1
|
Yu J, Hu JR, Tian Y, Lei YM, Hu HM, Lei BS, Zhang G, Sun Y, Ye HR. Nanosensitizer-assisted sonodynamic therapy for breast cancer. J Nanobiotechnology 2025; 23:281. [PMID: 40197318 PMCID: PMC11978163 DOI: 10.1186/s12951-025-03311-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Despite advancements in therapeutic modalities, its prognosis remains poor owing to complex clinical, pathological, and molecular characteristics. Sonodynamic therapy (SDT) is a promising approach for tumor elimination, using sonosensitizers that preferentially accumulate in tumor tissues and are activated by low-intensity ultrasound to produce reactive oxygen species. However, the clinical translation of SDT faces challenges, including the limited efficiency of sonosensitizers and resistance posed by the tumor microenvironment. The emergence of nanomedicine offers innovative strategies to address these obstacles. This review discusses strategies for enhancing the efficacy of SDT using sonosensitizers, including rational structural modifications, improved tumor-targeted enrichment, tumor microenvironment remodeling, and imaging-guided therapy. Additionally, SDT-based multimodal therapies, such as sono-chemotherapy, sono-immunotherapy, and sono-photodynamic therapy, and their potential applications in breast cancer treatment are summarized. The underlying mechanisms of SDT in breast cancer are briefly outlined. Finally, this review highlights current challenges and prospects for the clinical translation of SDT, providing insights into future advancements that may improve therapeutic outcomes for breast cancer.
Collapse
Affiliation(s)
- Jing Yu
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Jun-Rui Hu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yi Tian
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yu-Meng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Hai-Man Hu
- Department of Electrical and Electronic Engineering, Hubei University of Technology, Wuhan, 430068, China
| | - Bing-Song Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| | - Yao Sun
- National Key Laboratory of Green Pesticides, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Hua-Rong Ye
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
| |
Collapse
|
2
|
Zhang C, Shi Y, Zhu Z, Yang T, Wang Y, Hu S, Wu Q, Yang H, Liu J, Zhu WH, Wang Q. Electrostatically Stabilized Light-Activated Membrane Delivery System: Overcoming Membrane Flexibility and Self-Repair to Enhance Tumor Therapy. ACS NANO 2025; 19:12119-12137. [PMID: 40106788 DOI: 10.1021/acsnano.4c18518] [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: 03/22/2025]
Abstract
Cell membrane-coated nanoparticle-based delivery systems often struggle with inevitable drug leakage during the delivery process and inefficient drug release at the tumor site, resulting in unsatisfactory antitumor outcomes. Here, we present an electrostatically stabilized light-activated membrane delivery system (Hybrid membrane nanoparticles, [Hm]@NPs) for leak-free drug delivery, coupled with precisely site-specific and controllable drug release, to elevate cancer treatment. [Hm]@NPs are constructed by encapsulating an aggregation-induced emission (AIE) photosensitizer (Phenalen-1-one-quinoline malonitrile-thiophene tribenamine, Phe-Qui-T) into a positively charged reactive oxygen species (ROS)-responsive polymer (F127-TP-U11) to form a positively charged nanoparticle and then coating it with a negatively charged hybrid membrane containing red blood cell membrane and Panc-1 cell membrane. [Hm]@NPs with high stability effectively prevent drug leakage through electrostatic interaction between the hybrid membrane and nanoparticle. Simultaneously, the photosensitizer Phe-Qui-T with light-controlled ROS generation efficiently destroys both the ROS-responsive polymer and the hybrid membrane, ensuring precise and sufficient drug release while enabling photodynamic therapy (PDT), thereby augmenting antitumor efficacy. [Hm]@NPs show impressive tumor inhibition in pancreatic cancer mouse models, highlighting the potential of this light-controlled membrane-disruption strategy for advanced cell membrane-coated nanodelivery system design.
Collapse
Affiliation(s)
- Cuiyun Zhang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Yiqi Shi
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Zhirong Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P.R. China
| | - Ting Yang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Yuwei Wang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Shanshan Hu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Qi Wu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Haojian Yang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Jihong Liu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Wei-Hong Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
- Center of Photosensitive Chemicals Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Qi Wang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Shanghai Key Laboratory of Functional Materials Chemistry, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| |
Collapse
|
3
|
Cao X, Mao L, Tian Y, Yan L, Geng B, Zhou Y, Zhu J. In situ construction of heterojunctions to regulate the biodegradation behavior of copper carriers for tumor-specific cuproptosis-enhanced sono-immunotherapy. J Nanobiotechnology 2025; 23:246. [PMID: 40128745 PMCID: PMC11934600 DOI: 10.1186/s12951-025-03334-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: 02/09/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Cuproptosis, a novel approach utilizing copper carriers to trigger programmed cell death, exhibits promise for enhancing traditional therapies and activating robust adaptive immune responses. However, the uncontrolled release of Cu ions risks triggering cuproptosis in healthy tissues, potentially causing irreversible damage. To address this, we report on the use of a Cu-MOF (copper metal-organic framework) protective layer to regulate the biodegradation of copper-based nanomaterials. In situ formation of Cu-MOF on Cu2O nanocubes not only stabilizes the material under physiological conditions but also enhances its sonodynamic therapy (SDT) capabilities by establishing a Z-Scheme heterojunction. Upon SDT activation, the targeted Cu ion release at the tumor site triggers a cascade of reactions, generating reactive oxygen species (ROS) via Fenton-like processes and depleting glutathione (GSH). This ROS surge, combined with effective cuproptosis, modulates the immunosuppressive tumor microenvironment, inducing immunogenic cell death to eliminate primary tumors and inhibit metastasis. This study offers a new paradigm for the controlled integration of SDT, chemodynamic therapy (CDT), cuproptosis, and immunotherapy, achieving precise tumor-targeted treatment via controlled copper nanomaterial degradation.
Collapse
Affiliation(s)
- Xiqian Cao
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang Province, 316004, China
| | - Lingwei Mao
- Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Yijun Tian
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Lang Yan
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, 200433, China
| | - Bijiang Geng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Yingtang Zhou
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang Province, 316004, China.
| | - Jiangbo Zhu
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
4
|
Samanta R, Haldar N, Pamecha A, Gajbhiye V. Cell membrane-camouflaged nanocarriers: A cutting-edge biomimetic technology to develop cancer immunotherapy. Int J Pharm 2025; 672:125336. [PMID: 39947362 DOI: 10.1016/j.ijpharm.2025.125336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
The development and growth of many diseases are significantly influenced by immune dysregulation. Similarly, uncontrolled tumor growth occurs in cancer because the immune system is unable to identify and eradicate cancer cells. Therefore, to address this issue, cancer immunotherapy plays a crucial role in detecting tumors and inhibiting their growth. This immune-oncotherapy has gained significant interest over the last decade because of its relevant success in biomedical applications. The fundamental goal of immunotherapy in the war against cancer is to develop potent immunotherapies that have minimal side effects and excellent tumor selectivity. To develop these characteristics, nanotechnology offered promising opportunities for cancer immunotherapy. Cell membrane-coated nanoparticles (CMNPs) have recently evolved, which has a tremendous advantage over other nanoparticles (NPs). The CMNPs can be formed by wrapping cell membranes, which can camouflage the specific cell type, allowing these NPs to survive like "self" during blood circulation and escape immune cell capture. These provide NPs with increased biocompatibility, minimal immunogenicity, longer circulation, and targeted tumor therapy. These advantages have made CMNPs a potential delivery vehicle for immunostimulatory drugs, which can induce immunological responses and lead to cancer immunotherapy. Surface modification of CMNPs using cutting-edge genetic engineering techniques revolutionizes cancer immunotherapy to produce new nano-formulations with greater effectiveness. In this review, we briefly discuss the relationship between cancer and the immune system, various techniques of CMNPs synthesis, and the use of naturally occurring and genetically modified CMNPs for cancer immunotherapy.
Collapse
Affiliation(s)
- Rajkumar Samanta
- Nanobioscience, Agharkar Research Institute, Pune 411004 India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007 India
| | - Niladri Haldar
- Nanobioscience, Agharkar Research Institute, Pune 411004 India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007 India
| | - Anchal Pamecha
- Place of Work, Nanobioscience Group, Agharkar Research Institute, Pune 411004 India
| | - Virendra Gajbhiye
- Nanobioscience, Agharkar Research Institute, Pune 411004 India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007 India.
| |
Collapse
|
5
|
Zhang X, Liu J, Cao Y, Wang W, Lin H, Yu Y. Multi-omic and machine learning analysis of mitochondrial RNA modification genes in lung adenocarcinoma for prognostic and therapeutic implications. Transl Oncol 2025; 53:102306. [PMID: 39908965 PMCID: PMC11847145 DOI: 10.1016/j.tranon.2025.102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related deaths, driven by complex pathogenesis and poor prognosis. Recognizing the pivotal role of mitochondrial RNA modifications (MRM) in cancer progression, this study aims to provide a comprehensive analysis of MRM-related genes and their clinical relevance in lung adenocarcinoma (LUAD). Integrating multi-omic datasets, we systematically explored the molecular features of MRM-related genes across various cancers and identified distinct expression patterns and prognostic associations. Single-cell analysis further reveals MRM-driven cell-cell interactions and pathway activation, particularly in cycling and epithelial cells. Using advanced machine learning techniques, we developed a novel prognostic signature-the Mitochondrial RNA Modification-related Signature (MRMS)-comprising nine genes: TXN, LDHA, HMGA1, SFTPB, KRT8, ALG3, S100A16, HSPD1, and ALDOA. The MRMS demonstrates superior predictive performance for LUAD survival compared to previously reported models. Our findings uniquely link MRMS to increased tumor mutational burden, genetic instability, and an immunosuppressive tumor microenvironment characterized by reduced immune cell infiltration and elevated tumor purity. Additionally, MRMS is associated with immunotherapy-related features, suggesting its potential in predicting treatment response. Experimental validation identified ALG3 as an oncogenic driver in LUAD, influencing tumor cell proliferation, migration, and invasion. In conclusion, this study establishes MRMS as a robust prognostic biomarker and highlights its dual role in shaping the tumor immune microenvironment and guiding therapeutic strategies. These findings provide novel insights into mitochondrial RNA modifications and their potential applications in personalized treatment for LUAD.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Jiatao Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Yaolin Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Haoran Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Yue Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
6
|
Liang J, Cheng G, Qiu L, Xue L, Xu H, Qiao X, Guo N, Xiang H, Chen Y, Ding H. Activatable Sulfur Dioxide Nanosonosensitizer Enables Precisely Controllable Sono-Gaseous Checkpoint Trimodal Therapy for Orthotopic Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409442. [PMID: 39679828 PMCID: PMC11791957 DOI: 10.1002/advs.202409442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/28/2024] [Indexed: 12/17/2024]
Abstract
Immune checkpoint blockade (ICB) is combined with sonodynamic therapy (SDT) to increase response rates and enhance anticancer efficacy. However, the "always on" property of most sonosensitizers in reducing tumor microenvironment (TME) compromises the therapeutic outcome of sonoimmunotherapy and exacerbates adverse side effects. Precisely controllable strategies combining sulfur dioxide (SO2) gas therapy with cancer immunotherapy can address these issues but remain lacking. Herein an "activatable SO2 nanosonosensitizer" for precise sono-gaseous checkpoint trimodal therapy of orthotopic hepatocellular carcinoma (HCC) is reported, whose full activity is initiated by ultrasound (US) irradiation in the reducing TME. This "activatable SO2 nanosonosensitizer," Aza-DNBS nanoparticles (NPs), are established by self-assembling Aza-boron-dipyrromethene based sonosensitizer molecules and 2,4-dinitrobenzenesulfonate (DNBS)-caged SO2 prodrug. The activity of Aza-DNBS NPs is initially silenced, and the sonodynamic, gaseous, and immunosuppressive TME reprogramming activities are precisely awakened under US irradiation. Due to the glutathione-responsiveness of Aza-DNBS NPs, Aza-DNBS NPs can generate large amounts of SO2 for gas therapy-enhanced SDT, which triggers robust immunogenic cell death activation and reprogramming of the immunosuppressive TME, thereby significantly suppressing orthotopic tumor growth and delaying lung metastasis. Thus, this study represents a strategy for designing a generic nanoplatform for precisely combined immunotherapy of orthotopic HCC.
Collapse
Affiliation(s)
- Jing Liang
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Guangwen Cheng
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Luping Qiu
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Liyun Xue
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Huning Xu
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Xiaohui Qiao
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| | - Na Guo
- Department of PathologyZhejiang Cancer HospitalHangzhouZhejiang310022China
| | - Huijing Xiang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai2000444China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai2000444China
| | - Hong Ding
- Department of UltrasoundHuashan HospitalFudan UniversityShanghai200040China
| |
Collapse
|
7
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [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: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
8
|
Zhang X, Cao Y, Liu J, Wang W, Yan Q, Wang Z. Comprehensive Analysis of m6A-Related Programmed Cell Death Genes Unveils a Novel Prognostic Model for Lung Adenocarcinoma. J Cell Mol Med 2025; 29:e70255. [PMID: 39828988 PMCID: PMC11743404 DOI: 10.1111/jcmm.70255] [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: 09/14/2024] [Revised: 10/25/2024] [Accepted: 11/21/2024] [Indexed: 01/22/2025] Open
Abstract
Lung adenocarcinoma (LUAD) involves complex dysregulated cellular processes, including programmed cell death (PCD), influenced by N6-methyladenosine (m6A) RNA modification. This study integrates bulk RNA and single-cell sequencing data to identify 43 prognostically valuable m6A-related PCD genes, forming the basis of a 13-gene risk model (m6A-related PCD signature [mPCDS]) developed using machine-learning algorithms, including CoxBoost and SuperPC. The mPCDS demonstrated significant predictive performance across multiple validation datasets. In addition to its prognostic accuracy, mPCDS revealed distinct genomic profiles, pathway activations, associations with the tumour microenvironment and potential for predicting drug sensitivity. Experimental validation identified RCN1 as a potential oncogene driving LUAD progression and a promising therapeutic target. The mPCDS offers a new approach for LUAD risk stratification and personalised treatment strategies.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yaolin Cao
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jiatao Liu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Wei Wang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Qiuyue Yan
- Department of Respiratory DiseasesThe Affiliated Huai'an Hospital of Xuzhou Medical UniversityHuai'anJiangsuChina
| | - Zhibo Wang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
9
|
Yao J, Cui Z, Zhang F, Li H, Tian L. Biomaterials enhancing localized cancer therapy activated anti-tumor immunity: a review. J Mater Chem B 2024; 13:117-136. [PMID: 39544081 DOI: 10.1039/d4tb01995d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Localized cancer therapies such as radiotherapy, phototherapy, and chemotherapy are precise cancer treatment strategies aimed at minimizing systemic side effects. However, cancer metastasis remains the primary cause of mortality among cancer patients in clinical settings, and localized cancer treatments have limited efficacy against metastatic cancer. Therefore, researchers are exploring strategies that combine localized therapy with immunotherapy to activate robust anti-tumor immune responses, thereby eradicating metastatic cancer. Biomaterials, as novel materials, exhibit great potential in biomedical applications and have achieved great progress in clinic translation. This review introduces biomaterials and their applications in research focused on enhancing localized cancer treatment activated anti-tumor immunity. Additionally, the current challenges and future directions of biomaterials are also discussed, providing insights and references for related research.
Collapse
Affiliation(s)
- Jipeng Yao
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Zhencun Cui
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- Department of Nuclear Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Feifei Zhang
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Haidong Li
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Longlong Tian
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| |
Collapse
|
10
|
Zhao J, Zhang H, Zhao Y, Lin Z, Lin F, Wang Z, Mo Q, Lu G, Zhao G, Wang G. Exploratory Research for HIF-1α Overexpression Tumor Antigen in the Activation of Dendritic Cells and the Potent Anti-Tumor Immune Response. Cancer Manag Res 2024; 16:1813-1822. [PMID: 39713567 PMCID: PMC11662640 DOI: 10.2147/cmar.s482363] [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: 07/12/2024] [Accepted: 11/24/2024] [Indexed: 12/24/2024] Open
Abstract
Background Tumor-specific antigens play an important role in dendritic cell (DC)-based immunotherapy. The acquisition of tumor-specific antigens, which are essential for DC-based immunotherapy, poses a significant challenge. This study aimed to explore the efficacy of hypoxia inducible factor-1α (HIF-1α) overexpression tumor antigens in DC-based immunotherapy. Methods An HIF-1α over-expression cell line was constructed to prepare HIF-1α overexpression tumor antigens. The expression of CD14, CD40, CD80, CD86, and HLA-DR on the surface of dendritic cells derived from monocytes was assessed using flow cytometry after stimulation with tumor antigens enriched in HIF-1α. T cell proliferation was analyzed by CFSE division following incubation with mature DCs. The apoptotic tumor cells were detected through annexin V/PI staining following coculture with dendritic cells (DCs) stimulated by HIF-1α enriched antigens. The detection of damage-associated molecular pattern molecules (DAMPs) HMGB1 and calreticulin (CALR) was performed using Western blotting. Results The results demonstrated that HIF-1α-enriched tumor antigens significantly upregulated the expression of CD40, CD80, CD86, and HLA-DR in DCs compared to normal tumor antigens. Furthermore, co-incubation with HIF-1α-enriched tumor antigen-activated DCs enhanced T cell proliferation and stimulated the T cell-mediated cytotoxicity. Notably, the expression of DAMPs, such as HMGB1 and CALR, was elevated in HIF-1α-enriched tumor antigens. Conclusion Our findings demonstrate that tumor antigens enriched with HIF-1α may encompass tumor-specific antigens capable of stimulating DC activation, thereby enhancing T cell proliferation and cytotoxicity. These results provide support for the further advancement of HIF-1α enriched tumor antigens in preclinical and clinical investigations pertaining to tumor treatment.
Collapse
Affiliation(s)
- Jinjin Zhao
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
- Key Laboratory of Nano-Drug Delivery System Construction and Application in Xinxiang City, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Haiguang Zhang
- Department of Gynecology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Yilin Zhao
- Department of Cardiology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Zhiqiang Lin
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Fei Lin
- Department of Cardiology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
- Cardiovascular Repair Engineering Technology Research Center, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Zhiyin Wang
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Qingjiang Mo
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Guangjian Lu
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Guoan Zhao
- Department of Cardiology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
- Cardiovascular Repair Engineering Technology Research Center, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Guoqiang Wang
- Clinical Laboratory, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| |
Collapse
|
11
|
Zhao H, Du F, Xiang X, Tang Y, Feng Z, Wang Z, Rong X, Qiu L. Progress in application of nanomedicines for enhancing cancer sono-immunotherapy. ULTRASONICS SONOCHEMISTRY 2024; 111:107105. [PMID: 39427436 PMCID: PMC11533716 DOI: 10.1016/j.ultsonch.2024.107105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/22/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Cancer immunotherapy has significant potential as a cancer treatment since it boosts the immune system and prevents immune escape to get rid of or fight cancers. However, its clinical applicability is still limited because of the low response rate and immune-related side effects. Recently ultrasound has been shown to alter the tumor immune microenvironment, enhance the effectiveness of other antitumor therapies, and cause tumors to become more sensitive to immunotherapy, thus providing new insights into cancer treatment. Nanomedicines are also anticipated to have a positive impact on improving the immunological effects and enhancing ultrasound effect for cancer therapy. Therefore, designing effective nanomedicines enhanced ultrasound effect for augmenting sono-immunotherapy has been a pivot on anticancer therapy. In this review, the immunological impacts of various ultrasound therapeutic modalities, ultrasound parameters, and their underlying mechanisms are discussed. Moreover, we highlight the recent progress of nanomedicines synergistically enhancing sono-immunotherapy. Finally, we put forward opportunities and challenges on sono-immunotherapy.
Collapse
Affiliation(s)
- Hongxin Zhao
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fangxue Du
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xi Xiang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Tang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyan Feng
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyao Wang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiao Rong
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Li Qiu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
12
|
Wang X, Wang H, Li Y, Sun Z, Liu J, Sun C, Cao X. Engineering macrophage membrane-camouflaged nanoplatforms with enhanced macrophage function for mediating sonodynamic therapy of ovarian cancer. NANOSCALE 2024; 16:19048-19061. [PMID: 39310965 DOI: 10.1039/d4nr01307g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Cancer immunotherapy has demonstrated remarkable efficacy in the treatment of cancer, and it has been successfully applied in the treatment of various solid tumors. However, the response rates to immunotherapy in patients with ovarian cancer remain modest because of the immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages (TAMs) represent the predominant myeloid cell population within the TME, which adopt the protumorigenic M2 phenotype and are blinded by the "don't eat me" signals from tumor cells. These characteristics of TAMs result in insufficient phagocytic activation. In this study, we constructed a SIM@TR-NP-mediated combination therapy of sonodynamic and immunotherapy. SIM@TR-NPs were modified by engineered macrophage membranes with overexpressed sialic acid-binding Ig-like lectin 10 (Siglec-10), and were internally loaded with sonosensitizer 4,4',4'',4'''-(porphine-5,10,15,20-tetrayl)tetrakis(benzoic acid) and immune adjuvant resiquimod. SIM@TR-NPs can block "don't eat me" signals to enhance macrophage phagocytosis and trigger the polarization of TAMs toward the M1 phenotype, thereby improving the immunosuppressive TME. Simultaneously, upon ultrasound irradiation, SIM@TR-NP-mediated sonodynamic therapy (SDT) triggered immunogenic cell death in tumor cells, in combination with TAM-based immunotherapy, transforming the "immune cold tumor" into an "immune hot tumor". SIM@TR-NP-mediated sonodynamic immunotherapy exhibited potent antitumor efficacy in ovarian cancer and exhibited substantial potential for improving the immunosuppressive TME. This study presents an emerging therapeutic regimen for ovarian cancer that synergizes TAM-based antitumor immunotherapy and SDT.
Collapse
Affiliation(s)
- Xiaofei Wang
- Yantai Yuhuangding hospital, Shandong University, Yantai, 264000, P.R. China.
| | - Hongling Wang
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P.R. China.
| | - Yansheng Li
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P.R. China.
| | - Zhihong Sun
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P.R. China.
| | - Jie Liu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P.R. China.
| | - Chengming Sun
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P.R. China.
| | - Xiaoli Cao
- Yantai Yuhuangding hospital, Shandong University, Yantai, 264000, P.R. China.
| |
Collapse
|
13
|
Chen Z, Chen L, Lyu TD, Weng S, Xie Y, Jin Y, Wu O, Jones M, Kwan K, Makvnadi P, Li B, Sharopov F, Ma C, Li H, Wu A. Targeted mitochondrial nanomaterials in biomedicine: Advances in therapeutic strategies and imaging modalities. Acta Biomater 2024; 186:1-29. [PMID: 39151665 DOI: 10.1016/j.actbio.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/15/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
Mitochondria, pivotal organelles crucial for energy generation, apoptosis regulation, and cellular metabolism, have spurred remarkable advancements in targeted material development. This review surveys recent breakthroughs in targeted mitochondrial nanomaterials, illuminating their potential in drug delivery, disease management, and biomedical imaging. This review approaches from various application perspectives, introducing the specific applications of mitochondria-targeted materials in cancer treatment, probes and imaging, and diseases treated with mitochondria as a therapeutic target. Addressing extant challenges and elucidating potential therapeutic mechanisms, it also outlines future development trajectories and obstacles. By comprehensively exploring the diverse applications of targeted mitochondrial nanomaterials, this review aims to catalyze innovative treatment modalities and diagnostic approaches in medical research. STATEMENT OF SIGNIFICANCE: This review presents the latest advancements in mitochondria-targeted nanomaterials for biomedical applications, covering diverse fields such as cancer therapy, bioprobes, imaging, and the treatment of various systemic diseases. The novelty and significance of this work lie in its systematic analysis of the intricate relationship between mitochondria and different diseases, as well as the ingenious design strategies employed to harness the therapeutic potential of nanomaterials. By providing crucial insights into the development of mitochondria-targeted nanomaterials and their applications, this review offers a valuable resource for researchers working on innovative treatment modalities and diagnostic approaches. The scientific impact and interest to the readership lie in the identification of promising avenues for future research and the potential for clinical translation of these cutting-edge technologies.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China
| | - Linjie Chen
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Tai Dong Lyu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Shoutao Weng
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Yihao Xie
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Yuxin Jin
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Ouqiang Wu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Morgan Jones
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham B31 2AP, UK
| | - Kenny Kwan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Pooyan Makvnadi
- University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India; Centre of Research Impact and Outreach, Chitkara University, Rajpura, Punjab 140417, India
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College Soochow University, PR China
| | - Farukh Sharopov
- V.I. Nikitin Chemistry Institute of Tajikistan National Academy of Sciences, Dushanbe 734063, Tajikistan
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, PR China
| | - Huaqiong Li
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China.
| | - Aimin Wu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China.
| |
Collapse
|
14
|
Ma C, Cheng Z, Tan H, Wang Y, Sun S, Zhang M, Wang J. Nanomaterials: leading immunogenic cell death-based cancer therapies. Front Immunol 2024; 15:1447817. [PMID: 39185425 PMCID: PMC11341423 DOI: 10.3389/fimmu.2024.1447817] [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: 06/12/2024] [Accepted: 07/24/2024] [Indexed: 08/27/2024] Open
Abstract
The field of oncology has transformed in recent years, with treatments shifting from traditional surgical resection and radiation therapy to more diverse and customized approaches, one of which is immunotherapy. ICD (immunogenic cell death) belongs to a class of regulatory cell death modalities that reactivate the immune response by facilitating the interaction between apoptotic cells and immune cells and releasing specific signaling molecules, and DAMPs (damage-associated molecular patterns). The inducers of ICD can elevate the expression of specific proteins to optimize the TME (tumor microenvironment). The use of nanotechnology has shown its unique potential. Nanomaterials, due to their tunability, targeting, and biocompatibility, have become powerful tools for drug delivery, immunomodulators, etc., and have shown significant efficacy in clinical trials. In particular, these nanomaterials can effectively activate the ICD, trigger a potent anti-tumor immune response, and maintain long-term tumor suppression. Different types of nanomaterials, such as biological cell membrane-modified nanoparticles, self-assembled nanostructures, metallic nanoparticles, mesoporous materials, and hydrogels, play their respective roles in ICD induction due to their unique structures and mechanisms of action. Therefore, this review will explore the latest advances in the application of these common nanomaterials in tumor ICD induction and discuss how they can provide new strategies and tools for cancer therapy. By gaining a deeper understanding of the mechanism of action of these nanomaterials, researchers can develop more precise and effective therapeutic approaches to improve the prognosis and quality of life of cancer patients. Moreover, these strategies hold the promise to overcome resistance to conventional therapies, minimize side effects, and lead to more personalized treatment regimens, ultimately benefiting cancer treatment.
Collapse
Affiliation(s)
- Changyu Ma
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Beijing, China
| | - Zhe Cheng
- Department of Forensic Medicine, Harbin Medical University, Harbin, China
| | - Haotian Tan
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Beijing, China
| | - Yihan Wang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Clinical College, Peking University Health Science Center, Beijing, China
| | - Shuzhan Sun
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Clinical College, Peking University Health Science Center, Beijing, China
| | - Mingxiao Zhang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
| | - Jianfeng Wang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
15
|
Tousian B, Khosravi AR, Ghasemi MH, Kadkhodaie M. Biomimetic functionalized metal organic frameworks as multifunctional agents: Paving the way for cancer vaccine advances. Mater Today Bio 2024; 27:101134. [PMID: 39027676 PMCID: PMC11255118 DOI: 10.1016/j.mtbio.2024.101134] [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: 04/06/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Biomimetic functionalized metal-organic frameworks (Fn-MOFs) represent a cutting-edge approach in the realm of cancer vaccines. These multifunctional agents, inspired by biological systems, offer unprecedented opportunities for the development of next-generation cancer vaccines. The vast surface area, tunable pore size, and diverse chemistry of MOFs provide a versatile scaffold for the encapsulation and protection of antigenic components, crucial for vaccine stability and delivery. This work delves into the innovative design and application of Fn-MOFs, highlighting their role as carriers for immune enhancement and their potential to revolutionize vaccine delivery. By mimicking natural processes, Fn-MOFs, with their ability to be functionalized with a myriad of chemical and biological entities, exhibit superior biocompatibility and stimuli-responsive behavior and facilitate targeted delivery to tumor sites. This review encapsulates the latest advancements in Fn-MOF technology, from their synthesis and surface modification to their integration into stimuli-responsive and combination therapies. It underscores the significance of biomimetic approaches in overcoming current challenges in cancer vaccine development, such as antigen stability and immune evasion. By leveraging the biomimetic nature of Fn-MOFs, this work paves the way for innovative strategies in cancer vaccines, aiming to induce potent and long-lasting immune responses against malignancies.
Collapse
Affiliation(s)
- Bushra Tousian
- Department of Microbiology and Immunology, Veterinary Medicine Faculty, University of Tehran, PO Box 1419963111, Tehran, Iran
| | - Ali Reza Khosravi
- Department of Microbiology and Immunology, Veterinary Medicine Faculty, University of Tehran, PO Box 1419963111, Tehran, Iran
| | - Mohammad Hadi Ghasemi
- Applied Chemistry Research Group, ACECR-Tehran Organization, PO Box 13145-186, Tehran, Iran
| | - Majid Kadkhodaie
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
16
|
Rix A, Heinrichs H, Porte C, Leenaars C, Bleich A, Kiessling F. Ultrasound-induced immune responses in tumors: A systematic review and meta-analysis. J Control Release 2024; 371:146-157. [PMID: 38777126 DOI: 10.1016/j.jconrel.2024.05.030] [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: 02/16/2024] [Revised: 04/29/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Ultrasound is widely used in the diagnosis and therapy of cancer. Tumors can be treated by thermal or mechanical tissue ablation. Furthermore, tumors can be manipulated by hyperthermia, sonodynamic therapy and sonoporation, e.g., by increasing tumor perfusion or the permeability of biological barriers to enhance drug delivery. These treatments induce various immune responses in tumors. However, conflicting data and high heterogeneity between experimental settings make it difficult to generalize the effects of ultrasound on tumor immunity. Therefore, we performed a systematic review to answer the question: "Does ultrasound alter the immune reaction of peripheral solid tumors in humans and animals compared to control conditions without ultrasound?" A systematic literature search was performed in PubMed, EMBASE, and Web of Science and 24,401 potentially relevant publications were identified. Of these, 96 publications were eligible for inclusion in the systematic review. Experiments were performed in humans, rats, and mice and focused on different tumor types, primarily breast and melanoma. We collected data on thermal and non-thermal ultrasound settings, the use of sono-sensitizers or sono-enhancers, and anti-tumor therapies. Six meta-analyses were performed to quantify the effect of ultrasound on tumor infiltration by T cells (cytotoxic, helper, and regulatory T cells) and on blood cytokines (interleukin-6, interferon-γ, tumor necrosis factor-α). We provide robust scientific evidence that ultrasound alters T cell infiltration into tumors and increases blood cytokine concentrations. Furthermore, we identified significant differences in immune cell infiltration based on tumor type, ultrasound settings, and mouse age. Stronger effects were observed using hyperthermia in combination with sono-sensitizers and in young mice. The latter may impair the translational impact of study results as most cancer patients are older. Thus, our results may help refining ultrasound parameters to enhance anti-tumor immune responses for therapeutic use and to minimize immune effects in diagnostic applications.
Collapse
Affiliation(s)
- Anne Rix
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Helen Heinrichs
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Céline Porte
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Cathalijn Leenaars
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Medical Faculty, RWTH Aachen University, Aachen, Germany; Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany.
| |
Collapse
|
17
|
Rahmat JN, Liu J, Chen T, Li Z, Zhang Y. Engineered biological nanoparticles as nanotherapeutics for tumor immunomodulation. Chem Soc Rev 2024; 53:5862-5903. [PMID: 38716589 DOI: 10.1039/d3cs00602f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Biological nanoparticles, or bionanoparticles, are small molecules manufactured in living systems with complex production and assembly machinery. The products of the assembly systems can be further engineered to generate functionalities for specific purposes. These bionanoparticles have demonstrated advantages such as immune system evasion, minimal toxicity, biocompatibility, and biological clearance. Hence, bionanoparticles are considered the new paradigm in nanoscience research for fabricating safe and effective nanoformulations for therapeutic purposes. Harnessing the power of the immune system to recognize and eradicate malignancies is a viable strategy to achieve better therapeutic outcomes with long-term protection from disease recurrence. However, cancerous tissues have evolved to become invisible to immune recognition and to transform the tumor microenvironment into an immunosuppressive dwelling, thwarting the immune defense systems and creating a hospitable atmosphere for cancer growth and progression. Thus, it is pertinent that efforts in fabricating nanoformulations for immunomodulation are mindful of the tumor-induced immune aberrations that could render cancer nanotherapy inoperable. This review systematically categorizes the immunosuppression mechanisms, the regulatory immunosuppressive cellular players, and critical suppressive molecules currently targeted as breakthrough therapies in the clinic. Finally, this review will summarize the engineering strategies for affording immune moderating functions to bionanoparticles that tip the tumor microenvironment (TME) balance toward cancer elimination, a field still in the nascent stage.
Collapse
Affiliation(s)
- Juwita N Rahmat
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117585, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yong Zhang
- Department of Biomedical Engineering, College of Engineering, The City University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
18
|
Chen W, Tang C, Chen G, Li J, Li N, Zhang H, Di L, Wang R. Boosting Checkpoint Immunotherapy with Biomimetic Nanodrug Delivery Systems. Adv Healthc Mater 2024; 13:e2304284. [PMID: 38319961 DOI: 10.1002/adhm.202304284] [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: 12/04/2023] [Revised: 01/26/2024] [Indexed: 02/08/2024]
Abstract
Immune checkpoint blockade (ICB) has achieved unprecedented progress in tumor immunotherapy by blocking specific immune checkpoint molecules. However, the high biodistribution of the drug prevents it from specifically targeting tumor tissues, leading to immune-related adverse events. Biomimetic nanodrug delivery systems (BNDSs) readily applicable to ICB therapy have been widely developed at the preclinical stage to avoid immune-related adverse events. By exploiting or mimicking complex biological structures, the constructed BNDS as a novel drug delivery system has good biocompatibility and certain tumor-targeting properties. Herein, the latest findings regarding the aforementioned therapies associated with ICB therapy are highlighted. Simultaneously, prospective bioinspired engineering strategies can be designed to overcome the four-level barriers to drug entry into lesion sites. In future clinical translation, BNDS-based ICB combination therapy represents a promising avenue for cancer treatment.
Collapse
Affiliation(s)
- Wenjing Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Chenlu Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Guijin Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Hanwen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| |
Collapse
|
19
|
Yang Y, Wang N, Yan F, Shi Z, Feng S. Metal-organic frameworks as candidates for tumor sonodynamic therapy: Designable structures for targeted multifunctional transformation. Acta Biomater 2024; 181:67-97. [PMID: 38697383 DOI: 10.1016/j.actbio.2024.04.037] [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: 01/27/2024] [Revised: 03/25/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Sonodynamic therapy (SDT), utilizing ultrasound (US) as the trigger, has gained popularity recently as a therapeutic approach with significant potential for treating various diseases. Metal-organic frameworks (MOFs), characterized by structural flexibility, are prominently emerging in the SDT realm as an innovative type of sonosensitizer, offering functional tunability and biocompatibility. However, due to the inherent limitations of MOFs, such as low reactivity to reactive oxygen species and challenges posed by the complex tumor microenvironment, MOF-based sonosensitizers with singular functions are unable to demonstrate the desired therapeutic efficacy and may pose risks of toxicity, limiting their biological applications to superficial tissues. MOFs generally possess distinctive crystalline structures and properties, and their controlled coordination environments provide a flexible platform for exploring structure-effect relationships and guiding the design and development of MOF-based nanomaterials to unlock their broader potential in biological fields. The primary focus of this paper is to summarize cases involving the modification of different MOF materials and the innovative strategies developed for various complex conditions. The paper outlines the diverse application areas of functionalized MOF-based sonosensitizers in tumor synergistic therapies, highlighting the extensive prospects of SDT. Additionally, challenges confronting SDT are briefly summarized to stimulate increased scientific interest in the practical application of MOFs and the successful clinical translation of SDT. Through these discussions, we strive to foster advancements that lead to early-stage clinical benefits for patients. STATEMENT OF SIGNIFICANCE: 1. An overview for the progresses in SDT explored from a novel and fundamental perspective. 2. Different modification strategies to improve the MOFs-mediated SDT efficacy are provided. 3. Guidelines for the design of multifunctional MOFs-based sonosensitizers are offered. 4. Powerful tumor ablation potential is reflected in SDT-led synergistic therapies. 5. Future challenges in the field of MOFs-based SDT in clinical translation are suggested.
Collapse
Affiliation(s)
- Yilin Yang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Ning Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China.
| | - Zhan Shi
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China.
| | - Shouhua Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| |
Collapse
|
20
|
Jiao X, Li X, Du Y, Cong Y, Yang S, Chen D, Zhang T, Feng M, Hong H. Positron emission tomography guided synergistic treatment of melanoma using multifunctional zirconium-hematoporphyrin nanosonosensitizers. J Control Release 2024; 370:95-109. [PMID: 38642859 DOI: 10.1016/j.jconrel.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/01/2024] [Accepted: 04/18/2024] [Indexed: 04/22/2024]
Abstract
Sonodynamic therapy (SDT) has emerged as a useful approach for tumor treatment. However, its widespread application is impeded by poor pharmacokinetics of existing sonosensitizers. Here we developed a metal-organic nanoplatform, wherein a small-molecule sonosensitizer (hematoporphyrin monomethyl ether, HMME) was ingeniously coordinated with zirconium, resulting in a multifunctional nanosonosensitizer termed Zr-HMME. Through post-synthetic modifications involving PEGylation and tumor-targeting peptide (F3) linkage, a nanoplatform capable of homing on melanoma was produced, which could elicit robust immune responses to suppress tumor lung metastasis in the host organism. Importantly, after seamless incorporation of positron-emitting 89Zr into this nanosonosensitizer, positron emission tomography (PET) could be used to monitor its in vivo pharmacokinetics. PET imaging studies revealed that this nanoplatform exhibited potent tumor accumulation and strong in vivo stability. Using intrinsic fluorescence from HMME, a dual-modal diagnostic capability (fluorescence and PET) was confirmed for this nanosonosensitizer. In addition, the mechanisms of how this nanoplatform interacted with immune system were also investigated. The collective data proved that the coordination structure between small-molecule drug cargos and metals may enhance the functions of each other while mitigating their weaknesses. This straightforward approach can expand the potential applications of suitable drug molecules.
Collapse
Affiliation(s)
- Xiaodan Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Xiaoyang Li
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Yan Du
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Yiyang Cong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Shuyang Yang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Daiqin Chen
- Innovation Academy for Precision Measurement Science and Technology, China Academy of Sciences, West No.30 Xiao Hong Shan, Wuhan 430071, China
| | - Tao Zhang
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Translation Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Min Feng
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing 210093, China.
| |
Collapse
|
21
|
Liu B, Du F, Feng Z, Xiang X, Guo R, Ma L, Zhu B, Qiu L. Ultrasound-augmented cancer immunotherapy. J Mater Chem B 2024; 12:3636-3658. [PMID: 38529593 DOI: 10.1039/d3tb02705h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Cancer is a growing worldwide health problem with the most broadly studied treatments, in which immunotherapy has made notable advancements in recent years. However, innumerable patients have presented a poor response to immunotherapy and simultaneously experienced immune-related adverse events, with failed therapeutic results and increased mortality rates. Consequently, it is crucial to develop alternate tactics to boost therapeutic effects without producing negative side effects. Ultrasound is considered to possess significant therapeutic potential in the antitumor field because of its inherent characteristics, including cavitation, pyrolysis, and sonoporation. Herein, this timely review presents the comprehensive and systematic research progress of ultrasound-enhanced cancer immunotherapy, focusing on the various ultrasound-related mechanisms and strategies. Moreover, this review summarizes the design and application of current sonosensitizers based on sonodynamic therapy, with an attempt to provide guidance on new directions for future cancer therapy.
Collapse
Affiliation(s)
- Bingjie Liu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Fangxue Du
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ziyan Feng
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xi Xiang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ruiqian Guo
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lang Ma
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bihui Zhu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Li Qiu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
22
|
Wang S, Wang R, Hu D, Zhang C, Cao P, Huang J. Machine learning reveals diverse cell death patterns in lung adenocarcinoma prognosis and therapy. NPJ Precis Oncol 2024; 8:49. [PMID: 38409471 PMCID: PMC10897292 DOI: 10.1038/s41698-024-00538-5] [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: 07/13/2023] [Accepted: 02/08/2024] [Indexed: 02/28/2024] Open
Abstract
Cancer cell growth, metastasis, and drug resistance pose significant challenges in the management of lung adenocarcinoma (LUAD). However, there is a deficiency in optimal predictive models capable of accurately forecasting patient prognoses and guiding the selection of targeted treatments. Programmed cell death (PCD) pathways play a pivotal role in the development and progression of various cancers, offering potential as prognostic indicators and drug sensitivity markers for LUAD patients. The development and validation of predictive models were conducted by integrating 13 PCD patterns with comprehensive analysis of bulk RNA, single-cell RNA transcriptomics, and pertinent clinicopathological details derived from TCGA-LUAD and six GEO datasets. Utilizing the machine learning algorithms, we identified ten critical differentially expressed genes associated with PCD in LUAD, namely CHEK2, KRT18, RRM2, GAPDH, MMP1, CHRNA5, TMPRSS4, ITGB4, CD79A, and CTLA4. Subsequently, we conducted a programmed cell death index (PCDI) based on these genes across the aforementioned cohorts and integrated this index with relevant clinical features to develop several prognostic nomograms. Furthermore, we observed a significant correlation between the PCDI and immune features in LUAD, including immune cell infiltration and the expression of immune checkpoint molecules. Additionally, we found that patients with a high PCDI score may exhibit resistance to immunotherapy and standard adjuvant chemotherapy regimens; however, they may benefit from other FDA-supported drugs such as docetaxel and dasatinib. In conclusion, the PCDI holds potential as a prognostic signature and can facilitate personalized treatment for LUAD patients.
Collapse
Affiliation(s)
- Shun Wang
- Department of Respiratory Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, China
| | - Ruohuang Wang
- Department of Otolaryngology, the Second Affiliated Hospital of the Naval Military Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Dingtao Hu
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China
| | - Caoxu Zhang
- Department of Molecular Diagnostics, The Core Laboratory in Medical Center of Clinical Research, Department of Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Peng Cao
- Department of Interventional Pulmonology, Anhui Chest Hospital, Hefei, Anhui, 230022, China
| | - Jie Huang
- Department of Respiratory Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
23
|
Wang S, Liu Y, Quan C, Luan S, Shi H, Wang L. A metal-organic framework-integrated composite for piezocatalysis-assisted tumour therapy: design, related mechanisms, and recent advances. Biomater Sci 2024; 12:896-906. [PMID: 38234222 DOI: 10.1039/d3bm01944f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
With the growing need for more effective tumour treatment, piezocatalytic therapy has emerged as a promising approach due to its distinctive capacities to generate ROS through stress induction and regulate the hypoxic state of the TME. MOF-based piezocatalysts not only possess the benefits of piezocatalysis but also exhibit several advantages associated with MOFs, such as tunable pore size, large specific surface area, and good biocompatibility. Therefore, they are expected to become a powerful promoter of piezocatalytic therapy. This review elaborates on the fundamental principles of piezocatalysis and summarises recent advances in the piezocatalytic therapy and combination therapies of tumours, generalising the strategies for constructing piezocatalytic systems based on MOFs. Finally, the challenges confronted and future opportunities for the design and application of piezocatalytic MOF anticancer systems have been discussed.
Collapse
Affiliation(s)
- Shuteng Wang
- State Key Laboratory of Polymer Physics and Chemistry, 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
| | - Yifan Liu
- State Key Laboratory of Polymer Physics and Chemistry, 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
| | - Chunhua Quan
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002, P. R. China.
| | - Shifang Luan
- State Key Laboratory of Polymer Physics and Chemistry, 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
| | - Hengchong Shi
- State Key Laboratory of Polymer Physics and Chemistry, 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
| | - Lei Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
| |
Collapse
|
24
|
Duan Q, Li H, Xue J, Zhang Q, Gao J, Wang X, Zhang Q, Guo X, Guo L, Li P, Wang X, Sang S, Xi Y. Effective Combination of Targeted Therapies with Sonodynamic Treatment for Use in Exploring Differences in Therapeutic Efficacy across Organelle Targets. Mol Pharm 2024; 21:760-769. [PMID: 38175712 DOI: 10.1021/acs.molpharmaceut.3c00899] [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: 01/05/2024]
Abstract
Acoustic kinetic therapy systems that target specific organelles can improve the precision of a sonosensitizer, which is a perfect combination of targeted therapy and sonodynamic therapy (SDT) and plays an important role in current acoustic kinetic therapy. In this study, we loaded PpIX, a sonosensitizer, on targeted-functional carbon dots (CDs) via an amide reaction and then generated the mitochondria-targeted system (Mit-CDs-PpIX) and nucleus-targeted system (Nuc-CDs-PpIX), respectively, to deliver the sonosensitizer. Both systems exhibited minimal cytotoxicity in the absence of ultrasound stimulation. The efficacy of the targeted SDT systems was investigated using methylthiazol tetrazolium (MTT) assays, live/dead staining, flow cytometry, etc. Compared with the free PpIX and mitochondria-targeted system, the nucleus-targeted system is more potent in killing effect under ultrasound stimulation and induces apoptosis with higher intensity. To achieve the equal killing effect, the effective concentration of Nuc-CDs-PpIX is just one third of that of Mit-CDs-PpIX.
Collapse
Affiliation(s)
- Qianqian Duan
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Huaqian Li
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Juanjuan Xue
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Qi Zhang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jing Gao
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Xiaoyuan Wang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Qiang Zhang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Xing Guo
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Li Guo
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xiaojuan Wang
- Department of Gynecology, Pathology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, Shanxi China
| | - Shengbo Sang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yanfeng Xi
- Department of Gynecology, Pathology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, Shanxi China
| |
Collapse
|
25
|
Xie X, Zhang J, Wang Y, Shi W, Tang R, Tang Q, Sun S, Wu R, Xu S, Wang M, Liang X, Cui L. Nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. Mater Today Bio 2024; 24:100926. [PMID: 38179429 PMCID: PMC10765306 DOI: 10.1016/j.mtbio.2023.100926] [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: 10/05/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024] Open
Abstract
Immunotherapy as a milestone in cancer treatment has made great strides in the past decade, but it is still limited by low immune response rates and immune-related adverse events. Utilizing bioeffects of ultrasound to enhance tumor immunotherapy has attracted more and more attention, including sonothermal, sonomechanical, sonodynamic and sonopiezoelectric immunotherapy. Moreover, the emergence of nanomaterials has further improved the efficacy of ultrasound mediated immunotherapy. However, most of the summaries in this field are about a single aspect of the biological effects of ultrasound, which is not comprehensive and complete currently. This review proposes the recent progress of nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. The concept of immunotherapy and the application of bioeffects of ultrasound in cancer immunotherapy are initially introduced. Then, according to different bioeffects of ultrasound, the representative paradigms of nanomaterial augmented sono-immunotherapy are described, and their mechanisms are discussed. Finally, the challenges and application prospects of nanomaterial augmented ultrasound mediated cancer immunotherapy are discussed in depth, hoping to pave the way for cancer immunotherapy and promote the clinical translation of ultrasound mediated cancer immunotherapy through the reasonable combination of nanomaterials augmented ultrasonic bioeffects.
Collapse
Affiliation(s)
- Xinxin Xie
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Jinxia Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Yuan Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Wanrui Shi
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Rui Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Qingshuang Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ruiqi Wu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Shuyu Xu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Mengxin Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ligang Cui
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| |
Collapse
|
26
|
Peng X, Zheng J, Liu T, Zhou Z, Song C, Geng Y, Wang Z, Huang Y. Tumor Microenvironment Heterogeneity, Potential Therapeutic Avenues, and Emerging Therapies. Curr Cancer Drug Targets 2024; 24:288-307. [PMID: 37537777 DOI: 10.2174/1568009623666230712095021] [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: 04/17/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVE This review describes the comprehensive portrait of tumor microenvironment (TME). Additionally, we provided a panoramic perspective on the transformation and functions of the diverse constituents in TME, and the underlying mechanisms of drug resistance, beginning with the immune cells and metabolic dynamics within TME. Lastly, we summarized the most auspicious potential therapeutic strategies. RESULTS TME is a unique realm crafted by malignant cells to withstand the onslaught of endogenous and exogenous therapies. Recent research has revealed many small-molecule immunotherapies exhibiting auspicious outcomes in preclinical investigations. Furthermore, some pro-immune mechanisms have emerged as a potential avenue. With the advent of nanosystems and precision targeting, targeted therapy has now transcended the "comfort zone" erected by cancer cells within TME. CONCLUSION The ceaseless metamorphosis of TME fosters the intransigent resilience and proliferation of tumors. However, existing therapies have yet to surmount the formidable obstacles posed by TME. Therefore, scientists should investigate potential avenues for therapeutic intervention and design innovative pharmacological and clinical technologies.
Collapse
Affiliation(s)
- Xintong Peng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jingfan Zheng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Tianzi Liu
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ziwen Zhou
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Chen Song
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Geng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Zichuan Wang
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Huang
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
27
|
Li J, Zeng H, Li L, Yang Q, He L, Dong M. Advanced Generation Therapeutics: Biomimetic Nanodelivery System for Tumor Immunotherapy. ACS NANO 2023; 17:24593-24618. [PMID: 38055350 DOI: 10.1021/acsnano.3c10212] [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: 12/08/2023]
Abstract
Tumor immunotherapy is a safe and effective strategy for precision medicine. However, immunotherapy for most cancer cases still ends in failure, with the root causes of the immunosuppressive and extraordinary heterogeneity of the solid tumors microenvironment. The emerging biomimetic nanodelivery system provides a promising tactic to improve the immunotherapy effect while reducing the adverse reactions on nontarget cells. Herein, we summarize the relationship between tumor occurrence and tumor immune microenvironment, mechanism of tumor immune escape, immunotherapy classification (including adoptive cellular therapy, cytokines, cancer vaccines, and immune checkpoint inhibitors) and recommend target cells for immunotherapy first, and then emphatically introduce the recent advances and applications of the latest biomimetic nanodelivery systems (e.g., immune cells, erythrocytes, tumor cells, platelets, bacteria) in tumor immunotherapy. Meanwhile, we separately summarize the application of tumor vaccines. Finally, the predictable challenges and perspectives in a forward exploration of biomimetic nanodelivery systems for tumor immunotherapy are also discussed.
Collapse
Affiliation(s)
- Jie Li
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611135, Sichuan, China
- Cancer Prevention and Institute of Chengdu, Department of Oncology, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611130, Sichuan, China
| | - Huamin Zeng
- Department of Pathology, Chengdu Fifth People's Hospital (The Second Clinical Medical Colloge, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611130, Sichuan, China
| | - Luwei Li
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Qiu Yang
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611135, Sichuan, China
| | - Lang He
- Cancer Prevention and Institute of Chengdu, Department of Oncology, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611130, Sichuan, China
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611135, Sichuan, China
| |
Collapse
|
28
|
Ma D, Wang G, Lu J, Zeng X, Cheng Y, Zhang Z, Lin N, Chen Q. Multifunctional nano MOF drug delivery platform in combination therapy. Eur J Med Chem 2023; 261:115884. [PMID: 37862817 DOI: 10.1016/j.ejmech.2023.115884] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/11/2023] [Accepted: 10/14/2023] [Indexed: 10/22/2023]
Abstract
Recent preclinical and clinical studies have demonstrated that for cancer treatment, combination therapies are more effective than monotherapies in reducing drug-related toxicity, decreasing drug resistance, and improving therapeutic efficacy. With the rapid development of nanotechnology, the combination of metal-organic frameworks (MOFs) and multi-mode therapy offers a realistic possibility to further improve the shortcomings of cancer treatment. This article focuses on the latest developments, achievements, and treatment strategies of representative multifunctional MOF combination therapies for cancer treatment in recent years, which include not only bimodal combination therapies, but also multi-modal synergistic therapies, further demonstrating the effectiveness and superiority of the MOF drug delivery systems in cancer treatment.
Collapse
Affiliation(s)
- Dongwei Ma
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China
| | - Gang Wang
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China
| | - Jingsheng Lu
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China
| | - Xiaoxuan Zeng
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China
| | - Yanwei Cheng
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China
| | - Zhenwei Zhang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China
| | - Ning Lin
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China.
| | - Qing Chen
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Nanning, 530200, China.
| |
Collapse
|
29
|
Luo Z, Sheng Y, Jiang C, Pan Y, Wang X, Nezamzadeh-Ejhieh A, Ouyang J, Lu C, Liu J. Recent advances and prospects of metal-organic frameworks in cancer therapies. Dalton Trans 2023; 52:17601-17622. [PMID: 37953742 DOI: 10.1039/d3dt02543h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Metal-organic frameworks (MOFs) have been broadly applied in biomedical and other fields. MOFs have high porosity, a large comparative area, and good biostability and have attracted significant attention, especially in cancer therapies. This paper presents the latest applications of MOFs in chemodynamic therapy (CDT), sonodynamic therapy (SDT), photodynamic therapy (PDT), photothermal therapy (PTT), immunotherapy (IT), and combination therapy for breast cancer. A combination therapy is the combination of two different treatment modalities, such as CDT and PDT combination therapy, and is considered more effective than separate therapies. Herein, we have also discussed the advantages and disadvantages of combination therapy in the treatment of breast cancer. This paper aims to illustrate the potential of MOFs in new cancer therapeutic approaches, discuss their potential advantages, and provide some reflections on the latest research results.
Collapse
Affiliation(s)
- Zhiying Luo
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan, 523808, China.
| | - Yu Sheng
- Tungwah High School of Dongguan City (Dongcheng Campus), 1st Guangming Road, 523125 Dongguan, Guangdong, China
| | - Chenyi Jiang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan, 523808, China.
| | - Ying Pan
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan, 523808, China.
| | - Xiaoxiong Wang
- School of Materials and Environmental Engineering, Shenzhen Polytechnic University, Shenzhen, Guangdong, 518055, PR China
| | - Ali Nezamzadeh-Ejhieh
- Chemistry Department, Shahreza Branch, Islamic Azad University, Shahreza, Isfahan, Iran
| | - Jie Ouyang
- Key Laboratory for Breast Cancer Prevention and Treatment of Dongguan, Department of Breast Surgery, Dongguan Tungwah Hospital, Dongguan, China
| | - Chengyu Lu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan, 523808, China.
| | - Jianqiang Liu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan, 523808, China.
| |
Collapse
|
30
|
Dai X, Du Y, Li Y, Yan F. Nanomaterials-based precision sonodynamic therapy enhancing immune checkpoint blockade: A promising strategy targeting solid tumor. Mater Today Bio 2023; 23:100796. [PMID: 37766898 PMCID: PMC10520454 DOI: 10.1016/j.mtbio.2023.100796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Burgeoning is an evolution from conventional photodynamic therapy (PDT). Thus, sonodynamic therapy (SDT) regulated by nanoparticles (NPs) possesses multiple advantages, including stronger penetration ability, better biological safety, and not reactive oxygen species (ROS)-dependent tumor-killing effect. However, the limitation to tumor inhibition instead of shrinkage and the incapability of eliminating metastatic tumors hinder the clinical potential for SDT. Fortunately, immune checkpoint blockade (ICB) can revive immunological function and induce a long-term immune memory against tumor rechallenges. Hence, synergizing NPs-based SDT with ICB can provide a promising therapeutic outcome for solid tumors. Herein, we briefly reviewed the progress in NPs-based SDT and ICB therapy. We highlighted the synergistic anti-tumor mechanisms and summarized the representative preclinical trials on SDT-assisted immunotherapy. Compared to other reviews, we provided comprehensive and unique perspectives on the innovative sonosensitizers in each trial. Moreover, we also discussed the current challenges and future corresponding solutions.
Collapse
Affiliation(s)
- Xinlun Dai
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Yangyang Du
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yumei Li
- Department of Pediatric Intensive Care Unit, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
31
|
Wang X, Xu X, Yang Z, Xu X, Han S, Zhang H. Improvement of the effectiveness of sonodynamic therapy: by optimizing components and combination with other treatments. Biomater Sci 2023; 11:7489-7511. [PMID: 37873617 DOI: 10.1039/d3bm00738c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Sonodynamic therapy (SDT) is an emerging treatment method. In comparison with photodynamic therapy (PDT), SDT exhibits deep penetration, high cell membrane permeability, and free exposure to light capacity. Unfortunately, owing to inappropriate ultrasound parameter selection, poor targeting of sonosensitizers, and the complex tumor environment, SDT is frequently ineffective. In this review, we describe the approaches for selecting ultrasound parameters and how to develop sonosensitizers to increase targeting and improve adverse tumor microenvironments. Furthermore, the potential of combining SDT with other treatment methods, such as chemotherapy, chemodynamic therapy, photodynamic therapy, photothermal therapy, and immunotherapy, is discussed to further increase the treatment efficiency of SDT.
Collapse
Affiliation(s)
- Xiangting Wang
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Xiaohong Xu
- Department of Ultrasound, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhe Yang
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Xuanshou Xu
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Shisong Han
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| | - Heng Zhang
- Zhuhai Institute of Translational Medicine, Department of Ultrasound and Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), The First School of Clinical Medicine of Guangdong Medical University, Zhuhai 519000, China.
| |
Collapse
|
32
|
Lei J, Zhang W, Ma L, He Y, Liang H, Zhang X, Li G, Feng X, Tan L, Yang C. Sonodynamic amplification of cGAS-STING activation by cobalt-based nanoagonist against bone and metastatic tumor. Biomaterials 2023; 302:122295. [PMID: 37666101 DOI: 10.1016/j.biomaterials.2023.122295] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/19/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023]
Abstract
The therapeutic effect of cancer immunotherapy is restrained by limited patient response rate caused by 'cold' tumors with an intrinsically immunosuppressive tumor microenvironment (TME). Activating stimulator of interferon genes (STING) confers promising antitumor immunity even in 'cold' tumors, but the further promotion of STING agonists is hindered by undesirable toxicity, low specificity and lack of controllability. Herein, an ultrasound-controllable cGAS-STING amplifying nanoagonist was constructed by coordinating mitochondria-targeting ligand triphenylphosphonium (TPP) to sonodynamic cobalt organic framework nanosheets (TPP@CoTCPP). The Co ions specifically amplify STING activation only when cytosolic mitochondrial DNA leakage is caused by sonocatalysis-induced ROS production and sensed by cGAS. A series of downstream innate immune proinflammatory responses induced by local cGAS-STING pathway activation under spatiotemporal ultrasound stimulation efficiently prime the antitumor T-cell response against bone metastatic tumor, a typical immunosuppressive tumor. We also found that the coordination of TPP augments the sonodynamic effect of CoTCPP nanosheets by reducing the band gap, improving O2 adsorption and enhancing electron transfer. Overall, our study demonstrates that the targeted and amplified cGAS-STING activation in cancer cell controlled by spatiotemporal ultrasound irradiation boosts high-efficiency sonodynamic-ionicimmunotherapy against immunosuppressive tumor.
Collapse
Affiliation(s)
- Jie Lei
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Weifeng Zhang
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Liang Ma
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Yaqi He
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Huaizhen Liang
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Xiaoguang Zhang
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Gaocai Li
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Xiaobo Feng
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China.
| | - Lei Tan
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China.
| | - Cao Yang
- Orthopaedic Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China.
| |
Collapse
|
33
|
Li S, Mok GSP, Dai Y. Lipid bilayer-based biological nanoplatforms for sonodynamic cancer therapy. Adv Drug Deliv Rev 2023; 202:115110. [PMID: 37820981 DOI: 10.1016/j.addr.2023.115110] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/01/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
Sonodynamic therapy (SDT) has been developed as a promising alternative therapeutic modality for cancer treatment, involving the synergetic application of sonosensitizers and low-intensity ultrasound. However, the antitumor efficacy of SDT is significantly limited due to the poor performance of conventional sonosensitizers in vivo and the constrained tumor microenvironment (TME). Recent breakthroughs in lipid bilayer-based nanovesicles (LBBNs), including multifunctional liposomes, exosomes, and isolated cellular membranes, have brought new insights into the advancement of SDT. Despite their distinct sources and preparation methods, the lipid bilayer structure in common allows them to be functionalized in many comparable ways to serve as ideal nanocarriers against challenges arising from the tumor-specific sonosensitizer delivery and the complicated TME. In this review, we provide a comprehensive summary of the recent advances in LBBN-based SDT, with particular attention on how LBBNs can be engineered to improve the delivery efficiency of sonosensitizers and overcome physical, biological, and immune barriers within the TME for enhanced sonodynamic cancer therapy. We anticipate that this review will offer valuable guidance in the construction of LBBN-based nanosonosensitizers and contribute to the development of advanced strategies for next-generation sonodynamic cancer therapy.
Collapse
Affiliation(s)
- Songhao 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
| | - Greta S P Mok
- Biomedical Imaging Laboratory (BIG), Department of Electrical and Computer Engineering, Faculty of Science and Technology, 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
|
34
|
Ding H, Xia Q, Shen J, Zhu C, Zhang Y, Feng N. Advances and prospects of tumor immunotherapy mediated by immune cell-derived biomimetic metal-organic frameworks. Colloids Surf B Biointerfaces 2023; 232:113607. [PMID: 39491916 DOI: 10.1016/j.colsurfb.2023.113607] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/18/2023] [Indexed: 11/05/2024]
Abstract
The clinical translational success of nanomedicine and immunotherapy has already proved the immense potential in the field of nanotechnology and immunization. However, the development of nanomedicine is confronted with challenges such as potential toxicity and unclear nano-bio interactions. The efficacy of immunotherapy is limited to only a few groups. Combining immunotherapy with nanomedicine for multi-modal treatment effectively compensates for the limitations of the above single therapy. Immune cell membrane camouflaged metal-organic frameworks (ICM-MOFs) have emerged as a simple yet promising multimodal treatment strategy that possess multifunctional nanoscale properties and exhibit immune cell-like behaviors of stealth, targeting and immunomodulation. Here, we comprehensively discuss the latest advancements in ICM-MOFs, with a focus on the challenges of mono-immunotherapy, the superiority of biomimetic coating for MOF functionalization, preparation methods, related action mechanisms and biomedical applications. Finally, we address the challenges and prospects for clinical translation.
Collapse
Affiliation(s)
- Huining Ding
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qing Xia
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqi Shen
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chunyun Zhu
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yongtai Zhang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
35
|
Desai DN, Mahal A, Varshney R, Obaidullah AJ, Gupta B, Mohanty P, Pattnaik P, Mohapatra NC, Mishra S, Kandi V, Rabaan AA, Mohapatra RK. Nanoadjuvants: Promising Bioinspired and Biomimetic Approaches in Vaccine Innovation. ACS OMEGA 2023; 8:27953-27968. [PMID: 37576639 PMCID: PMC10413842 DOI: 10.1021/acsomega.3c02030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023]
Abstract
Adjuvants are the important part of vaccine manufacturing as they elicit the vaccination effect and enhance the durability of the immune response through controlled release. In light of this, nanoadjuvants have shown unique broad spectrum advantages. As nanoparticles (NPs) based vaccines are fast-acting and better in terms of safety and usability parameters as compared to traditional vaccines, they have attracted the attention of researchers. A vaccine nanocarrier is another interesting and promising area for the development of next-generation vaccines for prophylaxis. This review looks at the various nanoadjuvants and their structure-function relationships. It compiles the state-of-art literature on numerous nanoadjuvants to help domain researchers orient their understanding and extend their endeavors in vaccines research and development.
Collapse
Affiliation(s)
- Dhruv N. Desai
- Department
of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ahmed Mahal
- Department
of Medical Biochemical Analysis, College of Health Technology, Cihan University−Erbil, Erbil, Kurdistan Region, Iraq
| | - Rajat Varshney
- Department
of Veterinary Microbiology, FVAS, Banaras
Hindu University, Mirzapur 231001, India
| | - Ahmad J. Obaidullah
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Bhawna Gupta
- School
of Biotechnology, KIIT Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Pratikhya Mohanty
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | | | | | - Snehasish Mishra
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Venkataramana Kandi
- Department
of Microbiology, Prathima Institute of Medical
Sciences, Karimnagar 505 417, Telangana, India
| | - Ali A. Rabaan
- Molecular
Diagnostic Laboratory, Johns Hopkins Aramco
Healthcare, Dhahran 31311, Saudi Arabia
- College
of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department
of Public Health and Nutrition, The University
of Haripur, Haripur 22610, Pakistan
| | - Ranjan K. Mohapatra
- Department
of Chemistry, Government College of Engineering, Keonjhar 758002, Odisha, India
| |
Collapse
|
36
|
Liang S, Yao J, Liu D, Rao L, Chen X, Wang Z. Harnessing Nanomaterials for Cancer Sonodynamic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211130. [PMID: 36881527 DOI: 10.1002/adma.202211130] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/12/2023] [Indexed: 06/18/2023]
Abstract
Immunotherapy has made remarkable strides in cancer therapy over the past decade. However, such emerging therapy still suffers from the low response rates and immune-related adverse events. Various strategies have been developed to overcome these serious challenges. Therein, sonodynamic therapy (SDT), as a non-invasive treatment, has received ever-increasing attention especially in the treatment of deep-seated tumors. Significantly, SDT can effectively induce immunogenic cell death to trigger systemic anti-tumor immune response, termed sonodynamic immunotherapy. The rapid development of nanotechnology has revolutionized SDT effects with robust immune response induction. As a result, more and more innovative nanosonosensitizers and synergistic treatment modalities are established with superior efficacy and safe profile. In this review, the recent advances in cancer sonodynamic immunotherapy are summarized with a particular emphasis on how nanotechnology can be explored to harness SDT for amplifying anti-tumor immune response. Moreover, the current challenges in this field and the prospects for its clinical translation are also presented. It is anticipated that this review can provide rational guidance and facilitate the development of nanomaterials-assisted sonodynamic immunotherapy, helping to pave the way for next-generation cancer therapy and eventually achieve a durable response in patients.
Collapse
Affiliation(s)
- Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jianjun Yao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, 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
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, 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
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
37
|
Chen P, Zhang P, Shah NH, Cui Y, Wang Y. A Comprehensive Review of Inorganic Sonosensitizers for Sonodynamic Therapy. Int J Mol Sci 2023; 24:12001. [PMID: 37569377 PMCID: PMC10418994 DOI: 10.3390/ijms241512001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/06/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Sonodynamic therapy (SDT) is an emerging non-invasive cancer treatment method in the field of nanomedicine, which has the advantages of deep penetration, good therapeutic efficacy, and minimal damage to normal tissues. Sonosensitizers play a crucial role in the process of SDT, as their structure and properties directly determine the treatment outcome. Inorganic sonosensitizers, with their high stability and longer circulation time in the human body, have great potential in SDT. In this review, the possible mechanisms of SDT including the ultrasonic cavitation, reactive oxygen species generation, and activation of immunity are briefly discussed. Then, the latest research progress on inorganic sonosensitizers is systematically summarized. Subsequently, strategies for optimizing treatment efficacy are introduced, including combination therapy and image-guided therapy. The challenges and future prospects of sonodynamic therapy are discussed. It is hoped that this review will provide some guidance for the screening of inorganic sonosensitizers.
Collapse
Affiliation(s)
- Peng Chen
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Ping Zhang
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Navid Hussain Shah
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
| | - Yanyan Cui
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
| | - Yaling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| |
Collapse
|
38
|
Songca SP. Combinations of Photodynamic Therapy with Other Minimally Invasive Therapeutic Technologies against Cancer and Microbial Infections. Int J Mol Sci 2023; 24:10875. [PMID: 37446050 DOI: 10.3390/ijms241310875] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The rapid rise in research and development following the discovery of photodynamic therapy to establish novel photosensitizers and overcome the limitations of the technology soon after its clinical translation has given rise to a few significant milestones. These include several novel generations of photosensitizers, the widening of the scope of applications, leveraging of the offerings of nanotechnology for greater efficacy, selectivity for the disease over host tissue and cells, the advent of combination therapies with other similarly minimally invasive therapeutic technologies, the use of stimulus-responsive delivery and disease targeting, and greater penetration depth of the activation energy. Brought together, all these milestones have contributed to the significant enhancement of what is still arguably a novel technology. Yet the major applications of photodynamic therapy still remain firmly located in neoplasms, from where most of the new innovations appear to launch to other areas, such as microbial, fungal, viral, acne, wet age-related macular degeneration, atherosclerosis, psoriasis, environmental sanitization, pest control, and dermatology. Three main value propositions of combinations of photodynamic therapy include the synergistic and additive enhancement of efficacy, the relatively low emergence of resistance and its rapid development as a targeted and high-precision therapy. Combinations with established methods such as chemotherapy and radiotherapy and demonstrated applications in mop-up surgery promise to enhance these top three clinical tools. From published in vitro and preclinical studies, clinical trials and applications, and postclinical case studies, seven combinations with photodynamic therapy have become prominent research interests because they are potentially easily applied, showing enhanced efficacy, and are rapidly translating to the clinic. These include combinations with chemotherapy, photothermal therapy, magnetic hyperthermia, cold plasma therapy, sonodynamic therapy, immunotherapy, and radiotherapy. Photochemical internalization is a critical mechanism for some combinations.
Collapse
Affiliation(s)
- Sandile Phinda Songca
- School of Chemistry and Physics, College of Agriculture Engineering and Science, Pietermaritzburg Campus, University of KwaZulu-Natal, Pietermaritzburg 3209, South Africa
| |
Collapse
|
39
|
Wang F, Pu K, Li J. Activating Nanomedicines with Electromagnetic Energy for Deep-Tissue Induction of Immunogenic Cell Death in Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201083. [PMID: 36316270 DOI: 10.1002/smtd.202201083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Indexed: 05/17/2023]
Abstract
Immunotherapy is an attractive approach for cancer therapy, while its antitumor efficacy is still limited, especially for non-immunogenic tumors. Nanomedicines can be utilized to convert the non-immunogenic "cold" tumors to immunogenic "hot" tumors via inducing immunogenic cell death (ICD), thereby promoting the antitumor immune response. Some nanomedicines that can produce local heat and reactive oxygen species upon the stimulation of electromagnetic energy are the main candidates for inducing the ICD effect. However, their applications are often restricted due to the poor tissue penetration depths of electromagnetic energy, such as light. By contrast, ultrasound, X-ray, alternating magnetic field, and microwave show excellent tissue penetration depths and thereby can be used for sonodynamic therapy, radiotherapy, magnetic hyperthermia therapy, and microwave ablation therapy, all of which can effectively induce ICD. Herein, the combination of deep-tissue electromagnetic energy with nanomedicines for inducing ICD and cancer immunotherapy are summarized. In particular, the designs of nanomedicines to amplify ICD effect in the presence of deep-tissue electromagnetic energy and sensitize tumors to various immunotherapies will be discussed. At the end of this review, a brief conclusion and discussion of current challenges and further perspectives in this subfield are provided.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
40
|
Meng Q, Ding B, Ma P, Lin J. Interrelation between Programmed Cell Death and Immunogenic Cell Death: Take Antitumor Nanodrug as an Example. SMALL METHODS 2023; 7:e2201406. [PMID: 36707416 DOI: 10.1002/smtd.202201406] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/20/2022] [Indexed: 05/17/2023]
Abstract
Programmed cell death (PCD, mainly including apoptosis, necrosis, ferroptosis, pyroptosis, and autophagy) and immunogenic cell death (ICD), as important cell death mechanisms, are widely reported in cancer therapy, and understanding the relationship between the two is significant for clinical tumor treatments. Considering that vast nanodrugs are developed to induce tumor PCD and ICD simultaneously, in this review, the interrelationship between PCD and ICD is described using nanomedicines as examples. First, an overview of PCD patterns and focus on the morphological differences and interconnections among them are provided. Then the interrelationship between apoptosis and ICD in terms of endoplasmic reticulum stress is described by introducing various cancer treatments and the recent developments of nanomedicines with inducible immunogenicity. Next, the crosstalk between non-apoptotic (including necrosis, ferroptosis, pyroptosis, and autophagy) signaling pathways and ICD is introduced and their relationship through various nanomedicines as examples is further illustrated. Finally, the relationship between PCD and ICD and its application prospects in the development of new ICD nanomaterials are summarized. This review is believed to deepen the understanding of the relationship between PCD and ICD, extend the biomedical applications of various nanodrugs, and promote the progress of clinical tumor therapy.
Collapse
Affiliation(s)
- Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
41
|
Lin Q, Peng Y, Wen Y, Li X, Du D, Dai W, Tian W, Meng Y. Recent progress in cancer cell membrane-based nanoparticles for biomedical applications. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:262-279. [PMID: 36895440 PMCID: PMC9989677 DOI: 10.3762/bjnano.14.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Immune clearance and insufficient targeting have limited the efficacy of existing therapeutic strategies for cancer. Toxic side effects and individual differences in response to treatment have further limited the benefits of clinical treatment for patients. Biomimetic cancer cell membrane-based nanotechnology has provided a new approach for biomedicine to overcome these obstacles. Biomimetic nanoparticles exhibit various effects (e.g., homotypic targeting, prolonging drug circulation, regulating the immune system, and penetrating biological barriers) after encapsulation by cancer cell membranes. The sensitivity and specificity of diagnostic methods will also be improved by utilizing the properties of cancer cell membranes. In this review, different properties and functions of cancer cell membranes are presented. Utilizing these advantages, nanoparticles can exhibit unique therapeutic capabilities in various types of diseases, such as solid tumors, hematological malignancies, immune system diseases, and cardiovascular diseases. Furthermore, cancer cell membrane-encapsulated nanoparticles show improved effectiveness and efficiency in combination with current diagnostic and therapeutic methods, which will contribute to the development of individualized treatments. This strategy has promising clinical translation prospects, and the associated challenges are discussed.
Collapse
Affiliation(s)
- Qixiong Lin
- The Ninth Clinical Medical School of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yueyou Peng
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Yanyan Wen
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaoqiong Li
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Donglian Du
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Weibin Dai
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| | - Wei Tian
- Department of General Surgery, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, China
| | - Yanfeng Meng
- Department of MRI, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi 030009, China
| |
Collapse
|
42
|
Nguyen Cao TG, Truong Hoang Q, Hong EJ, Kang SJ, Kang JH, Ravichandran V, Kang HC, Ko YT, Rhee WJ, Shim MS. Mitochondria-targeting sonosensitizer-loaded extracellular vesicles for chemo-sonodynamic therapy. J Control Release 2023; 354:651-663. [PMID: 36682729 DOI: 10.1016/j.jconrel.2023.01.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/04/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
Sonodynamic therapy (SDT) has emerged as an effective therapeutic modality as it employs ultrasound (US) to eradicate deep-seated tumors noninvasively. However, the therapeutic efficacy of SDT in clinical settings remains limited owing to the low aqueous stability and poor pharmacokinetic properties of sonosensitizers. In this study, extracellular vesicles (EVs), which have low systemic toxicity, were used as clinically available nanocarriers to effectively transfer a sonosensitizer to cancer cells. Chlorin e6 (Ce6), a sonosensitizer, was conjugated to a mitochondria-targeting triphenylphosphonium (TPP) moiety and loaded into EVs to enhance the efficacy of SDT, because mitochondria are critical subcellular organelles that regulate cell survival and death. Additionally, piperlongumine (PL), a pro-oxidant and cancer-specific chemotherapeutic agent, was co-encapsulated into EVs to achieve efficient and selective anticancer activity. The EVs substantially amplified the cellular internalization of TPP-conjugated Ce6 (TPP-Ce6), resulting in the enhanced generation of intracellular reactive oxygen species (ROS) in MCF-7 human breast cancer cells upon US exposure. Importantly, EVs encapsulating TPP-Ce6 effectively destroyed the mitochondria under irradiation with US, leading to efficient anticancer activity. The co-encapsulation of pro-oxidant PL into EVs significantly enhanced the SDT efficacy in MCF-7 cells through the excessive generation of ROS. Moreover, the EV co-encapsulating TPP-Ce6 and PL [EV(TPP-Ce6/PL)] exhibited cancer-specific cell death owing to the cancer-selective apoptosis triggered by PL. In vivo study using MCF-7 tumor-xenograft mice revealed that EV(TPP-Ce6/PL) effectively accumulated in tumors after intravenous injection. Notably, treatment with EV(TPP-Ce6/PL) and US inhibited tumor growth significantly without causing systemic toxicity. This study demonstrated the feasibility of using EV(TPP-Ce6/PL) for biocompatible and cancer-specific chemo-SDT.
Collapse
Affiliation(s)
- Thuy Giang Nguyen Cao
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Quan Truong Hoang
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Eun Ji Hong
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Su Jin Kang
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Ji Hee Kang
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Vasanthan Ravichandran
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Han Chang Kang
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Gyeonggi-do 14662, Republic of Korea
| | - Young Tag Ko
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Won Jong Rhee
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; Research Center for Bio Materials & Process Development, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea.
| |
Collapse
|
43
|
Yang Y, Huang J, Liu M, Qiu Y, Chen Q, Zhao T, Xiao Z, Yang Y, Jiang Y, Huang Q, Ai K. Emerging Sonodynamic Therapy-Based Nanomedicines for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204365. [PMID: 36437106 PMCID: PMC9839863 DOI: 10.1002/advs.202204365] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/25/2022] [Indexed: 05/08/2023]
Abstract
Cancer immunotherapy effect can be greatly enhanced by other methods to induce immunogenic cell death (ICD), which has profoundly affected immunotherapy as a highly efficient paradigm. However, these treatments have significant limitations, either by causing damage of the immune system or limited to superficial tumors. Sonodynamic therapy (SDT) can induce ICD to promote immunotherapy without affecting the immune system because of its excellent spatiotemporal selectivity and low side effects. Nevertheless, SDT is still limited by low reactive oxygen species yield and the complex tumor microenvironment. Recently, some emerging SDT-based nanomedicines have made numerous attractive and encouraging achievements in the field of cancer immunotherapy due to high immunotherapeutic efficiency. However, this cross-cutting field of research is still far from being widely explored due to huge professional barriers. Herein, the characteristics of the tumor immune microenvironment and the mechanisms of ICD are firstly systematically summarized. Subsequently, the therapeutic mechanism of SDT is fully summarized, and the advantages and limitations of SDT are discussed. The representative advances of SDT-based nanomedicines for cancer immunotherapy are further highlighted. Finally, the application prospects and challenges of SDT-based immunotherapy in future clinical translation are discussed.
Collapse
Affiliation(s)
- Yunrong Yang
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jia Huang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Min Liu
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Yige Qiu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yuqi Yang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yitian Jiang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Qiong Huang
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| |
Collapse
|
44
|
Xu T, Liu Z, Huang L, Jing J, Liu X. Modulating the tumor immune microenvironment with nanoparticles: A sword for improving the efficiency of ovarian cancer immunotherapy. Front Immunol 2022; 13:1057850. [PMID: 36532066 PMCID: PMC9751906 DOI: 10.3389/fimmu.2022.1057850] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
With encouraging antitumor effects, immunotherapy represented by immune checkpoint blockade has developed into a mainstream cancer therapeutic modality. However, only a minority of ovarian cancer (OC) patients could benefit from immunotherapy. The main reason is that most OC harbor a suppressive tumor immune microenvironment (TIME). Emerging studies suggest that M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), myeloid-derived suppressor cells (MDSCs), and cancer-associated fibroblasts (CAFs) are enriched in OC. Thus, reversing the suppressive TIME is considered an ideal candidate for improving the efficiency of immunotherapy. Nanoparticles encapsulating immunoregulatory agents can regulate immunocytes and improve the TIME to boost the antitumor immune response. In addition, some nanoparticle-mediated photodynamic and photothermal therapy can directly kill tumor cells and induce tumor immunogenic cell death to activate antigen-presenting cells and promote T cell infiltration. These advantages make nanoparticles promising candidates for modulating the TIME and improving OC immunotherapy. In this review, we analyzed the composition and function of the TIME in OC and summarized the current clinical progress of OC immunotherapy. Then, we expounded on the promising advances in nanomaterial-mediated immunotherapy for modulating the TIME in OC. Finally, we discussed the obstacles and challenges in the clinical translation of this novel combination treatment regimen. We believe this resourceful strategy will open the door to effective immunotherapy of OC and benefit numerous patients.
Collapse
Affiliation(s)
| | | | | | - Jing Jing
- *Correspondence: Xiaowei Liu, ; Jing Jing,
| | | |
Collapse
|
45
|
Zhang Y, Zhang X, Li H, Liu J, Wei W, Gao J. Membrane-Coated Biomimetic Nanoparticles: A State-of-the-Art Multifunctional Weapon for Tumor Immunotherapy. MEMBRANES 2022; 12:membranes12080738. [PMID: 36005653 PMCID: PMC9412372 DOI: 10.3390/membranes12080738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022]
Abstract
The advent of immunotherapy, which improves the immune system’s ability to attack and eliminate tumors, has brought new hope for tumor treatment. However, immunotherapy regimens have seen satisfactory results in only some patients. The development of nanotechnology has remarkably improved the effectiveness of tumor immunotherapy, but its application is limited by its passive immune clearance, poor biocompatibility, systemic immunotoxicity, etc. Therefore, membrane-coated biomimetic nanoparticles have been developed by functional, targeting, and biocompatible cell membrane coating technology. Membrane-coated nanoparticles have the advantages of homologous targeting, prolonged circulation, and the avoidance of immune responses, thus remarkably improving the therapeutic efficacy of tumor immunotherapy. Herein, this review explores the recent advances and future perspectives of cell membrane-coated nanoparticles for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China;
| | - Xinyi Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China;
| | - Haitao Li
- Department of Vascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefangdadao Road, Wuhan 430022, China; (H.L.); (J.L.)
| | - Jianyong Liu
- Department of Vascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefangdadao Road, Wuhan 430022, China; (H.L.); (J.L.)
| | - Wei Wei
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China;
- Correspondence: (W.W.); (J.G.)
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China;
- Correspondence: (W.W.); (J.G.)
| |
Collapse
|
46
|
Gareev KG, Grouzdev DS, Koziaeva VV, Sitkov NO, Gao H, Zimina TM, Shevtsov M. Biomimetic Nanomaterials: Diversity, Technology, and Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2485. [PMID: 35889709 PMCID: PMC9316400 DOI: 10.3390/nano12142485] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/04/2023]
Abstract
Biomimetic nanomaterials (BNMs) are functional materials containing nanoscale components and having structural and technological similarities to natural (biogenic) prototypes. Despite the fact that biomimetic approaches in materials technology have been used since the second half of the 20th century, BNMs are still at the forefront of materials science. This review considered a general classification of such nanomaterials according to the characteristic features of natural analogues that are reproduced in the preparation of BNMs, including biomimetic structure, biomimetic synthesis, and the inclusion of biogenic components. BNMs containing magnetic, metal, or metal oxide organic and ceramic structural elements (including their various combinations) were considered separately. The BNMs under consideration were analyzed according to the declared areas of application, which included tooth and bone reconstruction, magnetic and infrared hyperthermia, chemo- and immunotherapy, the development of new drugs for targeted therapy, antibacterial and anti-inflammatory therapy, and bioimaging. In conclusion, the authors' point of view is given about the prospects for the development of this scientific area associated with the use of native, genetically modified, or completely artificial phospholipid membranes, which allow combining the physicochemical and biological properties of biogenic prototypes with high biocompatibility, economic availability, and scalability of fully synthetic nanomaterials.
Collapse
Affiliation(s)
- Kamil G. Gareev
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia; (N.O.S.); (T.M.Z.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Denis S. Grouzdev
- SciBear OU, Tartu mnt 67/1-13b, Kesklinna Linnaosa, 10115 Tallinn, Estonia;
| | - Veronika V. Koziaeva
- Research Center of Biotechnology of the Russian Academy of Sciences, Institute of Bioengineering, 119071 Moscow, Russia;
| | - Nikita O. Sitkov
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia; (N.O.S.); (T.M.Z.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
| | - Tatiana M. Zimina
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia; (N.O.S.); (T.M.Z.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
- Center of Translational Cancer Research (TranslaTUM), Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
- National Center for Neurosurgery, Nur-Sultan 010000, Kazakhstan
| |
Collapse
|