1
|
Wang F, Wang Z, Huang S, Yang J, Chen Y, Chao H. A Ruthenium(II)-DNA Nanophotosensitizer for cGAS-STING-Mediated Photodynamic Immunotherapy Against Hypoxic Cisplatin-Resistant Tumor Cells. Chemistry 2025:e202501016. [PMID: 40326977 DOI: 10.1002/chem.202501016] [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: 03/13/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/07/2025]
Abstract
Plasmid DNAs intercalated with Ru(II) complexes were encapsulated in liposomes to create nano-photosensitizers. Upon light exposure, the ruthenium complex photo-cleavages DNA. The DNA fragments specifically stimulate the cGAS-STING pathway and induce immunogenic death. This study presents a convenient strategy for photodynamic immunotherapy against cisplatin-resistant tumor cells under hypoxia.
Collapse
Affiliation(s)
- Fa Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 51006, People's Republic of China
| | - Zhiyang Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 51006, People's Republic of China
| | - Shuqi Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 51006, People's Republic of China
| | - Jinrong Yang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 51006, People's Republic of China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 51006, People's Republic of China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 51006, People's Republic of China
- MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 400201, People's Republic of China
| |
Collapse
|
2
|
Wang K, Zhang X, Li A, Qiao X, Xu Y. The mechanism of action and therapeutic potential of tumor-associated macrophages in tumor immune evasion. Front Immunol 2025; 16:1545928. [PMID: 40330472 PMCID: PMC12052954 DOI: 10.3389/fimmu.2025.1545928] [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: 12/16/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Tumor-associated macrophages (TAMs) play a multifaceted role in tumor progression. As specialized immune cells, macrophages are capable of phagocytosis and digesting foreign substances, as well as removing harmful substances including cellular debris and tumor cells. Under specific pathological conditions, circulating monocytes can be recruited into the tumor microenvironment and differentiate into TAMs. Macrophages are generally polarized into two distinct subpopulations: classically activated macrophages (M1) and alternatively activated macrophages (M2). TAMs constitute a significant proportion of the mononuclear leukocyte population in solid tumors, exhibiting a complex and dualistic relationship with tumor cells. Substantial evidence indicates that TAMs can interact with tumor cells, facilitating their immune evasion while promoting invasion and metastasis. This review focuses on the mechanism and regulation of macrophages in the immune response to tumor cells, as well as various macrophage-based tumor-targeted therapeutic strategies. It will provide a reference for research on macrophage-centered therapy strategies and their application in clinical practice.
Collapse
Affiliation(s)
- Kehua Wang
- Department of Vascular Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xu Zhang
- Department of Surgery Laboratory, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Aiqin Li
- Department of Surgery Laboratory, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xia Qiao
- Department of Surgery Laboratory, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yanan Xu
- Department of Surgery Laboratory, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
3
|
Sun K, Wei X, Han S, Sun Y, Xiao H, Wei D. Biotin Receptor-Targeting Pt IV Oxygen Carrying Prodrug Amphiphile for Alleviating Tumor Hypoxia Induced Immune Chemotherapy Suppression. ACS NANO 2025; 19:13300-13313. [PMID: 40035261 DOI: 10.1021/acsnano.5c00691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Platinum (Pt)-based chemotherapeutic agents, known for their potent cytotoxicity, are extensively used in clinical oncology. However, their therapeutic efficacy is severely limited by a variety of factors, particularly the hypoxic tumor microenvironment (TME), which not only impedes effective drug delivery but also triggers immune suppression, further diminishing the antitumor effects of Pt drugs. In response to these challenges, we have developed a biotin receptor (BR)-targeting oxaliplatin (OXA)-based PtIV prodrug, named Lipo-OPtIV-BT, which could encapsulate hemoglobin (Hb) as an oxygen carrier, forming PtIV-loaded lipid nanoparticles (Hb@BTOPtIV). The design of the Hb@BTOPtIV aims to address the dual issues of poor drug delivery and immune suppression by effectively increasing local oxygen tension in the TME. Notably, our findings demonstrate that the cytotoxic effects of the BR-targeting PtIV prodrug and increased oxygen levels synergistically reverse the tumor immune microenvironment, leading to improved antitumor efficacy. We observed that Hb@BTOPtIV significantly improved the biodistribution of the drug, enabling it to preferentially accumulate in tumor regions. Importantly, the enhanced oxygenation within the TME also plays a critical role in reshaping the immune landscape of the tumor, promoting a more favorable immune environment for effective chemotherapy. This reversal of immune suppression is evidenced by increased infiltration of cytotoxic T cells and reduced levels of regulatory T cells (Tregs) within the tumor. These findings highlight the promising potential of using BR-targeting lipid PtIV prodrug amphiphiles to improve drug accumulation at tumor sites and counteract immunosuppression induced by tumor hypoxia.
Collapse
Affiliation(s)
- Kaichuang Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Xiaodan Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Dengshuai Wei
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| |
Collapse
|
4
|
Tian L, Piao S, Li X, Guo L, Huang L, Gao W. Functional Materials Targeted Regulation of Gasdermins: From Fundamentals to Functionalities and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500873. [PMID: 40273335 PMCID: PMC12021126 DOI: 10.1002/advs.202500873] [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: 01/14/2025] [Revised: 02/19/2025] [Indexed: 04/26/2025]
Abstract
Targeted regulation of pyroptosis to modulate the immune landscape has emerged as a novel design strategy for cancer immunotherapy and anti-inflammatory therapy. However, pyroptosis acts as a double-edged sword, making it important to optimize the design strategies of functional materials to appropriately activate pyroptosis for effective disease treatment. This paper summarizes and discusses the structure, pore formation, and molecular mechanisms of "executor" Gasdermins, as well as the events preceding and following these processes. Subsequently, the focus is on reviewing functional materials that directly regulate Gasdermin pore formation to target pyroptosis and those that indirectly regulate the events before and after Gasdermin pore formation to control pyroptosis activity. Finally, the advantages, disadvantages, and future prospects of designing such functional materials are provided, aiming to facilitate the precise design, pharmacological investigation, and clinical translation of pyroptosis-related functional materials.
Collapse
Affiliation(s)
- Luyao Tian
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Shuo Piao
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Xia Li
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Lanping Guo
- National Resource Center for Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700P. R. China
| | - Luqi Huang
- National Resource Center for Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700P. R. China
| | - Wenyuan Gao
- Key Laboratory of Pharmacology School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| |
Collapse
|
5
|
Sharma A, Al Amin M, Kshetri MB, Alqarni S, Jogadi W, Solmen J, Lin Z, Akter S, Zheng YR. PEGylation Effects on Amphiphilic Platinum(IV) Complexes: Influence on Uptake, Activation, and Cytotoxicity. Pharmaceutics 2025; 17:440. [PMID: 40284435 PMCID: PMC12030465 DOI: 10.3390/pharmaceutics17040440] [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: 03/03/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The utilization of amphiphilic Pt(IV) complexes as prodrugs offers a promising strategy to revolutionize Pt-based cancer therapy by enhancing drug delivery and activation. While PEGylation is widely used to optimize drug properties, its impact on the biological behavior of amphiphilic Pt(IV) complexes remains unclear. This study systematically investigates how the PEGylation of varying molecular weights influences their cytotoxicity, cellular uptake, and activation. Methods: Pt(IV) complexes were synthesized with PEG chains of different molecular weights using HATU-catalyzed amide bond formation and copper-free click chemistry. Their biological properties were assessed through cell-based analyses, focusing on cytotoxicity, cellular uptake, and activation by biological reductants. Results: Small PEG modifications retained the potent cytotoxicity of amphiphilic Pt(IV) prodrugs, whereas large PEG chains significantly reduced efficacy. The decrease in potency was linked to impaired cellular uptake and mitochondrial accumulation. Additionally, large PEG modifications slowed the reduction and activation of Pt(IV) prodrugs by biological reductants, further limiting their anticancer activities. Conclusions: These findings underscore the critical role of PEGylation in metallodrug design and provide key insights into optimizing PEGylation strategies for enhancing platinum-based cancer therapies.
Collapse
Affiliation(s)
- Arpit Sharma
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Md Al Amin
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Man B. Kshetri
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Suha Alqarni
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
- Department of Chemistry, University of Bisha, Bisha 67714, Saudi Arabia
| | - Wjdan Jogadi
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Jordan Solmen
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Zexin Lin
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Shirin Akter
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| | - Yao-Rong Zheng
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA
| |
Collapse
|
6
|
Yang J, Su T, Wang Q, Shi R, Ding J, Chen X. Glucose Metabolism-Targeted Poly(amino acid) Nanoformulation of Oxaliplatin(IV)-Aspirin Prodrug for Enhanced Chemo-Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419033. [PMID: 39950419 DOI: 10.1002/adma.202419033] [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: 12/05/2024] [Revised: 01/21/2025] [Indexed: 03/27/2025]
Abstract
Inappropriate glucose metabolism in cancer cells is associated with immunosuppressive tumor microenvironments (TMEs). Although glycolysis inhibition enhances T cell-mediated immune responses, the integrated platforms combining glycolysis inhibition with immunotherapy remain underdeveloped. To address this gap, a glucose metabolism-targeted poly(amino acid) nanoformulation of oxaliplatin(IV)-aspirin prodrug (NP/OXA-ASP2) is developed to improve chemo-immunotherapy by suppressing tumor glycolysis. This poly(amino acid) nanoparticle exhibits selective release, discharging 90.0% of OXA-ASP2 under reductive conditions within 36 h. Furthermore, over 80% of the prodrug converts to OXA and ASP within 12 h, promoting mitochondrial damage and glycolysis inhibition, which amplifies immunogenic cell death induced by OXA. In addition, suppressing glycolytic flux reduces lactate leakage, mitigating the immunosuppressive TMEs. Together, these mechanisms contribute to stronger chemo-immunotherapy efficacy. Compared to the OXA plus ASP formulation, NP/OXA-ASP2 demonstrates superior performances, reducing lactate levels at the tumor site by 25.4%, increasing the proportion of cytotoxic T lymphocytes by 1.53 times, decreasing the proportion of regulatory T cells by 2.20 times, and improving 1.39-fold of the tumor inhibition rate. These findings underscore that NP/OXA-ASP2 is a promising platform for integrating tumor metabolic regulation with immunomodulation and holds significant potential for advancing clinical chemo-immunotherapy.
Collapse
Affiliation(s)
- Jiazhen Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| | - Tianqi Su
- Department of Rehabilitation Medicine, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, P. R. China
| | - Qinqi Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| |
Collapse
|
7
|
Liang C, Yang H, Li T, Jiang X, Li X, Gao C, Hou L. On-Demand Controlled Release Multi-Drugs Delivery System for Spatiotemporally Synergizing Antitumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414233. [PMID: 39792614 PMCID: PMC11884579 DOI: 10.1002/advs.202414233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/09/2024] [Indexed: 01/12/2025]
Abstract
Although cytotoxic T lymphocytes (CTLs) activation combined with programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis blockade have emerged as an effective strategy to improve immunotherapeutic potency, it remains challenging to realize the spatiotemporal synergy of these two components. Herein, the study reports an engineered bacterial-based delivery system that can simultaneously promote CTLs infiltration and control PD-L1 binding protein (PD-L1 trap) release on demand at tumor site. The drug release button of this tumor targeting system is the specific temperature, which is accomplished by dual-modified melanin nanoparticles with photothermal conversion capacity on the engineered bacterial. These dual-modified nanoparticles can form in situ reservoir of heat supplier and antitumor immunity activator once arriving at tumor microenvironment (TME). Importantly, the study establishes the personalized administration regimen according to TME changes, and perform local laser irradiation to trigger PD-L1 trap production only in TME when infiltrated CTLs reach the highest level. This work provides a flexible platform for optimizing cancer immunotherapy.
Collapse
Affiliation(s)
- Chenglin Liang
- School of Pharmaceutical SciencesKey Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
| | - Hanxiao Yang
- School of Pharmaceutical SciencesKey Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
| | - Tongtong Li
- School of Pharmaceutical SciencesKey Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
| | - Xiaojuan Jiang
- School of Pharmaceutical SciencesKey Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
| | - Xinni Li
- School of Pharmaceutical SciencesKey Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
| | - Chen Gao
- School of Pharmaceutical SciencesKey Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
| | - Lin Hou
- School of Pharmaceutical SciencesKey Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
| |
Collapse
|
8
|
Zhang X, Tang D, Xiao H, Li B, Shang K, Zhao D. Activating the cGAS-STING Pathway by Manganese-Based Nanoparticles Combined with Platinum-Based Nanoparticles for Enhanced Ovarian Cancer Immunotherapy. ACS NANO 2025; 19:4346-4365. [PMID: 39846241 DOI: 10.1021/acsnano.4c12237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Recent research has demonstrated that activating the cGAS-STING pathway can enhance interferon production and the activation of T cells. A manganese complex, called TPA-Mn, was developed in this context. The reactive oxygen species (ROS)-sensitive nanoparticles (NPMn) loaded with TPA-Mn are developed. NPMn activates the cGAS-STING pathway via cGAS activation (i.e., 1.6-fold enhancement of P-STING), which in turn increases the secretion of pro-inflammatory cytokines (e.g., TNF-α, IL-6, and IL-2). This promotes dendritic cell maturation, enhances the infiltration of cytotoxic T lymphocytes, and reduces the percentage of immunosuppressive regulatory T cells. In addition, it is crucial to emphasize that cisplatin-induced DNA damage can potentially trigger activation of the cGAS-STING pathway. NPMn, in combination with low-dose NPPt, a carrier of a Cis(IV) prodrug capable of causing DNA damage, augments the cGAS-STING pathway activation and significantly activates the tumor immune microenvironment (TIME). Furthermore, combined with anti-PD-1 antibody, NPPt+NPMn shows synergistic efficacy in both ovarian cancer peritoneal metastases and recurrence models. It not only effectively eliminates tumors but also induces a strong immune memory response, providing a promising strategy for the clinical management of ovarian cancer. This work offers a design of manganese-based nanoparticles for immunotherapy.
Collapse
Affiliation(s)
- Xiangling Zhang
- Department of Gynecology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Bin Li
- Department of Gynecology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Kun Shang
- Department of Nuclear Medicine, Peking University People's Hospital, Beijing 100044, P. R. China
| | - Dan Zhao
- Department of Gynecology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| |
Collapse
|
9
|
Li L, Xu Q, Zhang X, Jiang Y, Zhang L, Guo J, Liu H, Jiang B, Li S, Peng Q, Jiang N, Wang J. AIEgen-self-assembled nanoparticles with anti-PD-L1 antibody functionalization realize enhanced synergistic photodynamic therapy and immunotherapy against malignant melanoma. Mater Today Bio 2025; 30:101387. [PMID: 39742147 PMCID: PMC11683329 DOI: 10.1016/j.mtbio.2024.101387] [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: 09/09/2024] [Revised: 11/17/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) become integral in clinical practice, yet their application in cancer therapy is constrained by low overall response rates and the primary resistance of cancers to ICIs. Herein, this study proposes aggregation-induced emission (AIE)-based nanoparticles (NPs) for a more effective and synergistic approach combining immunotherapy and photodynamic therapy (PDT) to achieve higher responses than anti-PD-L1 monotherapy. The TBP@aPD-L1 NPs are constructed by functionalizing azide group-modified TBP-2 (TBP-N3) with anti-PD-L1 antibodies via the DBCO-S-S-PEG2000-COOH linker. The anti-PD-L1 target the tumor cells and promote the TBP-N3 accumulation in tumors for enhanced PDT. Notably, the TBP-N3, featuring aggregation-induced emission, boosts reactive oxygen species (ROS) generation through both type I and type II processes for enhanced PDT. The TBP@aPD-L1-mediated PDT induces more powerful effects of direct tumor cell-killing and further elicits effective immunogenic cell death (ICD), which exerts anti-tumor immunity by activating T cells for ICI treatment and reshapes the tumor immune microenvironment (TIME), thereby enhancing the efficacy of PD-L1 blockade of anti-PD-L1. Consequently, TBP@aPD-L1 NPs demonstrated significantly enhanced inhibition of tumor growth in the mouse model of malignant melanoma (MM). Our NPs act as a facile and effective drug delivery platform for enhanced immunotherapy combined with enhanced PDT in treating MM.
Collapse
Affiliation(s)
- Lu Li
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qing Xu
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xiuzhen Zhang
- Hunan University of Medicine General Hospital, Hunan, 418000, PR China
| | - Yuan Jiang
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - La Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Jiao Guo
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Haichuan Liu
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Bin Jiang
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Shenglong Li
- School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qiling Peng
- Bijie Municipal Health Bureau, Guizhou, 551700, PR China
| | - Ning Jiang
- Department of Pathology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, PR China
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Jianwei Wang
- Department of Immunology, School of Basic Medical Science, Chongqing Medical University, Chongqing, 400016, PR China
| |
Collapse
|
10
|
Kong R, Huang J, Wu Y, Yan N, Chen X, Cheng H. Tricomponent immunoactivating nanomedicine to downregulate PD-L1 and polarize macrophage for photodynamic immunotherapy of colorectal cancer. Int J Pharm 2025; 668:124968. [PMID: 39561904 DOI: 10.1016/j.ijpharm.2024.124968] [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: 09/17/2024] [Revised: 11/05/2024] [Accepted: 11/16/2024] [Indexed: 11/21/2024]
Abstract
The unsatisfactory immunotherapeutic responses are primarily attributed to the insufficient immune recognition and the presence of an immunosuppressive tumor microenvironment (ITM). This study focuses on the development of a tricomponent immunoactivating nanomedicine called TIN that combines a photosensitizer, an inhibitor of epidermal growth factor receptor (EGFR) and a CSF-1R inhibitor to enable photodynamic immunotherapy by downregulating PD-L1 expression and repolarizing tumor-associated macrophages (TAMs). TIN is designed to facilitate the drug delivery and target specific pathways involved in tumor progression. By inhibiting the activity of EGFR and CSF-1R, TIN reduces PD-L1 expression on tumor cells and induces the TAMs polarization to M1 phenotype, restoring the immune recognition of T cells and the phagocytosis of macrophage to reshape the immunosuppressive microenvironment. Additionally, the photodynamic therapy (PDT) of TIN can greatly destroy the primary tumor and trigger immunogenic cell death (ICD). Importantly, the immune checkpoint blockade effect of TIN can enhance the immune response of PDT-induced ICD for metastatic tumor treatment. This study presents a self-assembling strategy for the development of an all-in-one nanomedicine, effectively integrating multiple therapeutic modalities to provide a comprehensive and systemic approach for tumor suppression.
Collapse
Affiliation(s)
- Renjiang Kong
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Jiaqi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Yeyang Wu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Ni Yan
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| | - Hong Cheng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
11
|
Hou DY, You Q, Zhang P, Li XP, Wu JC, Wang Y, You HH, Lv MY, Wu G, Liu X, Guo P, Cheng DB, Chen X, Xu W. Cascade-Activatable Nanoprodrug System Augments Sonochemotherapy of Bladder Cancer. ACS NANO 2024; 18:35507-35519. [PMID: 39686741 DOI: 10.1021/acsnano.4c12967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Sonochemotherapy (SCT) has emerged as a powerful modality for cancer treatment by triggering excessive production of reactive oxygen species (ROS) and controlled release of chemotherapeutic agents under ultrasound. However, achieving spatiotemporally controlled release of chemotherapeutic agents during ROS generation is still an enormous challenge. In this work, we developed a cascade-activated nanoprodrug (CAN) system that utilizes a reversible covalent Schiff base mixed with a hypoxia-activatable camptothecin (CPT) prodrug. Briefly, the designed fluorinated CAN system is self-assembled into nanoparticles under aqueous conditions, which could penetrate deep tumors to offer sufficient oxygen for ultrasound-triggered ROS production. Consequently, the nanoparticles substantially exacerbated the hypoxia of the tumor microenvironment (TME) by elevating oxygen consumption. The aggravated hypoxia in turn served as a positive amplifier to boost the tumor-specific CPT release of Azo-CPT prodrug, which made up for the insufficient treatment efficacy of sonodynamic therapy (SDT). On this basis, we observed a substantial reduction, approximately 3.5-fold, in the half-maximal inhibitory concentration (IC50) of the CAN system compared to that of free CPT in bladder cancer cell lines (T24). Furthermore, the CAN system demonstrated potent antitumor efficacy with reduced side effects, resulting in regression and eradication of T24 tumors in various mouse models. In summary, the CAN system can be easily extended by incorporating different chemotherapeutic agents, showing great potential to revolutionize the clinical management paradigm of bladder cancer.
Collapse
Affiliation(s)
- Da-Yong Hou
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of PET-CT/MRI, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Peng Zhang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Xiang-Peng Li
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Jiong-Cheng Wu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Yueze Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Hui-Hui You
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Mei-Yu Lv
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Gege Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xiao Liu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Pengyu Guo
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Wanhai Xu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| |
Collapse
|
12
|
Víg L, Zátonyi A, Csernyus B, Horváth ÁC, Bojtár M, Kele P, Madarász M, Rózsa B, Fürjes P, Hermann P, Hakkel O, Péter L, Fekete Z. Optically Controlled Drug Delivery Through Microscale Brain-Machine Interfaces Using Integrated Upconverting Nanoparticles. SENSORS (BASEL, SWITZERLAND) 2024; 24:7987. [PMID: 39771721 PMCID: PMC11680031 DOI: 10.3390/s24247987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025]
Abstract
The aim of this work is to incorporate lanthanide-cored upconversion nanoparticles (UCNP) into the surface of microengineered biomedical implants to create a spatially controlled and optically releasable model drug delivery device in an integrated fashion. Our approach enables silicone-based microelectrocorticography (ECoG) implants holding platinum/iridium recording sites to serve as a stable host of UCNPs. Nanoparticles excitable in the near-infrared (lower energy) regime and emitting visible (higher energy) light are utilized in a study. With the upconverted higher energy photons, we demonstrate the induction of photochemical (cleaving) reactions that enable the local release of specific dyes as a model system near the implant. The modified ECoG electrodes can be implanted in brain tissue to act as an uncaging system that releases small amounts of substance while simultaneously measuring the evoked neural response upon light activation. In this paper, several technological challenges like the surface modification of UCNPs, the immobilization of particles on the implantable platform, and measuring the stability of integrated UCNPs in in vitro and in vivo conditions are addressed in detail. Besides the chemical, mechanical, and optical characterization of the ready-to-use devices, the effect of nanoparticles on the original electrophysiological function is also evaluated. The results confirm that silicone-based brain-machine interfaces can be efficiently complemented with UCNPs to facilitate local model drug release.
Collapse
Affiliation(s)
- Levente Víg
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary; (L.V.)
| | - Anita Zátonyi
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary; (L.V.)
| | - Bence Csernyus
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary; (L.V.)
| | - Ágoston C. Horváth
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary; (L.V.)
| | - Márton Bojtár
- Chemical Biology Research Group, Institute of Organic Chemistry, HUN-REN Research Centre for Natural Sciences, H-1117 Budapest, Hungary
| | - Péter Kele
- Chemical Biology Research Group, Institute of Organic Chemistry, HUN-REN Research Centre for Natural Sciences, H-1117 Budapest, Hungary
| | | | - Balázs Rózsa
- BrainVisionCenter, H-1094 Budapest, Hungary; (M.M.); (B.R.)
- HUN-REN Institute of Experimental Medicine, H-1083 Budapest, Hungary
- Two-Photon Measurement Technology Research Group, The Faculty of Information Technology, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | - Péter Fürjes
- Microsystems Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, H-1121 Budapest, Hungary; (P.F.)
| | - Petra Hermann
- Microsystems Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, H-1121 Budapest, Hungary; (P.F.)
| | - Orsolya Hakkel
- Microsystems Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, H-1121 Budapest, Hungary; (P.F.)
| | - László Péter
- Complex Fluid Research Department, Institute of Solid-State Physics and Optics, HUN-REN Wigner Research Centre for Physics, H-1121 Budapest, Hungary;
| | - Zoltán Fekete
- Research Group for Implantable Microsystems, Faculty of Information Technology & Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary; (L.V.)
- Sleep Oscillation Research Group, Institute of Cognitive Neuroscience & Psychology, HUN-REN Research Center for Natural Sciences, H-1117 Budapest, Hungary
| |
Collapse
|
13
|
Yang S, Hu X, Yong Z, Dou Q, Quan C, Cheng HB, Zhang M, Wang J. GSH-responsive bithiophene Aza-BODIPY@HMON nanoplatform for achieving triple-synergistic photoimmunotherapy. Colloids Surf B Biointerfaces 2024; 242:114109. [PMID: 39047644 DOI: 10.1016/j.colsurfb.2024.114109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Photoimmunotherapy represents an innovative approach to enhancing the efficiency of immunotherapy in cancer treatment. This approach involves the fusion of immunotherapy and phototherapy (encompassing techniques like photodynamic therapy (PDT) and photothermal therapy (PTT)). Boron-dipyrromethene (BODIPY) has the potential to trigger immunotherapy owing to its excellent PD and PT efficiency. However, the improvements in water solubility, bioavailability, PD/PT combined efficiency, and tumor tissue targeting of BODIPY require introduction of suitable carriers for potential practical application. Herein, a disulfide bond-based hollow mesoporous organosilica (HMON) with excellent biocompatibility and GSH-responsive degradation properties was used as a carrier to load a bithiophene Aza-BODIPY dye (B5), constructing a sample chemotherapy reagent-free B5@HMON nanoplatform achieving triple-synergistic photoimmunotherapy. HMON, involving disulfide bond, is utilized to improve water solubility, tumor tissue targeting, and PD efficiency by depleting GSH and enhancing host-guest interaction between B5 and HMO. The study reveals that HMON's large specific surface area and porous properties significantly enhance the light collection and oxygen adsorption capacity. The HMON's rich mesoporous structure and internal cavity achieved a loading rate of B5 at 11 %. It was found that the triple-synergistic nanoplatform triggered a stronger anti-tumor immune response, including tumor invasion, cytokine production, calreticulin translocation, and dendritic cell maturation, eliciting specific tumor-specific immunological responses in vivo and in vitro. The BALB/c mouse model with 4T1 tumors was used to assess tumor suppression efficiency in vivo, showing that almost all tumors in the B5@HMON group disappeared after 14 days. Such a simple chemotherapy reagent-free B5@HMON nanoplatform achieved triple-synergistic photoimmunotherapy.
Collapse
Affiliation(s)
- Siao Yang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Xiaoxiao Hu
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Zhengze Yong
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, 15 North Third Ring Road, Beijing 100029, PR China
| | - Qingqing Dou
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Cuilu Quan
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Hong-Bo Cheng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, 15 North Third Ring Road, Beijing 100029, PR China; Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Mo Zhang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, PR China.
| | - Jing Wang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, PR China.
| |
Collapse
|
14
|
Chen X, Yong Z, Xiong Y, Yang H, Xu C, Wang X, Deng Q, Li J, Yang X, Li Z. Hydroxyethyl starch conjugates co-assembled nanoparticles promote photodynamic therapy and antitumor immunity by inhibiting antioxidant systems. Asian J Pharm Sci 2024; 19:100950. [PMID: 39497748 PMCID: PMC11532429 DOI: 10.1016/j.ajps.2024.100950] [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: 04/09/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 11/07/2024] Open
Abstract
Photodynamic therapy (PDT) can produce high levels of reactive oxygen species (ROS) to kill tumor cells and induce antitumor immunity. However, intracellular antioxidant systems, including glutathione (GSH) system and thioredoxin (Trx) system, limit the accumulation of ROS, resulting in compromised PDT and insufficient immune stimulation. Herein, we designed a nanomedicine PtHPs co-loading photosensitizer pyropheophorbide a (PPa) and cisplatin prodrug Pt-COOH(IV) (Pt (IV)) based on hydroxyethyl starch (HES) to inhibit both GSH and Trx antioxidant systems and achieve potent PDT as well as antitumor immune responses. Specifically, HES-PPa and HES-Pt were obtained by coupling HES with PPa and Pt (IV), and assembled into nanoparticle PtHPs by emulsification method to achieve the purpose of co-delivery of PPa and Pt (IV). PtHPs improved PPa photostability while retaining PPa photodynamic properties. In vitro experiments showed that PtHPs reduced GSH, inhibited Trx system and had better cell-killing effect and ROS generation ability. Subcutaneous tumor models showed that PtHPs had good safety and tumor inhibition effect. Bilateral tumor models suggested that PtHPs promoted the release of damage-associated molecular patterns and the maturation of dendritic cells, induced T cell-mediated immune responses, and thus suppressed the growth of both primary and distal tumors. This study reports a novel platinum-based nanomedicine and provides a new strategy for boosting PDT therapy-mediated antitumor immunity by overcoming intrinsic antioxidant systems.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhengtao Yong
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuxuan Xiong
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hai Yang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chen Xu
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xing Wang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingyuan Deng
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiayuan Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zifu Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
15
|
Cao Z, Liu C, Wen J, Lu Y. Innovative Formulation Platform: Paving the Way for Superior Protein Therapeutics with Enhanced Efficacy and Broadened Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403116. [PMID: 38819929 PMCID: PMC11571700 DOI: 10.1002/adma.202403116] [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: 02/29/2024] [Revised: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Protein therapeutics offer high therapeutic potency and specificity; the broader adoptions and development of protein therapeutics, however, have been constricted by their intrinsic limitations such as inadequate stability, immunogenicity, suboptimal pharmacokinetics and biodistribution, and off-target effects. This review describes a platform technology that formulates individual protein molecules with a thin formulation layer of crosslinked polymers, which confers the protein therapeutics with high activity, enhanced stability, controlled release capability, reduced immunogenicity, improved pharmacokinetics and biodistribution, and ability to cross the blood brain barriers. Based on currently approved protein therapeutics, this formulating platform affords the development of a vast family of superior protein therapeutics with improved efficacy and broadened indications at significantly reduced cost.
Collapse
Affiliation(s)
- Zheng Cao
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, 90095, USA
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, UCLA AIDS Institute, University of California, Los Angeles, CA, 90066, USA
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, 90095, USA
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
- Changping Laboratory, Beijing, 100871, P. R. China
| |
Collapse
|
16
|
Liang G, Cao W, Tang D, Zhang H, Yu Y, Ding J, Karges J, Xiao H. Nanomedomics. ACS NANO 2024; 18:10979-11024. [PMID: 38635910 DOI: 10.1021/acsnano.3c11154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Nanomaterials have attractive physicochemical properties. A variety of nanomaterials such as inorganic, lipid, polymers, and protein nanoparticles have been widely developed for nanomedicine via chemical conjugation or physical encapsulation of bioactive molecules. Superior to traditional drugs, nanomedicines offer high biocompatibility, good water solubility, long blood circulation times, and tumor-targeting properties. Capitalizing on this, several nanoformulations have already been clinically approved and many others are currently being studied in clinical trials. Despite their undoubtful success, the molecular mechanism of action of the vast majority of nanomedicines remains poorly understood. To tackle this limitation, herein, this review critically discusses the strategy of applying multiomics analysis to study the mechanism of action of nanomedicines, named nanomedomics, including advantages, applications, and future directions. A comprehensive understanding of the molecular mechanism could provide valuable insight and therefore foster the development and clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Ganghao Liang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wanqing Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|