1
|
Ma C, Gao L, Song K, Gu B, Wang B, Yu Y, Wang X, Li X, Hu J, Pu W, Wang Y, Wang N, Lu D, Han Z, Chen H. Targeted Dual-Responsive Liposomes Co-Deliver Jolkinolide B and Ce6 to Synergistically Enhance the Photodynamic/Immunotherapy Efficacy in Gastric Cancer through the PANoptosis Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e02289. [PMID: 40387011 DOI: 10.1002/advs.202502289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/20/2025] [Indexed: 05/20/2025]
Abstract
Improving the efficacy of gastric cancer (GC) treatment remains an ongoing challenge. Considering the increasing importance of PANoptosis, a novel form of programmed cell death, the current study integrates photodynamic therapy (PDT) and chemodynamic therapy (CDT) into nanoliposomes. This approach utilizes the ability of photosensitizer Chlorin e6 (Ce6) to generate reactive oxygen species (ROS) and the function of the natural targeting agent Jolkinolide B to activate the PANoptosis molecular switch, inducing the ROS-caspase8/PANoptosis pathway to promote GC cell death. The designed CJP-TiN liposome targets GC via internalizing RGD peptide (iRGD), and demonstrates ROS/pH dual responsiveness in the tumor microenvironment. In vitro and in vivo experiments show effective ROS generation ability under light exposure, killing tumor cells and triggers thioether bond cleavage for dual-controlled drug release. The combined therapy enhances antitumor effect, converting "cold tumors" into "hot tumors," thereby enhancing the success of immunotherapy. The role of CJP-TiN as a PANoptosis inducer in the tumor microenvironment is confirmed, thereby expanding its application potential as a molecularly targeted therapy for GC treatment, and providing a novel perspective for therapeutic strategies.
Collapse
Affiliation(s)
- Chenhui Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Lei Gao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Kewei Song
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Baohong Gu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Bofang Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Yang Yu
- Department of Thyroid Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xueyan Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Xuemei Li
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, Gansu, 730030, China
| | - Jike Hu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Weigao Pu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Yunpeng Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Na Wang
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China
| | - Dedai Lu
- Key Laboratory of Eco-Functional Polymer Materials of the Ministry of Education, Northwest Normal University, Lanzhou, 730070, China
| | - Zhijian Han
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, Gansu, 730030, China
| | - Hao Chen
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, Gansu, 730030, China
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| |
Collapse
|
2
|
Xu Y, Ren M, Deng R, Meng J, Xu L, Zhao W, Ni Y, Mao C, Zhang S. UCNPs@PVP-Hemin-GOx@CaCO 3 Nanoplatform for Ferroptosis Self-Amplification Combined with Calcium Overload. Adv Healthc Mater 2025; 14:e2404215. [PMID: 40072332 DOI: 10.1002/adhm.202404215] [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: 10/28/2024] [Revised: 02/27/2025] [Indexed: 04/26/2025]
Abstract
Due to the complexity of the tumor microenvironment (TME), current tumor treatments cannot achieve satisfactory results. A nanocomposite material, UCNPs@PVP-Hemin-GOx@CaCO3 (UPHGC NPs) is developed that responds to the TME and controls release to achieve multimodal synergistic therapy in tumor tissues. UPHGC NPs mediate photodynamic therapy (PDT), chemodynamic therapy (CDT), and starvation therapy (ST) synergistically, ultimately inducing self-amplification of ferroptosis. The Hemin loaded in UPHGC NPs exhibits peroxidase (POD) activity, which can react with H2O2 to produce ·OH (CDT) and generate the maximum amount of ·O2 - (PDT) under UV excitation from upconversion materials. Hemin can also consume glutathione (GSH), downregulate glutathione peroxidase 4 (GPX4), and combine with PDT/CDT to induce lipid peroxidation (LPO), leading to ferroptosis. In addition, Glucose oxidase (GOx) provides sufficient H2O2 for the ·OH production, amplifying ROS generation to further enhance ferroptosis. The gluconic acid produced by GOx during the ST process synergizes with the TME's acidic conditions to promote Ca2+ release, induce intracellular calcium overload, enhance oxidative stress, lead to mitochondrial dysfunction, and ultimately kill tumor cells through mitochondrial damage. Furthermore, the externally mineralized calcium carbonate can prevent premature drug release in normal tissues.
Collapse
Affiliation(s)
- Yuping Xu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Mingming Ren
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Runzhi Deng
- Nanjing Stomatological Hospital, Affliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jiajia Meng
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Lingxia Xu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Wenbo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Yanhong Ni
- Nanjing Stomatological Hospital, Affliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Shirong Zhang
- Molecular Diagnostic Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China
| |
Collapse
|
3
|
Yang L, Zhang WH, Li Y, An YL, Wu YM, Gu N, Teng GJ. Oxygen Nanobubbles Enhance ICG/Fe(III)-Mediated Dual-Modal Therapy To Induce Ferroptosis in Tumor Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11718-11730. [PMID: 39963718 DOI: 10.1021/acsami.4c19604] [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: 02/28/2025]
Abstract
Noninvasive therapies such as photodynamic therapy (PDT) and chemodynamic therapy (CDT), which rely on reactive oxygen species (ROS), are gaining attention for their low toxicity. However, single-modal treatments have individual limitations that restrict the therapeutic efficacy. Fe(III) can coordinate with the hydrophilic regions of indocyanine green (ICG) molecules to form the ICG/Fe(III) complex, making it a promising dual-modal agent for combined PDT and CDT. However, coordination with Fe(III) leads to the aggregation quenching of ICG, hindering its application in dual-modal therapy. We innovatively utilize oxygen nanobubbles, prepared solely from water and oxygen, to significantly reverse the aggregation-induced quenching of the ICG/Fe(III) complex, thereby enhancing its stability in aqueous environments. In this system, Fe(III) assembles at the nanobubble interface, coordinating with ICG's hydrophilic regions to form the ICG/Fe(III)-NBs. The oxygen nanobubbles boost PDT efficiency by improving the ICG/Fe(III) complex stability and oxygen content, while Fe(III) achieves CDT by generating hydroxyl radicals (•OH) through the Fenton reaction. This dual-modality treatment significantly disrupts the tumor's redox balance, induces ferroptosis, and demonstrates strong antitumor efficacy, reducing tumor volume to 34% of its initial size in mice. The strategy offers a promising and clinically viable approach to cancer treatment.
Collapse
Affiliation(s)
- Li Yang
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Wei-Hua Zhang
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Yan Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210009, China
| | - Yan-Li An
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Ye-Ming Wu
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Ning Gu
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210093, China
| | - Gao-Jun Teng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| |
Collapse
|
4
|
Yang J, Chen M, Li R, Sun Y, Ye P, Fang K, Wang C, Shi S, Dong C. A responsive cocktail nano-strategy breaking the immune excluded state enhances immunotherapy for triple negative breast cancer. NANOSCALE 2025; 17:4610-4623. [PMID: 39810651 DOI: 10.1039/d4nr03054k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The exclusion of immune cells from the tumor can limit the effectiveness of immunotherapy in triple negative breast cancer (TNBC). The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway plays a crucial role in priming adaptive anti-tumor immunity through the production of type I interferons (IFNs), facilitating the maturation of dendritic cells (DCs) and the function of T cells. Although the increased expression of programmed death-ligand 1 (PD-L1) upon STING activation is favorable for amplifying the efficacy of immune checkpoint inhibitors (ICIs) and realizing combination therapy, the penetration barrier remains a major obstacle. Herein, we fabricated a smart-responsive nanosystem (B&V@ZB-MCL) by integrating the extracellular matrix (ECM)-degrading drug losartan with a STING agonist (Vadimezan, abbreviated to Vad) and a PD-L1 inhibitor (BMS-1). Losartan was first released in the acidic tumor microenvironment to overcome the physical barrier, enhancing the penetration of immunotherapeutic components. Under the triggering of 1O2 generated by a photosensitizer (Ce6), the reactive oxygen species (ROS)-sensitive degradation of the nanocore ensured the site-directed release of Vad and BMS-1. The released Vad and damaged tumor DNA activated immune responses through the cGAS-STING pathway, while the elevated expression level of PD-L1 promoted the anti-tumor effect of BMS-1. Significant degradation of collagen fibers, restoration of immune effector cells, and lower tumor volume were observed in this integrated triple drug sequential therapy, which provides a promising prospect for TNBC treatment.
Collapse
Affiliation(s)
- Jingxian Yang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Mengyao Chen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Yanting Sun
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Pingting Ye
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Kang Fang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Chunhui Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Shuo Shi
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| |
Collapse
|
5
|
Yang L, Shi L, Liu Y, Liu Z, Tian Z, Li H, Zhang J, He J, Liu Y. ROS-mediated Therapeutics Combined with Metal-based Porphyrin Nanoparticles and their Applications in Tumor Treatment. Curr Med Chem 2025; 32:627-646. [PMID: 37859412 DOI: 10.2174/0109298673264765231006062032] [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: 06/02/2023] [Revised: 08/04/2023] [Accepted: 09/11/2023] [Indexed: 10/21/2023]
Abstract
High concentrations of reactive oxygen species (ROS) can disrupt cell structure and induce apoptosis and necrosis of tumor cells. Photodynamic therapy (PDT) and chemodynamic therapy (CDT) are two cancer treatments mediated by reactive oxygen species. Oxygen molecules (O2) are one of the indispensable factors in PDT and hypoxic tumor sites limit its application. However, another ROS-mediated method, CDT, can generate •OH and O2in situ by Fenton reaction or Fenton-like reaction. Synergistic PDT/CDT therapy is a strategy to overcome the limitations of tumor microenvironment therapy. In this review, PDT and CDT therapies are briefly introduced, with an emphasis on metal-basrd porphyrin nanoparticles constructed in different ways for PDT/CDT dual-mode therapy. By introducing the history and latest design schemes of the treatment model, it provides ideas for researchers engaged in ROS-mediated cancer therapies.
Collapse
Affiliation(s)
- Lingyan Yang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Lei Shi
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Yihui Liu
- The Second Hospital, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Zhenhua Liu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Zejie Tian
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Hui Li
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Jiayao Zhang
- Institute of Chemistry & Chemical Engineering, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Jun He
- Institute of Chemistry & Chemical Engineering, University of South China, Hengyang City, Hunan Province, 421001, China
| | - Yunmei Liu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, Hunan Province, 421001, China
| |
Collapse
|
6
|
Shen L, Zhang QL, Yao Y, Huang YL, Zheng Z, Li M, Xu H, Tan L, Liao X, Xia B, Li L, Redshaw C, Bai Y, Yang C. Alkyl chain length-regulated in situ intelligent nano-assemblies with AIE-active photosensitizers for photodynamic cancer therapy. Asian J Pharm Sci 2024; 19:100967. [PMID: 39640060 PMCID: PMC11617974 DOI: 10.1016/j.ajps.2024.100967] [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: 12/22/2023] [Revised: 03/08/2024] [Accepted: 07/03/2024] [Indexed: 12/07/2024] Open
Abstract
Photodynamic therapy (PDT) brings new hope for the treatment of breast cancer due to few side effects and highly effective cell killing; however, the low bioavailability of traditional photosensitizers (PSs) and their dependence on oxygen severely limits their application. Aggregation-induced emission (AIE) PSs can dramatically facilitate the photosensitization effect, which can have positive impacts on tumor PDT. To-date, most AIE PSs lack tumor targeting capability and possess poor cell delivery, resulting in their use in large quantities that are harmful to healthy tissues. In this study, a series of AIE PSs based on pyridinium-substituted triphenylamine salts ( TTPAs 1-6) with different alkyl chain lengths are synthesized. Results reveal that TTPAs 1-6 promote the generation of type I and II ROS, including ·OH and 1O2. In particular, the membrane permeability and targeting of TTPAs 4-6 bearing C8-C10 side-chains are higher than TTPAs 1-3 bearing shorter alkyl chains. Additionally, they can assemble with albumin, thereby forming nanoparticles (TTPA 4-6 NPs) in situ in blood, which significantly facilitates mitochondrial-targeting and strong ROS generation ability. Moreover, the TTPA 4-6 NPs are pH-responsive, allowing for increased accumulation or endocytosis of the tumor and enhancing the imaging or therapeutic effect. Therefore, the in vivo distributions of TTPA 4-6 NPs are visually enriched in tumor sites and exhibited excellent PDT efficacy. This work demonstrates a novel strategy for AIE PDT and has the potential to play an essential role in clinical applications using nano-delivery systems.
Collapse
Affiliation(s)
- Lingyi Shen
- School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Qi-Long Zhang
- School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Yongchao Yao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ya-Li Huang
- School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Zhichang Zheng
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Ming Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Hong Xu
- School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Lin Tan
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Xukun Liao
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Binyi Xia
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Lin Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Carl Redshaw
- Chemistry, School of Natural Sciences, University of Hull, Yorkshire HU6 7RX, UK
| | - Yang Bai
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Chengli Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
7
|
Wang Y, Wang Q, Zhong Q, Xu Y, Zheng C, Li M, Tao Y, Ju E. Immunomodulatory microneedle patch for enhanced Ferroptosis and immunogenic cell death in postoperative tumor therapy. J Control Release 2024; 376:766-776. [PMID: 39437964 DOI: 10.1016/j.jconrel.2024.10.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Microneedle technologies have emerged as a promising transdermal drug delivery platform for postoperative tumor therapy. Despite their potential, enhancing intracellular drug delivery to tumor cells and boosting the therapeutic efficiency of microneedles pose significant challenges. Herein, we develop a nanomedicine-loaded microneedle to enhance the induction of ferroptosis and immunogenic cell death for postoperative tumor therapy. This advancement is achieved by pre-formulating small molecule drugs with transition metal and protein templates into nanomedicine. Upon insertion into the tumors, the microneedle rapidly dissolves, facilitating the release and subsequent cellular uptake of the nanomedicine by tumor cells. Notably, the nanomedicine can release Mn ions and ferroptosis-inducer sulfasalazine (SAS) under acidic conditions. Furthermore, the released Mn ions can produce reactive oxygen species, which decrease the levels of glutathione (GSH) and glutathione peroxidase 4 (GPX4) with increased lipid peroxidation and enhanced induction of ferroptosis. Besides, the treatment stimulates immunogenic cell death through the cell surface exposure of calreticulin (CRT) and release of high-mobility group box 1 (HMGB1), which further stimulates dendric cell maturation, T cell infiltration, and macrophage polarization towards the M1 phenotype. Consequently, this strategy significantly inhibits postoperative tumor regrowth and extends overall survival. Our study indicates the potential of the combination of nanomedicine and microneedle to improve postoperative therapeutic efficiency.
Collapse
Affiliation(s)
- Yuqin Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Quanmin Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
8
|
Chen M, Xu C, Wang C, Huang N, Bian Z, Xiao Y, Ruan J, Sun F, Shi S. Three Birds with One Stone: Copper Ions Assisted Synergistic Cuproptosis/Chemodynamic/Photothermal Therapy by a Three-Pronged Approach. Adv Healthc Mater 2024:e2401567. [PMID: 38962848 DOI: 10.1002/adhm.202401567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Copper is indispensable to organisms, while its homeostatic imbalance may interference normal cellular physiological processes and even induce cell death. Artificially regulating cellular copper content provides a viable strategy to activate antineoplastic effect. In light of this, a copper ions homeostasis perturbator (CuP-CL) with cinnamaldehyde (Cin) packaging and thermosensitive liposome coating is reported. Following laser exposure, the doping of Cu2+ in polydopamine initiates enhanced photothermal therapy (PTT) and unlocks the outer layer of liposome, leading to the release of copper ions and Cin in tumor microenvironment with mild acidity and high glutathione (GSH) levels. The liberative Cu2+ can evoke cuproptosis and chemodynamic therapy (CDT). Meanwhile, leveraging the merits of H2O2 supply and GSH consumption, Cin serves as a tumor microenvironment regulator to amplify Cu2+ mediated cuproptosis and CDT. Additionally, the positive feedback effects of "laser-triggered PTT, PTT accelerates reactive oxygen species (ROS) generation, ROS amplifies lipid peroxide (LPO) accumulation, LPO mediates heat shock proteins (HSPs) clearance, down-regulated HSPs promote PTT" entailed the overall benefit to therapeutic outcomes. Both in vitro and in vivo results corroborate the remarkable antineoplastic performance of CuP-CL by the synergy of cuproptosis/CDT/PTT. Collectively, based on the three-pronged approach, this work plots a viable multimodal regimen for cancer therapy.
Collapse
Affiliation(s)
- Mengyao Chen
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Chang Xu
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Chunhui Wang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Nan Huang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Zhixuan Bian
- Department of Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yixuan Xiao
- Department of Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Juan Ruan
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Fenyong Sun
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Shuo Shi
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
9
|
Huang Y, Liu H, Zhao Y, Chen H, Li Q, Li X, Hua S, Cao D, Chang Y. Disrupting redox homeostasis for tumor therapy based on PDT/chemo/ferroptosis therapeutic hybrid liposomes. RSC Adv 2024; 14:20152-20162. [PMID: 38915327 PMCID: PMC11195642 DOI: 10.1039/d4ra03361b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/09/2024] [Indexed: 06/26/2024] Open
Abstract
Synergistic photodynamic therapy (PDT) with other therapeutic modalities can enhance the therapeutic efficacy of tumor treatment and reduce the adverse effects associated with drug leakage and off-target accumulation. However, shaping combined strategies for synergistic therapy remains challenging. Herein, we developed versatile hybrid liposomes self-assembled from Ce6-lipid conjugates and loaded with the chemo drug doxorubicin (DOX) and ferroptosis inducer Fe3O4 nanoparticles for synergistic PDT/chemo/ferroptosis therapy. Abundant ROS are generated by PDT upon 650 nm light irradiation, Fe3O4-mediated Fenton reaction, and DOX-induced apoptosis. Furthermore, amplifying oxidative stress in cancer cells to disrupt cellular redox homeostasis could accelerate tumor cell death through oxidative damage to lipids, proteins, and DNA. Overall, this work highlights liposome-based therapeutic nanoformulations, thus offering a breakthrough redox homeostasis-based synergistic PDT/chemo/ferroptosis therapy for lung cancer.
Collapse
Affiliation(s)
- Yuanping Huang
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| | - Hongsen Liu
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000 China
| | - Yanfei Zhao
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
| | - Haoran Chen
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| | - Qiqing Li
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| | - Xiaodan Li
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
| | - Shucheng Hua
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
| | - Dianbo Cao
- Department of Radiology, The First Hospital of Jilin University Changchun 130021 China
| | - Yulei Chang
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| |
Collapse
|
10
|
Ma J, Li Y, Ying Y, Wu B, Liu Y, Zhou J, Hu L. Progress of Mesoporous Silica Coated Gold Nanorods for Biological Imaging and Cancer Therapy. ChemMedChem 2024; 19:e202300374. [PMID: 37990850 DOI: 10.1002/cmdc.202300374] [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: 07/18/2023] [Revised: 11/19/2023] [Accepted: 11/19/2023] [Indexed: 11/23/2023]
Abstract
For unique surface plasmon absorption and fluorescence characteristics, gold nanorods have been developed and widely employed in the biomedical field. However, limitations still exist due their low specific surface area, instability and tendency agglomerate in cytoplasm. Mesoporous silica materials have been broadly applied in field of catalysts, adsorbents, nanoreactors, and drug carriers due to its unique mesoporous structure, highly comparative surface area, good stability and biocompatibility. Therefore, coating gold nanorods with a dendritic mesopore channels can effectively prevent particle agglomeration, while increasing the specific surface area and drug loading efficiency. This review discusses the advancements of GNR@MSN in synthetic process, bio-imaging technique and tumor therapy. Additionally, the further application of GNR@MSN in imaging-guided treatment modalities is explored, while its promising superior application prospect is highlighted. Finally, the issues related to in vivo studies are critically examined for facilitating the transition of this promising nanoplatform into clinical trials.
Collapse
Affiliation(s)
- Jiaying Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| | - Yongzhen Li
- Department of Pharmacy, School of Pharmacy, University of South China, Hengyang, 421001, PR China
| | - Yunfei Ying
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| | - Baibei Wu
- Department of Clinical Medicine, University of South China, Hengyang, 421001, PR China
| | - Yanmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| | - Juan Zhou
- School of Mechanical Engineering, University of South China, Hengyang, 421001, PR China
| | - Lidan Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| |
Collapse
|
11
|
Zhang JA, Haddleton D, Wilson P, Zhu LH, Dai CY, Zhao LL. pH-Responsive Amphiphilic Triblock Fluoropolymers as Assemble Oxygen Nanoshuttles for Enhancing PDT against Hypoxic Tumor. Bioconjug Chem 2024; 35:400-411. [PMID: 38366969 DOI: 10.1021/acs.bioconjchem.4c00029] [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: 02/19/2024]
Abstract
Photodynamic therapy (PDT) is a cancer treatment strategy that utilizes photosensitizers to convert oxygen within tumors into reactive singlet oxygen (1O2) to lyse tumor cells. Nevertheless, pre-existing tumor hypoxia and oxygen consumption during PDT can lead to an insufficient oxygen supply, potentially reducing the photodynamic efficacy. In response to this issue, we have devised a pH-responsive amphiphilic triblock fluorinated polymer (PDP) using copper-mediated RDRP. This polymer, composed of poly(ethylene glycol) methyl ether acrylate, 2-(diethylamino)ethyl methacrylate, and (perfluorooctyl)ethyl acrylate, self-assembles in an aqueous environment. Oxygen, chlorine e6 (Ce6), and doxorubicin (DOX) can be codelivered efficiently by PDP. The incorporation of perfluorocarbon into the formulation enhances the oxygen-carrying capacity of PDP, consequently extending the lifetime of 1O2. This increased lifetime, in turn, amplifies the PDT effect and escalates the cellular cytotoxicity. Compared with PDT alone, PDP@Ce6-DOX-O2 NPs demonstrated significant inhibition of tumor growth. This study proposes a novel strategy for enhancing the efficacy of PDT.
Collapse
Affiliation(s)
- Jun-An Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - David Haddleton
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | - Paul Wilson
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
| | - Lin-Hua Zhu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
- Key Laboratory of Functional Organic Polymers of Haikou, Tropical Functional Polymer Materials Engineering Research Center of Hainan, Haikou 571158, China
| | - Chun-Yan Dai
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
- Department of Chemistry, Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
- Key Laboratory of Functional Organic Polymers of Haikou, Tropical Functional Polymer Materials Engineering Research Center of Hainan, Haikou 571158, China
| | - Lin-Lu Zhao
- College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
12
|
Wang SK, Zhang XT, Jiang XY, Geng BJ, Qing TL, Li L, Chen Y, Li JF, Zhang XF, Xu SG, Zhu JB, Zhu YP, Wang MT, Chen JK. Activation of Piezo1 increases the sensitivity of breast cancer to hyperthermia therapy. Open Med (Wars) 2024; 19:20240898. [PMID: 38463518 PMCID: PMC10921451 DOI: 10.1515/med-2024-0898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/23/2023] [Accepted: 12/15/2023] [Indexed: 03/12/2024] Open
Abstract
Photothermal therapy (PTT) of nanomaterials is an emerging novel therapeutic strategy for breast cancer. However, there exists an urgent need for appropriate strategies to enhance the antitumor efficacy of PTT and minimize damage to surrounding normal tissues. Piezo1 might be a promising novel photothermal therapeutic target for breast cancer. This study aims to explore the potential role of Piezo1 activation in the hyperthermia therapy of breast cancer cells and investigate the underlying mechanisms. Results showed that the specific agonist of Piezo1 ion channel (Yoda1) aggravated the cell death of breast cancer cells triggered by heat stress in vitro. Reactive oxygen species (ROS) production was significantly increased following heat stress, and Yoda1 exacerbated the rise in ROS release. GSK2795039, an inhibitor of NADPH oxidase 2 (NOX2), reversed the Yoda1-mediated aggravation of cellular injury and ROS generation after heat stress. The in vivo experiments demonstrate the well photothermal conversion efficiency of TiCN under the 1,064 nm laser irradiation, and Yoda1 increases the sensitivity of breast tumors to PTT in the presence of TiCN. Our study reveals that Piezo1 activation might serve as a photothermal sensitizer for PTT, which may develop as a promising therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Shao-Kang Wang
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiao-Ting Zhang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xuan-Yao Jiang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Bi-Jiang Geng
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Tao-Lin Qing
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Lei Li
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Emergency, The Second Naval Hospital of Southern Theater Command of PLA, Hainan, China
- Heatstroke Treatment and Research Center of PLA, Hainan, China
| | - Yun Chen
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jin-Feng Li
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Xiao-Fang Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Shuo-Gui Xu
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jiang-Bo Zhu
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Yu-Ping Zhu
- Basic Medical Experimental Teaching Center, Basic Medical College, Naval Medical University, No 800, Xiangyin Road, Shanghai, 200433, China
| | - Mei-Tang Wang
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ji-Kuai Chen
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
13
|
Lv L, Fu Z, You Q, Xiao W, Wang H, Wang C, Yang Y. Enhanced photodynamic therapy through multienzyme-like MOF for cancer treatment. Front Bioeng Biotechnol 2024; 11:1338257. [PMID: 38312507 PMCID: PMC10834778 DOI: 10.3389/fbioe.2023.1338257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/29/2023] [Indexed: 02/06/2024] Open
Abstract
Overcoming resistance to apoptosis is a major challenge in cancer therapy. Recent research has shown that manipulating mitochondria, the organelles critical for energy metabolism in tumor cells, can increase the effectiveness of photodynamic therapy and trigger apoptosis in tumor cells. However, there is currently insufficient research and effective methods to exploit mitochondrial damage to induce apoptosis in tumor cells and improve the effectiveness of photodynamic therapy. In this study, we present a novel nanomedicine delivery and therapeutic system called PyroFPSH, which utilizes a nanozymes-modified metal-organic framework as a carrier. PyroFPSH exhibits remarkable multienzyme-like activities, including glutathione peroxidase (GPx) and catalase (CAT) mimicry, allowing it to overcome apoptosis resistance, reduce endogenous glutathione levels, and continuously generate reactive oxygen species (ROS). In addition, PyroFPSH can serve as a carrier for the targeted delivery of sulfasalazine, a drug that can induce mitochondrial depolarization in tumor cells, thereby reducing oxygen consumption and energy supply in the mitochondria of tumor cells and weakening resistance to other synergistic treatment approaches. Our experimental results highlight the potential of PyroFPSH as a versatile nanoplatform in cancer treatment. This study expands the biomedical applications of nanomaterials as platforms and enables the integration of various novel therapeutic strategies to synergistically improve tumor therapy. It deepens our understanding of multienzyme-mimicking active nanocarriers and mitochondrial damage through photodynamic therapy. Future research can further explore the potential of PyroFPSH in clinical cancer treatment and improve its drug loading capacity, biocompatibility and targeting specificity. In summary, PyroFPSH represents a promising therapeutic approach that can provide new insights and possibilities for cancer treatment.
Collapse
Affiliation(s)
- Letian Lv
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhao Fu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Wei Xiao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Zhang J, Sun Y, Ren L, Chen L, Nie L, Shavandi A, Yunusov KE, Aharodnikau UE, Solomevich SO, Jiang G. Red Blood Cell Membrane-Camouflaged Polydopamine and Bioactive Glass Composite Nanoformulation for Combined Chemo/Chemodynamic/Photothermal Therapy. ACS Biomater Sci Eng 2024; 10:442-454. [PMID: 38047725 DOI: 10.1021/acsbiomaterials.3c01239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Combinations of different therapeutic strategies, including chemotherapy (CT), chemodynamic therapy (CDT), and photothermal therapy (PTT), are needed to effectively address evolving drug resistance and the adverse effects of traditional cancer treatment. Herein, a camouflage composite nanoformulation (TCBG@PR), an antitumor agent (tubercidin, Tub) loaded into Cu-doped bioactive glasses (CBGs) and subsequently camouflaged by polydopamine (PDA), and red blood cell membranes (RBCm), was successfully constructed for targeted and synergetic antitumor therapies by combining CT of Tub, CDT of doped copper ions, and PTT of PDA. In addition, the TCBG@PRs composite nanoformulation was camouflaged with a red blood cell membrane (RBCm) to improve biocompatibility, longer blood retention times, and excellent cellular uptake properties. It integrated with long circulation and multimodal synergistic treatment (CT, CDT, and PTT) with the benefit of RBCms to avoid immune clearance for efficient targeted delivery to tumor locations, producing an "all-in-one" nanoplatform. In vivo results showed that the TCBG@PRs composite nanoformulation prolonged blood circulation and improved tumor accumulation. The combination of CT, CDT, and PTT therapies enhanced the antitumor therapeutic activity, and light-triggered drug release reduced systematic toxicity and increased synergistic antitumor effects.
Collapse
Affiliation(s)
- Junhao Zhang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers, Hangzhou 310018, China
| | - Yanfang Sun
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Luping Ren
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers, Hangzhou 310018, China
| | - Lianxu Chen
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers, Hangzhou 310018, China
| | - Lei Nie
- College of Life Sciences, Xinyang Normal University, Xinyang 464000, China
| | - Amin Shavandi
- École polytechnique de Bruxelles, Université libre de Bruxelles (ULB), 3BIO10 BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, Brussels 1050, Belgium
| | - Khaydar E Yunusov
- Institute of Polymer Chemistry and Physics, Uzbekistan Academy of Sciences, Tashkent 100128, Uzbekistan
| | - Uladzislau E Aharodnikau
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk 220030, Belarus
| | - Sergey O Solomevich
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk 220030, Belarus
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers, Hangzhou 310018, China
| |
Collapse
|
15
|
Zou J, Sun R, He M, Chen Y, Cheng Y, Xia C, Ma Y, Zheng S, Fu X, Yuan Z, Lan M, Lou K, Chen X, Gao F. Sequential Rocket-Mode Bioactivating Ticagrelor Prodrug Nanoplatform Combining Light-Switchable Diphtherin Transgene System for Breast Cancer Metastasis Inhibition. ACS APPLIED MATERIALS & INTERFACES 2023; 15:53198-53216. [PMID: 37942626 DOI: 10.1021/acsami.3c11594] [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: 11/10/2023]
Abstract
The increased risk of breast cancer metastasis is closely linked to the effects of platelets. Our previously light-switchable diphtheria toxin A fragment (DTA) gene system, known as the LightOn system, has demonstrated significant therapeutic potential; it lacks antimetastatic capabilities. In this study, we devised an innovative system by combining cell membrane fusion liposomes (CML) loaded with the light-switchable transgene DTA (pDTA) and a ticagrelor (Tig) prodrug. This innovative system, named the sequential rocket-mode bioactivating drug delivery system (pDTA-Tig@CML), aims to achieve targeted pDTA delivery while concurrently inhibiting platelet activity through the sequential release of Tig triggered by reactive oxygen species with the tumor microenvironment. In vitro investigations have indicated that pDTA-Tig@CML, with its ability to sequentially release Tig and pDTA, effectively suppresses platelet activity, resulting in improved therapeutic outcomes and the mitigation of platelet driven metastasis in breast cancer. Furthermore, pDTA-Tig@CML exhibits enhanced tumor aggregation and successfully restrains tumor growth and metastasis. It also reduces the levels of ADP, ATP, TGF-β, and P-selectin both in vitro and in vivo, underscoring the advantages of combining the bioactivating Tig prodrug nanoplatform with the LightOn system. Consequently, pDTA-Tig@CML emerges as a promising light-switchable DTA transgene system, offering a novel bioactivating prodrug platform for breast cancer treatment.
Collapse
Affiliation(s)
- Jiafeng Zou
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Rui Sun
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Muye He
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - You Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Cheng
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Chuanhe Xia
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ying Ma
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Shulei Zheng
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiuzhi Fu
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zeting Yuan
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyan Lou
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xianjun Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Research Unit of New Techniques for Live-Cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Feng Gao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
16
|
Zhao S, Li Y, Cheng B. A tumor microenvironment-responsive microneedle patch for chemodynamic therapy of oral squamous cell carcinoma. NANOSCALE ADVANCES 2023; 5:6162-6169. [PMID: 37941950 PMCID: PMC10629002 DOI: 10.1039/d3na00527e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/16/2023] [Indexed: 11/10/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors of the head and neck, and this disease has become a threat to public health due to its poor prognosis and high fatality rate. Chemodynamic therapy (CDT) is an emerging oncology treatment based on the Fenton reaction. However, the lack of endogenous hydrogen peroxide (H2O2) in tumor cells and the high concentration of glutathione (GSH) that depletes toxic hydroxyl radicals (·OH) significantly impair the efficacy of CDT. Here, we developed a polyvinyl alcohol (PVA)-based soluble microneedle patch (denoted as Fe3O4 + VC-MN) loaded with Fe3O4 nanoparticles (NPs) and vitamin C (VC) for the effective treatment of OSCC. When Fe3O4 + VC-MNs are inserted into the OSCC tissue, the Fe3O4 NPs and VC loaded in the tip of the needle are released in a targeted manner. After VC is converted into oxidized vitamin C (DHA), it can consume GSH in tumor cells and generate sufficient intracellular H2O2in situ. Moreover, by virtue of their peroxidase-like activity, Fe3O4 NPs can induce the generation of lethal ·OH through the Fenton reaction with the aforementioned H2O2, leading to tumor cell ferroptosis and apoptosis, thus achieving CDT. Collectively, this functional microneedle patch provides a more efficient and minimally invasive targeted drug delivery solution for the treatment of OSCC.
Collapse
Affiliation(s)
- Siyu Zhao
- Department of Stomatology, Zhongnan Hospital of Wuhan University No. 169, Donghu Road, Wuchang District Wuhan 430071 China
| | - Yue Li
- Department of Stomatology, Zhongnan Hospital of Wuhan University No. 169, Donghu Road, Wuchang District Wuhan 430071 China
| | - Bo Cheng
- Department of Stomatology, Zhongnan Hospital of Wuhan University No. 169, Donghu Road, Wuchang District Wuhan 430071 China
| |
Collapse
|
17
|
Ashrafizadeh M, Zarrabi A, Bigham A, Taheriazam A, Saghari Y, Mirzaei S, Hashemi M, Hushmandi K, Karimi-Maleh H, Nazarzadeh Zare E, Sharifi E, Ertas YN, Rabiee N, Sethi G, Shen M. (Nano)platforms in breast cancer therapy: Drug/gene delivery, advanced nanocarriers and immunotherapy. Med Res Rev 2023; 43:2115-2176. [PMID: 37165896 DOI: 10.1002/med.21971] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/09/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023]
Abstract
Breast cancer is the most malignant tumor in women, and there is no absolute cure for it. Although treatment modalities including surgery, chemotherapy, and radiotherapy are utilized for breast cancer, it is still a life-threatening disease for humans. Nanomedicine has provided a new opportunity in breast cancer treatment, which is the focus of the current study. The nanocarriers deliver chemotherapeutic agents and natural products, both of which increase cytotoxicity against breast tumor cells and prevent the development of drug resistance. The efficacy of gene therapy is boosted by nanoparticles and the delivery of CRISPR/Cas9, Noncoding RNAs, and RNAi, promoting their potential for gene expression regulation. The drug and gene codelivery by nanoparticles can exert a synergistic impact on breast tumors and enhance cellular uptake via endocytosis. Nanostructures are able to induce photothermal and photodynamic therapy for breast tumor ablation via cell death induction. The nanoparticles can provide tumor microenvironment remodeling and repolarization of macrophages for antitumor immunity. The stimuli-responsive nanocarriers, including pH-, redox-, and light-sensitive, can mediate targeted suppression of breast tumors. Besides, nanoparticles can provide a diagnosis of breast cancer and detect biomarkers. Various kinds of nanoparticles have been employed for breast cancer therapy, including carbon-, lipid-, polymeric- and metal-based nanostructures, which are different in terms of biocompatibility and delivery efficiency.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, Italy
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yalda Saghari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hassan Karimi-Maleh
- School of Resources and Environment, University of Electronic Science and Technology of China, Chengdu, PR China
| | | | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mingzhi Shen
- Department of Cardiology, Hainan Hospital of PLA General Hospital, Sanya, China
| |
Collapse
|
18
|
Guo Y, Ma R, Zhang M, Cao Y, Zhang Z, Yang W. Nanotechnology-Assisted Immunogenic Cell Death for Effective Cancer Immunotherapy. Vaccines (Basel) 2023; 11:1440. [PMID: 37766117 PMCID: PMC10534761 DOI: 10.3390/vaccines11091440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Tumor vaccines have been used to treat cancer. How to efficiently induce tumor-associated antigens (TAAs) secretion with host immune system activation is a key issue in achieving high antitumor immunity. Immunogenic cell death (ICD) is a process in which tumor cells upon an external stimulus change from non-immunogenic to immunogenic, leading to enhanced antitumor immune responses. The immune properties of ICD are damage-associated molecular patterns and TAA secretion, which can further promote dendritic cell maturation and antigen presentation to T cells for adaptive immune response provocation. In this review, we mainly summarize the latest studies focusing on nanotechnology-mediated ICD for effective cancer immunotherapy as well as point out the challenges.
Collapse
Affiliation(s)
- Yichen Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (R.M.); (M.Z.); (Y.C.)
| | - Rong Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (R.M.); (M.Z.); (Y.C.)
| | - Mengzhe Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (R.M.); (M.Z.); (Y.C.)
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (R.M.); (M.Z.); (Y.C.)
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (R.M.); (M.Z.); (Y.C.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.G.); (R.M.); (M.Z.); (Y.C.)
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| |
Collapse
|
19
|
Wang Y, Wang Q, Wang X, Yao P, Dai Q, Qi X, Yang M, Zhang X, Huang R, Yang J, Wang Q, Xia P, Zhang D, Sun F. Docetaxel-loaded pH/ROS dual-responsive nanoparticles with self-supplied ROS for inhibiting metastasis and enhancing immunotherapy of breast cancer. J Nanobiotechnology 2023; 21:286. [PMID: 37608285 PMCID: PMC10464340 DOI: 10.1186/s12951-023-02013-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/18/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Although stimuli-responsive nanoplatforms were developed to deliver immunogenic cell death (ICD) inducers to enhance cancer immunotherapy, the complete release of ICD inducers into the tumor microenvironment (TME) was limited by the inadequate supplementation of endogenous stimulus (e.g., reactive oxygen species (ROS)). To address this issue, we synthesized a self-responsive nanomaterial with self-supplied ROS, which mainly consists of a ROS responsive moiety HPAP and cinnamaldehyde (CA) as the ROS-generating agent. The endogenous ROS can accelerate the degradation of HPAP in materials to release docetaxel (DTX, an ICD inducer). In intracellular acidic environment, the pH-sensitive acetal was cleaved to release CA. The released CA in turn induces the generation of more ROS through mitochondrial damage, resulting in amplified DTX release. Using this self-cycling and self-responsive nanomaterial as a carrier, DTX-loaded pH/ROS dual-responsive nanoparticles (DTX/FA-CA-Oxi-αCD NPs) were fabricated and evaluated in vitro and in vivo. RESULTS In vitro experiments validated that the NPs could be effectively internalized by FA-overexpressed cells and completely release DTX in acidic and ROS microenvironments to induce ICD effect. These NPs significantly blocked 4T1 cell migration and decreased cell invasion. In vivo experiments demonstrated that the tumor-targeted NPs significantly inhibited tumor growth and blocked tumor metastasis. More importantly, these NPs significantly improved immunotherapy through triggering effector T-cell activation and relieving the immunosuppressive state of the TME. CONCLUSIONS Our results demonstrated that DTX/FA-CA-Oxi-αCD NPs displayed great potential in preventing tumor metastasis, inhibiting tumor growth, and improving the efficacy of anti-PD-1antibody.
Collapse
Affiliation(s)
- Yu Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qianmei Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaowen Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Pu Yao
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qing Dai
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xiao Zhang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Rong Huang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jing Yang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qian Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Peiyuan Xia
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Department of Urology, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing, 400038, China.
| | - Fengjun Sun
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
20
|
Liu S, Han S, Song Y, Sun R, Zhao L, Yan C. Disulfide-Bridged Dendritic Organosilicas-Based Biodegradable Molecularly Imprinted Polymers for Multiple Targeting and pH/Redox-Responsive Drug Release toward Chemical/Photodynamic Synergistic Tumor Therapy. Adv Healthc Mater 2023; 12:e2300184. [PMID: 36943098 DOI: 10.1002/adhm.202300184] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
In this study, a sialic acid (SA) and transferrin (TF) imprinted biodegradable disulfide bridging organosilicas-based drug delivery system (SS-DMONS/DOX-Ce6@MIPs) for targeted cancer therapy is constructed, for the first time. Disulfide bridged dendritic mesoporous organosilicas nanoparticles (SS-DMONs) not only enhance drug loading as the drug repository, but also provide enough specific surface area for the molecular imprinting shell to expose more degradation and imprinted sites on the surface. In addition, SS can be disturbed in a highly reducing tumor microenvironment to achieve degradation. The biodegradable imprinting film, prepared with customized 2-amino-N-(3,4-dihydroxyphenethyl)-3-mercaptopropanamide and 4-mercaptophenylboronic acid as functional monomers, endows SS-DMONs with active targeting capacity, and responsive drug release through degradation under acidic and highly reductive tumor microenvironment. SS-DMONS/DOX-Ce6@MIPs after binding of TF can target tumor cells actively through multiple interactions, including the affinity between antigen and antibody, and the specific recognition between molecularly imprinted polymers and template molecules. Under laser irradiation the loaded chlorin e6 (Ce6) that can produce toxic reactive oxygen, combined with the doxorubicin (DOX), achieves chemical/photodynamic synergistic anticancer effects. SS-DMONS/DOX-Ce6@MIPs present excellent tumor targeting and dual-responsive drug release, which provides an effective strategy for chemical/photodynamic antitumor therapy.
Collapse
Affiliation(s)
- Shiwei Liu
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
| | - Shuang Han
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
- Heilongjiang Provincial Key Laboratory of Catalytic Synthesis for Fine Chemicals, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
| | - Yuzhuo Song
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
| | - Ruonan Sun
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
| | - Le Zhao
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
| | - Chen Yan
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar, Heilongjiang, 161006, China
| |
Collapse
|
21
|
Yang R, Chen L, Wang Y, Zhang L, Zheng X, Yang Y, Zhu Y. Tumor microenvironment responsive metal nanoparticles in cancer immunotherapy. Front Immunol 2023; 14:1237361. [PMID: 37575228 PMCID: PMC10413122 DOI: 10.3389/fimmu.2023.1237361] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Malignant tumors have a unique tumor microenvironment (TME), which includes mild acidity, hypoxia, overexpressed reactive oxygen species (ROS), and high glutathione (GSH) levels, among others. Recently, TME regulation approaches have attracted widespread attention in cancer immunotherapy. Nanoparticles as drug delivery systems have ability to modulate the hydrophilicity of drugs to affect drug uptake and efflux in tumor. Especially, the metal nanoparticles have been extensive applied for tumor immunotherapy due to their unique physical properties and elaborate design. However, the potential deficiencies of metal nanoparticles due to their low biodegradability, toxicity and treatment side effects restrict their clinical application. In this review, we briefly introduce the feature characteristics of the TME and the recent advances in tumor microenvironment responsive metal nanoparticles for tumor immunotherapy. In addition, nanoparticles could be combined with other treatments, such as chemotherapy, radiotherapy and photodynamic therapy also is presented. Finally, the challenges and outlook for improving the antitumor immunotherapy efficiency, side effect and potential risks of metal nanoparticles has been discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Yong Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuxuan Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
22
|
Dirersa WB, Getachew G, Wibrianto A, Rasal AS, Gurav VS, Zakki Fahmi M, Chang JY. Molybdenum-oxo-sulfide quantum dot-based nanocarrier: Efficient generation of reactive oxygen species via photo/chemodynamic therapy and stimulus-induced drug release. J Colloid Interface Sci 2023:S0021-9797(23)00890-1. [PMID: 37230831 DOI: 10.1016/j.jcis.2023.05.099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
The fabrication of multifunctional nano-therapies has increased gradually to strengthen the therapeutic performance and minimize adverse effects of traditional cancer treatment strategies. Currently, we have designed a facile preparation drug-loaded nanocarrier for multimodal cancer therapy upon external stimuli. First, defect-rich molybdenum oxo-sulfide (MoOxS2-x) quantum dots (QDs) was synthesized via rapid biomineralization techniques with superior optical quantum yield reaching upto 37.28%. The presence of the Fenton ion, Mo+IV/+VI, enables MoOxS2-x QDs to efficiently catalyze peroxide solutions to produce •OH radicals for chemodynamic treatment (CDT) and also deactivate the intracellular glutathione (GSH) enzymes through redox reaction for boosted reactive oxygen species (ROS)-mediated therapies. In addition, upon laser combination, MoOxS2-x QDs generate ROS for photodynamic therapy (PDT). Also, due to a large amount of sulfide content, MoOxS2-x QDs showed excellent H2S gas release in acidic pH for cancer gas therapy. Then, MoOxS2-x QDs was further conjugated with ROS-responsive thioketal linked Camptothecin (CPT-TK-COOH) drug, forming a multitargeted MoOxS2-xCPT anticancer agent with better drug-loading efficiency (38.8%). After triggering the ROS generation through the CDT and PDT mechanisms, the thioketal linkage was disrupted, releasing up to 79% of the CPT drug in 48 h. Besides, in vitro experiments verified that MoOxS2-x QDs possess higher biocompatibility with 4T1 and HeLa cells but also showed considerable toxicity in the presence of laser/H2O2, resulting in 84.45% cell death through PDT/CDT and chemotherapeutic effects. Therefore, the designed MoOxS2-xCPT exhibited outstanding therapeutic benefits for image-guided cancer therapy.
Collapse
Affiliation(s)
- Worku Batu Dirersa
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan, Republic of China
| | - Girum Getachew
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan, Republic of China
| | - Aswandi Wibrianto
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan, Republic of China
| | - Akash S Rasal
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan, Republic of China
| | - Vivek S Gurav
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan, Republic of China
| | | | - Jia-Yaw Chang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan, Republic of China.
| |
Collapse
|
23
|
Su Y, Jin G, Zhou H, Yang Z, Wang L, Mei Z, Jin Q, Lv S, Chen X. Development of stimuli responsive polymeric nanomedicines modulating tumor microenvironment for improved cancer therapy. MEDICAL REVIEW (2021) 2023; 3:4-30. [PMID: 37724108 PMCID: PMC10471091 DOI: 10.1515/mr-2022-0048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 02/16/2023] [Indexed: 09/20/2023]
Abstract
The complexity of the tumor microenvironment (TME) severely hinders the therapeutic effects of various cancer treatment modalities. The TME differs from normal tissues owing to the presence of hypoxia, low pH, and immune-suppressive characteristics. Modulation of the TME to reverse tumor growth equilibrium is considered an effective way to treat tumors. Recently, polymeric nanomedicines have been widely used in cancer therapy, because their synthesis can be controlled and they are highly modifiable, and have demonstrated great potential to remodel the TME. In this review, we outline the application of various stimuli responsive polymeric nanomedicines to modulate the TME, aiming to provide insights for the design of the next generation of polymeric nanomedicines and promote the development of polymeric nanomedicines for cancer therapy.
Collapse
Affiliation(s)
- Yuanzhen Su
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Materials Science and Engineering, Peking University, Beijing, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Guanyu Jin
- School of Materials Science and Engineering, Peking University, Beijing, China
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Huicong Zhou
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Zhaofan Yang
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Lanqing Wang
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Zi Mei
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Qionghua Jin
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Materials Science and Engineering, Peking University, Beijing, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
24
|
Zhang H, Chen W, Wang J, Du W, Wang B, Song L, Hu Y, Ma X. A novel ROS-activable self-immolative prodrug for tumor-specific amplification of oxidative stress and enhancing chemotherapy of mitoxantrone. Biomaterials 2023; 293:121954. [PMID: 36538847 DOI: 10.1016/j.biomaterials.2022.121954] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) as well-known endogenous stimuli has been widely used to activate drug delivery systems (DDSs) for tumor-specific therapy. Unfortunately, endogenous ROS in the tumor microenvironment (TME) is not enough to achieve effective therapeutic efficacy and cancer cells have adapted to high oxidative stress by upregulating glutathione (GSH) level. Herein, we devised a novel ROS-activable self-immolative prodrug CASDB with both GSH-depletion ability and ROS self-supply competence. Then, an stimuli-responsive nanoplatform integrating CASDB with clinical chemotherapeutics mitoxantrone (MTO) and PLGA was fabricated (denoted as CMPs) through nanoprecipitation method. The CMPs could achieve desired accumulation at tumor tissues through enhanced permeability and retention (EPR) effects. Then the accumulated CMPs could induce tumor cell apoptosis efficiently. Especially, ROS in tumor sites could trigger the immolation of CASDB to generate CA and quinone methide (QM). Then CA and QM cooperatively promoted damage of mitochondria due to oxidative stress and led to cancer cells more sensitive to MTO. Accordingly, MTO could perturb cellular microenvironment of cancer cells then promote the degradation of CASDB. The experiment results demonstrated that CMPs were ideal for desirable synergetic tumor-specific anticancer therapy with negligible systemic toxicity. The half-maximal inhibitory concentrations (IC50) value of CMPs was 6.53 μM, while the IC50 values of MTO was 14.76 μM. And the CMPs group showed the strongest tumor suppressor effect with the tumor sizes increased to 1.2-fold (Control group: 20.6-fold, MTO only: 3.0-fold). This study should be inspirational for designing efficient prodrugs to overcome the handicaps of traditional chemotherapy.
Collapse
Affiliation(s)
- Hongjie Zhang
- CAS Key Lab of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, PR China
| | - Weijian Chen
- CAS Key Lab of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, PR China
| | - Jing Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, PR China
| | - Wenxiang Du
- CAS Key Lab of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, PR China
| | - Bibo Wang
- CAS Key Lab of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, PR China
| | - Lei Song
- CAS Key Lab of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, PR China.
| | - Yuan Hu
- CAS Key Lab of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, PR China.
| | - Xiaopeng Ma
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, PR China.
| |
Collapse
|
25
|
Wu H, Du X, Xu J, Kong X, Li Y, Liu D, Yang X, Ye L, Ji J, Xi Y, Zhai G. Multifunctional biomimetic nanoplatform based on photodynamic therapy and DNA repair intervention for the synergistic treatment of breast cancer. Acta Biomater 2023; 157:551-565. [PMID: 36513248 DOI: 10.1016/j.actbio.2022.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/27/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Photodynamic therapy (PDT) is a minimally invasive and locally effective treatment method, which has been used in the clinical treatment of a variety of superficial tumors. In recent years, PDT has received extensive attention due to its induction of immunogenic cell death (ICD). However, the repair mechanism of tumor cells and low immune response limit the further development of PDT. To this end, a multifunctional biomimetic nanoplatform 4T1Mem@PGA-Ce6/Ola (MPCO) is developed to co-deliver the photosensitizer Chlorin e6 (Ce6) and Olaparib (Ola) with the function of preventing DNA repair. The nanoplatform shows efficient tumor targeting and cellular internalization properties due to cell membrane camouflage, and Ce6 and Ola produce a significant synergistic anti-tumor effect under laser irradiation. Meanwhile, the nanoplatform can also activate the cyclic guanosine monophosphate-adenosine monophosphate synthase-interferon gene stimulator signaling (cGAS-STING) pathway to produce cytokines. The damage-associated molecular patterns induced by ICD can work with these cytokines to recruit and stimulate the maturation of dendritic cells and induce the systemic anti-tumor immune response. Overall, this multifunctional biomimetic nanoplatform integrating PDT, chemotherapy, and immunotherapy is highlighted here to boost anti-tumor therapy. STATEMENT OF SIGNIFICANCE: Self-repair of DNA damage is the most important reason for the failure of primary tumor eradication and the formation of secondary and metastatic tumors. To address this issue, a multifunctional biomimetic nanoplatform 4T1Mem@PGA-Ce6/Ola (MPCO) was developed to integrate a photosensitizer Chlorine a6 and a poly (ADP-ribose) polymerase inhibitor Olaparib. With tumor targeting ability and controlled release of drugs, the MPCO was expected to enhance tumor immunogenicity and facilitate antitumor immunity through the induction of immunogenic cell death as well as the activation of the cGAS-STING pathway. This study develops a promising combination strategy against tumors and has substantial implications for the prognosis of patients with breast cancer.
Collapse
Affiliation(s)
- Hang Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xiyou Du
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Jiangkang Xu
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xinru Kong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yingying Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Dongzhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yanwei Xi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| |
Collapse
|
26
|
Kola P, Nagesh PKB, Roy PK, Deepak K, Reis RL, Kundu SC, Mandal M. Innovative nanotheranostics: Smart nanoparticles based approach to overcome breast cancer stem cells mediated chemo- and radioresistances. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023:e1876. [PMID: 36600447 DOI: 10.1002/wnan.1876] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
The alarming increase in the number of breast cancer patients worldwide and the increasing death rate indicate that the traditional and current medicines are insufficient to fight against it. The onset of chemo- and radioresistances and cancer stem cell-based recurrence make this problem harder, and this hour needs a novel treatment approach. Competent nanoparticle-based accurate drug delivery and cancer nanotheranostics like photothermal therapy, photodynamic therapy, chemodynamic therapy, and sonodynamic therapy can be the key to solving this problem due to their unique characteristics. These innovative formulations can be a better cargo with fewer side effects than the standard chemotherapy and can eliminate the stability problems associated with cancer immunotherapy. The nanotheranostic systems can kill the tumor cells and the resistant breast cancer stem cells by novel mechanisms like local hyperthermia and reactive oxygen species and prevent tumor recurrence. These theranostic systems can also combine with chemotherapy or immunotherapy approaches. These combining approaches can be the future of anticancer therapy, especially to overcome the breast cancer stem cells mediated chemo- and radioresistances. This review paper discusses several novel theranostic systems and smart nanoparticles, their mechanism of action, and their modifications with time. It explains their relevance and market scope in the current era. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Prithwish Kola
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Rui Luis Reis
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
27
|
Zhang Q, Guo L, Li H, Huang J, Li Z, Hong W, Wang J, Bai Z, Zhu J. Biosensor based on bimetallic/graphene composite for non-enzymatic detection of hydrogen peroxide in living tumor cells. Biotechnol Appl Biochem 2022. [PMID: 36427331 DOI: 10.1002/bab.2417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/15/2022] [Indexed: 11/26/2022]
Abstract
A highly sensitive electrochemical biosensor was manufactured with triple synergistic catalysis to detect hydrogen peroxide (H2 O2 ). In this study, a highly sensitive biosensor based on Prussian blue-chitosan/graphene-hemin nanomaterial/platinum and palladium nanoparticles (PB-CS/HGNs/Pt&Pd biosensor) was fabricated for the detection of H2 O2 . The materials described above were modified on the electrode surface and applied to catalyze the breakdown of hydrogen peroxide. The current response of the biosensor presented a linear relationship with H2 O2 concentration from 6 × 10-2 to 20 μM (R2 = 0.9766) and with the logarithm of H2 O2 concentration from 20 to 9×103 μM (R2 = 0.9782), the low detection limit of 25 nM was obtained at the signal/noise (S/N) ratio of 3. Besides, the biosensor showed an outstanding anti-interference ability and acceptable reproducibility. PB-CS/HGNs/Pt&Pd electrodes are effective in measuring H2 O2 from living tumor cells, which implies that the biosensor has the potential to assess reactive oxygen species in various living tumor cells.
Collapse
Affiliation(s)
- Qiyan Zhang
- Blood Purification Centre, Chun'an First People's Hospital, Zhejiang Provincial People's Hospital Chun'an Branch, Hangzhou Medical College Affiliated Chun'an Hospital, Hangzhou, Zhejiang, P.R. China
| | - Lianshan Guo
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Haoyu Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Jianfeng Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Zhengzhao Li
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Wenzhong Hong
- Clinical Laboratory, Chun'an First People's Hospital, Zhejiang Provincial People's Hospital Chun'an Branch, Hangzhou Medical College Affiliated Chun'an Hospital, Hangzhou, Zhejiang, P.R. China
| | - Jian Wang
- Clinical Laboratory, Chun'an First People's Hospital, Zhejiang Provincial People's Hospital Chun'an Branch, Hangzhou Medical College Affiliated Chun'an Hospital, Hangzhou, Zhejiang, P.R. China
| | - Zhihao Bai
- College of Chemistry & Chemical Engineering, Guangxi University, Nanning, Guangxi, P.R. China
| | - Jianmeng Zhu
- Clinical Laboratory, Chun'an First People's Hospital, Zhejiang Provincial People's Hospital Chun'an Branch, Hangzhou Medical College Affiliated Chun'an Hospital, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
28
|
Han W, Wei Z, Feng L, Yao M, Zhang H, Zhang S. Single-Site Fe-N-C Atom Based Carbon Nanotubes for Mutually Promoted and Synergistic Oncotherapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48356-48367. [PMID: 36281918 DOI: 10.1021/acsami.2c11809] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A carbon nanotube (CNT) supported single-site Fe-N-C catalyst (CNTs/Fe-N-C) exhibited attractive properties in peroxidase (POD)-like activity and photothermal effect. Herein, we designed a therapeutic platform by wrapping doxorubicin (DOX) in mesoporous CNTs/Fe-N-C with the cell membrane (CM) of breast cancer. The ultimate nanoagent (CNTs/Fe-N-C/DOX/CM) exhibited high antitumor activity on account of its efficient catalytic ability, increased drug release rates, and significant photothermal effect. Due to the POD-like activity, CNTs/Fe-N-C could effectively catalyze hydrogen peroxide (H2O2) into cytotoxic hydroxyl radicals (•OH) for chemodynamic therapy (CDT) of the tumor. Besides, the released DOX not only merely induced the diagnosis of the tumor cells for chemotherapy (CT) but also generated H2O2 to promote CDT. Moreover, the photothermal effect of the nanoagent could use for photothermal therapy (PTT). The increasing temperature was conducive to the release of DOX from micropore into the cell, which indirectly enhanced CT and CDT effects. As an intelligent and multifunctional drug delivery platform, the present CNTs/Fe-N-C/DOX/CM nanoagent could be engineered with synergistic treatments and favorable biosafety, which provides a promising paradigm in site-specific antitumor treatment and biomedicine.
Collapse
Affiliation(s)
- Wenxiu Han
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Zizhen Wei
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Lu Feng
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Mei Yao
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Huairong Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| |
Collapse
|
29
|
Abstract
Alginates have been widely explored due to their salient advantages of hydrophilicity, biocompatibility, mucoadhesive features, bioavailability, environmentally-benign properties, and cost-effectiveness. They are applied for designing micro- and nanosystems for controlled and targeted drug delivery and cancer therapy as alginate biopolymers find usage in encapsulating anticancer drugs to improve their bioavailability, sustained release, pharmacokinetics, and bio-clearance. Notably, these nanomaterials can be applied for photothermal, photodynamic, and chemodynamic therapy of cancers/tumors. Future explorations ought to be conducted to find novel alginate-based (nano)systems for targeted cancer therapy using advanced drug delivery techniques with benefits of non-invasiveness, patient compliance, and convenience of drug administration. Thus, some critical parameters such as mucosal permeability, stability in the gastrointestinal tract environment, and drug solubility ought to be considered. In addition, the comprehensive clinical translational studies along with the optimization of synthesis techniques still need to be addressed. Herein, we present an overview of the current state of knowledge and recent developments pertaining to the applications of alginate-based micro- and nanosystems for targeted cancer therapy based on controlled drug delivery, photothermal therapy, and chemodynamic/photodynamic therapy approaches, focusing on important challenges and future directions.
Collapse
|
30
|
Abe C, Miyazawa T, Miyazawa T. Current Use of Fenton Reaction in Drugs and Food. Molecules 2022; 27:molecules27175451. [PMID: 36080218 PMCID: PMC9457891 DOI: 10.3390/molecules27175451] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Iron is the most abundant mineral in the human body and plays essential roles in sustaining life, such as the transport of oxygen to systemic organs. The Fenton reaction is the reaction between iron and hydrogen peroxide, generating hydroxyl radical, which is highly reactive and highly toxic to living cells. “Ferroptosis”, a programmed cell death in which the Fenton reaction is closely involved, has recently received much attention. Furthermore, various applications of the Fenton reaction have been reported in the medical and nutritional fields, such as cancer treatment or sterilization. Here, this review summarizes the recent growing interest in the usefulness of iron and its biological relevance through basic and practical information of the Fenton reaction and recent reports.
Collapse
|
31
|
Stimuli-responsive nanoassemblies for targeted delivery against tumor and its microenvironment. Biochim Biophys Acta Rev Cancer 2022; 1877:188779. [PMID: 35977690 DOI: 10.1016/j.bbcan.2022.188779] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 02/06/2023]
Abstract
Despite the emergence of various cancer treatments, such as surgery, chemotherapy, radiotherapy, and immunotherapy, their use remains restricted owing to their limited tumor elimination efficacy and side effects. The use of nanoassemblies as delivery systems in nanomedicine for tumor diagnosis and therapy is flourishing. These nanoassemblies can be designed to have various shapes, sizes, and surface charges to meet the requirements of different applications. It is crucial for nanoassemblies to have enhanced delivery of payloads while inducing minimal to no toxicity to healthy tissues. In this review, stimuli-responsive nanoassemblies capable of combating the tumor microenvironment (TME) are discussed. First, various TME characteristics, such as hypoxia, oxidoreduction, adenosine triphosphate (ATP) elevation, and acidic TME, are described. Subsequently, the unique characteristics of the vascular and stromal TME are differentiated, and multiple barriers that have to be overcome are discussed. Furthermore, strategies to overcome these barriers for successful drug delivery to the targeted site are reviewed and summarized. In conclusion, the possible challenges and prospects of using these nanoassemblies for tumor-targeted delivery are discussed. This review aims at inspiring researchers to develop stimuli-responsive nanoassemblies for tumor-targeted delivery for clinical applications.
Collapse
|
32
|
Zhang J, Ha E, Li D, Wang L, Hu J. Ultrasmall AgBiSe 2 nanodots for CT/thermal imaging-guided photothermal tumor therapy in the NIR-II biowindow. NANOSCALE 2022; 14:10750-10760. [PMID: 35797993 DOI: 10.1039/d2nr02908a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Stimulus-responsive ternary chalcogenide nanomaterials are regarded as promising 'all-in-one' nanotheranostics agents on account of their tunable band structures and multi-metal intrinsic properties. Herein, ultrasmall AgBiSe2 nanodots are prepared by a simple thermal injection method. It shows a narrow band gap of 0.91 eV and high absorption coefficient in the NIR-II biowindow, resulting in excellent photothermal performance. Under the irradiation of a 1064 nm laser, AgBiSe2 can induce the overexpression of intracellular heat shock protein (Hsp70) and cell apoptosis to inhibit the growth of tumor cells. The strong signal from CT/thermal imaging also provides guidance for tumor diagnosis. Importantly, AgBiSe2 can be rapidly excreted from the body, thus avoiding long term toxicity. This study presents the first biomedical application of AgBiSe2 nanodots in cancer treatment and extends the development of ternary chalcogenide-based semiconductor nanomedicine.
Collapse
Affiliation(s)
- Jingge Zhang
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, P. R. China.
| | - Enna Ha
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, P. R. China.
| | - Danyang Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, P. R. China.
| | - Luyang Wang
- College of New Materials and New Energies, Shenzhen Technology University, Shenzhen 518118, P. R. China
| | - Junqing Hu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, P. R. China.
- Shenzhen Bay Laboratory, Shenzhen, 518132, P. R. China
| |
Collapse
|
33
|
Sun Y, Fang K, Hu X, Yang J, Jiang Z, Feng L, Li R, Rao Y, Shi S, Dong C. NIR-light-controlled G-quadruplex hydrogel for synergistically enhancing photodynamic therapy via sustained delivery of metformin and catalase-like activity in breast cancer. Mater Today Bio 2022; 16:100375. [PMID: 35983175 PMCID: PMC9379686 DOI: 10.1016/j.mtbio.2022.100375] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 12/27/2022] Open
Abstract
Severely hypoxic condition of tumour represents a notable obstacle against the efficiency of photodynamic therapy (PDT). While mitochondria targeted therapy by metformin has been considered as a promising strategy for reducing oxygen consumption in tumours, its low treatment sensitivity, short half-life and narrow absorption window in vivo remain the intractable challenges. In this report, 5′-guanosine monophosphate (5′GMP), indocyanine green (ICG), hemin and metformin, were combined to construct a smart G-quadruplex (G4) hydrogel named HMI@GEL for breast cancer (BC) treatment. Benefiting from the photothermal (PTT) effect of ICG, HMI@GEL exhibited excellent characteristics of NIR-light-triggered and persistent drug delivery to maintain high intratumoral concentration of metformin. Furthermore, drug loading concentration of metformin reached an amazing 300 mg mL−1 in HMI@GEL. To our knowledge, it might be the highest loading efficiency in the reported literatures. With the combination of catalase-mimicking Hemin@mil88, metformin could inhibit tumour mitochondrial respiratory significantly, which sequentially permitted in situ efficient oxygen generation. Remarkable apoptosis and necrosis were achieved by the combination of PTT and synergistically enhanced PDT as well as the activated tumour immunotherapy. Collectively, the HMI@GEL in situ injectable platform showed a promising strategy for enhanced PDT by metformin, and opened new perspectives for treating BC versatilely. A NIR-light-controlled G-quadruplex hydrogel HMI@GEL loading metformin was prepared for precision breast cancer therapy. The extremely high drug loading capacity (300 mg mL−1) and persistent delivery of metformin was realized for the first time. The combination of catalase-mimicking Hemin@mil88 and metformin dual enhanced intracellular ROS generation. The tumour immune microenvironment was dramatically reshaped by synthetic photodynamic/photothermal therapy.
Collapse
Affiliation(s)
- Yanting Sun
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Kang Fang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Xiaochun Hu
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Jingxian Yang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Zhengyang Jiang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Lei Feng
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Ruihao Li
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Yiming Rao
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Shuo Shi
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
- Corresponding author.
| | - Chunyan Dong
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
- Corresponding author.
| |
Collapse
|
34
|
Zhang H, Ma W, Wang Z, Wu X, Zhang H, Fang W, Yan R, Jin Y. Self-Supply Oxygen ROS Reactor via Fenton-like Reaction and Modulating Glutathione for Amplified Cancer Therapy Effect. NANOMATERIALS 2022; 12:nano12142509. [PMID: 35889731 PMCID: PMC9319594 DOI: 10.3390/nano12142509] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive oxidant molecules that can kill cancer cells through irreversible damage to biomacromolecules. ROS-mediated cancer therapies, such as chemodynamic (CDT) and photodynamic therapy (PDT), are often limited by the hypoxia tumor microenvironment (TME) with high glutathione (GSH) level. This paper reported the preparation, characterization, in vitro and in vivo antitumor bioactivity of a meso-tetra(4-carboxyphenyl)porphine (TCPP)-based therapeutic nanoplatform (CMMFTP) to overcome the limitations of TME. Using Cu2+ as the central ion and TCPP as the ligand, the 2D metal-organic framework Cu-TCPP was synthesized by the solvothermal method, then CMMFTP was prepared by modifying MnO2, folic acid (FA), triphenylphosphine (TPP), and poly (allylamine hydrochloride) (PAH) on the surface of Cu-TCPP MOFs. CMMFTP was designed as a self-oxygenating ROS nanoreactor based on the PDT process of TCPP MOFs and the CDT process by Cu(II) and MnO2 components (mainly through Fenton-like reaction). The in vitro assay suggested CMMFTP caused a 96% lethality rate against Hela cells (MTT analysis) in specific response to TME stimulation. Moreover, the Cu(II) and MnO2 in CMMFTP efficiently depleted the glutathione (80%) in tumor cells and consequently amplified ROS levels to improve CDT/PDT effects. The FA-induced tumor targeting and TPP-induced mitochondria targeting further enhanced the antitumor activity. Therefore, the nanoreactor based on dual targeting and self-oxygenation-enhanced ROS mechanism provided a new strategy for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Wen Fang
- Correspondence: (W.F.); (R.Y.); (Y.J.)
| | - Rui Yan
- Correspondence: (W.F.); (R.Y.); (Y.J.)
| | | |
Collapse
|
35
|
Li R, Hu X, Shang F, Wu W, Zhang H, Wang Y, Pan J, Shi S, Dong C. Treatment of triple negative breast cancer by near infrared light triggered mild temperature photothermal therapy combined with oxygen-independent cytotoxic free radicals. Acta Biomater 2022; 148:218-229. [DOI: 10.1016/j.actbio.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 11/01/2022]
|
36
|
Foglizzo V, Marchiò S. Nanoparticles as Physically- and Biochemically-Tuned Drug Formulations for Cancers Therapy. Cancers (Basel) 2022; 14:cancers14102473. [PMID: 35626078 PMCID: PMC9139219 DOI: 10.3390/cancers14102473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/26/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Conventional antitumor drugs have limitations, including poor water solubility and lack of targeting capability, with consequent non-specific distribution, systemic toxicity, and low therapeutic index. Nanotechnology promises to overcome these drawbacks by exploiting the physical properties of diverse nanocarriers that can be linked to moieties with binding selectivity for cancer cells. The use of nanoparticles as therapeutic formulations allows a targeted delivery and a slow, controlled release of the drug(s), making them tunable modules for applications in precision medicine. In addition, nanoparticles are also being developed as cancer vaccines, offering an opportunity to increase both cellular and humoral immunity, thus providing a new weapon to beat cancer. Abstract Malignant tumors originate from a combination of genetic alterations, which induce activation of oncogenes and inactivation of oncosuppressor genes, ultimately resulting in uncontrolled growth and neoplastic transformation. Chemotherapy prevents the abnormal proliferation of cancer cells, but it also affects the entire cellular network in the human body with heavy side effects. For this reason, the ultimate aim of cancer therapy remains to selectively kill cancer cells while sparing their normal counterparts. Nanoparticle formulations have the potential to achieve this aim by providing optimized drug delivery to a pathological site with minimal accumulation in healthy tissues. In this review, we will first describe the characteristics of recently developed nanoparticles and how their physical properties and targeting functionalization are exploited depending on their therapeutic payload, route of delivery, and tumor type. Second, we will analyze how nanoparticles can overcome multidrug resistance based on their ability to combine different therapies and targeting moieties within a single formulation. Finally, we will discuss how the implementation of these strategies has led to the generation of nanoparticle-based cancer vaccines as cutting-edge instruments for cancer immunotherapy.
Collapse
Affiliation(s)
- Valentina Foglizzo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Serena Marchiò
- Department of Oncology, University of Torino, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
- Correspondence: ; Tel.: +39-01199333239
| |
Collapse
|
37
|
Yu XT, Sui SY, He YX, Yu CH, Peng Q. Nanomaterials-based photosensitizers and delivery systems for photodynamic cancer therapy. BIOMATERIALS ADVANCES 2022; 135:212725. [PMID: 35929205 DOI: 10.1016/j.bioadv.2022.212725] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022]
Abstract
The increasing cancer morbidity and mortality requires the development of high-efficiency and low-toxicity anticancer approaches. In recent years, photodynamic therapy (PDT) has attracted much attention in cancer therapy due to its non-invasive features and low side effects. Photosensitizer (PS) is one of the key factors of PDT, and its successful delivery largely determines the outcome of PDT. Although a few PS molecules have been approved for clinical use, PDT is still limited by the low stability and poor tumor targeting capacity of PSs. Various nanomaterial systems have shown great potentials in improving PDT, such as metal nanoparticles, graphene-based nanomaterials, liposomes, ROS-sensitive nanocarriers and supramolecular nanomaterials. The small molecular PSs can be loaded in functional nanomaterials to enhance the PS stability and tumor targeted delivery, and some functionalized nanomaterials themselves can be directly used as PSs. Herein, we aim to provide a comprehensive understanding of PDT, and summarize the recent progress of nanomaterials-based PSs and delivery systems in anticancer PDT. In addition, the concerns of nanomaterials-based PDT including low tumor targeting capacity, limited light penetration, hypoxia and nonspecific protein corona formation are discussed. The possible solutions to these concerns are also discussed.
Collapse
Affiliation(s)
- Xiao-Tong Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shang-Yan Sui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yu-Xuan He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chen-Hao Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|