1
|
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
|
2
|
Chen G, Li B, Li T, Lin M, Zhong H, Xie X, Zhang Q, Chen Q, Meng X, Xiao Z, Shuai X. Core-Satellite Nanoassembly Overcomes Spatial Heterogeneity of Dendric Cell Distribution in Pancreatic Tumors for Effective Chemoimmunotherapy. ACS NANO 2025; 19:4739-4753. [PMID: 39834130 DOI: 10.1021/acsnano.4c15444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Pancreatic cancer therapies such as chemotherapy and immunotherapy are hindered by the dense extracellular matrix known as physical barriers, leading to heterogeneity impeding the effective penetration of chemotherapeutic agents and activation of antitumor immune responses. To address this challenge, we developed a hybrid nanoassembly with a distinct core-satellite-like heterostructure, PLAF@P/T-PD, which is responsive to both internal pH/redox and external ultrasound stimulations. This heterostructural nanoassembly features a polymersome core encapsulating an ultrasound contrast agent perfluoropentane and a chemotherapeutic agent Taxol (PLAF@P/T) electrostatically coated with satellite-like polyplexes carrying an immune agonist dsDNA (PD), which brings about synergistic functions inside the pancreatic tumor. The PLAF@P/T core functions as an enhancer for intratumor delivery through size enlargement and charge conversion in response to reactive oxygen species (ROS) and low pH, which triggers polyplex release and enables ultrasound-assisted tumor-penetrating Taxol delivery. Meanwhile, the released cationic polyplexes function as nucleic nanomedicine preferentially engulfed by peripheral dendritic cells (DCs) for immune modulation. Animal studies in mouse orthotopic pancreatic tumor model demonstrated exceptional therapeutic efficacy against both primary and metastatic tumors, which underlines the potential of this heterostructural nanoplatform for overcoming the therapeutic challenges associated with the heterogeneous physical barrier hindering intratumor drug delivery in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Gengjia Chen
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Department of Radiology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Bo Li
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Tan Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Minzhao Lin
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Huihai Zhong
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiaoxue Xie
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Qiaoyun Zhang
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Qi Chen
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaochun Meng
- Department of Radiology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
3
|
Shen J, Feng K, Yu J, Zhao Y, Chen R, Xiong H, Ruan Y, Xu Z, Zhang T, Sun X. Responsive and traceless assembly of iron nanoparticles and 131I labeled radiopharmaceuticals for ferroptosis enhanced radio-immunotherapy. Biomaterials 2025; 313:122795. [PMID: 39232333 DOI: 10.1016/j.biomaterials.2024.122795] [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: 05/17/2024] [Revised: 08/07/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Ferroptosis is an iron-dependent form of programmed cell death with the potential to reverse traditional cancer therapy resistance. The combination of ferroptosis with chemotherapy, photodynamic therapy and X-ray therapy has demonstrated remarkably improved therapeutic efficiency. Radiopharmaceutical therapy (RPT) is an emerging approach that achieves precise radiation to diseased tissues via radionuclide delivery. However, insufficient accumulation and retention of therapeutic radiopharmaceuticals in tumor region as well as cancer radioresistance impact treatment efficacy. Here, a nanoassembly of renal clearable ultrasmall iron nanoparticles (USINPs) and 131I-aPD-L1 is prepared via the affinity of fluorophenylboronic acid modified on the USINPs with 131I-aPD-L1. The 150 nm USINAs(131I-aPD-L1) nanoassembly is stable in blood circulation, effectively targets to the tumor and disassembles in the presence of ATP in the tumor microenvironment. Both in vitro and in vivo experiments prove that USINPs-induced ferroptosis boosted the tumor radiosensitization to 131I while 131I-mediated RPT further enhanced ferroptosis. Meanwhile, the immunogenic cell death caused by RPT and ferroptosis combined with PD-L1 immune checkpoint blockade therapy exhibits a strong antitumor immunity. This study provides a novel way to improve the tumor accumulation of ferroptosis inducer and radiopharmaceuticals, insights into the interaction between RPT and ferroptosis and an effective SPECT-guided ferroptosis-enhanced radio-immunotherapy.
Collapse
Affiliation(s)
- Jingjing Shen
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kai Feng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jing Yu
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yaxuan Zhao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ruifang Chen
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hehua Xiong
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yiling Ruan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhengtao Xu
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Tao Zhang
- Northern Jiangsu Institute of Clinical Medicine, Department of Radiopharmaceuticals, Nuclear Medicine Clinical Translation Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
4
|
Yan L, Chang L, Tian Y, Hu J, Cao Z, Guo X, Geng B. Graphene Quantum Dot Sensitized Heterojunctions Induce Tumor-Specific Cuproptosis to Boost Sonodynamic and Chemodynamic Enhanced Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410606. [PMID: 39716968 PMCID: PMC11831527 DOI: 10.1002/advs.202410606] [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: 09/01/2024] [Revised: 12/01/2024] [Indexed: 12/25/2024]
Abstract
Cuproptosis that utilizes copper ionophore to induce programmed cell death holds promise for enhancing the effectiveness of conventional anticancer therapies and triggering efficient adaptive immune responses. However, the non-tumor-specific release of Cu ions can induce cuproptosis and cause irreversible damage to normal tissues. To maximize the therapeutic effects of tumor-specific cuproptosis, this work reports for the first time the regulation of degradation behaviors of Cu-based nanomaterials using graphene quantum dots (GQDs) as a protection layer. The deposition of GQDs not only avoids the degradation of Cu2O nanocubes under normal physiological conditions, but also sensitizes their sonodynamic activity due to the formation of Z-scheme heterojunctions. The tumor-specific released Cu ions achieve the cascade amplification of reactive oxygen species (ROS) generation through Cu+-mediated Fenton-like reaction and Cu2+-facilitated GSH depletion. More importantly, the immunosuppressive tumor microenvironment (TME) can be reversed by the greatly enhanced ROS levels and high-efficiency cuproptosis, ultimately inducing immunogenic cell death that promotes robust systemic immune responses for the eradication of primary tumors and suppression of distant tumors. This work provides a novel paradigm for the integration of SDT, CDT, cuproptosis, and immunotherapy in a controlled manner to achieve tumor-specific antitumor therapy by controlling the degradation behaviors of Cu-based nanomaterials.
Collapse
Affiliation(s)
- Lang Yan
- Department of Health ToxicologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433China
| | - Liang Chang
- Department of Emergency and Critical CareShanghai Changzheng HospitalSecond Affiliated HospitalNaval Medical UniversityShanghai200003China
| | - Yijun Tian
- Department of Health ToxicologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433China
| | - Jinyan Hu
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444China
| | - Zhi Cao
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghai200433China
| | - Xiang Guo
- Department of OrthopaedicsShanghai Changzheng HospitalSecond Affiliated HospitalNaval Medical UniversityShanghai200003China
| | - Bijiang Geng
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444China
| |
Collapse
|
5
|
Hong K, Cao J, Jiang W, Deng W, Huang G, Huang T, Fang J, Wang Y. A nanodrug provokes antitumor immune responses via synchronous multicellular regulation for enhanced cancer immunotherapy. J Colloid Interface Sci 2025; 678:750-762. [PMID: 39265345 DOI: 10.1016/j.jcis.2024.09.016] [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: 05/27/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
Hepatocellular carcinoma (HCC) exhibits a low response to immunotherapy due to the dense extracellular matrix (ECM) filled with immunosuppressive cells including dendritic cells (DCs) of blocked maturation. Herein, we develop a nanoprodrug self-assembled from polyethylene glycol-poly-4-borono-l-phenylalanine (mPEG-PBPA) conjugating with quercetin (QUE) via boronic ester bonds. In addition, an immune adjuvant of imiquimod (R837) is incorporated. The nanodrug (denoted as Q&R@NPs) is prepared from a simple mixing means with a high loading content of QUE reaching more than 30%. Owing to the acid and reactive oxygen species (ROS) sensitivities of boronic ester bonds, Q&R@NPs can respond to the tumor microenvironment (TME) and release QUE and R837 to synchronously exert multicellular regulation functions. Specifically, QUE inhibits the activation state of hepatic stellate cells and reduces highly expressed programmed death receptor ligand 1 (PD-L1) on tumor cells, meanwhile R837 exposes calreticulin on tumor cell surface as an "eat me" signal and leads to a large number of DCs maturing for enhanced antigen presentation. Consequently, the cooperative immune regulation results in a remodeled TME with high infiltration of cytotoxic T lymphocytes for enhanced HCC immunotherapy, which demonstrates an effective immunotherapy paradigm for dense ECM characterized solid tumors with high PD-L1 expression.
Collapse
Affiliation(s)
- Keze Hong
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Jianrong Cao
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Weiting Jiang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Wei Deng
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Guohong Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Tao Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| | - Jin Fang
- Department of Radiology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
6
|
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
|
7
|
Liu Y, Guo Q, Shi Y, Cui M, Jing F. Research progress of novel anti-tumor drug formulations. Front Oncol 2024; 14:1507958. [PMID: 39737395 PMCID: PMC11683012 DOI: 10.3389/fonc.2024.1507958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Cancers have become the second leading cause of death worldwide, following cardiovascular diseases.Traditional anti- cancer strategies, including radiotherapy chemotherapy, surgery, and targeted therapies, have been widely used but are often reassessed due to their significant side effects and relatively low cure rate. Recently, the development of novel formulations for anti-tumor drugs has gained considerable attention, marking a pivotal step forward in cancer treatment advancements. These innovative formulations aim to enhance the therapeutic efficacy of anti-tumor drugs by employing advanced drug formulation technologies and delivery systems. In particular, nano-drug delivery systems (NDDS) have emerged as a promising approach to improve drug targeting, reduce side effects, and overcome drug resistance. This review highlights recent progress in NDDS for anti-tumor drug development and explores the future prospects of these advanced formulations in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pharmacology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - YunYan Shi
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - MengNa Cui
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - FanBo Jing
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
8
|
Chauhan A, Kamal R, Bhatia R, Singh TG, Awasthi A. From Bench to Bedside: ROS-Responsive Nanocarriers in Cancer Therapy. AAPS PharmSciTech 2024; 26:10. [PMID: 39668268 DOI: 10.1208/s12249-024-03011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024] Open
Abstract
Reactive oxygen species (ROS) play a dual role in cancer, acting as both signaling molecules that promote tumour growth and as agents that can inhibit tumour progression through cytotoxic effects. In cancer therapy, ROS-responsive drug delivery systems take advantage of the elevated ROS levels found in tumors compared to healthy tissues. These systems are engineered to release drugs precisely in response to increased ROS levels in tumour cells, allowing targeted and controlled treatment, minimizing side effects, and enhancing therapeutic outcomes. ROS generation in cancer cells is linked to metabolic changes, mitochondrial dysfunction, and oncogenic signaling, leading to increased oxidative stress. Tumour cells manage this by upregulating antioxidant defenses to prevent ROS from reaching harmful levels. This balance between ROS production and neutralization is critical for cancer cell survival, making ROS both a challenge and an opportunity for targeted therapies. ROS also connect inflammation and cancer. Chronic inflammation leads to elevated ROS, which can damage DNA and proteins, promoting mutations and cancer development. Additionally, ROS contribute to protein degradation, affecting essential cellular functions. Therapeutic strategies targeting ROS aim to either increase ROS beyond tolerable levels for cancer cells or inhibit their antioxidant defenses. Nanocarriers responsive to ROS show great potential in improving the precision of cancer treatments by releasing drugs specifically in high ROS environments, like tumors. This review discusses the mechanisms of ROS in cancer, its role in inflammation and protein degradation, and the advances in ROS-targeted nanocarrier therapies across different cancer types.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- School of Pharmacy, Desh Bhagat University, 147301, Punjab, India, Mandi Gobindgarh
| | - Rohit Bhatia
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Ankit Awasthi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
9
|
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
|
10
|
Li L, Bian L, Kou N, Yuan Y, Zou H. Special immune-related adverse events and subsequent photodynamic therapy in tislelizumab treatment for esophageal cancer: a case report. Front Immunol 2024; 15:1497259. [PMID: 39654898 PMCID: PMC11625816 DOI: 10.3389/fimmu.2024.1497259] [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: 09/16/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
This case report highlights the immune-related adverse events (irAEs) that occurred during the treatment of esophageal cancer with Tislelizumab and discusses management strategies, indicating that photodynamic therapy (PDT) may be an optimal adjunctive treatment option. Following Tislelizumab therapy, the patient demonstrated significant tumor reduction; however, subsequent irAEs related to immunotherapy emerged, including eyelid muscle weakness and myocardial and skeletal muscle injury. Methylprednisolone successfully alleviated these symptoms, with early intervention being crucial for controlling irAEs. The patient then underwent PDT, which not only further helped manage irAEs but also inhibited tumor progression. This case underscores the specific adverse reactions, such as eyelid ptosis, skeletal muscle, and myocardial damage associated with Tislelizumab, and the importance of early corticosteroid intervention. It also emphasizes the significance of PDT as an adjunctive treatment for controlling tumors and alleviating immune-related adverse reactions.
Collapse
Affiliation(s)
- Longzhao Li
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Integrative Traditional Chinese and Western Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Lingjie Bian
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Na Kou
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Yuan
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Heng Zou
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
11
|
Zhang M, Sun D, Huang H, Yang D, Song X, Feng W, Jing X, Chen Y. Nanosonosensitizer Optimization for Enhanced Sonodynamic Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409663. [PMID: 39308222 DOI: 10.1002/adma.202409663] [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: 07/05/2024] [Revised: 08/16/2024] [Indexed: 11/16/2024]
Abstract
Low-intensity ultrasound-mediated sonodynamic therapy (SDT), which, by design, integrates sonosensitizers and molecular oxygen to generate therapeutic substances (e.g., toxic hydroxyl radicals, superoxide anions, or singlet oxygen) at disease sites, has shown enormous potential for the effective treatment of a variety of diseases. Nanoscale sonosensitizers play a crucial role in the SDT process because their structural, compositional, physicochemical, and biological characteristics are key determinants of therapeutic efficacy. In particular, advances in materials science and nanotechnology have invigorated a series of optimization strategies for augmenting the therapeutic efficacy of nanosonosensitizers. This comprehensive review systematically summarizes, discusses, and highlights state-of-the-art studies on the current achievements of nanosonosensitizer optimization in enhanced sonodynamic disease treatment, with an emphasis on the general design principles of nanosonosensitizers and their optimization strategies, mainly including organic and inorganic nanosonosensitizers. Additionally, recent advancements in optimized nanosonosensitizers for therapeutic applications aimed at treating various diseases, such as cancer, bacterial infections, atherosclerosis, and autoimmune diseases, are clarified in detail. Furthermore, the biological effects of the improved nanosonosensitizers for versatile SDT applications are thoroughly discussed. The review concludes by highlighting the current challenges and future opportunities in this rapidly evolving research field to expedite its practical clinical translation and application.
Collapse
Affiliation(s)
- Min Zhang
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Dandan Sun
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Hui Huang
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Dayan Yang
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Xinran Song
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Wei Feng
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiangxiang Jing
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Yu Chen
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| |
Collapse
|
12
|
Qiao K, Pan Y, Zhang S, Shi G, Yang J, Zhang Z, Wang K, Chen X, Ning S. Cold Exposure Therapy Sensitizes Nanodrug-Mediated Radioimmunotherapy of Breast Cancer. ACS NANO 2024; 18:29689-29703. [PMID: 39401104 DOI: 10.1021/acsnano.4c09021] [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: 10/30/2024]
Abstract
Cold exposure (CE) therapy can quickly induce tumor starvation by brown adipose tissue (BAT) thermogenesis. Exploring the combined antitumor mechanism of CE and traditional therapies (such as radiotherapy (RT)) is exciting and promising. In this study, we investigated the effect of CE in combination with nitric oxide (NO) gas therapy on sensitizing tumors to RT and promoting tumor radio-immunotherapy. We first constructed a liposome (SL) loaded with the NO prodrug S-nitroso-N-acetylpenicillamine (SNAP). When SL is injected, the glutathione (GSH) within the tumor region promotes the release of NO from SNAP. Subsequently, the superoxide anion produced by RT reacts with NO to generate peroxynitrite (ONOO-), which has strong oxidative properties and induces cell death. Meanwhile, the mice were exposed to a CE environment of 4 °C. CE-mediated BAT thermogenesis induced tumor starvation, which led to a decrease in ATP and GSH content within the tumor as well as an improvement in the hypoxic microenvironment and a decrease in myeloid-derived suppressor cells. All of the above have promoted the effectiveness of RT and activated the systemic antitumor immunity. In the bilateral tumor experiment, treatment of the primary tumor inhibited the growth of the distant tumor and promoted the infiltration of CD8+ T cells into the tumor. These findings reveal that the synergy of CE, NO gas therapy, and RT could confer high effective anticancer effects, providing possibilities in personalized cancer treatment.
Collapse
Affiliation(s)
- Kun Qiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - You Pan
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Shiyuan Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Guangfu Shi
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Jinglin Yang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Zhenlin Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| |
Collapse
|
13
|
Yu N, Zhou J, Xu H, Wang F, Wang X, Tang L, Li J, Wang X, Lu X. Near-infrared photoactivatable three-in-one nanoagents to aggravate hypoxia and enable amplified photo-chemotherapy. BIOMATERIALS ADVANCES 2024; 163:213962. [PMID: 39032435 DOI: 10.1016/j.bioadv.2024.213962] [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: 04/28/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Solid tumors create a hypoxic microenvironment and this character can be utilized for cancer therapy, but the hypoxia levels are insufficient to achieve satisfactory therapeutic benefits. Some tactics have been used to improve hypoxia, which however will cause side effects due to the uncontrolled drug release. We herein report near-infrared (NIR) photoactivatable three-in-one nanoagents (PCT) to aggravate tumor hypoxia and enable amplified photo-combinational chemotherapy. PCT are formed based on a thermal-responsive liposome nanoparticle containing three therapeutic agents: a hypoxia responsive prodrug tirapazamine (TPZ) for chemotherapy, a vascular targeting agent combretastatin A-4 (CA4) for vascular disturbance and a semiconducting polymer for both photodynamic therapy (PDT) and photothermal therapy (PTT). With NIR laser irradiation, PCT generate heat for PTT and destructing thermal-responsive liposomes to achieve activatable releases of TPZ and CA4. Moreover, PCT produce singlet oxygen (1O2) for PDT via consuming tumor oxygen. CA4 can disturb the blood vessels in tumor microenvironment to aggravate the hypoxic microenvironment, which results in the activation of TPZ for amplified chemotherapy. PCT thus enable PTT, PDT and hypoxia-amplified chemotherapy to afford a high therapeutic efficacy to almost absolutely eradicate subcutaneous 4 T1 tumors and effectively inhibit tumor metastases in lung and liver. This work presents an activatable three-in-one therapeutic nanoplatform with remotely controllable and efficient therapeutic actions to treat cancer.
Collapse
Affiliation(s)
- Ningyue Yu
- Department of Nuclear Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, China; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Jianhui Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Haiming Xu
- Anorectal surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - 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
| | - Xing Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Liming Tang
- Gastrointestinal Surgery Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China.
| | - 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.
| | - Xiaoying Wang
- Office of Hospital Infection and Disease Control and Prevention, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Xia Lu
- Department of Nuclear Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
14
|
Chen Z, Liu Z, Zhou Y, Rao K, Lin J, Zhu D, Ning S, Wang H. Bionic aggregation-induced emission photosensitizer for enhanced cancer immunotherapy. Mater Today Bio 2024; 28:101217. [PMID: 39285944 PMCID: PMC11402640 DOI: 10.1016/j.mtbio.2024.101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/08/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Cold exposure therapy (CE), as an inexpensive method, has shown great potential in cancer therapy. Exploring the combined anti-tumor mechanism of CE and traditional therapies (such as photodynamic therapy (PDT)) is exciting and promising. Here, a bionic aggregation-induced emission photosensitizer system (named THL) is designed for combined CE to enhance anti-tumor immunotherapy. THL inherits the homologous targeting ability of tumor derived exosomes, promoting the enrichment of THL at the tumor site. Under external illumination, THL generates hydroxyl radicals and superoxide anions through type I PDT. In addition, mice are pretreated with cold exposure, which promotes THL mediated PDT and reactive oxygen species (ROS) generation by reducing the production of ATP and GSH in tumor tissue. This combination therapy increases production of ROS within the tumor, inhibits the growth of distant tumors, recurrent and rechallenged tumors and increases the number of cytotoxic CD8+T cells and memory T cells. Compared to PDT alone, combination therapy shows greater advantages in tumor immunotherapy. The combination therapy strategy provides new ideas for cancer immunotherapy.
Collapse
Affiliation(s)
- Zhongxian Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zeming Liu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yingguang Zhou
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Orthopaedic Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Kexiang Rao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Daoming Zhu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Hongbin Wang
- The Second Ward of Breast Surgery, Cancer Hospital Affiliated to Harbin Medical University, Harbin, 150000, China
| |
Collapse
|
15
|
Bharadwaj D, Mandal M. Tumor microenvironment: A playground for cells from multiple diverse origins. Biochim Biophys Acta Rev Cancer 2024; 1879:189158. [PMID: 39032537 DOI: 10.1016/j.bbcan.2024.189158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Tumor microenvironment is formed by various cellular and non-cellular components which interact with one another and form a complex network of interactions. Some of these cellular components also attain a secretory phenotype and release growth factors, cytokines, chemokines etc. in the surroundings which are capable of inducing even greater number of signalling networks. All these interactions play a decisive role in determining the course of tumorigenesis. The treatment strategies against cancer also exert their impact on the local microenvironment. Such interactions and anticancer therapies have been found to induce more deleterious outcomes like immunosuppression and chemoresistance in the process of tumor progression. Hence, understanding the tumor microenvironment is crucial for dealing with cancer and chemoresistance. This review is an attempt to develop some understanding about the tumor microenvironment and different factors which modulate it, thereby contributing to tumorigenesis. Along with summarising the major components of tumor microenvironment and various interactions taking place between them, it also throws some light on how the existing and potential therapies exert their impact on these dynamics.
Collapse
Affiliation(s)
- Deblina Bharadwaj
- Department of Biotechnology, KIT-Kalaignarkarunanidhi Institute of Technology, Coimbatore, Tamil Nadu, India.
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
| |
Collapse
|
16
|
Soltanmohammadi F, Gharehbaba AM, Zangi AR, Adibkia K, Javadzadeh Y. Current knowledge of hybrid nanoplatforms composed of exosomes and organic/inorganic nanoparticles for disease treatment and cell/tissue imaging. Biomed Pharmacother 2024; 178:117248. [PMID: 39098179 DOI: 10.1016/j.biopha.2024.117248] [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: 05/18/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024] Open
Abstract
Exosome-nanoparticle hybrid nanoplatforms, can be prepared by combining exosomes with different types of nanoparticles. The main purpose of combining exosomes with nanoparticles is to overcome the limitations of using each of them as drug delivery systems. Using nanoparticles for drug delivery has some limitations, such as high immunogenicity, poor cellular uptake, low biocompatibility, cytotoxicity, low stability, and rapid clearance by immune cells. However, using exosomes as drug delivery systems also has its own drawbacks, such as poor encapsulation efficiency, low production yield, and the inability to load large molecules. These limitations can be addressed by utilizing hybrid nanoplatforms. Additionally, the use of exosomes allows for targeted delivery within the hybrid system. Exosome-inorganic/organic hybrid nanoparticles may be used for both therapy and diagnosis in the future. This may lead to the development of personalized medicine using hybrid nanoparticles. However, there are a few challenges associated with this. Surface modifications, adding functional groups, surface charge adjustments, and preparing nanoparticles with the desired size are crucial to the possibility of preparing exosome-nanoparticle hybrids. Additional challenges for the successful implementation of hybrid platforms in medical treatments and diagnostics include scaling up the manufacturing process and ensuring consistent quality and reproducibility across various batches. This review focuses on various types of exosome-nanoparticle hybrid systems and also discusses the preparation and loading methods for these hybrid nanoplatforms. Furthermore, the potential applications of these hybrid nanocarriers in drug/gene delivery, disease treatment and diagnosis, and cell/tissue imaging are explained.
Collapse
Affiliation(s)
- Fatemeh Soltanmohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Mahmoudi Gharehbaba
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Rajabi Zangi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khosro Adibkia
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Javadzadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Yu H, Huang Y, Nong Z, Lin X, Tang K, Cai Z, Huang K, Yu T, Lan H, Zhang Q, Wang Q, Yang L, Zhu J, Wu L, Luo H. In-Situ Grown Nanocrystal TiO 2 on 2D Ti 3C 2 Nanosheets with Anti-Tumor Activity from Photo-Sonodynamic Treatment and Immunology. Int J Nanomedicine 2024; 19:7963-7981. [PMID: 39130689 PMCID: PMC11316479 DOI: 10.2147/ijn.s457112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Traditional cancer treatment strategies often have severe toxic side effects and poor therapeutic efficacy. To address the long-standing problems related to overcoming the complexity of tumors, we develop a novel nanozyme based on the in situ oxidation of 2D Ti3C2 structure to perform simultaneous phototherapy and sonodynamic therapy on tumors. Ti3C2 nanozymes exhibit multi-enzyme activity, including intrinsic peroxidase (POD) activities, which can react with H2O2 in the tumor microenvironment. This new material can construct Ti3C2/TiO2 heterostructures in vivo. Methods Photothermal (PTT), sonodynamic (SDT) effects, and photoacoustic (PA) image-guided synergy therapy can be achieved. Finally, anticancer immune responses occur with this nanozyme. In vivo experiments revealed that the Ti3C2/TiO2 heterostructure inhibited tumor growth. Results Complementarily, our results showed that the Ti3C2/TiO2 heterostructure enhanced the immunogenic activity of tumors by recruiting cytotoxic T cells, thereby enhancing the tumor ablation effect. Mechanistic studies consistently indicated that Reactive Oxygen Species (ROS) regulates apoptosis of HCC cells by modulating NRF2/OSGIN1 signaling both in vitro and in vivo. As a result, Ti3C2 nanozyme effectively inhibited tumor through its synergistic ability to modulate ROS and enhance immune infiltration of cytotoxic T cells in the tumor microenvironment. Discussion These findings open up new avenues for enhancing 2D Ti3C2 nanosheets and suggest a new way to develop more effective sonosensitizers for the treatment of cancer.
Collapse
Affiliation(s)
- Hailing Yu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Yongquan Huang
- Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Zhisheng Nong
- School of Materials Science and Engineering, Shenyang Aerospace University, Shenyang, Liaoning, People’s Republic of China
| | - Xi Lin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Kexin Tang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Zeyu Cai
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Kaichen Huang
- Department of Clinical laboratory, The Third People’s Hospital of Zhuhai, Zhuhai, Guangdong, People’s Republic of China
| | - Ting Yu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Huimin Lan
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Qianqian Zhang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Qiang Wang
- The Green Aerotechnics Research Institute of Chongqing Jiaotong University, Chongqing, People’s Republic of China
| | - Lei Yang
- Center for Composite Materials and Structures, Harbin Institute of Technology, Harbin, Heilongjiang, People’s Republic of China
| | - Jingchuan Zhu
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, People’s Republic of China
| | - Lili Wu
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, School of Physics and Electronic Engineering, Harbin Normal University, Harbin, Heilongjiang, People’s Republic of China
| | - Hui Luo
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| |
Collapse
|
18
|
Lai JM, Chen PL, Shi QY, Xie YQ, Jiaerheng G, Liu LH. A Self-Delivery Nanodrug Simultaneously Inhibits COX-2/PGE 2 Mediated Inflammation and Downregulates PD-L1 to Boost Photoimmunotherapy. Adv Healthc Mater 2024; 13:e2400367. [PMID: 38704750 DOI: 10.1002/adhm.202400367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/11/2024] [Indexed: 05/07/2024]
Abstract
Phototherapy promotes anti-tumor immunity by inducing immunogenic cell death (ICD), However, the accompanying inflammatory responses also trigger immunosuppression, attenuating the efficacy of photo-immunotherapy. Herein, they co-assembled a cell-membrane targeting chimeric peptide C16-Cypate-RRKK-PEG8-COOH (CCP) and anti-inflammatory diclofenac (DA) to develop a nanodrug (CCP@DA) that both enhances the immune effect of phototherapy and weakens the inflammation-mediated immunosuppression. CCP@DA achieves cell membrane-targeting photodynamic and photothermal synergistic therapies to damage programmed death ligand 1 (PD-L1) and induce a strong ICD to activate anti-tumor response. Simultaneously, the released DA inhibits the cycoperoxidase-2 (COX-2)/prostaglandin E2 (PGE2) pathway in tumor cells to inhibit pro-tumor inflammation and further down-regulate PD-L1 expression to relieve the immunosuppressive microenvironment. CCP@DA significantly inhibited tumor growth and inflammation both in vitro and in vivo, while maintaining a potent anti-tumor immune response. Additionally, it exhibits excellent anti-metastatic capabilities and prolongs mouse survival time with a single dose and low levels of near-infrared (NIR) light exposure. This work provides a valuable strategy to control the therapy-induced inflammation for high-efficiency photoimmunotherapy.
Collapse
Affiliation(s)
- Jin-Mei Lai
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Pei-Ling Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Qun-Ying Shi
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yong-Qi Xie
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - GuliJiayina Jiaerheng
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Li-Han Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
19
|
He L, Chen Q, Lu Q, Yang M, Xie B, Chen T, Wang X. Autophagy-Inducing MoO 3-x Nanowires Boost Photothermal-Triggered Cancer Immunotherapy. Angew Chem Int Ed Engl 2024; 63:e202404822. [PMID: 38687056 DOI: 10.1002/anie.202404822] [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: 03/10/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/02/2024]
Abstract
Autophagy could play suppressing role in cancer therapy by facilitating release of tumor antigens from dying cells and inducing immunogenic cell death (ICD). Therefore, discovery and rational design of more effective inducers of cytotoxic autophagy is expected to develop new strategies for finding innovative drugs for precise and successful cancer treatment. Herein, we develop MoO3-x nanowires (MoO3-x NWs) with high oxygen vacancy and strong photothermal responsivity to ablate tumors through hyperthermia, thus promote the induction of cytotoxic autophagy and severe ICD. As expected, the combination of MoO3-x NWs and photothermal therapy (PTT) effectively induces autophagy to promote the release of tumor antigens from the ablated cells, and induces the maturation and antigen presentation of dendritic cells (DCs), subsequently activates cytotoxic T lymphocytes (CTLs)-mediated adaptive immunity. Furthermore, the combination treatment of MoO3-x NWs with immune checkpoint blockade of PD-1 could promote the tumor-associated macrophages (TAMs) polarization into tumor-killing M1 macrophages, inhibit infiltration of Treg cells at tumor sites, and alleviate immunosuppression in the tumor microenvironment, finally intensify the anti-tumor activity in vivo. This study provides a strategy and preliminary elucidation of the mechanism of using MoO3-x nanowires with high oxygen vacancy to induce autophagy and thus enhance photothermal immunotherapy.
Collapse
Affiliation(s)
- Lizhen He
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Qi Chen
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Qichen Lu
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Meijin Yang
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Bin Xie
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Xun Wang
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
20
|
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
|
21
|
Zhang N, Ping W, Rao K, Zhang Z, Huang R, Zhu D, Li G, Ning S. Biomimetic copper-doped polypyrrole nanoparticles induce glutamine metabolism inhibition to enhance breast cancer cuproptosis and immunotherapy. J Control Release 2024; 371:204-215. [PMID: 38810704 DOI: 10.1016/j.jconrel.2024.05.045] [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: 03/24/2024] [Revised: 05/10/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Cuproptosis, a newly discovered mechanism of inducing tumor cell death, primarily relies on the intracellular accumulation of copper ions. The utilization of Cu-based nanomaterials to induce cuproptosis holds promising prospects in future biomedical applications. However, the presence of high levels of glutathione (GSH) within tumor cells hinders the efficacy of cuproptosis. In this study, we have developed a BPTES-loaded biomimetic Cu-doped polypyrrole nanoparticles (CuP) nanosystem (PCB) for enhanced cuproptosis and immune modulation. PCB comprises an internal BPTES and CuP core and an external platelet membrane (PM) that facilitates active targeting to tumor sites following intravenous administration. Subsequently, PCB effectively suppresses glutaminase (GLS1) activity, thereby reducing GSH content. Moreover, CuP catalyze intracellular H2O2, amplifying oxidative stress while simultaneously inducing dihydrolipoyl transacetylase (DLAT) oligomerization through released Cu2+, resulting in cuproptosis. PCB not only inhibits primary tumors but also exhibits inhibitory effects on abscopal tumors. This work represents the first instance where GLS inhibition has been employed to enhance cuproptosis and immunotherapy. It also provides valuable insights into further investigations on cuproptosis.
Collapse
Affiliation(s)
- Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Ping
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kexiang Rao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhenlin Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Rong Huang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Daoming Zhu
- Department of Electronic Science and Technology, School of Physics and Technology, Wuhan University, Wuhan, 430072, China; Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Guoxin Li
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No.168 Litang Road, Changping District, Beijing, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China.
| |
Collapse
|
22
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
23
|
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
|
24
|
Shan T, Wang W, Fan M, Bi J, He T, Sun Y, Zheng M, Yan D. Effective glioblastoma immune sonodynamic treatment mediated by macrophage cell membrane cloaked biomimetic nanomedicines. J Control Release 2024; 370:866-878. [PMID: 38685386 DOI: 10.1016/j.jconrel.2024.04.043] [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: 12/04/2023] [Revised: 04/16/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Glioblastoma (GBM) as one of the most lethal brain tumours, remains poor therapeutic index due to its typical characters including heterogeneous, severe immune suppression as well as the existence of blood brain barrier (BBB). Immune sonodynamic (ISD) therapy combines noninvasive sonodynamic therapy with immunotherapy, which has great prospects for the combinational treatment of GBM. Herein, we develop macrophage cell membrane cloaked reactive oxygen species (ROS) responsive biomimetic nanoparticles, co-delivering of sonosensitizer Ce6 and JQ1 (a bromo-domain protein 4 (BRD4) inhibitor which can down-regulate PD-L1) and realizing potent GBM ISD therapy. The ApoE peptide decorated macrophage membrane coating endows these biomimetic nanoparticles with low immunogenicity, efficient BBB permeability, prolonged blood circulation half-live and good biocompatibility. The ROS responsive polymeric inner core could be readily degraded as triggered by excessive ROS under the ultrasound once they accumulated in tumour cells, fast release encapsulated drugs. The generation of ROS not only killed tumour cells via sonodynamic therapy, but also induced immunogenic cell death (ICD) and further activated the anti-tumour immune response. The released JQ1 inhibited tumour cell proliferation and augmented the immune activities by inhibiting the PD-L1 expression on the surface of tumour cells. The cascade sonodynamic and immune therapy resulted in significantly improved median survival time in both orthotopic GL261 and PTEN deficient immunosuppressive CT2A GBM mice models. Therefore, our developed biomimetic nanoparticle platform provides a promising combinational therapy strategy to treat immune suppressive GBM.
Collapse
Affiliation(s)
- Tikun Shan
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wendie Wang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Mengyu Fan
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiajia Bi
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Tengfei He
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yajing Sun
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| | - Dongming Yan
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China..
| |
Collapse
|
25
|
Hong L, Xu K, Yang M, Zhu L, Chen C, Xu L, Zhu W, Jin L, Wang L, Lin J, Wang J, Ren W, Wu A. VISTA antibody-loaded Fe 3O 4@TiO 2 nanoparticles for sonodynamic therapy-synergistic immune checkpoint therapy of pancreatic cancer. Mater Today Bio 2024; 26:101106. [PMID: 38883421 PMCID: PMC11176928 DOI: 10.1016/j.mtbio.2024.101106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Breaking the poor permeability of immune checkpoint inhibitors (ICIs) caused by the stromal barrier and reversing the immunosuppressive microenvironment are significant challenges in pancreatic cancer immunotherapy. In this study, we synthesized core-shell Fe3O4@TiO2 nanoparticles to act as carriers for loading VISTA monoclonal antibodies to form Fe3O4@TiO2@VISTAmAb (FTV). The nanoparticles are designed to target the overexpressed ICIs VISTA in pancreatic cancer, aiming to improve magnetic resonance imaging-guided sonodynamic therapy (SDT)-facilitated immunotherapy. Laser confocal microscopy and flow cytometry results demonstrate that FTV nanoparticles are specifically recognized and phagocytosed by Panc-2 cells. In vivo experiments reveal that ultrasound-triggered TiO2 SDT can induce tumor immunogenic cell death (ICD) and recruit T-cell infiltration within the tumor microenvironment by releasing damage-associated molecular patterns (DAMPs). Furthermore, ultrasound loosens the dense fibrous stroma surrounding the pancreatic tumor and increases vascular density, facilitating immune therapeutic efficiency. In summary, our study demonstrates that FTV nanoparticles hold great promise for synergistic SDT and immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Lu Hong
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Kaiwei Xu
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Ming Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Lubing Zhu
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Chunqu Chen
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Liu Xu
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
| | - Weihao Zhu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Lufei Jin
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
| | - Linwei Wang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Jie Lin
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, PR China
| | - Jianhua Wang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, PR China
- Health Science Center, Ningbo University, Ningbo, 315210, PR China
| | - Wenzhi Ren
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, PR China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, PR China
| |
Collapse
|
26
|
Feng Y, Yang Z, Wang J, Zhao H. Cuproptosis: unveiling a new frontier in cancer biology and therapeutics. Cell Commun Signal 2024; 22:249. [PMID: 38693584 PMCID: PMC11064406 DOI: 10.1186/s12964-024-01625-7] [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: 02/26/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024] Open
Abstract
Copper plays vital roles in numerous cellular processes and its imbalance can lead to oxidative stress and dysfunction. Recent research has unveiled a unique form of copper-induced cell death, termed cuproptosis, which differs from known cell death mechanisms. This process involves the interaction of copper with lipoylated tricarboxylic acid cycle enzymes, causing protein aggregation and cell death. Recently, a growing number of studies have explored the link between cuproptosis and cancer development. This review comprehensively examines the systemic and cellular metabolism of copper, including tumor-related signaling pathways influenced by copper. It delves into the discovery and mechanisms of cuproptosis and its connection to various cancers. Additionally, the review suggests potential cancer treatments using copper ionophores that induce cuproptosis, in combination with small molecule drugs, for precision therapy in specific cancer types.
Collapse
Affiliation(s)
- Ying Feng
- Department of Emergency, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China
| | - Zhibo Yang
- Department of Neurosurgery, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong, 723000, Shaanxi, China
| | - Jianpeng Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China.
| |
Collapse
|
27
|
Guo Z, Ye J, Cheng X, Wang T, Zhang Y, Yang K, Du S, Li P. Nanodrug Delivery Systems in Antitumor Immunotherapy. Biomater Res 2024; 28:0015. [PMID: 38840653 PMCID: PMC11045275 DOI: 10.34133/bmr.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/26/2024] [Indexed: 06/07/2024] Open
Abstract
Cancer has become one of the most important factors threatening human health, and the global cancer burden has been increasing rapidly. Immunotherapy has become another clinical research hotspot after surgery, chemotherapy, and radiotherapy because of its high efficiency and tumor metastasis prevention. However, problems such as lower immune response rate and immune-related adverse reaction in the clinical application of immunotherapy need to be urgently solved. With the development of nanodrug delivery systems, various nanocarrier materials have been used in the research of antitumor immunotherapy with encouraging therapeutic results. In this review, we mainly summarized the combination of nanodrug delivery systems and immunotherapy from the following 4 aspects: (a) nanodrug delivery systems combined with cytokine therapy to improve cytokines delivery in vivo; (b) nanodrug delivery systems provided a suitable platform for the combination of immune checkpoint blockade therapy with other tumor treatments; (c) nanodrug delivery systems helped deliver antigens and adjuvants for tumor vaccines to enhance immune effects; and (d) nanodrug delivery systems improved tumor treatment efficiency and reduced toxicity for adoptive cell therapy. Nanomaterials chosen by researchers to construct nanodrug delivery systems and their function were also introduced in detail. Finally, we discussed the current challenges and future prospects in combining nanodrug delivery systems with immunotherapy.
Collapse
Affiliation(s)
- Zishuo Guo
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinhong Ye
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xuehao Cheng
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tieshan Wang
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- YiDu Central Hospital of Weifang, Weifang, Shandong 262500, China
| | - Kaili Yang
- Beijing University of Chinese Medicine, Beijing 102488, China
| | | | - Pengyue Li
- Address correspondence to: (P.L.); (S.D.)
| |
Collapse
|
28
|
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
|
29
|
Huang Y, Ouyang W, Lai Z, Qiu G, Bu Z, Zhu X, Wang Q, Yu Y, Liu J. Nanotechnology-enabled sonodynamic therapy against malignant tumors. NANOSCALE ADVANCES 2024; 6:1974-1991. [PMID: 38633037 PMCID: PMC11019498 DOI: 10.1039/d3na00738c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/09/2024] [Indexed: 04/19/2024]
Abstract
Sonodynamic therapy (SDT) is an emerging approach for malignant tumor treatment, offering high precision, deep tissue penetration, and minimal side effects. The rapid advancements in nanotechnology, particularly in cancer treatment, have enhanced the efficacy and targeting specificity of SDT. Combining sonodynamic therapy with nanotechnology offers a promising direction for future cancer treatments. In this review, we first systematically discussed the anti-tumor mechanism of SDT and then summarized the common nanotechnology-related sonosensitizers and their recent applications. Subsequently, nanotechnology-related therapies derived using the SDT mechanism were elaborated. Finally, the role of nanomaterials in SDT combined therapy was also introduced.
Collapse
Affiliation(s)
- Yunxi Huang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Yat-sen Supercomputer Intelligent Medical Joint Research Institute, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University 510120 Guangzhou China
| | - Zijia Lai
- First Clinical Medical College, Guangdong Medical University 524000 Zhanjiang China
| | - Guanhua Qiu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Zhaoting Bu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Xiaoqi Zhu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Qin Wang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Yat-sen Supercomputer Intelligent Medical Joint Research Institute, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University 510120 Guangzhou China
- Faculty of Medicine, Macau University of Science and Technology Taipa Macao PR China
| | - Junjie Liu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital 77 He Di Road 530021 Nanning China
| |
Collapse
|
30
|
Lu XX, Xue C, Dong JH, Zhang YZ, Gao F. Nanoplatform-based strategies for enhancing the lethality of current antitumor PDT. J Mater Chem B 2024; 12:3209-3225. [PMID: 38497405 DOI: 10.1039/d4tb00008k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Photodynamic therapy (PDT) exhibits great application prospects in future clinical oncology due to its spatiotemporal controllability and good biosafety. However, the antitumor efficacy of PDT is seriously hindered by many factors, including tumor hypoxia, limited light penetration ability, and strong defense mechanisms of tumors. Considering that it is difficult to completely solve the first two problems, enhancing the lethality of antitumor PDT has become a good idea to extend its clinical application. Herein, we summarize the nanoplatform-involved strategies to effectively amplify the tumoricidal capability of current PDT and then discuss the present bottlenecks and prospects of the nanoplatform-based PDT sensitization strategies in tumor therapy. We hope this review will provide some references for others to design high-performance PDT nanoplatforms for tumor therapy.
Collapse
Affiliation(s)
- Xin-Xin Lu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Chun Xue
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Jian-Hui Dong
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Yi-Zhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| |
Collapse
|
31
|
Tian S, Li J, Wang D, Han Y, Dai H, Yan L. Sonodynamic-chemotherapy synergy with chlorin e6-based carrier-free nanoparticles for non-small cell lung cancer. J Mater Chem B 2024; 12:3282-3291. [PMID: 38487900 DOI: 10.1039/d4tb00009a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Sonodynamic therapy (SDT), an emerging cancer treatment with significant potential, offers the advantages of non-invasiveness and deep tissue penetrability. The method involves activating sonosensitizers with ultrasound to generate reactive oxygen species (ROS) capable of eradicating cancer cells, addressing the challenge faced by photodynamic therapy (PDT) where conventional light sources struggle to penetrate deep tissues, impacting treatment efficacy. This study addresses prevalent challenges in numerous nanodiagnostic and therapeutic agents, such as intricate synthesis, poor repeatability, low stability, and high cost, by introducing a streamlined one-step assembly method for nanoparticle preparation. Specifically, the sonosensitizer Chlorin e6 (Ce6) and the chemotherapy drug erlotinib are effortlessly combined and self-assembled under sonication, yielding carrier-free nanoparticles (EC-NPs) for non-small cell lung cancer (NSCLC) treatment. The resulting EC-NPs exhibit optimal drug loading capacity, a simplified preparation process, and robust stability both in vitro and in vivo, owing to their carrier-free characteristics. Under the synergistic treatment of sonodynamic therapy and chemotherapy, EC-NPs induce an excess of reactive oxygen in tumor tissue, prompting apoptosis of cancer cells and reducing their proliferative capacity. Both in vitro and in vivo experiments demonstrate superior therapeutic effects of EC-NPs under ultrasound conditions compared to free Ce6. In summary, our research findings highlight that the innovatively designed carrier-free sonosensitizer EC-NPs present a therapeutic option with commendable efficacy and minimal side effects.
Collapse
Affiliation(s)
- Shuangyu Tian
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Canter of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Jinghang Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Canter of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Dongdong Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Canter of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Yingchao Han
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Canter of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Canter of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Canter of Hubei Province, Wuhan University of Technology, Wuhan 430070, China.
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
- Wuhan University of Technology Advanced Engineering Technology Research Institute of Zhongshan City, Zhongshan 528400, China
| |
Collapse
|
32
|
Huang X, Chen Y, Zhong F, Gui B, Hu Y, Guo Y, Deng Q, Zhou Q. Targeted Ultrasound Nanobubbles Therapy for Prostate Cancer via Immuno-Sonodynamic Effect. Int J Nanomedicine 2024; 19:2793-2806. [PMID: 38525011 PMCID: PMC10959301 DOI: 10.2147/ijn.s451179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/07/2024] [Indexed: 03/26/2024] Open
Abstract
Background Prostate cancer (PCa) poses a significant global health threaten. Immunotherapy has emerged as a novel strategy to augment the inhibition of tumor proliferation. However, the sole use of anti-PD-L1 Ab for PCa has not yielded improvements, mirroring outcomes observed in other tumor types. Methods This study employed the thin film hydration method to develop lipid nanobubbles (NBs) encapsulating chlorin e6 (Ce6) and anti-PD-L1 Ab (Ce6@aPD-L1 NBs). Our experimental approach included cellular assays and mouse immunization, providing a comprehensive evaluation of Ce6@aPD-L1 NBs' impact. Results The Ce6@aPD-L1 NBs effectively induced reactive oxygen species generation, leading to tumor cells death. In mice, they demonstrated a remarkable enhancement of immune responses compared to control groups. These immune responses encompassed immunogenic cell death induced by sonodynamic therapy and PD-1/PD-L1 blockade, activating dendritic cells maturation and effectively stimulating CD8+T cells. Conclusion Ce6@aPD-L1 NBs facilitate tumor-targeted delivery, activating anti-tumor effects through direct sonodynamic therapy action and immune system reactivation in the tumor microenvironment. Ce6@aPD-L1 NBs exhibit substantial potential for achieving synergistic anti-cancer effects in PCa.
Collapse
Affiliation(s)
- Xin Huang
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Yueying Chen
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Fanglu Zhong
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Bin Gui
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Yugang Hu
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Yuxin Guo
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Qing Deng
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Qing Zhou
- Department of Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| |
Collapse
|
33
|
Li T, Guo L, Li J, Mu X, Liu L, Song S, Luo N, Zhang Q, Zheng B, Jin G. Precision USPIO-PEG-SLe x Nanotheranostic Agent Targeted Photothermal Therapy for Enhanced Anti-PD-L1 Immunotherapy to Treat Immunotherapy Resistance. Int J Nanomedicine 2024; 19:1249-1272. [PMID: 38348177 PMCID: PMC10859766 DOI: 10.2147/ijn.s445879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024] Open
Abstract
Background The anti-Programmed Death-Ligand 1 (termed aPD-L1) immune checkpoint blockade therapy has emerged as a promising treatment approach for various advanced solid tumors. However, the effect of aPD-L1 inhibitors limited by the tumor microenvironment makes most patients exhibit immunotherapy resistance. Methods We conjugated the Sialyl Lewis X with a polyethylene glycol-coated ultrasmall superparamagnetic iron oxide (USPIO-PEG) to form UPS nanoparticles (USPIO-PEG-SLex, termed UPS). The physicochemical properties of UPS were tested and characterized. Transmission electron microscopy and ICP-OES were used to observe the cellular uptake and targeting ability of UPS. Flow cytometry, mitochondrial membrane potential staining, live-dead staining and scratch assay were used to verify the in vitro photothermal effect of UPS, and the stimulation of UPS on immune-related pathways at the gene level was analyzed by sequencing. Biological safety analysis and pharmacokinetic analysis of UPS were performed. Finally, the amplification effect of UPS-mediated photothermal therapy on aPD-L1-mediated immunotherapy and the corresponding mechanism were studied. Results In vitro experiments showed that UPS had strong photothermal therapy ability and was able to stimulate 5 immune-related pathways. In vivo, when the PTT assisted aPD-L1 treatment, it exhibited a significant increase in CD4+ T cell infiltration by 14.46-fold and CD8+ T cell infiltration by 14.79-fold, along with elevated secretion of tumor necrosis factor-alpha and interferon-gamma, comparing with alone aPD-L1. This PTT assisted aPD-L1 therapy achieved a significant inhibition of both primary tumors and distant tumors compared to the alone aPD-L1, demonstrating a significant difference. Conclusion The nanotheranostic agent UPS has been introduced into immunotherapy, which has effectively broadened its application in biomedicine. This photothermal therapeutic approach of the UPS nanotheranostic agent enhancing the efficacy of aPD-L1 immune checkpoint blockade therapy, can be instructive to address the challenges associated with immunotherapy resistance, thereby offering potential for clinical translation.
Collapse
Affiliation(s)
- Ting Li
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Lianshan Guo
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People’s Republic of China
| | - Jiaxu Li
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, College of Chemistry and Materials, Graduate School, Nanning Normal University, Nanning, 530001, People’s Republic of China
| | - Xingyu Mu
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Lijuan Liu
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Shulin Song
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Ningbin Luo
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Qi Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, People’s Republic of China
| | - Bin Zheng
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Guanqiao Jin
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| |
Collapse
|
34
|
Chang M, Zhang L, Wang Z, Chen L, Dong Y, Yang J, Chen Y. Nanomedicine/materdicine-enabled sonocatalytic therapy. Adv Drug Deliv Rev 2024; 205:115160. [PMID: 38110153 DOI: 10.1016/j.addr.2023.115160] [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: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/20/2023]
Abstract
The advent of numerous treatment modalities with desirable therapeutic efficacy has been made possible by the fast development of nanomedicine and materdicine, among which the ultrasound (US)-triggered sonocatalytic process as minimal or non-invasive method has been frequently employed for diagnostic and therapeutic purposes. In comparison to phototherapeutic approaches with inherent penetration depth limitations, sonocatalytic therapy shatters the depth limit of photoactivation and offers numerous remarkable prospects and advantages, including mitigated side effects and appropriate tissue-penetration depth. Nevertheless, the optimization of sonosensitizers and therapies remains a significant issue in terms of precision, intelligence and efficiency. In light of the fact that nanomedicine and materdicine can effectively enhance the theranostic efficiency, we herein aim to furnish a cutting-edge review on the latest progress and development of nanomedicine/materdicine-enabled sonocatalytic therapy. The design methodologies and biological features of nanomedicine/materdicine-based sonosensitizers are initially introduced to reveal the underlying relationship between composition/structure, sonocatalytic function and biological effect, in accompany with a thorough discussion of nanomedicine/materdicine-enabled synergistic therapy. Ultimately, the facing challenges and future perspectives of this intriguing sonocatalytic therapy are highlighted and outlined to promote technological advancements and clinical translation in efficient disease treatment.
Collapse
Affiliation(s)
- Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, PR China
| | - Lu Zhang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Hebei University, Baoding 071000, PR China
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yang Dong
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| | - Jishun Yang
- Naval Medical Center of PLA, Medical Security Center, Shanghai 200052, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
35
|
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
|
36
|
Zu M, Ma Y, Zhang J, Sun J, Shahbazi MA, Pan G, Reis RL, Kundu SC, Liu J, Xiao B. An Oral Nanomedicine Elicits In Situ Vaccination Effect against Colorectal Cancer. ACS NANO 2024; 18:3651-3668. [PMID: 38241481 DOI: 10.1021/acsnano.3c11436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Abstract
Oral administration is the most preferred approach for treating colon diseases, and in situ vaccination has emerged as a promising cancer therapeutic strategy. However, the lack of effective drug delivery platforms hampered the application of in situ vaccination strategy in oral treatment of colorectal cancer (CRC). Here, we construct an oral core-shell nanomedicine by preparing a silk fibroin-based dual sonosensitizer (chlorin e6, Ce6)- and immunoadjuvant (imiquimod, R837)-loaded nanoparticle as the core, with its surface coated with plant-extracted lipids and pluronic F127 (p127). The resultant nanomedicines (Ce6/R837@Lp127NPs) maintain stability during their passage through the gastrointestinal tract and exert improved locomotor activities under ultrasound irradiation, achieving efficient colonic mucus infiltration and specific tumor penetration. Thereafter, Ce6/R837@Lp127NPs induce immunogenic death of colorectal tumor cells by sonodynamic treatment, and the generated neoantigens in the presence of R837 serve as a potent in situ vaccine. By integrating with immune checkpoint blockades, the combined treatment modality inhibits orthotopic tumors, eradicates distant tumors, and modulates intestinal microbiota. As the first oral in situ vaccination, this work spotlights a robust oral nanoplatform for producing a personalized vaccine against CRC.
Collapse
Affiliation(s)
- Menghang Zu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Ya Ma
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Jun Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7LD, U.K
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco 4805-017, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga 4800-058, Guimarães, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco 4805-017, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga 4800-058, Guimarães, Portugal
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bo Xiao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
37
|
Liu T, Chai S, Li M, Chen X, Xie Y, Zhao Z, Xie J, Yu Y, Gao F, Zhu F, Yang L. A nanoparticle-based sonodynamic therapy reduces Helicobacter pylori infection in mouse without disrupting gut microbiota. Nat Commun 2024; 15:844. [PMID: 38286999 PMCID: PMC10825188 DOI: 10.1038/s41467-024-45156-8] [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: 03/29/2023] [Accepted: 01/15/2024] [Indexed: 01/31/2024] Open
Abstract
Infection by Helicobacter pylori, a prevalent global pathogen, currently requires antibiotic-based treatments, which often lead to antimicrobial resistance and gut microbiota dysbiosis. Here, we develop a non-antibiotic approach using sonodynamic therapy mediated by a lecithin bilayer-coated poly(lactic-co-glycolic) nanoparticle preloaded with verteporfin, Ver-PLGA@Lecithin, in conjunction with localized ultrasound exposure of a dosage permissible for ultrasound medical devices. This study reveals dual functionality of Ver-PLGA@Lecithin. It effectively neutralizes vacuolating cytotoxin A, a key virulence factor secreted by H. pylori, even in the absence of ultrasound. When coupled with ultrasound exposure, it inactivates H. pylori by generating reactive oxygen species, offering a potential solution to overcome antimicrobial resistance. In female mouse models bearing H. pylori infection, this sonodynamic therapy performs comparably to the standard triple therapy in reducing gastric infection. Significantly, unlike the antibiotic treatments, the sonodynamic therapy does not negatively disrupt gut microbiota, with the only major impact being upregulation of Lactobacillus, which is a bacterium widely used in yogurt products and probiotics. This study presents a promising alternative to the current antibiotic-based therapies for H. pylori infection, offering a reduced risk of antimicrobial resistance and minimal disturbance to the gut microbiota.
Collapse
Affiliation(s)
- Tao Liu
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Shuang Chai
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Mingyang Li
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xu Chen
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yutao Xie
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Zehui Zhao
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Jingjing Xie
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yunpeng Yu
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Feng Gao
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Feng Zhu
- Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Lihua Yang
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China.
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China.
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China.
| |
Collapse
|
38
|
Cui LW, Fan LY, Shen ZY. Application Research Progress of Nanomaterial Graphene and its Derivative Complexes in Tumor Diagnosis and Therapy. Curr Med Chem 2024; 31:6436-6459. [PMID: 38299292 DOI: 10.2174/0109298673251648231106112354] [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: 03/16/2023] [Revised: 08/05/2023] [Accepted: 10/05/2023] [Indexed: 02/02/2024]
Abstract
Functional nanomaterial graphene and its derivatives have attracted considerable attention in many fields because of their unique physical and chemical properties. Most notably, graphene has become a research hotspot in the biomedical field, especially in relation to malignant tumors. In this study, we briefly review relevant research from recent years on graphene and its derivatives in tumor diagnosis and antitumor therapy. The main contents of the study include the graphene-derivative diagnosis of tumors in the early stage, graphene quantum dots, photodynamics, MRI contrast agent, acoustic dynamics, and the effects of ultrasonic cavitation and graphene on tumor therapy. Moreover, the biocompatibility of graphene is briefly described. This review provides a broad overview of the applications of graphene and its derivatives in tumors. Conclusion, graphene and its derivatives play an important role in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Li Wen Cui
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, No. 30, North Tong-yang Road, Pingchao Town, Tongzhou District, Nantong, Jiangsu 226361, China
| | - Lu Yao Fan
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, No. 30, North Tong-yang Road, Pingchao Town, Tongzhou District, Nantong, Jiangsu 226361, China
| | - Zhi Yong Shen
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, No. 30, North Tong-yang Road, Pingchao Town, Tongzhou District, Nantong, Jiangsu 226361, China
| |
Collapse
|
39
|
Zhou Z, Wang H, Li J, Jiang X, Li Z, Shen J. Recent progress, perspectives, and issues of engineered PD-L1 regulation nano-system to better cure tumor: A review. Int J Biol Macromol 2024; 254:127911. [PMID: 37939766 DOI: 10.1016/j.ijbiomac.2023.127911] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Currently, immune checkpoint blockade (ICB) therapies that target the programmed cell death ligand-1 (PD-L1) have been used as revolutionary cancer treatments in the clinic. Apart from restoring the antitumor response of cytotoxic T cells by blocking the interaction between PD-L1 on tumor cells and programmed cell death-1 (PD-1) on T cells, PD-L1 proteins were also newly revealed to possess the capacity to accelerate DNA damage repair (DDR) and enhance tumor growth through multiple mechanisms, leading to the impaired efficacy of tumor therapies. Nevertheless, current free anti-PD-1/PD-L1 therapy still suffered from poor therapeutic outcomes in most solid tumors due to the non-selective tumor accumulation, ineludible severe cytotoxic effects, as well as the common occurrence of immune resistance. Recently, nanoparticles with efficient tumor-targeting capacity, tumor-responsive prosperity, and versatility for combination therapy were identified as new avenues for PD-L1 targeting cancer immunotherapies. In this review, we first summarized the multiple functions of PD-L1 protein in promoting tumor growth, accelerating DDR, as well as depressing immunotherapy efficacy. Following this, the effects and mechanisms of current clinically widespread tumor therapies on tumor PD-L1 expression were discussed. Then, we reviewed the recent advances in nanoparticles for anti-PD-L1 therapy via using PD-L1 antibodies, small interfering RNA (siRNA), microRNA (miRNA), clustered, regularly interspaced, short palindromic repeats (CRISPR), peptide, and small molecular drugs. At last, we discussed the challenges and perspectives to promote the clinical application of nanoparticles-based PD-L1-targeting therapy.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jie Li
- College of Pharmacy, Wenzhou Medical University, Wenzhou 325000, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhangping Li
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| |
Collapse
|
40
|
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
|
41
|
Wang W, Mo W, Hang Z, Huang Y, Yi H, Sun Z, Lei A. Cuproptosis: Harnessing Transition Metal for Cancer Therapy. ACS NANO 2023; 17:19581-19599. [PMID: 37820312 DOI: 10.1021/acsnano.3c07775] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Transition metal elements, such as copper, play diverse and pivotal roles in oncology. They act as constituents of metalloenzymes involved in cellular metabolism, function as signaling molecules to regulate the proliferation and metastasis of tumors, and are integral components of metal-based anticancer drugs. Notably, recent research reveals that excessive copper can also modulate the occurrence of programmed cell death (PCD), known as cuprotosis, in cancer cells. This modulation occurs through the disruption of tumor cell metabolism and the induction of proteotoxic stress. This discovery uncovers a mode of interaction between transition metals and proteins, emphasizing the intricate link between copper homeostasis and tumor metabolism. Moreover, they provide innovative therapeutic strategies for the precise diagnosis and treatment of malignant tumors. At the crossroads of chemistry and oncology, we undertake a comprehensive review of copper homeostasis in tumors, elucidating the molecular mechanisms underpinning cuproptosis. Additionally, we summarize current nanotherapeutic approaches that target cuproptosis and provide an overview of the available laboratory and clinical methods for monitoring this process. In the context of emerging concepts, challenges, and opportunities, we emphasize the significant potential of nanotechnology in the advancement of this field.
Collapse
Affiliation(s)
- Wuyin Wang
- 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, Wuhan University, Wuhan 430079, P. R. China
| | - Wentao Mo
- 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, Wuhan University, Wuhan 430079, P. R. China
| | - Zishan Hang
- 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, Wuhan University, Wuhan 430079, P. R. China
| | - Yueying Huang
- 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, Wuhan University, Wuhan 430079, P. R. China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), Wuhan University, Wuhan 430072, P. R. China
| | - Zhijun Sun
- 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, Wuhan University, Wuhan 430079, P. R. China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, P. R. China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
42
|
Chen Z, Yue Z, Yang K, Shen C, Cheng Z, Zhou X, Li S. Four Ounces Can Move a Thousand Pounds: The Enormous Value of Nanomaterials in Tumor Immunotherapy. Adv Healthc Mater 2023; 12:e2300882. [PMID: 37539730 DOI: 10.1002/adhm.202300882] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 07/17/2023] [Indexed: 08/05/2023]
Abstract
The application of nanomaterials in healthcare has emerged as a promising strategy due to their unique structural diversity, surface properties, and compositional diversity. In particular, nanomaterials have found a significant role in improving drug delivery and inhibiting the growth and metastasis of tumor cells. Moreover, recent studies have highlighted their potential in modulating the tumor microenvironment (TME) and enhancing the activity of immune cells to improve tumor therapy efficacy. Various types of nanomaterials are currently utilized as drug carriers, immunosuppressants, immune activators, immunoassay reagents, and more for tumor immunotherapy. Necessarily, nanomaterials used for tumor immunotherapy can be grouped into two categories: organic and inorganic nanomaterials. Though both have shown the ability to achieve the purpose of tumor immunotherapy, their composition and structural properties result in differences in their mechanisms and modes of action. Organic nanomaterials can be further divided into organic polymers, cell membranes, nanoemulsion-modified, and hydrogel forms. At the same time, inorganic nanomaterials can be broadly classified as nonmetallic and metallic nanomaterials. The current work aims to explore the mechanisms of action of these different types of nanomaterials and their prospects for promoting tumor immunotherapy.
Collapse
Affiliation(s)
- Ziyin Chen
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Ziqi Yue
- Department of Forensic Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Kaiqi Yang
- Clinical Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Congrong Shen
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Zhe Cheng
- Department of Forensic Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Xiaofeng Zhou
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 110042, Shenyang, P. R. China
- The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, Shenyang, 110042, China
| |
Collapse
|
43
|
Zhang W, Li J, Chen L, Chen H, Zhang L. Palladium-based multifunctional nanoparticles for combined chemodynamic/photothermal and calcium overload therapy of tumors. Colloids Surf B Biointerfaces 2023; 230:113529. [PMID: 37708713 DOI: 10.1016/j.colsurfb.2023.113529] [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: 05/22/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023]
Abstract
Due to the high mortality and incidence rates associated with tumors and the specificity of the tumor microenvironment (TME), it is difficult to achieve a complete cure for tumors using a single therapy. In this study, calcium carbonate-modified palladium hydride nanoparticles (PdH@CaCO3) were prepared and utilized for the combined treatment of tumors through chemodynamic therapy (CDT)/photothermal therapy (PTT) and calcium overload therapy. After entering tumor cells, PdH@CaCO3 releases calcium ions (Ca2+) and PdH once it reaches the TME due to the pH reactivity of the calcium carbonate coating. The mitochondrial membrane potential is lowered by the Ca2+, leading to irreversible cell damage. Meanwhile, PdH reacts with excessive hydrogen peroxide (H2O2) in the TME via the Fenton reaction, generating hydroxyl radicals (·OH). Moreover, PdH is an excellent photothermal agent that can kill tumor cells under laser irradiation, leading to significant anti-tumor effects. In vitro and in vivo studies have demonstrated that PdH@CaCO3 could combine CDT/PTT and calcium overload therapy, exhibiting great clinical potential in the treatment of tumors.
Collapse
Affiliation(s)
- Wenge Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Jiangyong Li
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Lamei Chen
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Huan Chen
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Liangke Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
44
|
Wang Y, Lei H, Yan B, Zhang S, Xu B, Lin M, Shuai X, Huang J, Pang J. Tumor acidity-activatable macromolecule autophagy inhibitor and immune checkpoint blockade for robust treatment of prostate cancer. Acta Biomater 2023; 168:593-605. [PMID: 37474083 DOI: 10.1016/j.actbio.2023.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Immune checkpoint blockade (ICB) antibody such as anti-PD-L1 (aPD-L1) activates cytotoxic T cells (CTLs) to combat cancer, but they showed poor efficacy in prostate cancer (PCa). Lysosome-dependent autophagy is utilized by cancer cells to degrade their MHC-I and to lower their vulnerability to TNF-α and CTLs. Lysosomal pH-sensitive polymeric nanoparticle as a drug delivery carrier may also be a novel autophagy inhibitor to boost immunotherapy, but such an important effect has not been investigated. Herein, we developed a unique tumor acidity-activatable macromolecular nanodrug (called P-PDL1-CP) with the poly(2-diisopropylaminoethyl methacrylate) (PDPA) core and the conjugations of both aPD-L1 and long-chain polyethylene glycol (PEG) coating. The PDPA core was demonstrated to disturb lysosome to block the autophagic flux, thus elevating the cancer cell's MHC-I expression and vulnerability to the TNF-α and CTLs. Long-chain PEG facilitated a good tumor accumulation of P-PDL1-CP nanodrug. Furthermore, P-PDL1-CP nanodrug inhibited tumor autophagy, which synergized with aPD-L1 to promote the tumor-infiltrating CTLs and DCs maturation, to elevate intratumoral TNF-α and IFN-γ levels, and to elicit an anti-tumor immune memory effect in mice for PCa growth inhibition with low side effects. This study verified the synergistic anti-PCa treatment between autophagy inhibition and PD-L1 blockade and meantime broadened the application of pH-sensitive macromolecular nanodrug. STATEMENT OF SIGNIFICANCE: A macromolecular nanodrug, comprising the PDPA core and the surface conjugation of both aPD-L1 antibodies and long-chain PEG coating via a tumor acidity-labile α-carboxy-dimethylmaleic anhydride amine bond, was developed. Tumoral acidity triggered the release of aPD-L1 for immunotherapy. Meantime, the charge switch of the remanent nanodrug enhanced the cancer cell uptake of PDPA, which disturbed the lysosomes to inhibit autophagy. This advanced nanodrug promoted the tumor-infiltrating CTLs and DCs maturation, elevated the intratumoral TNF-α and IFN-γ levels, and elicited the robust anti-tumor immune memory effect. This study demonstrated that the pH-sensitive PDPA macromolecule could serve as a carrier for the aPD-L1 delivery and as an efficient autophagy inhibitor to boost the immunotherapy of prostate cancer.
Collapse
Affiliation(s)
- Yiyao Wang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, PR China
| | - Hanqi Lei
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, PR China
| | - Binyuan Yan
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, PR China
| | - Shiqiang Zhang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, PR China
| | - Bin Xu
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, PR China
| | - Minzhao Lin
- Nanomedicine Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, PR China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, PR China.
| | - Jinsheng Huang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, PR China.
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, PR China.
| |
Collapse
|
45
|
Xu X, Wu Q, Tan L, Men X, Huang Y, Li H. Biomimetic Metal-Chalcogenide Agents Enable Synergistic Cancer Therapy via Microwave Thermal-Dynamic Therapy and Immune Cell Activation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:42182-42195. [PMID: 37651685 DOI: 10.1021/acsami.3c05728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Microwave thermal dynamic therapy (MTDT), which combines thermal effects and reactive oxygen species (ROS) by microwave activation, seems to be a promising anticancer therapeutic method. A multifunctional agent for achieving synergistic localized cancer treatment is the key to exploit the strategy to inhibit tumor cell recurrence and metastasis. In the study, a ZIF-67 based theranostic agent loaded with metal-chalcogenide open framework 3 (MCOF3) and heat shock protein 70 (HSP70) as the inner component was studied, coupled with targeting cancer cell membrane (TCM) as the biomimetic outer shell. We found that metal ions in MCOF3 enabled the composite agents to show peroxide-like activity to produce •OH and destroy cancer cells. And then, the microwave (MW) thermal sensitizer of ZIF-67 was used to specifically convert the MW energy into thermal energy and selectively heat the tumor via the cell's targeting. Additionally, the effect of continuous MW thermal therapy has been shown to promote the expression of HSP70, and further activate the effector of CD4 T cell and CD8α T cell. As such, the agents effectively inhibit the tumor cell growth under MW irradiation in vitro and in vivo due to the synergistic effects of MTDT and immune cell activation. The study provides an emerging strategy to ablation cancer effectively.
Collapse
Affiliation(s)
- Xiaomu Xu
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, China
| | - Qiong Wu
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Longfei Tan
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianwei Men
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yue Huang
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Hong Li
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, China
| |
Collapse
|
46
|
Li J, Hu Z, Zhu J, Lin X, Gao X, Lv G. Antitumor Effects of Pegylated Zinc Protoporphyrin-Mediated Sonodynamic Therapy in Ovarian Cancer. Pharmaceutics 2023; 15:2275. [PMID: 37765244 PMCID: PMC10534787 DOI: 10.3390/pharmaceutics15092275] [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: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Sonodynamic therapy (SDT) induces reactive oxygen species (ROS) to kill tumor cells. Heme oxygenase-1 (HO-1), as an important antioxidant enzyme, resists killing by scavenging ROS. Zinc protoporphyrin (ZnPP) not only effectively inhibits HO-1 activity, but also becomes a potential sonosensitizer. However, its poor water solubility limits its applications. Herein, we developed an improved water-soluble method. It was proved that pegylated zinc protoporphyrin-mediated SDT (PEG-ZnPP-SDT) could significantly enhance ROS production by destroying the HO-1 antioxidant system in ovarian cancer. Increased ROS could cause mitochondrial membrane potential collapse, release cytochrome c from mitochondria to the cytoplasm, and trigger the mitochondrial-caspase apoptotic pathway. In conclusion, our results demonstrated that PEG-ZnPP-SDT, as a novel sonosensitizer, could improve the antitumor effects by destroying the HO-1 antioxidant system. It provided a new therapeutic strategy for SDT to treat cancers, especially those with higher HO-1 expression.
Collapse
Affiliation(s)
- Jia Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| | - Zheng Hu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China;
| | - Jiwei Zhu
- Department of Forensic Medicine, Harbin Medical University, Harbin 150086, China;
| | - Xin Lin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| | - Guixiang Lv
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin 150086, China; (J.L.); (X.L.); (X.G.)
| |
Collapse
|
47
|
Huang T, Zhang Q, Yi J, Wang R, Zhang Z, Luo P, Zeng R, Wang Y, Tu M. PEG-Sheddable Nanodrug Remodels Tumor Microenvironment to Promote Effector T Cell Infiltration and Revise Their Exhaustion for Breast Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301749. [PMID: 37211704 DOI: 10.1002/smll.202301749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/30/2023] [Indexed: 05/23/2023]
Abstract
Low infiltration of cytotoxic T lymphocytes and their exhaustion manifest the two concurrent main hurdles for achieving effective tumor immunotherapy of triple-negative breast cancer. It is found that Galectin-9 blockage can revise the exhaustion of effector T cells, meanwhile the repolarization of protumoral M2 tumor-associated macrophages (TAMs) into tumoricidal M1-like ones can recruit effector T cells infiltrating into tumor to boost immune responses. Herein, a sheddable PEG-decorated and M2-TAMs targeted nanodrug incorporating Signal Transducer and Activator of Transcription 6 inhibitor (AS) and anti-Galectin-9 antibody (aG-9) is prepared. The nanodrug responds to acidic tumor microenvironment (TME) with the shedding of PEG corona and the release of aG-9, exerting local blockade of PD-1/Galectin-9/TIM-3 interaction to augment effector T cells via exhaustion reversing. Synchronously, targeted repolarization of M2-TAMs into M1 phenotype by AS-loaded nanodrug is achieved, which promotes tumor infiltration of effector T cells and thus synergizes with aG-9 blockade to boost the therapeutic efficacy. Besides, the PEG-sheddable approach endows nanodrug with stealth ability to reduce immune-related adverse effects caused by AS and aG-9. This PEG sheddable nanodrug holds the potential to reverse the immunosuppressive TME and increase effector T cell infiltration, which dramatically enhances immunotherapy in highly malignant breast cancer.
Collapse
Affiliation(s)
- Tao Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Qiaoyun Zhang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Jing Yi
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Rongze Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Zekun Zhang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Pin Luo
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Rong Zeng
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Mei Tu
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
48
|
Wang RX, Zheng RR, Cai H, Yang N, Chen ZX, Zhao LP, Huang YK, Li PF, Cheng H, Chen AL, Li SY, Xu L. Coordination-Driven Self-Assembly of Biomedicine to Enhance Photodynamic Therapy by Inhibiting Proteasome and Bcl-2. Adv Healthc Mater 2023; 12:e2300711. [PMID: 37166979 DOI: 10.1002/adhm.202300711] [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: 03/06/2023] [Revised: 05/04/2023] [Indexed: 05/12/2023]
Abstract
Tumor cells resist oxidative damage and apoptosis by activating defense mechanisms. Herein, a self-delivery biomedicine (designated as BSC) is developed by the self-assembly of Bortezomib (BTZ), Sabutoclax (Sab) and Chlorin e6 (Ce6). Interestingly, BTZ can be coordinated with Sab to promote the assembly of uniform ternary biomedicine through non-covalent intermolecular interactions. Moreover, BTZ as a proteasome inhibitor can prevent tumor cells from scavenging damaged proteins to reduce their oxidative resistance. Sab can downregulate B-cell lymphoma 2 (Bcl-2) to decrease the antiapoptotic protein. Both the proteasome and Bcl-2 inhibitions contribute to increasing cell apoptosis and amplifying photodynamic therapy (PDT) efficacy of Ce6. Encouragingly, carrier-free BSC receives all biological activities of these assembly elements, including photodynamic performance as well as inhibitory capabilities of proteasome and Bcl-2. Besides, BSC has a preferable cellular uptake ability and tumor retention property, which increase the drug delivery efficiency and bioavailability. In vitro and in vivo research demonstrate the superior PDT efficiency of BSC by proteasome and Bcl-2 inhibitions. Of special note, the coordination-driven self-assembly of BSC is pH-responsive, which can be disassembled for controlled drug release upon tumor acidic microenvironment. This study will expand the applicability of self-delivery nanomedicine with sophisticated mechanisms for tumor treatment.
Collapse
Affiliation(s)
- Rui-Xin Wang
- Department of Geriatric Cardiology, General Hospital of the Southern Theatre Command, People's Liberation Army (PLA), Guangzhou, 510010, P. R. China
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Rong-Rong Zheng
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Hua Cai
- Department of Geriatric Cardiology, General Hospital of the Southern Theatre Command, People's Liberation Army (PLA), Guangzhou, 510010, P. R. China
| | - Ni Yang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Zu-Xiao Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Lin-Ping Zhao
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Yue-Kang Huang
- Department of Geriatric Cardiology, General Hospital of the Southern Theatre Command, People's Liberation Army (PLA), Guangzhou, 510010, P. R. China
| | - Peng-Fei Li
- Department of Geriatric Cardiology, General Hospital of the Southern Theatre Command, People's Liberation Army (PLA), Guangzhou, 510010, P. R. China
| | - Hong Cheng
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - A-Li Chen
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Shi-Ying Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Lin Xu
- Department of Geriatric Cardiology, General Hospital of the Southern Theatre Command, People's Liberation Army (PLA), Guangzhou, 510010, P. R. China
| |
Collapse
|
49
|
He S, Jia X, Feng S, Hu J. Three Strategies in Engineering Nanomedicines for Tumor Microenvironment-Enabled Phototherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300078. [PMID: 37226364 DOI: 10.1002/smll.202300078] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/29/2023] [Indexed: 05/26/2023]
Abstract
Canonical phototherapeutics have several limitations, including a lack of tumor selectivity, nondiscriminatory phototoxicity, and tumor hypoxia aggravation. The tumor microenvironment (TME) is characterized by hypoxia, acidic pH, and high levels of H2 O2 , GSH, and proteases. To overcome the shortcomings of canonical phototherapy and achieve optimal theranostic effects with minimal side effects, unique TME characteristics are employed in the development of phototherapeutic nanomedicines. In this review, the effectiveness of three strategies for developing advanced phototherapeutics based on various TME characteristics is examined. The first strategy involves targeted delivery of phototherapeutics to tumors with the assistance of TME-induced nanoparticle disassembly or surface modification. The second strategy involves near-infrared absorption increase-induced phototherapy activation triggered by TME factors. The third strategy involves enhancing therapeutic efficacy by ameliorating TME. The functionalities, working principles, and significance of the three strategies for various applications are highlighted. Finally, possible challenges and future perspectives for further development are discussed.
Collapse
Affiliation(s)
- Shiliang He
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, China
| | - Xiao Jia
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Sai Feng
- Jiangxi Key Laboratory of Bioprocess Engineering and Co-Innovation Center for In-Vitro Diagnostic Reagents and Devices of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Junqing Hu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, 518118, China
- Shenzhen Bay Laboratory, Shenzhen, 518132, China
| |
Collapse
|
50
|
Dong HQ, Fu XF, Wang MY, Zhu J. Research progress on reactive oxygen species production mechanisms in tumor sonodynamic therapy. World J Clin Cases 2023; 11:5193-5203. [PMID: 37621595 PMCID: PMC10445077 DOI: 10.12998/wjcc.v11.i22.5193] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/22/2023] [Indexed: 08/04/2023] Open
Abstract
In recent years, because of the growing desire to improve the noninvasiveness and safety of tumor treatments, sonodynamic therapy has gradually become a popular research topic. However, due to the complexity of the therapeutic process, the relevant mechanisms have not yet been fully elucidated. One of the widely accepted possibilities involves the effect of reactive oxygen species. In this review, the mechanism of reactive oxygen species production by sonodynamic therapy (SDT) and ways to enhance the sonodynamic production of reactive oxygen species are reviewed. Then, the clinical application and limitations of SDT are discussed. In conclusion, current research on sonodynamic therapy should focus on the development of sonosensitizers that efficiently produce active oxygen, exhibit biological safety, and promote the clinical transformation of sonodynamic therapy.
Collapse
Affiliation(s)
- He-Qin Dong
- School of Medicine, Shaoxing University, Shaoxin 312000, Zhejiang Province, China
| | - Xiao-Feng Fu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Min-Yan Wang
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Jiang Zhu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|