner
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
| | - Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
46
|
Peng Y, Liang S, Liu D, Ma K, Yun K, Zhou M, Hai L, Xu M, Chen Y, Wang Z. Multi-Metallic Nanosheets Reshaping Immunosuppressive Tumor Microenvironment through Augmenting cGAS-STING Innate Activation and Adaptive Immune Responses for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403347. [PMID: 39120546 PMCID: PMC11481177 DOI: 10.1002/advs.202403347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/14/2024] [Indexed: 08/10/2024]
Abstract
The highly immunosuppressive tumor microenvironment (TME) restricts the efficient activation of immune responses. To restore the surveillance of the immune system for robust activation, vast efforts are devoted to normalizing the TME. Here, a manganese-doped layered double hydroxide (Mn-LDH) is developed for potent anti-tumor immunity by reversing TME. Mn-LDH is synthesized via a one-step hydrothermal method. In addition to the inherent proton neutralization capacity of LDH, the introduction of manganese oxide endows LDH with an additional ability to produce oxygen. Mn-LDH effectively releases Mn2+ and Mg2+ upon exposure to TME with high levels of H+ and H2O2, which activates synthase-stimulator of interferon genes pathway and maintains the cytotoxicity of CD8+ T cells respectively, achieving a cascade-like role in innate and adaptive immunity. The locally administered Mn-LDH facilitated a "hot" network consisting of mature dendritic cells, M1-phenotype macrophages, as well as cytotoxic and helper T cells, significantly inhibiting the growth of primary and distal tumors. Moreover, the photothermal conversion capacity of Mn-LDH sparks more robust therapeutic effects in large established tumor models with a single administration and irradiation. Overall, this study guides the rational design of TME-modulating immunotherapeutics for robust immune activation, providing a clinical candidate for next-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Kaiqing Yun
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Linna Hai
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Mengdi Xu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Yiyang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| |
Collapse
|
47
|
Hajdu KL, Rousseau L, Ho PC. A pinch of salt boosts T cell function. Nat Immunol 2024; 25:1772-1774. [PMID: 39251800 DOI: 10.1038/s41590-024-01946-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Affiliation(s)
- Karina L Hajdu
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Épalinges, Switzerland
| | - Lorène Rousseau
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Épalinges, Switzerland
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Épalinges, Switzerland.
| |
Collapse
|
48
|
Scirgolea C, Sottile R, De Luca M, Susana A, Carnevale S, Puccio S, Ferrari V, Lise V, Contarini G, Scarpa A, Scamardella E, Feno S, Camisaschi C, De Simone G, Basso G, Giuliano D, Mazza EMC, Gattinoni L, Roychoudhuri R, Voulaz E, Di Mitri D, Simonelli M, Losurdo A, Pozzi D, Tsui C, Kallies A, Timo S, Martano G, Barberis E, Manfredi M, Rescigno M, Jaillon S, Lugli E. NaCl enhances CD8 + T cell effector functions in cancer immunotherapy. Nat Immunol 2024; 25:1845-1857. [PMID: 39198631 DOI: 10.1038/s41590-024-01923-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 07/10/2024] [Indexed: 09/01/2024]
Abstract
CD8+ T cells control tumors but inevitably become dysfunctional in the tumor microenvironment. Here, we show that sodium chloride (NaCl) counteracts T cell dysfunction to promote cancer regression. NaCl supplementation during CD8+ T cell culture induced effector differentiation, IFN-γ production and cytotoxicity while maintaining the gene networks responsible for stem-like plasticity. Accordingly, adoptive transfer of tumor-specific T cells resulted in superior anti-tumor immunity in a humanized mouse model. In mice, a high-salt diet reduced the growth of experimental tumors in a CD8+ T cell-dependent manner by inhibiting terminal differentiation and enhancing the effector potency of CD8+ T cells. Mechanistically, NaCl enhanced glutamine consumption, which was critical for transcriptional, epigenetic and functional reprogramming. In humans, CD8+ T cells undergoing antigen recognition in tumors and predicting favorable responses to checkpoint blockade immunotherapy resembled those induced by NaCl. Thus, NaCl metabolism is a regulator of CD8+ T cell effector function, with potential implications for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Simone Puccio
- IRCCS Humanitas Research Hospital, Milan, Italy
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Milan, Italy
| | | | | | | | | | | | - Simona Feno
- IRCCS Humanitas Research Hospital, Milan, Italy
| | | | | | | | | | | | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
- University of Regensburg, Regensburg, Germany
- Center for Immunomedicine in Transplantation and Oncology (CITO), University Hospital Regensburg, Regensburg, Germany
| | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Cambridge, UK
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Emanuele Voulaz
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Division of Thoracic, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Matteo Simonelli
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Davide Pozzi
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Carlson Tsui
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | - Sara Timo
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Giuseppe Martano
- IRCCS Humanitas Research Hospital, Milan, Italy
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Milan, Italy
| | - Elettra Barberis
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Sebastien Jaillon
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Enrico Lugli
- IRCCS Humanitas Research Hospital, Milan, Italy.
| |
Collapse
|
49
|
Li Y, Wang Y, Zhao L, Stenzel MH, Jiang Y. Metal ion interference therapy: metal-based nanomaterial-mediated mechanisms and strategies to boost intracellular "ion overload" for cancer treatment. MATERIALS HORIZONS 2024; 11:4275-4310. [PMID: 39007354 DOI: 10.1039/d4mh00470a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Metal ion interference therapy (MIIT) has emerged as a promising approach in the field of nanomedicine for combatting cancer. With advancements in nanotechnology and tumor targeting-related strategies, sophisticated nanoplatforms have emerged to facilitate efficient MIIT in xenografted mouse models. However, the diverse range of metal ions and the intricacies of cellular metabolism have presented challenges in fully understanding this therapeutic approach, thereby impeding its progress. Thus, to address these issues, various amplification strategies focusing on ionic homeostasis and cancer cell metabolism have been devised to enhance MIIT efficacy. In this review, the remarkable progress in Fe, Cu, Ca, and Zn ion interference nanomedicines and understanding their intrinsic mechanism is summarized with particular emphasis on the types of amplification strategies employed to strengthen MIIT. The aim is to inspire an in-depth understanding of MIIT and provide guidance and ideas for the construction of more powerful nanoplatforms. Finally, the related challenges and prospects of this emerging treatment are discussed to pave the way for the next generation of cancer treatments and achieve the desired efficacy in patients.
Collapse
Affiliation(s)
- Yutang Li
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, P. R. China.
| | - Yandong Wang
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, P. R. China.
| | - Li Zhao
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, P. R. China.
| | - Martina H Stenzel
- School of Chemistry, University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Yanyan Jiang
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, P. R. China.
| |
Collapse
|
50
|
Porte S, Audemard-Verger A, Wu C, Durand A, Level T, Giraud L, Lombès A, Germain M, Pierre R, Saintpierre B, Lambert M, Auffray C, Peyssonnaux C, Goldwasser F, Vaulont S, Alves-Guerra MC, Dentin R, Lucas B, Martin B. Iron Boosts Antitumor Type 1 T-cell Responses and Anti-PD1 Immunotherapy. Cancer Immunol Res 2024; 12:1252-1267. [PMID: 38912762 DOI: 10.1158/2326-6066.cir-23-0739] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 04/02/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
Cancers only develop if they escape immunosurveillance, and the success of cancer immunotherapies relies in most cases on their ability to restore effector T-cell functions, particularly IFNγ production. Revolutionizing the treatment of many cancers, immunotherapies targeting immune checkpoints such as PD1 can increase survival and cure patients. Unfortunately, although immunotherapy has greatly improved the prognosis of patients, not all respond to anti-PD1 immunotherapy, making it crucial to identify alternative treatments that could be combined with current immunotherapies to improve their effectiveness. Here, we show that iron supplementation significantly boosts T-cell responses in vivo and in vitro. The boost was associated with a metabolic reprogramming of T cells in favor of lipid oxidation. We also found that the "adjuvant" effect of iron led to a marked slowdown of tumor cell growth after tumor cell line transplantation in mice. Specifically, our results suggest that iron supplementation promotes antitumor responses by increasing IFNγ production by T cells. In addition, iron supplementation improved the efficacy of anti-PD1 cancer immunotherapy in mice. Finally, our study suggests that, in patients with cancer, the quality and efficacy of the antitumor response following anti-PD1 immunotherapy may be modulated by plasma ferritin levels. In summary, our results suggest the benefits of iron supplementation on the reactivation of antitumor responses and support the relevance of a fruitful association between immunotherapy and iron supplementation.
Collapse
Affiliation(s)
- Sarah Porte
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | | | - Christian Wu
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Aurélie Durand
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Théo Level
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Léa Giraud
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Amélie Lombès
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Mathieu Germain
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Rémi Pierre
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Benjamin Saintpierre
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Mireille Lambert
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Cédric Auffray
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Carole Peyssonnaux
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - François Goldwasser
- Department of Medical Oncology, Cochin Hospital, Paris Cancer Institute CARPEM, Université Paris Cité, APHP.Centre, Paris, France
| | - Sophie Vaulont
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Marie-Clotilde Alves-Guerra
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Renaud Dentin
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Bruno Lucas
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Bruno Martin
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| |
Collapse
|