1
|
Liu H, Wang L, Jin H, Tao K, Zhu X, Zhang M, Hou Y, Liu S, Zhang H. FeOOH-Assisted Formation of Hybrid Polymer Nanospindles for Efficient Iron Delivery and Ferroptosis Tumor Therapy. Bioconjug Chem 2025; 36:464-475. [PMID: 40045454 DOI: 10.1021/acs.bioconjchem.4c00546] [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: 03/20/2025]
Abstract
Exogenous iron delivery using iron-containing nanomaterials is an alternative strategy for enhancing the efficacy in ferroptosis tumor therapy but limited by the problems of low iron content, low tumor enrichment, low cellular uptake, and uncontrolled release of iron ions. To solve the problems, an FeOOH-assisted approach is demonstrated to produce iron hybrid polymer nanospindles (IHPNSs) for efficient iron delivery and ferroptosis tumor therapy. The IHPNSs are prepared through the cohydrolysis of FeCl3·6H2O with aniline, pyrrole, or amino-pyrrole. On the one hand, the hydrolysis of Fe3+ generates FeOOH particles, which further act as the templates to form fusiform architectures. On the other hand, Fe3+ triggers the oxidative polymerization of aniline, pyrrole, or amino-pyrrole. The as-prepared polymers are capable of coordinating with excessive Fe3+ and locate on the FeOOH templates, thus producing Fe3+/polymer composite-coated FeOOH nanospindles. Systematic studies indicate that the one-dimension-like morphology facilitates tumor enrichment and cellular uptake of IHPNSs. Besides the high iron content of IHPNSs, the controlled release of Fe3+ stimulated by the overexpressed glutathione (GSH) in the tumor microenvironment is achieved. The released Fe3+ is further transformed to Fe2+ by scavenging GSH, which leads to excessive accumulation of reactive oxygen species and lipid peroxides and finally induces ferroptosis of tumor cells. As a proof of concept, the IHPNSs show good efficacy in the treatment of a rat model of bladder tumors in situ.
Collapse
Affiliation(s)
- Heng Liu
- Department of Urinary, The First Hospital of Jilin University, Changchun 130021, P. R. China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Lu Wang
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Hao Jin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Kepeng Tao
- Department of Urinary, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Xuanqi Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Mengsi Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yuchuan Hou
- Department of Urinary, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Shuwei Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
2
|
Zhang B, Fan K. Design and application of ferritin-based nanomedicine for targeted cancer therapy. Nanomedicine (Lond) 2025; 20:481-500. [PMID: 39895329 PMCID: PMC11875477 DOI: 10.1080/17435889.2025.2459056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
Owing to its unique structure and favorable biocompatibility, ferritin has been widely studied as a promising drug carrier over the past two decades. Since the identification of its inherent tumor-targeting property due to unique recognition ablity of the transferrin receptor 1 (TfR1), ferritin-based nanomedicine has attracted widespread attention and triggered a research surge in the field of targeted cancer therapy. Along with progress in structure studies and modification technology, diverse strategies have been carried out to equip ferritin with on-demand functions, further improving the antitumor efficacy and in vivo safety of ferritin-based cancer therapy. In this review, we highlight the structure-based rational design of ferritin and summarize the design strategies in detail from two main perspectives: multifunctional modification and drug loading. In particular, the critical issues that need attention in the design are discussed in depth. Furthermore, we provide an overview of the latest advances in the application of ferritin-based nanomedicines in chemotherapy, phototherapy and immunotherapy, with particular emphasis on emerging therapeutic approaches among these therapies.
Collapse
Affiliation(s)
- Baoli Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Han W, Ding J, Qiao B, Yu Y, Sun H, Crespy D, Landfester K, Mao X, Jiang S. Self-Sustained Biophotocatalytic Nano-Organelle Reactors with Programmable DNA Switches for Combating Tumor Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415030. [PMID: 39797479 PMCID: PMC11881670 DOI: 10.1002/adma.202415030] [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: 10/02/2024] [Revised: 12/09/2024] [Indexed: 01/13/2025]
Abstract
Metastasis, the leading cause of mortality in cancer patients, presents challenges for conventional photodynamic therapy (PDT) due to its reliance on localized light and oxygen application to tumors. To overcome these limitations, a self-sustained organelle-mimicking nanoreactor is developed here with programmable DNA switches that enables bio-chem-photocatalytic cascade-driven starvation-photodynamic synergistic therapy against tumor metastasis. Emulating the compartmentalization and positional assembly strategies found in living cells, this nano-organelle reactor allows quantitative co-compartmentalization of multiple functional modules for the designed self-illuminating chemiexcited PDT system. Within the space-confined nanoreactor, biofuel glucose is converted to hydrogen peroxide (H2O2) which enhances luminol-based chemiluminescence (CL), consequently driving the generation of photochemical singlet oxygen (1O2) via chemiluminescence resonance energy transfer. Meanwhile, hemoglobin functions as a synchronized oxygen supplier for both glucose oxidation and PDT, while also exhibiting peroxidase-like activity to produce hydroxyl radicals (·OH). Crucially, the nanoreactor keeps switching off in normal tissues, with on-demand activation in tumors through toehold-mediated strand displacement. These findings demonstrate that this nanoreactor, which is self-sufficient in light and oxygen and precise in striking tumors, presents a promising paradigm for managing highly metastatic cancers.
Collapse
Affiliation(s)
- Wenshuai Han
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of ChinaQingdao266003P. R. China
- State Key Laboratory of Marine Food Processing and Safety ControlOcean University of ChinaQingdao266404P. R. China
- Laboratory for Marine Drugs and BioproductsQingdao Marine Science and Technology CenterQingdao266237P. R. China
| | - Jiayi Ding
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of ChinaQingdao266003P. R. China
- State Key Laboratory of Marine Food Processing and Safety ControlOcean University of ChinaQingdao266404P. R. China
- Laboratory for Marine Drugs and BioproductsQingdao Marine Science and Technology CenterQingdao266237P. R. China
| | - Bo Qiao
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of ChinaQingdao266003P. R. China
- State Key Laboratory of Marine Food Processing and Safety ControlOcean University of ChinaQingdao266404P. R. China
- Laboratory for Marine Drugs and BioproductsQingdao Marine Science and Technology CenterQingdao266237P. R. China
| | - Yingjie Yu
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of ChinaQingdao266003P. R. China
- State Key Laboratory of Marine Food Processing and Safety ControlOcean University of ChinaQingdao266404P. R. China
- Laboratory for Marine Drugs and BioproductsQingdao Marine Science and Technology CenterQingdao266237P. R. China
| | - Hao Sun
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of ChinaQingdao266003P. R. China
- State Key Laboratory of Marine Food Processing and Safety ControlOcean University of ChinaQingdao266404P. R. China
- Laboratory for Marine Drugs and BioproductsQingdao Marine Science and Technology CenterQingdao266237P. R. China
| | - Daniel Crespy
- Department of Materials Science and EngineeringSchool of Molecular Science and EngineeringVidyasirimedhi Institute of Science and Technology (VISTEC)Rayong21210Thailand
| | | | - Xiangzhao Mao
- State Key Laboratory of Marine Food Processing and Safety ControlOcean University of ChinaQingdao266404P. R. China
- Laboratory for Marine Drugs and BioproductsQingdao Marine Science and Technology CenterQingdao266237P. R. China
- College of Food Science and EngineeringOcean University of ChinaQingdao266003P. R. China
| | - Shuai Jiang
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of ChinaQingdao266003P. R. China
- State Key Laboratory of Marine Food Processing and Safety ControlOcean University of ChinaQingdao266404P. R. China
- Laboratory for Marine Drugs and BioproductsQingdao Marine Science and Technology CenterQingdao266237P. R. China
| |
Collapse
|
4
|
Jia R, Zhang S, Zhang J, Li Y. Laser-Free Photosensitive Systems in Cancer Therapy: A Comprehensive Review. Int J Mol Sci 2025; 26:1437. [PMID: 40003904 PMCID: PMC11855559 DOI: 10.3390/ijms26041437] [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: 12/30/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Photodynamic therapy (PDT) involves the use of photosensitizers (PSs) that, upon activation by specific wavelengths of light, generate reactive oxygen species (ROS), including singlet oxygen (1O2) and hydroxyl radicals (·OH), within the targeted tissue, typically tumor cells. The generated ROS induces cellular damage, disrupts cellular processes, and ultimately leads to apoptosis or necrosis of the tumor cells. However, the clinical application of PDT is significantly hindered by the limited tissue penetration ability of light. To address this limitation, laser-free self-luminescent photosensitive systems have emerged as potential solutions for achieving deep-tissue PDT and imaging. This review provides a comprehensive analysis of various laser-independent photosensitive systems, with a particular emphasis on those based on resonance energy transfer (RET), chemically induced electron exchange luminescence (CIEEL), and Cherenkov radiation energy transfer (CRET). The aim is to offer a theoretical framework for the development of novel photodynamic systems and to reassess the application potential of certain previously overlooked photosensitizers (PSs).
Collapse
Affiliation(s)
| | | | | | - Yi Li
- Academy of Pharmacy, Xian-Jiaotong Liverpool University, Suzhou 215000, China; (R.J.); (S.Z.); (J.Z.)
| |
Collapse
|
5
|
Ling S, Yang H, Wu F, Yang X, Li T, Zhang Y, Jiang J, Li C, Wang Q. Rapid, Non-Invasive, Accurate Diagnosis and Efficient Clearance of Metastatic Lymph Nodes. Angew Chem Int Ed Engl 2025; 64:e202419988. [PMID: 39557612 DOI: 10.1002/anie.202419988] [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: 10/16/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/20/2024]
Abstract
Sentinel lymph node (SLN) biopsy is currently the standard procedure for clinical cancer diagnosis and treatment, but still faces the risks of false negatives and tumor metastasis, as well as time-consuming pathological evaluation procedure. Herein, we proposed a near-infrared-II (NIR-II, 1000-1700 nm) theranostic nanosystem (FLAGC) for rapid, non-invasive, accurate diagnosis and efficient clearance of metastatic lymph nodes in breast cancer. Initialized by chlorin e6 (Ce6), a pH-sensitive amphiphilic amino acid fluorenylmethoxycarbonyl-L-histidine (Fmoc-His) was assembled with Gd3+, luminol, and AgAuSe quantum dots (AAS QDs) to form FLAGC. In FLAGC, luminol and AAS QDs form a NIR-II chemical resonance energy transfer (CRET) system (Luminol-AAS); Ce6 initiates the assembly and also serves as a photosensitizer. Upon subcutaneous injection, FLAGC is easily drained into SLNs, achieving their precise localization. Subsequently, the acidity of tumor microenvironment triggers the rapid disassembly of FLAGC, exposing Luminol-AAS. myeloperoxidase (MPO) secreted by tumor-associated macrophages and neutrophils in SLNs mediates the oxidation of luminol, lighting up AAS QDs through the CRET process for precise diagnosis of metastatic lymph nodes. Moreover, highly efficient clearance of positive lymph nodes is achieved through Ce6-mediated photodynamic therapy. Our strategy provides a new paradigm for identifying and eliminating clinically metastatic lymph nodes.
Collapse
Grants
- 21934007, 22127808, 22174158, 22271308 National Natural Science Foundation of China
- 2021YFF0701804 National Key Research and Development Program of China
- YJKYYQ20200036 Research instrument and equipment development project
- ZDBS-LYSLH021 Key Research Program of Frontier Sciences,CAS
- BE2022753,BK20222016,BK20232046,BE2022745,BK20210128,BK20200254 Natural Science Foundation of Jiangsu Province
- SJC2022001, SZS201904, SZS2023006, ZXT2022007 Science and Technology Foundation of Suzhou
Collapse
Affiliation(s)
- Sisi Ling
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Hongchao Yang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Feng Wu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xiaohu Yang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Tuanwei Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yejun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jiang Jiang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Chunyan Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine andi-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| |
Collapse
|
6
|
Dai W, Zhou X, Zhao J, Lei L, Huang Y, Jia F, Tang Z, Ji J, Jin Q. Tumor microenvironment-modulated nanoparticles with cascade energy transfer as internal light sources for photodynamic therapy of deep-seated tumors. Biomaterials 2025; 312:122743. [PMID: 39111233 DOI: 10.1016/j.biomaterials.2024.122743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Photodynamic therapy (PDT) is an appealing modality for cancer treatments. However, the limited tissue penetration depth of external-excitation light makes PDT impossible in treating deep-seated tumors. Meanwhile, tumor hypoxia and intracellular reductive microenvironment restrain the generation of reactive oxygen species (ROS). To overcome these limitations, a tumor-targeted self-illuminating supramolecular nanoparticle T-NPCe6-L-N is proposed by integrating photosensitizer Ce6 with luminol and nitric oxide (NO) for chemiluminescence resonance energy transfer (CRET)-activated PDT. The high H2O2 level in tumor can trigger chemiluminescence of luminol to realize CRET-activated PDT without exposure of external light. Meanwhile, the released NO significantly relieves tumor hypoxia via vascular normalization and reduces intracellular reductive GSH level, further enhancing ROS abundance. Importantly, due to the different ROS levels between cancer cells and normal cells, T-NPCe6-L-N can selectively trigger PDT in cancer cells while sparing normal cells, which ensured low side effect. The combination of CRET-based photosensitizer-activation and tumor microenvironment modulation overcomes the innate challenges of conventional PDT, demonstrating efficient inhibition of orthotopic and metastatic tumors on mice. It also provoked potent immunogenic cell death to ensure long-term suppression effects. The proof-of-concept research proved as a new strategy to solve the dilemma of PDT in treatment of deep-seated tumors.
Collapse
Affiliation(s)
- Wenbin Dai
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Xianchi Zhou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Jinchao Zhao
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, PR China
| | - Lei Lei
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, PR China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Fan Jia
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, PR China.
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, PR China; Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
7
|
Hu Z, Tan H, Ye Y, Xu W, Gao J, Liu L, Zhang L, Jiang J, Tian H, Peng F, Tu Y. NIR-Actuated Ferroptosis Nanomotor for Enhanced Tumor Penetration and Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2412227. [PMID: 39370589 DOI: 10.1002/adma.202412227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/18/2024] [Indexed: 10/08/2024]
Abstract
Ferroptosis nano-inducers have drawn considerable attention in the treatment of malignant tumors. However, low intratumoral hydrogen peroxide level and complex biological barriers hinder the ability of nanomedicines to generate sufficient reactive oxygen species (ROS) and achieve tumor penetration. Here a near-infrared (NIR)-driven ROS self-supplying nanomotor is successfully designed for synergistic tumor chemodynamic therapy (CDT) and photothermal therapy (PTT). Janus nanomotor is created by the asymmetrical modification of polydopamine (PDA) with zinc peroxide (ZnO2) and subsequent ferrous ion (Fe2+) chelation via the polyphenol groups from the PDA, here refer as ZnO2@PDA-Fe (Z@P-F). ZnO2 is capable of slowly releasing hydrogen peroxide (H2O2) into an acidic tumor microenvironment (TME) providing sufficient ingredients for the Fenton reaction necessary for ferroptosis. Upon NIR laser irradiation, the loaded Fe2+ is released and a thermal gradient is simultaneously formed owing to the asymmetric PDA coating, thus endowing the nanomotor with self-thermophoresis based enhanced diffusion for subsequent lysosomal escape and tumor penetration. Therefore, the release of ferrous ions (Fe2+), self-supplied H2O2, and self-thermophoresis of nanomotors with NIR actuation further improve the synergistic CDT/PTT efficacy, showing great potential for active tumor therapy.
Collapse
Affiliation(s)
- Ziwei Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Haixin Tan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yicheng Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenxin Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junbin Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lu Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lishan Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiamiao Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hao Tian
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
8
|
Tan H, Shen Z, Wang X, Shu S, Deng J, Lu L, Fan Z, Hu D, Cheng P, Cao X, Huang Q. Endoplasmic reticulum-targeted biomimetic nanoparticles induce apoptosis and ferroptosis by regulating endoplasmic reticulum function in colon cancer. J Control Release 2024; 375:422-437. [PMID: 39278355 DOI: 10.1016/j.jconrel.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Colorectal cancer (CRC) is a major threat to human health, as it is one of the most common malignancies with a high incidence and mortality rate. The cancer cell membrane (CCM) has significant potential in targeted tumor drug delivery due to its membrane antigen-mediated homologous targeting ability. The endoplasmic reticulum (ER) in cancer cells plays a crucial role in apoptosis and ferroptosis. In this study, we developed an ER-targeted peptide-modified CCM-biomimetic nanoparticle-delivered lovastatin (LOV) nanomedicine delivery system (EMPP-LOV) for cancer treatment. Both in vitro and in vivo experiments demonstrated that EMPP could effectively target cancer cells and localize within the ER. EMPP-LOV modulated ER function to promote apoptosis and ferroptosis in tumor cells. Furthermore, synergistic antitumor efficacy was observed in both in vitro and in vivo models. EMPP-LOV induced apoptosis in CRC cells by over-activating endoplasmic reticulum stress and promoted ferroptosis by inhibiting the mevalonate pathway, leading to synergistic tumor growth inhibition with minimal toxicity to major organs. Overall, the EMPP-LOV delivery system, with its subcellular targeting capability within tumor cells, presents a promising therapeutic platform for CRC treatment.
Collapse
Affiliation(s)
- Hongxin Tan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ziqi Shen
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Xiaohua Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Sicheng Shu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jie Deng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Li Lu
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Ziyan Fan
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Danni Hu
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Pu Cheng
- Department of Gynaecology, The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xi Cao
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China; Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Qi Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
9
|
Agrawal S, Singh GK, Tiwari S. Focused starvation of tumor cells using glucose oxidase: A comprehensive review. Int J Biol Macromol 2024; 281:136444. [PMID: 39389487 DOI: 10.1016/j.ijbiomac.2024.136444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Starvation therapy targets the high metabolic demand of tumor cells. It primarily leans over the consumption of intracellular glucose and simultaneous blockade of alternative metabolic pathways. The strategy involves the use of glucose oxidase (GOx) for catalyzing the conversion of glucose into gluconic acid and hydrogen peroxide. Under these conditions, metabolic re-programming of tumor cells enables the utilization of substrates such as amino acids, fatty acids and lipids. This can be overcome by co-administration of chemo-, photo- and immuno-therapeutics together with glucose oxidase. Targeted delivery of glucose oxidase at tumor site can be enabled with the use of nanoformulations. In this review, we highlight that the outcomes of starvation therapy can be improved using rationally developed nano-formulations. It is possible to load synergistically acting bioactives in these formulations and deliver in site-specific manner and hence achieve the elimination of tumors cells with greater efficacy.
Collapse
Affiliation(s)
- Shivanshu Agrawal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Gireesh K Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya 824236, India
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India.
| |
Collapse
|
10
|
Shen Q, Chen J, Jing X, Duan C. Modifying Parallel Excitations into One Framework for C(sp 3)─H Bond Activation with Energy Combined More Than Two Photons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404293. [PMID: 39052896 PMCID: PMC11423249 DOI: 10.1002/advs.202404293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/09/2024] [Indexed: 07/27/2024]
Abstract
Natural photosynthesis enzymes utilize energies of several photons for challenging oxidation of water, whereas artificial photo-catalysis typically involves only single-photon excitation. Herein, a multiphoton excitation strategy is reported that combines parallel photo-excitations with a photoinduced electron transfer process for the activation of C(sp3)─H bonds, including methane. The metal-organic framework Fe3-MOF is designed to consolidate 4,4',4″-nitrilotrisbenzoic units for the photoactivation of dioxygen and trinuclear iron clusters as the HAT precursor for photoactivating alkanes. Under visible light irradiation, the dyes and iron clusters absorbed parallel photons simultaneously to reach their excited states, respectively, generating 1O2 via energy transfer and chlorine radical via ligand-to-metal charge transfer. The further excitation of organic dyes leads to the reduction of 1O2 into O2 •- through a photoinduced electron transfer, guaranteeing an extra multiphoton oxygen activation manner. The chlorine radical abstracts a hydrogen atom from alkanes, generating the carbon radical for further oxidation transformation. Accordingly, the total oxidation conversion of alkane utilizing three photoexcitation processes combines the energies of more than two photons. This new platform synergistically combines a consecutive excited photoredox organic dye and a HAT catalyst to combine the energies of more than two photons, providing a promising multiphoton catalysis strategy under energy saving, and high efficiency.
Collapse
Affiliation(s)
- Qingbo Shen
- School of Chemistry, Dalian University of Technology, Dalian, 116024, China
| | - Jiali Chen
- School of Chemistry, Dalian University of Technology, Dalian, 116024, China
| | - Xu Jing
- School of Chemistry, Dalian University of Technology, Dalian, 116024, China
| | - Chunying Duan
- State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
11
|
Yang J, Fu Q, Jiang H, Zhong H, Qin HK, Miao X, Li Y, Liu M, Yao J. Blue light photobiomodulation induced osteosarcoma cell death by facilitating ferroptosis and eliciting an incomplete tumor cell stress response. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:113003. [PMID: 39121719 DOI: 10.1016/j.jphotobiol.2024.113003] [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: 05/09/2024] [Revised: 07/10/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
To investigate the potential of blue light photobiomodulation (PBM) in inducing ferroptosis, a novel form of regulated cell death, in OS cells, considering its known effectiveness in various cancer models. In this investigation, we exposed human OS cell lines, HOS and MG63, to different wavelengths (420, 460 and 480 nm) of blue light at varying irradiances, and examined cellular responses such as viability, apoptosis, levels of reactive oxygen species (ROS), and mitochondrial membrane potential (MMP). Transcriptome sequencing was employed to unravel the molecular mechanisms underlying blue light-induced effects, with validation via quantitative real-time PCR (qRT-PCR). Our findings revealed a wavelength- and time-dependent decrease in cell viability, accompanied by increased apoptosis and oxidative stress. Transcriptomic analysis identified differential expression of genes associated with ferroptosis, oxidative stress, and iron metabolism, further validated by qRT-PCR. These results implicated ferroptosis as a significant mechanism in the blue light-induced death of OS cells, potentially mediated by ROS generation and disruption of iron homeostasis. Also, An incomplete stress response was observed in MG63 cells induced by blue light exposure. Hence, blue light PBM holds promise as a therapeutic approach in OS clinical investigations; however, additional exploration of its underlying mechanisms remains imperative.
Collapse
Affiliation(s)
- Jiali Yang
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Qiqi Fu
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Hui Jiang
- Academy for Engineering and Technology, Fudan University, 220th Handan Road, Shanghai 200433, China
| | - Hongyu Zhong
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third School of Clinical Medicine, The Third Affiliated Hospital of Southern Medical University, No.183, Zhongshan Avenue West, Guangzhou 510515, China
| | - Hao Kuan Qin
- Academy for Engineering and Technology, Fudan University, 220th Handan Road, Shanghai 200433, China
| | - Xiaojing Miao
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Yinghua Li
- Shanghai Fifth People's Hospital, Fudan University, 801th Heqing Road, Shanghai 200240, China.
| | - Muqing Liu
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China; Zhongshan Fudan Joint Innovation Center, 6th Xiangxing Road, Zhongshan 28403, China.
| | - Jinghui Yao
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third School of Clinical Medicine, The Third Affiliated Hospital of Southern Medical University, No.183, Zhongshan Avenue West, Guangzhou 510515, China.
| |
Collapse
|
12
|
Mao YW, Chu KF, Song P, Wang AJ, Zhao T, Feng JJ. Atomically dispersed bimetallic active sites as H 2O 2 self-supplied nanozyme for effective chemodynamic therapy, chemotherapy and starvation therapy. BIOMATERIALS ADVANCES 2024; 162:213919. [PMID: 38861801 DOI: 10.1016/j.bioadv.2024.213919] [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: 03/12/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Tumor microenvironment (TME)-responsive chemodynamic therapy (CDT) is severely hindered by insufficient intracellular H2O2 level that seriously deteriorates antitumor efficacy, albeit with its extensively experimental and theoretical research. Herein, we designed atomically dispersed FeCo dual active sites anchored in porous carbon polyhedra (termed FeCo/PCP), followed by loading with glucose oxidase (GOx) and anticancer doxorubicin (DOX), named FeCo/PCP-GOx-DOX, which converted glucose into toxic hydroxyl radicals. The loaded GOx can either decompose glucose to self-supply H2O2 or provide fewer nutrients to feed the tumor cells. The as-prepared nanozyme exhibited the enhanced in vitro cytotoxicity at high glucose by contrast with those at less or even free of glucose, suggesting sufficient accumulation of H2O2 and continual transformation to OH for CDT. Besides, the FeCo/PCP-GOx-DOX can subtly integrate starvation therapy, the FeCo/PCP-initiated CDT, and DOX-inducible chemotherapy (CT), greatly enhancing the therapeutic efficacy than each monotherapy.
Collapse
Affiliation(s)
- Yan-Wen Mao
- Key laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, College of Life Science, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Kai-Fei Chu
- Key laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, College of Life Science, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Pei Song
- Key laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, College of Life Science, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, China; Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Ai-Jun Wang
- Key laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, College of Life Science, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Tiejun Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China.
| | - Jiu-Ju Feng
- Key laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, College of Life Science, College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua 321004, China.
| |
Collapse
|
13
|
Lu W, Wang N, Liu X, Chen D, Li Q, Rui J, Ning W, Shi X, Li C, Zhao Y, He A, Teng Z. Matrix-degrading soft-nanoplatform with enhanced tissue penetration for amplifying photodynamic therapeutic efficacy of breast cancer. J Mater Chem B 2024; 12:7837-7847. [PMID: 39016097 DOI: 10.1039/d4tb00894d] [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: 07/18/2024]
Abstract
The dense extracellular matrix (ECM) in the tumor microenvironment forms an abnormal physical barrier, which impedes the delivery and penetration of nanomedicines and hinders their therapeutic efficacy. Herein, we synthesize matrix-degrading soft-nanocapsules composed of human serum albumin (HSA) and hyaluronidase (HAase) for overcoming the obstruction of ECM in the tumor microenvironment. The matrix-degrading human serum albumin/hyaluronidase soft-nanocapsules, referred to as HSA/HAase SNCs, possess a uniform diameter, inward hollow structure, and wrinkled morphology. In vitro biocompatibility results indicate that the HSA/HAase SNCs display no adverse effects on the viability of human umbilical vein endothelial cells (HUVECs), smooth muscle cells (SMCs), and mouse breast cancer (4T1) cells and do not induce hemolysis towards red blood cells (RBCs). The HSA/HAase SNCs exhibit a 1.4-fold increase in tumor cellular uptake compared to the stiff-counterparts and enhanced penetration in 4T1-, mouse colon carcinoma 26- (CT26-), and mouse pancreatic cancer- (PanO2-) multicellular spheroids. Thanks to the advanced biological properties, a photodynamic platform prepared by loading Ce6 in the HSA/HAase SNCs (HSA/HAase@Ce6) shows improved reactive oxygen species production, a stronger killing effect for cancer cells, and deeper penetration in tumor tissues. In vivo experiments show that HSA/HAase@Ce6 effectively inhibits tumor growth in breast cancer mouse models. RNA-seq analysis of the mice that received the treatment of HSA/HAase@Ce6 shows enrichment of signaling pathways associated with ECM-degradation, which demonstrates that the matrix-degrading nanocapsules overcome the ECM-induced physical barriers in tumors. Overall, the matrix-degrading soft-nanoplatform represents a highly promising strategy to overcome ECM-induced physical barriers and enhance the therapeutic efficacy of nanomedicines.
Collapse
Affiliation(s)
- Wei Lu
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Ning Wang
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Xiao Liu
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Dong Chen
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Qiang Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, Jiangsu 210008, P. R. China
| | - Jianxin Rui
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Weiqing Ning
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Xuzhi Shi
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Chang Li
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| | - Yatong Zhao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 213161, P. R. China
| | - Ao He
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, Jiangsu 210008, P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P. R. China.
| |
Collapse
|
14
|
Luo Y, Bai XY, Zhang L, Hu QQ, Zhang N, Cheng JZ, Hou MZ, Liu XL. Ferroptosis in Cancer Therapy: Mechanisms, Small Molecule Inducers, and Novel Approaches. Drug Des Devel Ther 2024; 18:2485-2529. [PMID: 38919962 PMCID: PMC11198730 DOI: 10.2147/dddt.s472178] [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: 04/04/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Ferroptosis, a unique form of programmed cell death, is initiated by an excess of iron accumulation and lipid peroxidation-induced damage. There is a growing body of evidence indicating that ferroptosis plays a critical role in the advancement of tumors. The increased metabolic activity and higher iron levels in tumor cells make them particularly vulnerable to ferroptosis. As a result, the targeted induction of ferroptosis is becoming an increasingly promising approach for cancer treatment. This review offers an overview of the regulatory mechanisms of ferroptosis, delves into the mechanism of action of traditional small molecule ferroptosis inducers and their effects on various tumors. In addition, the latest progress in inducing ferroptosis using new means such as proteolysis-targeting chimeras (PROTACs), photodynamic therapy (PDT), sonodynamic therapy (SDT) and nanomaterials is summarized. Finally, this review discusses the challenges and opportunities in the development of ferroptosis-inducing agents, focusing on discovering new targets, improving selectivity, and reducing toxic and side effects.
Collapse
Affiliation(s)
- YiLin Luo
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xin Yue Bai
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Lei Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Qian Qian Hu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ning Zhang
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Jun Zhi Cheng
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Ming Zheng Hou
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| | - Xiao Long Liu
- Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China
| |
Collapse
|
15
|
Ren X, Yang Y, Kong X, Liu Z. Integrin α vβ 3-targeted self-assembled polypeptide nanomicelles for efficacious sonodynamic therapy against breast cancer. NANOSCALE 2024; 16:9953-9965. [PMID: 38693876 DOI: 10.1039/d4nr00794h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Sonodynamic therapy (SDT) is an advanced non-invasive cancer treatment strategy with moderate tissue penetration, less invasiveness and a reliable curative effect. However, due to the low stability, potential bio-toxicity and lack of tumor targeting capability of most sonosensitizers, the vast clinical application of SDT has been challenging and limited. Therefore, it is desirable to develop a novel approach to implement sonosensitizers to SDT for cancer treatments. In this study, an amphiphilic polypeptide was designed to effectively encapsulate rose bengal (RB) as a model sonosensitizer to form peptido-nanomicelles (REPNs). The as-fabricated REPNs demonstrated satisfactory tumor targeting and fluorescence performances, which made them superb imaging tracers in vivo. In the meantime, they generated considerable amounts of reactive oxygen species (ROS) to promote tumor cell apoptosis under ultrasound irradiation and showed excellent anti-tumor performance without obvious side effects. These engineered nanomicelles in combination with medical ultrasound may be used to achieve integrin αvβ3-targeted sonodynamic therapy against breast cancer, and it is also a promising non-invasive cancer treatment strategy for clinical translations.
Collapse
Affiliation(s)
- Xueli Ren
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| | - Yanxi Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| | - Xinru Kong
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| | - Zhe Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 300072, Tianjin, China.
| |
Collapse
|
16
|
Li Z, Xie HY, Nie W. Nano-Engineering Strategies for Tumor-Specific Therapy. ChemMedChem 2024; 19:e202300647. [PMID: 38356248 DOI: 10.1002/cmdc.202300647] [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: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/16/2024]
Abstract
Nanodelivery systems (NDSs) provide promising prospects for decreasing drug doses, reducing side effects, and improving therapeutic effects. However, the bioapplications of NDSs are still compromised by their fast clearance, indiscriminate biodistribution, and limited tumor accumulation. Hence, engineering modification of NDSs aiming at promoting tumor-specific therapy and avoiding systemic toxicity is usually needed. An NDS integrating various functionalities, including flexible camouflage, specific biorecognition, and sensitive stimuli-responsiveness, into one sequence would be "smart" and highly effective. Herein, we systematically summarize the related principles, methods, and progress. At the end of the review, we predict the obstacles to precise nanoengineering and prospects for the future application of NDSs.
Collapse
Affiliation(s)
- Zijin Li
- School of Life Science, Beijing Institute of Technology, No. 5, Zhongguancun South Street, Beijing, 100081, China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, No. 5, Zhongguancun South Street, Beijing, 100081, China
| |
Collapse
|
17
|
Yang J, Xiong W, Huang L, Li Z, Fan Q, Hu F, Duan X, Fan J, Li B, Feng J, Xu Y, Chen X, Shen Z. A mesoporous superparamagnetic iron oxide nanoparticle as a generic drug delivery system for tumor ferroptosis therapy. J Nanobiotechnology 2024; 22:204. [PMID: 38658948 PMCID: PMC11044424 DOI: 10.1186/s12951-024-02457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024] Open
Abstract
As a famous drug delivery system (DDS), mesoporous organosilica nanoparticles (MON) are degraded slowly in vivo and the degraded components are not useful for cell nutrition or cancer theranostics, and superparamagnetic iron oxide nanoparticles (SPION) are not mesoporous with low drug loading content (DLC). To overcome the problems of MON and SPION, we developed mesoporous SPIONs (MSPIONs) with an average diameter of 70 nm and pore size of 3.9 nm. Sorafenib (SFN) and/or brequinar (BQR) were loaded into the mesopores of MSPION, generating SFN@MSPION, BQR@MSPION and SFN/BQR@MSPION with high DLC of 11.5% (SFN), 10.1% (BQR) and 10.0% (SNF + BQR), demonstrating that our MSPION is a generic DDS. SFN/BQR@MSPION can be used for high performance ferroptosis therapy of tumors because: (1) the released Fe2+/3+ in tumor microenvironment (TME) can produce •OH via Fenton reaction; (2) the released SFN in TME can inhibit the cystine/glutamate reverse transporter, decrease the intracellular glutathione (GSH) and GSH peroxidase 4 levels, and thus enhance reactive oxygen species and lipid peroxide levels; (3) the released BQR in TME can further enhance the intracellular oxidative stress via dihydroorotate dehydrogenase inhibition. The ferroptosis therapeutic mechanism, efficacy and biosafety of MSPION-based DDS were verified on tumor cells and tumor-bearing mice.
Collapse
Affiliation(s)
- Jing Yang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Wei Xiong
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Lin Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Zongheng Li
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Qingdeng Fan
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Fang Hu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Xiaopin Duan
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, 510515, Guangdong, China
| | - Junbing Fan
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, 510515, Guangdong, China
| | - Bo Li
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, 510515, Guangdong, China
| | - Jie Feng
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Yikai Xu
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Clinical Imaging Research Centre, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119228, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore.
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
18
|
Qiu ZW, Zhong YT, Lu ZM, Yan N, Kong RJ, Huang JQ, Li ZF, Nie JM, Li R, Cheng H. Breaking Physical Barrier of Fibrotic Breast Cancer for Photodynamic Immunotherapy by Remodeling Tumor Extracellular Matrix and Reprogramming Cancer-Associated Fibroblasts. ACS NANO 2024; 18:9713-9735. [PMID: 38507590 DOI: 10.1021/acsnano.4c01499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Cancer-associated fibroblasts (CAFs) assist in breast cancer (BRCA) invasion and immune resistance by overproduction of extracellular matrix (ECM). Herein, we develop FPC@S, a photodynamic immunomodulator that targets the ECM, to improve the photodynamic immunotherapy for fibrotic BRCA. FPC@S combines a tumor ECM-targeting peptide, a photosensitizer (protoporphyrin IX) and an antifibrotic drug (SIS3). After anchoring to the ECM, FPC@S causes ECM remodeling and BRCA cell death by generating reactive oxygen species (ROS) in situ. Interestingly, the ROS-mediated ECM remodeling can normalize the tumor blood vessel to improve hypoxia and in turn facilitate more ROS production. Besides, upon the acidic tumor microenvironment, FPC@S will release SIS3 for reprograming CAFs to reduce their activity but not kill them, thus inhibiting fibrosis while preventing BRCA metastasis. The natural physical barrier formed by the dense ECM is consequently eliminated in fibrotic BRCA, allowing the drugs and immune cells to penetrate deep into tumors and have better efficacy. Furthermore, FPC@S can stimulate the immune system and effectively suppress primary, distant and metastatic tumors by combining with immune checkpoint blockade therapy. This study provides different insights for the development of fibrotic tumor targeted delivery systems and exploration of synergistic immunotherapeutic mechanisms against aggressive BRCA.
Collapse
Affiliation(s)
- Zi-Wen Qiu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ying-Tao Zhong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Zhen-Ming Lu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ni Yan
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ren-Jiang Kong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jia-Qi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Zhuo-Feng Li
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jun-Mei Nie
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Runqing Li
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Hong Cheng
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
19
|
Zhou X, Zhou Q, He Z, Xiao Y, Liu Y, Huang Z, Sun Y, Wang J, Zhao Z, Liu X, Zhou B, Ren L, Sun Y, Chen Z, Zhang X. ROS Balance Autoregulating Core-Shell CeO 2@ZIF-8/Au Nanoplatform for Wound Repair. NANO-MICRO LETTERS 2024; 16:156. [PMID: 38512388 PMCID: PMC10957853 DOI: 10.1007/s40820-024-01353-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/08/2024] [Indexed: 03/23/2024]
Abstract
Reactive oxygen species (ROS) plays important roles in living organisms. While ROS is a double-edged sword, which can eliminate drug-resistant bacteria, but excessive levels can cause oxidative damage to cells. A core-shell nanozyme, CeO2@ZIF-8/Au, has been crafted, spontaneously activating both ROS generating and scavenging functions, achieving the multi-faceted functions of eliminating bacteria, reducing inflammation, and promoting wound healing. The Au Nanoparticles (NPs) on the shell exhibit high-efficiency peroxidase-like activity, producing ROS to kill bacteria. Meanwhile, the encapsulation of CeO2 core within ZIF-8 provides a seal for temporarily limiting the superoxide dismutase and catalase-like activities of CeO2 nanoparticles. Subsequently, as the ZIF-8 structure decomposes in the acidic microenvironment, the CeO2 core is gradually released, exerting its ROS scavenging activity to eliminate excess ROS produced by the Au NPs. These two functions automatically and continuously regulate the balance of ROS levels, ultimately achieving the function of killing bacteria, reducing inflammation, and promoting wound healing. Such innovative ROS spontaneous regulators hold immense potential for revolutionizing the field of antibacterial agents and therapies.
Collapse
Affiliation(s)
- Xi Zhou
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Quan Zhou
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Zhaozhi He
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Yi Xiao
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Yan Liu
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Zhuohang Huang
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Yaoji Sun
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, School of Electronic Science and Engineering, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Jiawei Wang
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, School of Electronic Science and Engineering, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Zhengdong Zhao
- Department of Otorhinolaryngology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiaozhou Liu
- Department of Otorhinolaryngology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Bin Zhou
- NO.1 Middle School Affiliated to Central China Normal University, Wuhan, 430223, People's Republic of China
| | - Lei Ren
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| | - Zhiwei Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, School of Electronic Science and Engineering, Xiamen University, Xiamen, 361005, People's Republic of China.
| | - Xingcai Zhang
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
20
|
Li X, Sun T, Jiang C. Intelligent Delivery Systems in Tumor Metabolism Regulation: Exploring the Path Ahead. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309582. [PMID: 38105387 DOI: 10.1002/adma.202309582] [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: 09/16/2023] [Revised: 11/07/2023] [Indexed: 12/19/2023]
Abstract
Cancer metabolism plays multifaceted roles in the initiation and progression of tumors, and interventions in metabolism are considered fundamental approaches for cancer control. Within the vast metabolic networks of tumors, there exist numerous potential therapeutic targets, intricately interconnected with each other and with signaling networks related to immunity, metastasis, drug resistance, and more. Based on the characteristics of the tumor microenvironment, constructing drug delivery systems for multi-level modulation of the tumor microenvironment is proven as an effective strategy for achieving multidimensional control of cancer. Consequently, this article summarizes several features of tumor metabolism to provide insights into recent advancements in intelligent drug delivery systems for achieving multi-level regulation of the metabolic microenvironment in cancer, with the aim of offering a novel paradigm for cancer treatment.
Collapse
Affiliation(s)
- Xuwen Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tao Sun
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| |
Collapse
|
21
|
Lu J, Xu X, Sun X, Du Y. Protein and peptide-based renal targeted drug delivery systems. J Control Release 2024; 366:65-84. [PMID: 38145662 DOI: 10.1016/j.jconrel.2023.12.036] [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: 07/27/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Renal diseases have become an increasingly concerned public health problem in the world. Kidney-targeted drug delivery has profound transformative potential on increasing renal efficacy and reducing extra-renal toxicity. Protein and peptide-based kidney targeted drug delivery systems have garnered more and more attention due to its controllable synthesis, high biocompatibility and low immunogenicity. At the same time, the targeting methods based on protein/peptide are also abundant, including passive renal targeting based on macromolecular protein and active targeting mediated by renal targeting peptide. Here, we review the application and the drug loading strategy of different proteins or peptides in targeted drug delivery, including the ferritin family, albumin, low molecular weight protein (LMWP), different peptide sequence and antibodies. In addition, we summarized the factors influencing passive and active targeting in drug delivery system, the main receptors related to active targeting in different kidney diseases, and a variety of nano forms of proteins based on the controllable synthesis of proteins.
Collapse
Affiliation(s)
- Jingyi Lu
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xiaoling Xu
- College of Medical Sciences, Zhejiang Shuren University, 8 Shuren Street, Hangzhou, Zhejiang 310015, China.
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China.
| | - Yongzhong Du
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321299, China.
| |
Collapse
|
22
|
An Y, Xu D, Wen X, Chen C, Liu G, Lu Z. Internal Light Sources-Mediated Photodynamic Therapy Nanoplatforms: Hope for the Resolution of the Traditional Penetration Problem. Adv Healthc Mater 2024; 13:e2301326. [PMID: 37413664 DOI: 10.1002/adhm.202301326] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
Photodynamic therapy (PDT) is an alternative cancer treatment technique with a noninvasive nature, high selectivity, and minimal adverse effects. The indispensable light source used in PDT is a critical factor in determining the energy conversion of photosensitizers (PSs). Traditional light sources are primarily concentrated in the visible light region, severely limiting their penetration depth and making them prone to scattering and absorption when applied to biological tissues. For that reason, its efficacy in treating deep-seated lesions is often inadequate. Self-exciting PDT, also known as auto-PDT (APDT), is an attractive option for circumventing the limited penetration depth of traditional PDT and has acquired significant attention. APDT employs depth-independent internal light sources to excite PSs through resonance or radiative energy transfer. APDT has considerable potential for treating deep-tissue malignancies. To facilitate many researchers' comprehension of the latest research progress in this field and inspire the emergence of more novel research results. This review introduces internal light generation mechanisms and characteristics and provides an overview of current research progress based on the recently reported APDT nanoplatforms. The current challenges and possible solutions of APDT nanoplatforms are also presented and provide insights for future research in the final section of this article.
Collapse
Affiliation(s)
- Yibo An
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Dazhuang Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xiaofei Wen
- Department of Interventional Radiology, The First Affilited Hospital of Xiamen University, Xiamen, 361000, China
| | - Chuan Chen
- Department of Pharmacy, Xiamen Medical College, Xiamen, 361023, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhixiang Lu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
23
|
Yang M, Li H, Liu X, Huang L, Zhang B, Liu K, Xie W, Cui J, Li D, Lu L, Sun H, Yang B. Fe-doped carbon dots: a novel biocompatible nanoplatform for multi-level cancer therapy. J Nanobiotechnology 2023; 21:431. [PMID: 37978538 PMCID: PMC10655501 DOI: 10.1186/s12951-023-02194-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Tumor treatment still remains a clinical challenge, requiring the development of biocompatible and efficient anti-tumor nanodrugs. Carbon dots (CDs) has become promising nanomedicines for cancer therapy due to its low cytotoxicity and easy customization. RESULTS Herein, we introduced a novel type of "green" nanodrug for multi-level cancer therapy utilizing Fe-doped carbon dots (Fe-CDs) derived from iron nutrient supplement. With no requirement for target moieties or external stimuli, the sole intravenous administration of Fe-CDs demonstrated unexpected anti-tumor activity, completely suppressing tumor growth in mice. Continuous administration of Fe-CDs for several weeks showed no toxic effects in vivo, highlighting its exceptional biocompatibility. The as-synthesized Fe-CDs could selectively induce tumor cells apoptosis by BAX/Caspase 9/Caspase 3/PARP signal pathways and activate antitumoral macrophages by inhibiting the IL-10/Arg-1 axis, contributing to its significant tumor immunotherapy effect. Additionally, the epithelial-mesenchymal transition (EMT) process was inhibited under the treatment of Fe-CDs by MAPK/Snail pathways, indicating the capacity of Fe-CDs to inhibit tumor recurrence and metastasis. CONCLUSIONS A three-level tumor treatment strategy from direct killing to activating immunity to inhibiting metastasis was achieved based on "green" Fe-CDs. Our findings reveal the broad clinical potential of Fe-CDs as a novel candidate for anti-tumor nanodrugs and nanoplatform.
Collapse
Affiliation(s)
- Mingxi Yang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, 130031, People's Republic of China
| | - Haiqiu Li
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, 130031, People's Republic of China
| | - Xinchen Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Lei Huang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Boya Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Kexuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Wangni Xie
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Jing Cui
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China.
| | - Laijin Lu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China.
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, 130031, People's Republic of China.
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China.
| | - Bai Yang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
24
|
Ding C, Shi Z, Ou M, Li Y, Huang L, Wang W, Huang Q, Li M, Chen C, Zeng X, Chen H, Mei L. Dextran-based micelles for combinational chemo-photodynamic therapy of tumors via in vivo chemiluminescence. Carbohydr Polym 2023; 319:121192. [PMID: 37567697 DOI: 10.1016/j.carbpol.2023.121192] [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: 02/20/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 08/13/2023]
Abstract
Natural polysaccharides, represented by dextran, chitosan, and hyaluronic acid, are widely approved for use as pharmaceutical excipients and are important carrier materials for the design of advanced drug delivery systems, particularly in the field of anticancer drug delivery. The combination of stimuli-activable prodrug based chemotherapy and photodynamic therapy (PDT) has attracted increasing attention. Recent studies have verified the effectiveness of this strategy in the treatment of multiple aggressive cancers. However, in such combination, the stimuli-responsive chemotherapy and PDT have their own problems that need to be overcome. The uneven distribution of endogenous stimuli within tumor tissues makes it difficult for prodrug to be completely activated. And the inadequate tissue penetration depth of external light results in low efficiency of PDT. Aiming at these two bottlenecks, we designed a biocompatible dextran based - multi-component nanomedicine (PCL-NPs) that integrate a chemiluminescence agent luminol, a photosensitizer chlorine e6 (Ce6), and a reactive oxygen species (ROS)-activable thioketal-based paclitaxel (PTX) prodrug. The presence of overexpressed hydrogen peroxide (H2O2) inside tumor oxidizes the luminol moiety to generate in-situ light for PDT through chemiluminescence resonance energy transfer (CRET). The singlet oxygen (1O2) produced in this process not only directly kills tumor cells but also amplifies oxidative stress to accelerate the activation of PTX prodrug. We propose that the PCL-NPs have great therapeutic potential by simultaneously enhancing chemotherapy and PDT in a combination therapy.
Collapse
Affiliation(s)
- Chendi Ding
- Department of Oncology and Clinical Research Center, Maoming People's Hospital, Maoming 525000, China; Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; School of Medicine, Jinan University, Guangzhou 510632, China
| | - Zhaoqing Shi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Meitong Ou
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yingbang Li
- Department of Oncology and Clinical Research Center, Maoming People's Hospital, Maoming 525000, China
| | - Li Huang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Wenyan Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Qili Huang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Meihang Li
- Department of Oncology and Clinical Research Center, Maoming People's Hospital, Maoming 525000, China; School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chunbo Chen
- Department of Oncology and Clinical Research Center, Maoming People's Hospital, Maoming 525000, China.
| | - Xiaowei Zeng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Hongzhong Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
25
|
Zhong S, Zhang Z, Zhao Y, Wang S, Hu Q, Li L. Bismuth nanoclusters on nitrogen-doped porous carbon nanoenzyme for cancer therapy. NANOSCALE 2023; 15:16619-16625. [PMID: 37819091 DOI: 10.1039/d3nr03957a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Among the emerging cancer therapeutic methods, nanocatalytic therapy through the rational design of nanozymes is considered to be a promising strategy. However, high-performance nanozymes with the ability to catalyze the production of toxic substances to efficiently kill cancer cells are still highly desired. Herein, we fabricate bismuth nanoclusters loaded on nitrogen-doped porous carbon (Bi-NC) as a nanozyme for cancer therapy. The Bi-NC nanozyme displays both peroxidase (POD) and glutathione oxidase (GSHOx) biomimetic enzymatic activities, especially in a tumor microenvironment (TME), which catalyzes the production of hydroxyl radicals (·OH) and depletes antioxidant glutathione (GSH), simultaneously. Moreover, Bi-NC exhibits good photothermal conversion performance under near-infrared light irradiation. After surface modification with hyaluronic acid (HA) to improve the dispersity of nanoparticles and their accumulation in tumor tissues, Bi-NC@HA exhibits remarkable antitumor effects through the synergistic effect of catalytic and photothermal therapy. This work provides a new pathway for designing high-performance nanozymes for cancer catalytic therapy.
Collapse
Affiliation(s)
- Songjing Zhong
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, P. R. China.
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zeyu Zhang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, P. R. China.
- Center on Nanoenergy Research, Guangxi University, Nanning, 530004, China
| | - Yunchao Zhao
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, P. R. China.
- Center on Nanoenergy Research, Guangxi University, Nanning, 530004, China
| | - Shaobo Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, P. R. China.
- Center on Nanoenergy Research, Guangxi University, Nanning, 530004, China
| | - Quanhong Hu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, P. R. China.
- Center on Nanoenergy Research, Guangxi University, Nanning, 530004, China
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, P. R. China.
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Center on Nanoenergy Research, Guangxi University, Nanning, 530004, China
| |
Collapse
|
26
|
Zhu M, Zhu L, You Y, Sun M, Jin F, Song Y, Zhang J, Xu X, Ji J, Du Y. Positive Chemotaxis of CREKA-Modified Ceria@Polydopamine Biomimetic Nanoswimmers for Enhanced Penetration and Chemo-photothermal Tumor Therapy. ACS NANO 2023; 17:17285-17298. [PMID: 37595091 DOI: 10.1021/acsnano.3c05232] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2023]
Abstract
Tumor interstitial pressure represents the greatest barrier against drug diffusion into the depth of the tumor. Biometric nanomotors highlight the possibility of enhanced deep penetration and improve cellular uptake. However, control of their directionality remains difficult to achieve. Herein, we report cysteine-arginine-glutamic acid-lysine-alanine (CREKA)-modified ceria@polydopamine nanobowls as tumor microenvironment-fueled nanoscale motors for positive chemotaxis into the tumor depth or toward tumor cells. Upon laser irradiation, this nanoswimmer rapidly depletes the tumor microenvironment-specific hydrogen peroxide (H2O2) in the nanobowl, contributing to a self-generated gradient and subsequently propulsion (9.5 μm/s at 46 °C). Moreover, the asymmetrical modification of CREKA on nanobowls could automatically reconfigure the motion direction toward tumor depth or tumor cells in response to receptor-ligand interaction, leading to a deep penetration (70 μm in multicellular spheroids) and enhanced antitumor effects over conventional nanomedicine-induced chemo-photothermal therapy (tumor growth inhibition rate: 84.2% versus 56.9%). Thus, controlling the direction of nanomotors holds considerable potential for improved antitumor responses, especially in solid tumors with high tumor interstitial pressure.
Collapse
Affiliation(s)
- Minxia Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yuchan You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Mingchen Sun
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Feiyang Jin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yanling Song
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Jucong Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Jiansong Ji
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| |
Collapse
|
27
|
Zhang H, Yao M, Feng L, Wei Z, Wang Y, Han W, Zhang S. Escherichia coli-Based In Situ Triggerable Probe as an Amplifier for Sensitive Diagnosis and Penetrated Therapy of Cancer. Anal Chem 2023; 95:13073-13081. [PMID: 37610670 DOI: 10.1021/acs.analchem.3c01505] [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: 08/24/2023]
Abstract
Escherichia coli (E. coli) was used for cancer therapy due to the tumor-targeting, catalytic, and surface-reducing properties. Effective diagnosis combined with treatment of cancer based on E. coli, however, was rarely demonstrated. In this study, E. coli was used to surface reduce HAuCl4 and as a carrier to modify riboflavin (Rf) and luminol (E-Au@Rf@Lum). After targeted delivery to tumor, the E-Au@Rf@Lum probe could actively emit 425 nm blue-violet chemiluminescence (CL) to achieve cell imaging for cancer diagnosis. Furthermore, this light could in situ trigger the photosensitizer (Rf) through CL resonance energy transfer, which produces reactive oxygen species (ROS) for accurate photodynamic therapy. In return, the excessive ROS enhanced the blue-violet light which was further absorbed by Rf, and ROS production was cyclically amplified. Abundant ROS broke down the dense extracellular matrix network and penetrated deep into tumors. Besides, E. coli with excellent catalytic property could decompose H2O2 to O2 to relieve tumor hypoxia for a long time and enhance the photosensitized process of Rf. By self-illumination, effective penetration, and tumor hypoxia relief, this work opens a self-amplified therapy modality to tumor.
Collapse
Affiliation(s)
- Huairong Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Mei Yao
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Lu Feng
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Zizhen Wei
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yuqi Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Wenxiu Han
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Shusheng Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| |
Collapse
|
28
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
29
|
Lv S, Qiu Z, Yu D, Wu X, Yan X, Ren Y, Huang Y, Jiang G, Gao F. Custom-Made Piezoelectric Solid Solution Material for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300976. [PMID: 37066742 DOI: 10.1002/smll.202300976] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/25/2023] [Indexed: 06/19/2023]
Abstract
Piezoelectric material-mediated sonodynamic therapy (SDT) has received considerable research interest in cancer therapy. However, the simple applications of conventional piezoelectric materials do not realize the full potential of piezoelectric materials in medicine. Therefore, the energy band structure of a piezoelectric material is modulated in this study to meet the actual requirement for cancer treatment. Herein, an elaborate PEGylated piezoelectric solid solution 0.7BiFeO3 -0.3BaTiO3 nanoparticles (P-BF-BT NPs) is synthesized, and the resultant particles achieve excellent piezoelectric properties and their band structure is tuned via band engineering. The tuned band structure of P-BF-BT NPs is energetically favorable for the synchronous production of superoxide radicals (•O2 - ) and oxygen (O2 ) self-supply via water splitting by the piezoelectric effect. Besides, the P-BF-BT NPs can initiate the Fenton reaction to generate hydroxyl radical (•OH), and thus, chemodynamic therapy (CDT) can be augmented by ultrasound. Detailed in vitro and in vivo research has verified the promising effects of multimodal imaging-guided P-BF-BT NP-mediated synergistic SDT/CDT by the piezo-Fenton process in hypoxic tumor elimination, accompanied by high therapeutic biosafety. The current demonstrates a novel strategy for designing and synthesizing "custom-made" piezoelectric materials for cancer therapy in the future.
Collapse
Affiliation(s)
- Shanrong Lv
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 212002, P. R. China
| | - Zhili Qiu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Dehong Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- The Affiliated Pizhou Hospital of Xuzhou Medical University, Jiangsu, 221399, P. R. China
| | - Xiunan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Xiang Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 212002, P. R. China
| | - Yiping Ren
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 212002, P. R. China
| | - Yuqi Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 212002, P. R. China
| | - Guan Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 212002, P. R. China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Dermatology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 212002, P. R. China
| |
Collapse
|
30
|
Xu D, Wan Y, Xie Z, Du C, Wang Y. Hierarchically Structured Hydroxyapatite Particles Facilitate the Enhanced Integration and Selective Anti-Tumor Effects of Amphiphilic Prodrug for Osteosarcoma Therapy. Adv Healthc Mater 2023; 12:e2202668. [PMID: 36857811 DOI: 10.1002/adhm.202202668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Efficient delivery of cargo into target cells is a formidable challenge in modern medicine. Despite the great promise of biomimetic hydroxyapatite (HA) particles in tissue engineering, their potential applications in bone tumor therapy, particularly their structure-function relationships in cargo delivery to target cells, have not yet been well explored. In this study, biomimetic multifunctional composite microparticles (Bm-cMPs) are developed by integrating an amphiphilic prodrug of curcumin with hierarchically structured HA microspheres (Hs-hMPs). Then, the effects of the hierarchical structure of vehicles on the integration and delivery of cargo as well as the anti-osteosarcoma (OS) effect of the composite are determined. Different hierarchical structures of the vehicles strongly influence the self-assembly behavior of the prodrug. The flake-like crystals of Hs-hMPs enable the highest loading capacity and enhance the stability of the cargo. Compared to the normal cells, OS cells exhibit 3.56-times better uptake of flake-like Hs-hMPs, facilitating the selective anti-tumor effect of the prodrug. Moreover, Bm-cMPs suppress tumor growth and metastasis by promoting apoptosis and inhibiting cell proliferation and tumor vascularization. The findings shed light on the potential application of Bm-cMPs and suggest a feasible strategy for developing an effective targeted therapy platform using hierarchically structured minerals for OS treatment.
Collapse
Affiliation(s)
- Dong Xu
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yuxin Wan
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Zhenze Xie
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Chang Du
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yingjun Wang
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
31
|
Chen H, Cai Z, Gui J, Tang Y, Yin P, Zhu X, Zhang Y, Li H, Liu M, Yao S. A redox reaction-induced ratiometric fluorescence platform for the specific detection of ascorbic acid based on Ag 2S quantum dots and multifunctional CoOOH nanoflakes. J Mater Chem B 2023; 11:1279-1287. [PMID: 36651433 DOI: 10.1039/d2tb02438a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this work, a ratiometric fluorescent nanoplatform for the detection of ascorbic acid (AA) was constructed based on the Ag2S quantum dots (QDs) and multifunctional hydroxyl cobalt oxide nanoflakes (CoOOH NFs). Ag2S QDs can be assembled on the surface of CoOOH NFs by electrostatic adsorption, resulting in the quenching of the NIR fluorescence emission of Ag2S QDs at 680 nm effectively through the inner filter effect (IFE). o-Phenylenediamine (OPD), a common substrate of oxidase-like (OXD) mimic, is rapidly oxidized into the fluorescent product of 2,3-diaminophenazine (DAP) with the appearance of an emission peak at 575 nm under the catalysis of CoOOH NFs. After AA was added, the fluorescence emission of DAP declined because of the decline in the OXD-like activity of CoOOH NFs due to the transformation of Co2+. Simultaneously, Ag2S QDs were released, accompanied by the recovery of red fluorescence. These two fluorescent signals can be excited at the same excitation wavelength, simplifying the detection procedure. Using F575/F680 as the readout, the quantification of AA can be realized with the linear range and detection limit of 0.2 μM-20 mM and 0.014 μM, respectively. The ratiometric fluorescence sensor can be effectively used to determine the content of AA in real samples such as juice and serum. This work integrates the in-situ formation of the fluorescent species via the catalysis of the nanozyme and the redox reaction to destroy the CoOOH NFs nanozyme as well as the two dimensional nanoflake induced turn-off-on strategy for Ag2S QDs, which provides a specific strategy for the selective detection of AA and may offer a reliable approach for the construction of other biosensing platforms.
Collapse
Affiliation(s)
- Haoyu Chen
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Zifu Cai
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Jialing Gui
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Ying Tang
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Peng Yin
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Xiaohua Zhu
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Youyu Zhang
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Haitao Li
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Meiling Liu
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| | - Shouzhuo Yao
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China.
| |
Collapse
|
32
|
Liu S, Zhang M, Jin H, Wang Z, Liu Y, Zhang S, Zhang H. Iron-Containing Protein-Mimic Supramolecular Iron Delivery Systems for Ferroptosis Tumor Therapy. J Am Chem Soc 2023; 145:160-170. [PMID: 36542745 DOI: 10.1021/jacs.2c09139] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ferroptosis provides an innovative theoretical basis and method for tumor therapy but is limited by the low efficiency of conventional iron delivery systems. Herein, an efficient supramolecular iron delivery system (SIDS) is demonstrated upon the hydrolysis of FeCl3, condensation of amino acids, and self-assembly of iron-containing components. The as-assembled SIDS possesses a shuttle-like core/shell structure with β-FeOOH as the core and Fe3+/polyamino acid coordinated networks as shells. The iron content of SIDS is up to 42 wt %, which is greatly higher than that of ferritin. The iron-containing protein-mimic structure and shuttle-like morphology of SIDS facilitate tumor accumulation and cell internalization. Once exposed to the tumor microenvironment with overexpressed glutathione (GSH), the SIDS will disassemble, accompanied by the depletion of GSH and the release of Fe2+, leading to dual amplified ferroptosis. Primary studies indicate that SIDS exhibits outstanding antitumor efficacy on bladder cancer.
Collapse
Affiliation(s)
- Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Mengsi Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Hao Jin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China.,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Songling Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China.,Gynecolgical Oncology Division, Gynecology and Obstetrics Center, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Hao Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China.,State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China.,Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou 450001, P. R. China
| |
Collapse
|
33
|
Sun MC, Chen YF, Liu D, Xu XL, You YC, Lu W, Shi YJ, Ren MY, Fan YB, Du YZ, Tao XH. Effective decolonization strategy for mupirocin-resistant Staphylococcus aureus by TPGS-modified mupirocin-silver complex. Mater Today Bio 2023; 18:100534. [PMID: 36686036 PMCID: PMC9850068 DOI: 10.1016/j.mtbio.2022.100534] [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: 10/21/2022] [Revised: 12/11/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023]
Abstract
The widespread utilization of mupirocin to treat methicillin-resistant Staphylococcus aureus (MRSA)-caused infectious diseases has led to the emergence of mupirocin-resistant Staphylococcus aureus (MuRSA), posing a serious global medical threat. In order to counteract MuRSA, we develop a d-α-tocopherol polyethylene glycol 1000 succinate (TPGS) modified mupirocin and silver complex (TPGS/Mup-Ag) to combat MuRSA. The surfactivity of TPGS endows Mup-Ag with a homogeneous and small particle size (∼16 nm), which significantly enhances bacterial internalization. Silver ions are released from the mupirocin-Ag complex (Mup-Ag) to exert a synergistic antibacterial activity with mupirocin. Results manifest that our strategy reduces the concentration of mupirocin that induces 50% bacterial death from about 1000 μmol/mL to about 16 μmol/mL. In vitro bacterial infection model suggests that TPGS/Mup-Ag can not only eliminate both intracellular and inhibit bacterial adhesion, but also living cells are not affected. Results of in vivo experiments demonstrate that TPGS/Mup-Ag can effectively inhibit the progression of skin infection and accelerate wound healing, as well as alleviate systemic inflammation in both the subcutaneous infection model and the wound infection model. Furthermore, this study may contribute to the development of therapeutic agents for antibiotic-resistant bacteria and offer ideas for silver-based bactericides.
Collapse
Affiliation(s)
- Ming-Chen Sun
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, China,Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ying-Fang Chen
- HangZhou Xiaoshan District Skin Disease Hospital, Hangzhou, 311200, China
| | - Di Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Yu-Chan You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wei Lu
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, China
| | - Yun-Juan Shi
- Department of Graduate School, Bengbu Medical College, Bengbu, 233030, China
| | - Ming-Yang Ren
- Department of Graduate School, Bengbu Medical College, Bengbu, 233030, China
| | - Yi-Bin Fan
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China,Corresponding author. Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China.
| | - Xiao-Hua Tao
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, China,Corresponding author. Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People’s Hospital, 158 Shangtang Road, Hangzhou, 310014, China.
| |
Collapse
|
34
|
Integrated energy conversion units in nanoscale frameworks induce sustained generation and amplified lethality of singlet oxygen in oxidative therapy of tumor. VIEW 2022. [DOI: 10.1002/viw.20220051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
35
|
Wang X, Ding C, Zhang Z, Li C, Cao D, Zhao L, Deng G, Luo Y, Yuan C, Lu J, Liu X. Degradable nanocatalyst enables antitumor/antibacterial therapy and promotion of wound healing for diabetes via self-enhanced cascading reaction. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
Wang X, Liu Y, Xue C, Hu Y, Zhao Y, Cai K, Li M, Luo Z. A protein-based cGAS-STING nanoagonist enhances T cell-mediated anti-tumor immune responses. Nat Commun 2022; 13:5685. [PMID: 36167857 PMCID: PMC9515186 DOI: 10.1038/s41467-022-33301-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 09/13/2022] [Indexed: 12/01/2022] Open
Abstract
cGAS-STING pathway is a key DNA-sensing machinery and emerges as a promising target to overcome the immunoresistance of solid tumors. Here we describe a bovine serum albumin (BSA)/ferritin-based nanoagonist incorporating manganese (II) ions and β-lapachone, which cooperatively activates cGAS-STING signaling in dendritic cells (DCs) to elicit robust adaptive antitumor immunity. Mn2+-anchored mannose-modified BSAs and β-lapachone-loaded ferritins are crosslinked to afford bioresponsive protein nanoassemblies, which dissociate into monodispersive protein units in acidic perivascular tumor microenvironment (TME), thus enabling enhanced tumor penetration and spatiotemporally controlled Mn2+ and β-lapachone delivery to DCs and tumor cells, respectively. β-lapachone causes immunogenic tumor cell apoptosis and releases abundant dsDNA into TME, while Mn2+ enhances the sensitivity of cGAS to dsDNA and augments STING signaling to trigger downstream immunostimulatory signals. The cGAS-STING nanoagonist enhances the tumor-specific T cell-mediated immune response against poorly immunogenic solid tumors in vivo, offering a robust approach for immunotherapy in the clinics. Manganese has a crucial role in cGAS-STING-mediated DNA sensing and has emerged as a STING agonist. Here the authors report the design and characterization of a nanosystem incorporating manganese ions and the chemotherapeutic drug β-lapachone, inducing T-cell mediated anti-tumor immune responses in preclinical cancer models.
Collapse
Affiliation(s)
- Xuan Wang
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Yingqi Liu
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Chencheng Xue
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, P. R. China
| | - Yuanyuan Zhao
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China. .,Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, P. R. China.
| |
Collapse
|
37
|
Guan Y, Xing C, Tong T, Zhang X, Li J, Chen H, Zhu J, Kang Y, Pang J. Smart dual responsive nanocarriers with reactive oxygen species amplification assisted synergistic chemotherapy against prostate cancer. J Colloid Interface Sci 2022; 622:789-803. [DOI: 10.1016/j.jcis.2022.04.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
|
38
|
Shi J, Xu J, Li Y, Li B, Ming H, Nice EC, Huang C, Li Q, Wang C. Drug repurposing in cancer neuroscience: From the viewpoint of the autophagy-mediated innervated niche. Front Pharmacol 2022; 13:990665. [PMID: 36105204 PMCID: PMC9464986 DOI: 10.3389/fphar.2022.990665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Based on the bidirectional interactions between neurology and cancer science, the burgeoning field “cancer neuroscience” has been proposed. An important node in the communications between nerves and cancer is the innervated niche, which has physical contact with the cancer parenchyma or nerve located in the proximity of the tumor. In the innervated niche, autophagy has recently been reported to be a double-edged sword that plays a significant role in maintaining homeostasis. Therefore, regulating the innervated niche by targeting the autophagy pathway may represent a novel therapeutic strategy for cancer treatment. Drug repurposing has received considerable attention for its advantages in cost-effectiveness and safety. The utilization of existing drugs that potentially regulate the innervated niche via the autophagy pathway is therefore a promising pharmacological approach for clinical practice and treatment selection in cancer neuroscience. Herein, we present the cancer neuroscience landscape with an emphasis on the crosstalk between the innervated niche and autophagy, while also summarizing the underlying mechanisms of candidate drugs in modulating the autophagy pathway. This review provides a strong rationale for drug repurposing in cancer treatment from the viewpoint of the autophagy-mediated innervated niche.
Collapse
Affiliation(s)
- Jiayan Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jia Xu
- Department of Pharmacology, Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
| | - Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hui Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Qifu Li
- Department of Neurology and Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, The First Affiliated Hospital, Hainan Medical University, Haikou, China
- *Correspondence: Qifu Li, ; Chuang Wang,
| | - Chuang Wang
- Department of Pharmacology, Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
- *Correspondence: Qifu Li, ; Chuang Wang,
| |
Collapse
|
39
|
Zhang S, Zhang J, Fan X, Liu H, Zhu M, Yang M, Zhang X, Zhang H, Yu F. Ionizing Radiation-Induced Ferroptosis Based on Nanomaterials. Int J Nanomedicine 2022; 17:3497-3507. [PMID: 35966149 PMCID: PMC9364940 DOI: 10.2147/ijn.s372947] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death (RCD), that is associated with peroxidative damage to cellular membranes. A promising therapeutic method is to target ferroptosis. Nanomaterial-induced ferroptosis attracts enormous attention. Nevertheless, there are still certain shortcomings in ferroptosis, such as inadequate triggered immunogenic cell death to suit clinical demands. Various investigations have indicated that ionizing radiation (IR) can further induce ferroptosis. Consequently, it is a potential strategy for cancer therapy that combines nanomaterials and IR to induce ferroptosis. Initially, we discuss various ferroptosis inducers based on nanomaterials in this review. Furthermore, mechanisms of IR-induced ferroptosis are briefly introduced. Ultimately, we assess the feasibility of combining nanomaterials with IR to induce ferroptosis, paving the way for future research.
Collapse
Affiliation(s)
- Shenghong Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Clinical College, Anhui Medical University, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Xin Fan
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Hanhui Liu
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Clinical College, Anhui Medical University, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Mengqin Zhu
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Clinical College, Anhui Medical University, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Mengdie Yang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaoyi Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Han Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Fei Yu, Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China, Tel +86-021-66302721, Fax +86-021-66300588, Email
| |
Collapse
|
40
|
Zhao Y, Wang S, Ding Y, Zhang Z, Huang T, Zhang Y, Wan X, Wang ZL, Li L. Piezotronic Effect-Augmented Cu 2-xO-BaTiO 3 Sonosensitizers for Multifunctional Cancer Dynamic Therapy. ACS NANO 2022; 16:9304-9316. [PMID: 35699224 DOI: 10.1021/acsnano.2c01968] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ultrasound (US)-triggered sonodynamic therapy (SDT) based on semiconductor nanomaterials has attracted considerable attention for cancer therapy. However, most inorganic sonosensitizers suffer from low efficiency due to the rapid recombination of electron-hole pairs. Herein, the Cu2-xO-BaTiO3 piezoelectric heterostructure was fabricated as a sonosensitizer and chemodynamic agent, simultaneously, for improving reactive oxygen species (ROS) generation and cancer therapeutic outcome. Under US irradiation, the Cu2-xO-BaTiO3 heterojunction with a piezotronic effect exhibits high-performance singlet oxygen (1O2) and hydroxyl radical (•OH) generation to enhance SDT. Moreover, it possesses Fenton-like reaction activity to convert endogenous H2O2 into •OH for chemodynamic therapy (CDT). The integration of SDT and CDT substantially boosts ROS generation and cellular mitochondria damage, and the in vitro and in vivo results demonstrate high cytotoxicity and tumor inhibition on murine refractory breast cancer. This work realizes improvement in cancer therapy using piezoelectric heterostructures with piezotronic effects.
Collapse
Affiliation(s)
- Yunchao Zhao
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
| | - Shaobo Wang
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
| | - Yiming Ding
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
| | - Zeyu Zhang
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
| | - Tian Huang
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
| | - Yalong Zhang
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
| | - Xingyi Wan
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Zhong Lin Wang
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0245, United States
| | - Linlin Li
- Center on Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning 530004, P.R. China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100140, P.R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
41
|
Huang C, Zhan L. Network Pharmacology Identifies Therapeutic Targets and the Mechanisms of Glutathione Action in Ferroptosis Occurring in Oral Cancer. Front Pharmacol 2022; 13:851540. [PMID: 35359830 PMCID: PMC8963897 DOI: 10.3389/fphar.2022.851540] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/23/2022] [Indexed: 01/06/2023] Open
Abstract
Oral cancer (OC) is one of the most pernicious cancers with increasing incidence and mortality worldwide. Surgery is the primary approach for the treatment of early-stage OC, which reduces the quality of life of the patients. Therefore, there is an urgent need to discover novel treatments for OC. Targeting ferroptosis to induce cell death through the modulation of lipid oxidation has been used as a new approach to treat many cancers. Glutathione (GSH) is a coenzyme factor of GSH peroxidase 4, and it carries potential applicability in treating OC. By using network pharmacology and molecular docking followed by systematic bioinformatic analysis, we aimed to study GSH-targeting ferroptosis to treat OC. We identified 14 core molecular targets, namely, EGFR, PTGS2, HIF1A, VEGFA, TFRC, SLC2A1, CAV1, CDKN2A, SLC3A2, IFNG, NOX4, DDIT4, CA9, and DUSP1, involved in ferroptosis that were targeted by GSH for OC treatment. Functional characterization of these molecular targets showed their importance in the control of cell apoptosis, cell proliferation, and immune responses through various kinase activities such as the mitogen-activated protein kinase activity (e.g., ERK1 and ERK2 cascades) and modulation of TOR signaling (e.g., the HIF-1 signaling pathway). Molecular docking further revealed the direct binding of GSH with EGFR, PTGS2, and HIF1A proteins. These findings provide a novel insight into the targets of GSH in ferroptosis as well as possible molecular mechanisms involved, suggesting the possible use of GSH as a combined therapy for treating OC.
Collapse
Affiliation(s)
- Chen Huang
- The Center for Data Science in Health and Medicine, Business School, Qingdao University, Qingdao, China
| | - Lei Zhan
- Department of Ophthalmology, The Second People’s Hospital of Guilin, Guilin, China
| |
Collapse
|
42
|
Greer A. Violet-blue Light Induces "Natural" Photodynamic Plasma Disinfection with Endogenous Sensitizers. Photochem Photobiol 2022; 98:513-515. [PMID: 35020199 DOI: 10.1111/php.13591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/09/2022] [Indexed: 11/29/2022]
Abstract
Naturally, endogenous porphyrins can provide sensitized disinfection power, and to photobiologists' delight, violet-blue light has potential virtues. But progress is needed before violet-blue light treatment can used for microbe treatment of blood samples, and yet safeguard against plasma protein photooxidation. A report by Maclean et al. in this issue of Photochemistry & Photobiology on microbe reduction of blood samples showing negligible competing protein photooxidation may bring that goal a step closer.
Collapse
Affiliation(s)
- Alexander Greer
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York, 11210, United States.,Program in Chemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, New York, 10016, United States
| |
Collapse
|