1
|
Gómez-Pastor S, Maugard A, Walker HR, Elies J, Børsum KE, Grimaldi G, Reina G, Ruiz A. CD-44 targeted nanoparticles for combination therapy in an in vitro model of triple-negative breast cancer: Targeting the tumour inside out. Colloids Surf B Biointerfaces 2025; 249:114504. [PMID: 39817967 DOI: 10.1016/j.colsurfb.2025.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer defined by the lack of three key receptors: estrogen, progesterone, and HER2. This lack of receptors makes TNBC difficult to treat with hormone therapy or drugs, and so it is characterised by a poor prognosis compared to other kinds of breast cancer. This study explores photoactive Poly(lactic-co-glycolic acid) (PLGA) nanoparticles as a potential therapeutic strategy for TNBC. The nanoparticles are functionalised with hyaluronic acid (HA) for targeted delivery to CD-44 receptors overexpressed in TNBC cells, especially under hypoxic conditions. Additionally, we co-loaded the nanoparticles with Doxorubicin (Dox) and Indocyanine Green (ICG) to enable combinatorial chemo-photothermal therapy. After carefully optimising the formulation, we propose an effortless and reproducible preparation of the nanodrugs. We demonstrate that HA-conjugated nanoparticles effectively target TNBC cells and inhibit their proliferation while the treatment efficiency is enhanced during near-infrared light irradiation. We also prove that our treatment is effective in a 3D cell culture model, highlighting the importance of tumour architecture and the metabolic stage of the cells in the tumour microenvironment. This approach is promising for a tumour-targeted theragnostic for TNBC with improved efficacy in hypoxic microenvironments.
Collapse
Affiliation(s)
- Silvia Gómez-Pastor
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom; Departamento de Biología, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Auréane Maugard
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom
| | - Harriet R Walker
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom
| | - Jacobo Elies
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom
| | - Kaja E Børsum
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom
| | - Giulia Grimaldi
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom; School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom.
| | - Giacomo Reina
- Empa Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St. Gallen 9014, Switzerland.
| | - Amalia Ruiz
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom.
| |
Collapse
|
2
|
Cai Q, Cai X, Shubhra QTH. Mitochondrial transfer drives immune evasion in tumor microenvironment. Trends Cancer 2025:S2405-8033(25)00094-9. [PMID: 40268608 DOI: 10.1016/j.trecan.2025.04.002] [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: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Tumors subvert T cell metabolism through diverse mechanisms. Ikeda et al. reveal mitochondrial transfer as a tumor-driven immune evasion strategy, where cancer cells deliver dysfunctional mitochondria to T cells, impairing metabolism and inducing exhaustion. These findings highlight mitochondrial dynamics as a promising therapeutic target to improve immunotherapy outcomes.
Collapse
Affiliation(s)
- Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Xiaojun Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Quazi T H Shubhra
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Chemistry, University of Silesia in Katowice, Szkolna 9, 40-006 Katowice, Poland.
| |
Collapse
|
3
|
Qi Q, Zhang Z, Wang D. GSH-responsive paclitaxel prodrug used in chemotherapy in combination with photodynamic therapy. Bioorg Chem 2025; 157:108289. [PMID: 40007348 DOI: 10.1016/j.bioorg.2025.108289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025]
Abstract
The lack of targeting and poor solubility of anti-tumor drugs are two major limitations to the outcome of tumor therapy. To address the inherent drawbacks, we designed a novel prodrug of paclitaxel (PTX), HP-PTX. This HP-PTX prodrug contains a PEGylated heptamethylene cyanine dye (PEG-IR808-1) that was conjugated to PTX via a redox-sensitive disulfide bond. The moiety of IR808-1 acts as a tumor-targeting ligand, enabling HP-PTX not only to target tumor cells, but also to localize to mitochondria and generate ROS under 808 nm laser irradiation to wound cellular mitochondria thus exerting cytotoxic effect. Meanwhile, in vitro cellular uptake assays showed that HP-PTX possesses MCF-7 cell tumor targeting specificity which was attributed to the preferential uptake of heptamethine cyanine dye mediated by the overexpressed organic anion-transporting polypeptides (OATP) on MCF-7 cell membrane. Near-infrared in vivo imaging showed that incorporation of polyethylene glycol effectively prolonged prodrug's half-life in vivo. In addition, in vivo experiments showed that with combinational therapy strategy HP-PTX was able to kill cancer cells with high efficiency (69.52 %) without notable toxic side effects compared to PTX. These results are evidence of the potential of the tumor-targeting prodrug HP-PTX for the treatment of breast cancer.
Collapse
Affiliation(s)
- Qianqian Qi
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhanyu Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dun Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
4
|
Xu J, Zhang Y, Zheng Y, Wang T, Zhang H, Wang K, Wang Y, Williams GR, Zhu LM. A smart responsive NIR-operated chitosan-based nanoswitch to induce cascade immunogenic tumor ferroptosis via cytokine storm. Carbohydr Polym 2025; 351:123120. [PMID: 39779027 DOI: 10.1016/j.carbpol.2024.123120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
In this work we present a near-infrared (NIR)-operated nanoswitch based on chitosan nanoparticles (EpCAM-CS-co-PNVCL@IR780/IMQ NPs) that induces cascade immunogenic tumor ferroptosis via cytokine storm. The formulation was prepared by loading a photosensitiser (IR780) and an immunotherapeutic drug (imiquimod; IMQ) into temperature- and pH-responsive chitosan-based NPs functionalized with tumor-targeting aptamers. The EpCAM aptamer can chaperone the NPs selectively into cancer cells, and allow them to enter the cell nucleus. In vitro and in vivo assays revelaed that the NPs were able to effectively induce the immunogenic ferroptosis of cancer cells. Under NIR irradiation, EpCAM-CS-co-PNVCL@IR780/IMQ cause cell death in tumors via photothermal therapy. Moreover, IMQ promotes the maturation of dendritic cells (DCs), which then activate cytotoxic T-lymphocytes (CTLs); these T-cells go on to provide immunotherapy of metastatic tumor cells. The metastatic tumor cells can be induced to undergo ferroptosis by the addition of arachidonic acid (AA), which interacts with interferon cytokines (IFN-γ) released from CTLs.
Collapse
Affiliation(s)
- Jianxiang Xu
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Yanyan Zhang
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Yilu Zheng
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Tong Wang
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Huan Zhang
- International Union Laboratory on Acupuncture Based Target Discovery, International Joint Laboratory on Acupuncture Neuro-immunology, Shanghai Research Institute of Acupuncture and Meridian, Yue Yang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Kai Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu Wang
- International Union Laboratory on Acupuncture Based Target Discovery, International Joint Laboratory on Acupuncture Neuro-immunology, Shanghai Research Institute of Acupuncture and Meridian, Yue Yang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China; Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Li-Min Zhu
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China.
| |
Collapse
|
5
|
Shubhra QTH. Gasdermin D unlocks metabolic pathways to enhance tissue regeneration. Cell Mol Immunol 2025; 22:1-3. [PMID: 39572781 PMCID: PMC11686189 DOI: 10.1038/s41423-024-01239-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/10/2024] [Indexed: 01/01/2025] Open
Affiliation(s)
- Quazi T H Shubhra
- Institute of Chemistry, University of Silesia in Katowice, Katowice, Poland.
| |
Collapse
|
6
|
Sun W, Chai X, Zhang Y, Yu T, Wang Y, Zhao W, Liu Y, Yin D, Zhang C. Combination Using Magnetic Iron Oxide Nanoparticles and Magnetic Field for Cancer Therapy. CHEM REC 2024; 24:e202400179. [PMID: 39607378 DOI: 10.1002/tcr.202400179] [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: 09/05/2024] [Revised: 11/13/2024] [Indexed: 11/29/2024]
Abstract
Iron oxide nanoparticles (MNPs) demonstrate notable benefits in magnetic induction, attributed to their distinctive physical and chemical attributes. Emerging cancer treatment utilizing magnetic fields have also gathered increasing attention in the biomedical field. However, the defects of difficult dispersion and poor biocompatibility of MNPs seriously hinder their application. In order to overcome its inherent defects and maximize the therapeutic potential of MNPs, various functionalized MNPs have been developed, and numerous combined treatment methods based on MNPs have been widely studied. In this review, we compare and analyze the common nanoparticles based on MNPs with different sizes, shapes, and functional modifications. Additionally, we introduced the therapeutic mechanisms of the strategies, such as magnetically controlled targeting, magnetic hyperthermia, and magneto-mechanical effect, which based on the unique magnetic induction capabilities of MNPs. Finally, main challenges of MNPs as smart nanomaterials were also discussed. This review seeks to offer a thorough overview of MNPs in biomedicine and a new sight for their application in tumor treatment.
Collapse
Affiliation(s)
- Wenjun Sun
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
| | - Xiaoxia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
| | - Yuan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
| | - Tongyao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
| | - Yuhua Wang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
| | - Wenzhe Zhao
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
| | - Yanhua Liu
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Dachuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
| | - Chenyan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710100, PR China
- Research & Development Institute of, Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063, China
| |
Collapse
|
7
|
Agrawal S, Singh GK, Tiwari S. Focused starvation of tumor cells using glucose oxidase: A comprehensive review. Int J Biol Macromol 2024; 281:136444. [PMID: 39389487 DOI: 10.1016/j.ijbiomac.2024.136444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Starvation therapy targets the high metabolic demand of tumor cells. It primarily leans over the consumption of intracellular glucose and simultaneous blockade of alternative metabolic pathways. The strategy involves the use of glucose oxidase (GOx) for catalyzing the conversion of glucose into gluconic acid and hydrogen peroxide. Under these conditions, metabolic re-programming of tumor cells enables the utilization of substrates such as amino acids, fatty acids and lipids. This can be overcome by co-administration of chemo-, photo- and immuno-therapeutics together with glucose oxidase. Targeted delivery of glucose oxidase at tumor site can be enabled with the use of nanoformulations. In this review, we highlight that the outcomes of starvation therapy can be improved using rationally developed nano-formulations. It is possible to load synergistically acting bioactives in these formulations and deliver in site-specific manner and hence achieve the elimination of tumors cells with greater efficacy.
Collapse
Affiliation(s)
- Shivanshu Agrawal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Gireesh K Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya 824236, India
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India.
| |
Collapse
|
8
|
Wang H, Wang X, Wang L, Wang H, Zhang Y. Plant‐Derived Phytochemicals and Their Nanoformulations for Inducing Programed Cell Death in Cancer. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202400197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Indexed: 01/05/2025]
Abstract
AbstractPhytochemicals are a diverse class of compounds found in various plant‐based foods and beverages that have displayed the capacity to exert powerful anticancer effects through the induction of programed cell death (PCD) in malignancies. PCD is a sophisticated process that maintains in upholding tissue homeostasis and eliminating injured or neoplastic cells. Phytochemicals have shown the potential to induce PCD in malignant cells through various mechanisms, including modulation of cell signaling pathways, regulation of reactive oxygen species (ROS), and interaction with critical targets in cells such as DNA. Moreover, recent studies have suggested that nanomaterials loaded with phytochemicals may enhance cell death in tumors, which can also stimulate antitumor immunity. In this review, a comprehensive overview of the current understanding of the anticancer effects of phytochemicals and their potential as a promising approach to cancer therapy, is provided. The impacts of phytochemicals such as resveratrol, curcumin, apigenin, quercetin, and some approved plant‐derived drugs, such as taxanes on the regulation of some types of PCD, including apoptosis, pyroptosis, anoikis, autophagic cell death, ferroptosis, and necroptosis, are discussed. The underlying mechanisms and the potential of nanomaterials loaded with phytochemicals to enhance PCD in tumors are also explained.
Collapse
Affiliation(s)
- Haoyu Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Xiaoyang Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Long Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| | - Haifan Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Yuxing Zhang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| |
Collapse
|
9
|
Islam MR, Manir MS, Razzak M, Mamun MA, Mortuza MF, Islam MJ, Yang S, Pan H, Alam AKMM, Shubhra QTH. Silk-enriched hydrogels with ROS-scavenging dendrimers for advanced wound care. Int J Biol Macromol 2024; 280:135567. [PMID: 39288850 DOI: 10.1016/j.ijbiomac.2024.135567] [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: 03/02/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
This study explores the development of novel hydrogel composites for wound care, incorporating silk fibroin and reactive oxygen species (ROS)-scavenging dendrimers into a polyvinyl alcohol (PVA) matrix. Utilizing ionizing gamma radiation, we fabricated pristine PVA, silk-PVA (SPVA) binary, and dendrimer-silk-PVA (DSPVA) ternary hydrogel composites, with their composition confirmed via UV-visible absorption spectroscopy. Fourier-transform infrared (FTIR) and Raman spectroscopy analyses indicated complex interactions between the hydrogel components, enhancing their structural and biocompatible properties. Scanning electron microscopy (SEM) analysis revealed that dendrimer integration in DSPVA hydrogels significantly increased surface porosity, vital for tissue regeneration. The DSPVA hydrogels demonstrated effective ROS scavenging, reducing hydrogen peroxide (H2O2) concentrations by approximately 70 % within 24 h. In vivo wound healing studies in a diabetic mouse model showed enhanced wound closure in the DSPVA group, with a relative wound area reduction to 30 ± 4.3 % on day 10, compared to 56.5 ± 2.7 % in the control group. By the 16th day, the treated group exhibited near-complete wound contraction, markedly outperforming the control group. These findings underscore the potential of DSPVA hydrogels in diabetic wound management, combining silk fibroin's mechanical support, dendrimers' antioxidative properties, and PVA's structural benefits. Thus, DSPVA hydrogels are promising candidates for advanced wound care applications.
Collapse
Affiliation(s)
- M R Islam
- Institute of Radiation and Polymer Technology, AERE, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh
| | - M S Manir
- Institute of Radiation and Polymer Technology, AERE, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh
| | - M Razzak
- Institute of Radiation and Polymer Technology, AERE, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh
| | - M A Mamun
- Materials Science Division, AECD, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh
| | - M F Mortuza
- Gamma Source Division, IFRB, AERE, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh
| | - M J Islam
- Veterinary Drug Residue Analysis Division, IFRB, AERE, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh
| | - Shumin Yang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Huachun Pan
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - A K M M Alam
- Institute of Radiation and Polymer Technology, AERE, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh.
| | - Quazi T H Shubhra
- Institute of Radiation and Polymer Technology, AERE, Bangladesh Atomic Energy Commission, Dhaka 1207, Bangladesh; Institute of Chemistry, University of Silesia in Katowice, Szkolna 9, 40-003, Katowice, Poland; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
10
|
Shubhra QTH, Feczkó T, Cai Q. Beyond the danger signal: RNA aggregates orchestrate immunotherapy. Trends Mol Med 2024; 30:702-704. [PMID: 38866645 DOI: 10.1016/j.molmed.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024]
Abstract
Mendez-Gomez et al. recently demonstrated the transformative potential of RNA-lipid particle aggregates (RNA-LPAs) in immunotherapy. By reprogramming the tumor microenvironment (TME) and potentiating antitumor immunity, RNA-LPAs target primary tumors and elicit robust systemic immunity. This innovative platform holds promise for translating preclinical success into tangible clinical benefits.
Collapse
Affiliation(s)
- Quazi T H Shubhra
- Institute of Chemistry, University of Silesia in Katowice, Szkolna 9, 40-003 Katowice, Poland; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Tivadar Feczkó
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; Faculty of Engineering, University of Pannonia, Egyetem u. 10, H-8200, Veszprém, Hungary
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
11
|
Yan B, Li Y, He S. Aptamer-mediated therapeutic strategies provide a potential approach for cancer. Int Immunopharmacol 2024; 136:112356. [PMID: 38820957 DOI: 10.1016/j.intimp.2024.112356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
The treatment of tumors still faces considerable challenges. While conventional treatments such as surgery, chemotherapy, and radiation therapy provide some curative effects, their side effects and limitations highlight the importance of finding more precise treatment strategies. Aptamers have become an important target molecule in the field of drug delivery systems due to their good affinity and targeting, and they have gradually become an important link from basic research to clinical application. In this paper, we discussed the latest progress of aptamer-mediated nanodrugs, as well as aptamer-mediated photodynamic therapy, photothermal therapy, and immunotherapy strategies for tumor treatment, and explored the possibility of aptamer-mediated therapy for accurate tumor treatment. The purpose of this review is to provide novel insights for treating tumors with aptamer-mediated therapies by summarizing these innovative strategies, thereby ultimately enhancing the therapeutic efficacy for cancer patients.
Collapse
Affiliation(s)
- Bingshuo Yan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Yuting Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People's Republic of China.
| |
Collapse
|
12
|
Alsamarat R, Sunoqrot S. A Glucose Oxidase-Curcumin Composite Nanoreactor for Multimodal Synergistic Cancer Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4611-4621. [PMID: 38920441 DOI: 10.1021/acsabm.4c00479] [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: 06/27/2024]
Abstract
Glucose oxidase (GOx) selectively oxidizes β-d-glucose into gluconic acid and hydrogen peroxide; thus, it has emerged as a promising anticancer agent by tumor starvation and oxidative therapy. Here, we developed a nanoscale platform or "nanoreactor" that incorporates GOx and the bioactive natural product curcumin (CUR) to achieve a multimodal anticancer nanocomposite. The composite nanoreactor was formed by loading CUR in biodegradable polymeric nanoparticles (NPs) of poly(ethylene glycol)-b-poly(ε-caprolactone) (PEG-PCL). Prime-coating of the NPs with an iron(III)-tannic acid complex enabled facile immobilization of GOx on the NP surface. The NPs were monodisperse with a hydrodynamic diameter of 122 nm and a partially negative surface charge. The NPs were also associated with an excellent CUR loading efficiency and sustained release up to 96 h, which was accelerated by surface-immobilized GOx and followed supercase II transport. Viability assays were conducted on two model cancer cell lines, MCF-7 and MDA-MB-231 cells, as well as human dermal fibroblasts as a representative normal cell line. The assays revealed significantly improved potency of CUR in the composite nanoreactor, with up to 6000- and 1280-fold increase in MCF-7 and MDA-MB-231 cells, respectively, and lower toxicity toward normal cells. The NPs were also able to promote intracellular reactive oxygen species (ROS) generation and dissipation of the mitochondrial membrane potential, providing important clues on the mechanism of action of the nanoreactor. Further investigation of caspase-3 activity revealed that the nanoreactor had no effect or inhibited caspase-3 levels, signifying a caspase-independent mechanism of inducing apoptosis. Our findings present a promising nanocarrier platform that combines therapeutic agents with distinct mechanisms of action acting in synergy for more effective cancer therapy.
Collapse
Affiliation(s)
- Rama Alsamarat
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Suhair Sunoqrot
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| |
Collapse
|
13
|
Oehler JB, Rajapaksha W, Albrecht H. Emerging Applications of Nanoparticles in the Diagnosis and Treatment of Breast Cancer. J Pers Med 2024; 14:723. [PMID: 39063977 PMCID: PMC11278299 DOI: 10.3390/jpm14070723] [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: 05/18/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer remains the most prevalent cancer among women worldwide, driving the urgent need for innovative approaches to diagnosis and treatment. This review highlights the pivotal role of nanoparticles in revolutionizing breast cancer management through advancements of interconnected approaches including targeted therapy, imaging, and personalized medicine. Nanoparticles, with their unique physicochemical properties, have shown significant promise in addressing current treatment limitations such as drug resistance and nonspecific systemic distribution. Applications range from enhancing drug delivery systems for targeted and sustained release to developing innovative diagnostic tools for early and precise detection of metastases. Moreover, the integration of nanoparticles into photothermal therapy and their synergistic use with existing treatments, such as immunotherapy, illustrate their transformative potential in cancer care. However, the journey towards clinical adoption is fraught with challenges, including the chemical feasibility, biodistribution, efficacy, safety concerns, scalability, and regulatory hurdles. This review delves into the current state of nanoparticle research, their applications in breast cancer therapy and diagnosis, and the obstacles that must be overcome for clinical integration.
Collapse
Affiliation(s)
- Josephine B. Oehler
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4810, Australia
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical & Vet Sciences, James Cook University, Townsville, QLD 4810, Australia
| | - Weranga Rajapaksha
- Centre for Pharmaceutical Innovation (CPI), Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Hugo Albrecht
- Centre for Pharmaceutical Innovation (CPI), Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
14
|
Ji F, Shi C, Shu Z, Li Z. Nanomaterials Enhance Pyroptosis-Based Tumor Immunotherapy. Int J Nanomedicine 2024; 19:5545-5579. [PMID: 38882539 PMCID: PMC11178094 DOI: 10.2147/ijn.s457309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Abstract
Pyroptosis, a pro-inflammatory and lytic programmed cell death pathway, possesses great potential for antitumor immunotherapy. By releasing cellular contents and a large number of pro-inflammatory factors, tumor cell pyroptosis can promote dendritic cell maturation, increase the intratumoral infiltration of cytotoxic T cells and natural killer cells, and reduce the number of immunosuppressive cells within the tumor. However, the efficient induction of pyroptosis and prevention of damage to normal tissues or cells is an urgent concern to be addressed. Recently, a wide variety of nanoplatforms have been designed to precisely trigger pyroptosis and activate the antitumor immune responses. This review provides an update on the progress in nanotechnology for enhancing pyroptosis-based tumor immunotherapy. Nanomaterials have shown great advantages in triggering pyroptosis by delivering pyroptosis initiators to tumors, increasing oxidative stress in tumor cells, and inducing intracellular osmotic pressure changes or ion imbalances. In addition, the challenges and future perspectives in this field are proposed to advance the clinical translation of pyroptosis-inducing nanomedicines.
Collapse
Affiliation(s)
- Fujian Ji
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Chunyu Shi
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhenbo Shu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhongmin Li
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
15
|
Ban W, Chen Z, Zhang T, Du T, Huo D, Zhu G, He Z, Sun J, Sun M. Boarding pyroptosis onto nanotechnology for cancer therapy. J Control Release 2024; 370:653-676. [PMID: 38735396 DOI: 10.1016/j.jconrel.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Pyroptosis, a non-apoptotic programmed cellular inflammatory death mechanism characterized by gasdermin (GSDM) family proteins, has gathered significant attention in the cancer treatment. However, the alarming clinical trial data indicates that pyroptosis-mediated cancer therapeutic efficiency is still unsatisfactory. It is essential to integrate the burgeoning biomedical findings and innovations with potent technology to hasten the development of pyroptosis-based antitumor drugs. Considering the rapid development of pyroptosis-driven cancer nanotherapeutics, here we aim to summarize the recent advances in this field at the intersection of pyroptosis and nanotechnology. First, the foundation of pyroptosis-based nanomedicines (NMs) is outlined to illustrate the reliability and effectiveness for the treatment of tumor. Next, the emerging nanotherapeutics designed to induce pyroptosis are overviewed. Moreover, the cross-talk between pyroptosis and other cell death modalities are discussed, aiming to explore the mechanistic level relationships to provide guidance strategies for the combination of different types of antitumor drugs. Last but not least, the opportunities and challenges of employing pyroptosis-based NMs in potential clinical cancer therapy are highlighted.
Collapse
Affiliation(s)
- Weiyue Ban
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zhichao Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tao Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tengda Du
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Dianqiu Huo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Guorui Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| |
Collapse
|
16
|
Sun S, Han R, Sun Y, Chen W, Zhao L, Guan X, Zhang W. A minimalist cancer cell membrane-shielded biomimetic nanoparticle for nasopharyngeal carcinoma active-targeting therapy. Colloids Surf B Biointerfaces 2024; 238:113909. [PMID: 38599076 DOI: 10.1016/j.colsurfb.2024.113909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a common head and neck malignancy, which is characterized by high incidence and aggression with poor diagnosis and limited therapeutic opportunity. The innovative strategy for achieving precise NPC active-targeting drug delivery has emerged as a prominent focus in clinical research. Here, a minimalist cancer cell membrane (CCM) shielded biomimetic nanoparticle (NP) was designed for NPC active-targeting therapy. Chemotherapeutant model drug doxorubicin (DOX) was loaded in polyamidoamine (PAMAM) dendrimer. The PAMAM/DOX (PD) NP was further shielded by human CNE-2 NPC CCM. Characterization results verified that the biomimetic PAMAM/DOX@CCM (abbreviated as PDC) NPs had satisfactory physical properties with high DOX-loading and excellent stability. Cell experiments demonstrated that the CNE-2 membrane-cloaked PDC NPs presented powerful cellular uptake in the sourcing cells by homologous targeting and adhesive interaction. Further in vivo results confirmed that this biomimetic nanoplatform had extended circulation and remarkable tumor-targeting capability, and the PDC NPs effectively suppressed the progression of CNE-2 tumors by systemic administration. This CCM-shielded biomimetic NP displayed a minimalist paradigm nanoplatform for precise NPC therapy, and the strategy of CCM-shielded biomimetic drug delivery system (DDS) has great potential for extensive cancer active-targeting therapy.
Collapse
Affiliation(s)
- Shuo Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Rongrong Han
- Department of Otolaryngology, Weifang People's Hospital, Weifang 261000, China
| | - Yanju Sun
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Wenqiang Chen
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Limin Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Xiuwen Guan
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| | - Weifen Zhang
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
17
|
Mahmud MM, Pandey N, Winkles JA, Woodworth GF, Kim AJ. Toward the scale-up production of polymeric nanotherapeutics for cancer clinical trials. NANO TODAY 2024; 56:102314. [PMID: 38854931 PMCID: PMC11155436 DOI: 10.1016/j.nantod.2024.102314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Nanotherapeutics have gained significant attention for the treatment of numerous cancers, primarily because they can accumulate in and/or selectively target tumors leading to improved pharmacodynamics of encapsulated drugs. The flexibility to engineer the nanotherapeutic characteristics including size, morphology, drug release profiles, and surface properties make nanotherapeutics a unique platform for cancer drug formulation. Polymeric nanotherapeutics including micelles and dendrimers represent a large number of formulation strategies developed over the last decade. However, compared to liposomes and lipid-based nanotherapeutics, polymeric nanotherapeutics have had limited clinical translation from the laboratory. One of the key limitations of polymeric nanotherapeutics formulations for clinical translation has been the reproducibility in preparing consistent and homogeneous large-scale batches. In this review, we describe polymeric nanotherapeutics and discuss the most common laboratory and scale-up formulation methods, specifically those proposed for clinical cancer therapies. We also provide an overview of the major challenges and opportunities for scaling polymeric nanotherapeutics to clinical-grade formulations. Finally, we will review the regulatory requirements and challenges in advancing nanotherapeutics to the clinic.
Collapse
Affiliation(s)
- Md Musavvir Mahmud
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey A. Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Graeme F. Woodworth
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Anthony J. Kim
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| |
Collapse
|
18
|
Pan H, Yang S, Gao L, Zhou J, Cheng W, Chen G, Shuhang W, Li N, Veranič P, Musiol R, Cai Q, Shubhra QT. At the crossroad of nanotechnology and cancer cell membrane coating: Expanding horizons with engineered nanoplatforms for advanced cancer therapy harnessing homologous tumor targeting. Coord Chem Rev 2024; 506:215712. [DOI: 10.1016/j.ccr.2024.215712] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Zhang S, Hussain S, Tang Y, Wang K, Wang X, Zhang L, Liao Y, Wang C, Hao Y, Gao R. Enzyme-triggered on-demand release of a H 2O 2-self-supplying CuO 2@Fe 3O 4 nanoagent for enhanced chemodyamic antimicrobial therapy and wound healing. J Mater Chem B 2024; 12:3404-3416. [PMID: 38487992 DOI: 10.1039/d3tb02762g] [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: 04/04/2024]
Abstract
Nanoagents for chemodynamic therapy (CDT) hold a promising future in the field of antimicrobials, especially copper peroxide (CuO2) (CP) nanomaterials which have garnered significant attention due to their ability to self-supply H2O2. Nevertheless, the poor stability of CuO2 remains a critical challenge which restricts its practical application in the antibacterial field. In this study, an advanced nano-antimicrobial system HA-CP@Fe3O4 with enzyme-responsive properties is developed by coating hyaluronic acid (HA) on CuO2-loaded iron tetraoxide nanoparticles. The coating of HA not only stabilizes the CuO2 nanomaterials but also provides responsiveness towards the enzyme hyaluronidase, which is typically secreted by some bacteria. The outer layer of HA in HA-CP@Fe3O4 undergoes decomposition in the presence of hyaluronidase-secreting bacteria, resulting in the release of CuO2@Fe3O4. The released CuO2@Fe3O4 then self-supplies H2O2 and generates reactive oxygen species (ROS) within the infected microenvironment through Fenton and Russell effects, to ultimately achieve effective and precise antimicrobial activity. Simultaneously, the magnetic property provided by Fe3O4 allows the substance to be directed towards the infection site. Both in vitro and in vivo tests demonstrated that HA-CP@Fe3O4 exhibited excellent antimicrobial capabilities at low concentration (30 μg mL-1), exceptional biocompatibility and the ability to accelerate wound healing. The findings of this work offer a new and promising approach for targeted and precise CDT.
Collapse
Affiliation(s)
- Sijie Zhang
- School of Chemistry, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Sameer Hussain
- School of Chemistry, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| | - Yuhai Tang
- School of Chemistry, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| | - Kaili Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xingyan Wang
- School of Chemistry, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| | - Long Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuheng Liao
- Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chen Wang
- Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yi Hao
- School of Chemistry, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ruixia Gao
- School of Chemistry, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
20
|
Liu J, Chen T, Liu X, Li Z, Zhang Y. Engineering materials for pyroptosis induction in cancer treatment. Bioact Mater 2024; 33:30-45. [PMID: 38024228 PMCID: PMC10654002 DOI: 10.1016/j.bioactmat.2023.10.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer remains a significant global health concern, necessitating the development of innovative therapeutic strategies. This research paper aims to investigate the role of pyroptosis induction in cancer treatment. Pyroptosis, a form of programmed cell death characterized by the release of pro-inflammatory cytokines and the formation of plasma membrane pores, has gained significant attention as a potential target for cancer therapy. The objective of this study is to provide a comprehensive overview of the current understanding of pyroptosis and its role in cancer treatment. The paper discusses the concept of pyroptosis and its relationship with other forms of cell death, such as apoptosis and necroptosis. It explores the role of pyroptosis in immune activation and its potential for combination therapy. The study also reviews the use of natural, biological, chemical, and multifunctional composite materials for pyroptosis induction in cancer cells. The molecular mechanisms underlying pyroptosis induction by these materials are discussed, along with their advantages and challenges in cancer treatment. The findings of this study highlight the potential of pyroptosis induction as a novel therapeutic strategy in cancer treatment and provide insights into the different materials and mechanisms involved in pyroptosis induction.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - XianLing Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Oncology, Guilin Hospital of the Second Xiangya Hospital, Central South University, Guilin, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Zhang
- Department of Biomedical Engineering, The City University of Hong Kong, Hong Kong Special Administrative Region of China
| |
Collapse
|
21
|
Cai Q, Shubhra QTH. Gasdermin D triggers cardiolipin-driven mitochondrial damage and pyroptosis. Trends Immunol 2024; 45:75-77. [PMID: 38242759 DOI: 10.1016/j.it.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
In a remarkable recent study, Miao et al. reveal that gasdermin D N-terminal (GSDMD-NT) instigates mitochondrial damage in pyroptosis by forming pores in inner and outer mitochondrial membranes (OMMs). The authors highlight the key role of mitochondrial cardiolipin in the action of GSDMD-NT, and significantly advance our understanding of this inflammatory cell death mechanism.
Collapse
Affiliation(s)
- Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Quazi T H Shubhra
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Chemistry, University of Silesia in Katowice, 41-500 Chorzów, Poland.
| |
Collapse
|
22
|
Sun Q, Yang J, Wu Q, Shen W, Yang Y, Yin D. Targeting Lysosome for Enhanced Cancer Photodynamic/Photothermal Therapy in a "One Stone Two Birds" Pattern. ACS APPLIED MATERIALS & INTERFACES 2024; 16:127-141. [PMID: 38118049 DOI: 10.1021/acsami.3c13162] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Highly immunogenic programmed death of tumor cells, such as immunogenic cell death (ICD) and pyroptosis, strengthens antitumor responses and thus represents a promising target for cancer immunotherapy. However, the development of ICD and pyroptosis inducers remains challenging, and their efficiency is typically compromised by self-protective autophagy. Here, we report a potent ICD and pyroptosis-inducing strategy by coupling combined photodynamic/photothermal therapy (PTT/PDT) to biological processes in cancer cells. For this purpose, we rationally synthesize a lysosomal-targeting boron-dipyrromethene dimer (BDPd) with intense NIR absorption/emission, high reactive oxygen species (ROS) yield, and photothermal abilities, which can be self-assembled with Pluronic F127, producing lysosomal-acting nanomicelles (BDPd NPs) to facilitate cancer cell internalization of BDPd and generation of intracellular ROS. Owing to the favorable lysosomal-targeting ability of the morpholine group on BDPd, the intracellular BDPd NPs can accumulate in the lysosome and induce robust lysosomal damage in cancer cells upon 660 nm laser irradiation, which results in the synergetic induction of pyroptosis and ICD via activating NLRP3/GSDMD and caspase-3/GSDME pathways simultaneously. More importantly, PTT/PDT-induced self-protective autophagic degradation was blocked due to the dysfunction of lysosomes. Either intratumorally or intravenously, the injected BDPd NPs could markedly inhibit the growth of established tumor tissues upon laser activation, provoke local and systemic antitumor immune responses, and prolong the survival time in the mouse triple-negative breast cancer model. Collectively, this work represents a promising strategy to boost the therapeutic potential of PTT/PDT by coupling phototherapeutic reagents with the subcellular organelles, creating a "one stone two birds" pattern.
Collapse
Affiliation(s)
- Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Jinming Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Qinghua Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Provincial Key Laboratory of Research & Development of Chinese Medicine, Hefei 230021 ,China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
- Anhui Provincial Key Laboratory of Research & Development of Chinese Medicine, Hefei 230021 ,China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
| |
Collapse
|
23
|
Zhang T, Li N, Wang R, Sun Y, He X, Lu X, Chu L, Sun K. Enhanced therapeutic efficacy of doxorubicin against multidrug-resistant breast cancer with reduced cardiotoxicity. Drug Deliv 2023; 30:2189118. [PMID: 36919676 PMCID: PMC10026743 DOI: 10.1080/10717544.2023.2189118] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX), a commonly used anti-cancer drug, is limited by its cardiotoxicity and multidrug resistance (MDR) of tumor cells. Epigallocatechin gallate (EGCG), a natural antioxidant component, can effectively reduce the cardiotoxicity of DOX. Meanwhile, EGCG can inhibit the expression of P-glycoprotein (P-gp) and reverse the MDR of tumor cells. In this study, DOX is connected with low molecular weight polyethyleneimine (PEI) via hydrazone bond to get the pH-sensitive PEI-DOX, which is then combined with EGCG to prevent the cardiotoxicity of DOX and reverse the MDR of cancer cells. In addition, folic acid (FA) modified polyethylene glycol (PEG) (PEG-FA) is added to get the targeted system PEI-DOX/EGCG/FA. The MDR reversal and targeting ability of PEI-DOX/EGCG/FA is performed by cytotoxicity and in vivo anti-tumor activity on multidrug resistant MCF-7 cells (MCF-7/ADR). Additionally, we investigate the anti-drug resistant mechanism by Western Blot. The ability of EGCG to reduce DOX cardiotoxicity is confirmed by cardiotoxicity assay. In conclusion, PEI-DOX/EGCG/FA can inhibit the expression of P-gp and reverse the MDR in tumor cells. It also shows the ability of remove oxygen free radicals effectively to prevent the cardiotoxicity of DOX.
Collapse
Affiliation(s)
- Tianyu Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Nuannuan Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Ru Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Yiying Sun
- Yantai Saipute Analyzing Service Co. Ltd, Yantai, Shandong Province, China
| | - Xiaoyan He
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Xiaoyan Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Liuxiang Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Kaoxiang Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| |
Collapse
|
24
|
Zaman R, Cai X, Shubhra QTH. Hyperthermia-embolization-immunotherapy: a potent trio in advancing cancer treatment. Trends Mol Med 2023; 29:976-978. [PMID: 37863716 DOI: 10.1016/j.molmed.2023.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/22/2023]
Abstract
Yang et al. recently demonstrated the high potential of liquid metal microspheres (LM MSs) in cancer therapy. By amplifying the effects of magnetic hyperthermia and embolization, LM MSs not only target primary tumors, but also potentiate immune defenses. This dual-action approach effectively curtails distant tumor growth, marking a pivotal advancement in cancer immunotherapy.
Collapse
Affiliation(s)
- Rahela Zaman
- School of Healthy Aging, Faculty of Medicine and Health Sciences, UCSI University, Cheras 56000, Kuala Lumpur, Malaysia
| | - Xiaojun Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China.
| | - Quazi T H Shubhra
- Institute of Chemistry, University of Silesia in Katowice, 41-500 Chorzów, Poland; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
25
|
Alsaikhan F. Hyaluronic acid-empowered nanotheranostics in breast and lung cancers therapy. ENVIRONMENTAL RESEARCH 2023; 237:116951. [PMID: 37633628 DOI: 10.1016/j.envres.2023.116951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
Nanomedicine application in cancer therapy is an urgency because of inability of current biological therapies for complete removal of tumor cells. The development of smart and novel nanoplatforms for treatment of cancer can provide new insight in tumor suppression. Hyaluronic acid is a biopolymer that can be employed for synthesis of smart nanostructures capable of selective targeting CD44-overexpressing tumor cells. The breast and lung cancers are among the most malignant and common tumors in both females and males that environmental factors, lifestyle and genomic alterations are among the risk factors for their pathogenesis and development. Since etiology of breast and lung tumors is not certain and multiple factors participate in their development, preventative measures have not been completely successful and studies have focused on developing new treatment strategies for them. The aim of current review is to provide a comprehensive discussion about application of hyaluronic acid-based nanostructures for treatment of breast and lung cancers. The main reason of using hyaluronic acid-based nanoparticles is their ability in targeting breast and lung cancers in a selective way due to upregulation of CD44 receptor on their surface. Moreover, nanocarriers developed from hyaluronic acid or functionalized with hyaluronic acid have high biocompatibility and their safety is appreciated. The drugs and genes used for treatment of breast and lung cancers lack specific accumulation at cancer site and their cytotoxicity is low, but hyaluronic acid-based nanostructures provide their targeted delivery to tumor site and by increasing internalization of drugs and genes in breast and lung tumor cells, they improve their therapeutic index. Furthermore, hyaluronic acid-based nanostructures can be used for phototherapy-mediated breast and lung cancers ablation. The stimuli-responsive and smart kinds of hyaluronic acid-based nanostructures such as pH- and light-responsive can increase selective targeting of breast and lung cancers.
Collapse
Affiliation(s)
- Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| |
Collapse
|
26
|
Shubhra QTH. Iron oxide nanoparticles in magnetic drug targeting and ferroptosis-based cancer therapy. MEDICAL REVIEW (2021) 2023; 3:444-447. [PMID: 38283254 PMCID: PMC10811351 DOI: 10.1515/mr-2023-0029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/11/2023] [Indexed: 01/30/2024]
Abstract
Iron oxide (IO) nanoparticles (NPs) have gained significant attention in the field of biomedicine, particularly in drug targeting and cancer therapy. Their potential in magnetic drug targeting (MDT) and ferroptosis-based cancer therapy is highly promising. IO NPs serve as an effective drug delivery system (DDS), utilizing external magnetic fields (EMFs) to target cancer cells while minimizing damage to healthy organs. Additionally, IO NPs can generate reactive oxygen species (ROS) and induce ferroptosis, resulting in cytotoxic effects on cancer cells. This article explores how IO NPs can potentially revolutionize cancer research, focusing on their applications in MDT and ferroptosis-based therapy.
Collapse
Affiliation(s)
- Quazi T. H. Shubhra
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Institute of Chemistry, University of Silesia in KatowiceChorzów, Poland
| |
Collapse
|
27
|
Tan J, Zhuo Z, Si Y. Application of pyroptosis in tumor research (Review). Oncol Lett 2023; 26:376. [PMID: 37559585 PMCID: PMC10407856 DOI: 10.3892/ol.2023.13962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/27/2023] [Indexed: 08/11/2023] Open
Abstract
As a potent clinical strategy, cancer therapy has sparked an academic boom over the past few years. Immune checkpoint inhibitors (ICIs) have been demonstrated to be highly successful. These achievements have progressed cancer treatment and have made an indelible mark on cancer. However, the inherent complexity of cancer means that only part of the population can benefit from this treatment. Pyroptosis is a new suicidal cellular mechanism that induces inflammation by releasing immunogenic cellular components. Inflammatory signaling cascades mediated by pyroptosis commonly inspire numerous cell lysis in immune diseases. Contrariwise, this consequence may be a promising target in cancer research. Therefore, the present study briefly described programmed cell death processes and their potential roles in cancer. Because of the rapid development of bioengineering in cancer, the present study also examined the associated scaffolding available for cancer, highlighting advances in tumor engineering approaches. Ultimately, an improved understanding of pyroptosis and tumor scaffolding might shed light on a combination that can be manipulated for therapeutic purposes.
Collapse
Affiliation(s)
- Jianing Tan
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Suzhou, Jiangsu 215500, P.R. China
| | - Ziliang Zhuo
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Suzhou, Jiangsu 215500, P.R. China
| | - Yu Si
- Basic Research Laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
28
|
Pan H, Yang S, Cheng W, Cai Q, Shubhra QTH. Alternate-day fasting exacerbates doxorubicin cardiotoxicity in cancer chemotherapy. Trends Endocrinol Metab 2023:S1043-2760(23)00093-0. [PMID: 37246117 DOI: 10.1016/j.tem.2023.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/02/2023] [Accepted: 05/10/2023] [Indexed: 05/30/2023]
Abstract
Doxorubicin (Dox) is a highly potent chemotherapy drug. Despite its efficacy, Dox's clinical application is limited due to its association with significant complications, namely cardiotoxicity and the risk of heart failure. Recent intriguing findings by Ozcan et al. indicate that alternate-day fasting (ADF) significantly exacerbates the cardiotoxicity of Dox.
Collapse
Affiliation(s)
- Huachun Pan
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Shumin Yang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenke Cheng
- Medical Faculty, University of Leipzig, Leipzig 04103, Germany
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Quazi T H Shubhra
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Institute of Chemistry, University of Silesia in Katowice, 41-500 Chorzów, Poland.
| |
Collapse
|
29
|
Chen JW, Shen Y, Yu QS, Gan ZH. Paclitaxel Prodrug Nanomedicine for Potential CT-imaging Guided Breast Cancer Therapy. CHINESE JOURNAL OF POLYMER SCIENCE 2023. [DOI: 10.1007/s10118-023-2958-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
30
|
Chen C, Ye Q, Wang L, Zhou J, Xiang A, Lin X, Guo J, Hu S, Rui T, Liu J. Targeting pyroptosis in breast cancer: biological functions and therapeutic potentials on It. Cell Death Discov 2023; 9:75. [PMID: 36823153 PMCID: PMC9950129 DOI: 10.1038/s41420-023-01370-9] [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: 01/16/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Pyroptosis is a lytic and inflammatory type of programmed cell death that is mediated by Gasdermin proteins (GSDMs). Attractively, recent evidence indicates that pyroptosis involves in the development of tumors and can serve as a new strategy for cancer treatment. Here, we present a basic knowledge of pyroptosis, and an overview of the expression patterns and roles of GSDMs in breast cancer. In addition, we further summarize the available evidence of pyroptosis in breast cancer progression and give insight into the clinical potential of applying pyroptosis in anticancer strategies for breast cancer. This review will deepen our understanding of the relationship between pyroptosis and breast cancer, and provide a novel potential therapeutic avenue for breast cancer.
Collapse
Affiliation(s)
- Cong Chen
- grid.13402.340000 0004 1759 700XDepartment of Breast Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianwei Ye
- grid.13402.340000 0004 1759 700XDepartment of Breast Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linbo Wang
- grid.13402.340000 0004 1759 700XDepartment of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jichun Zhou
- grid.13402.340000 0004 1759 700XDepartment of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Aizhai Xiang
- grid.13402.340000 0004 1759 700XDepartment of Breast Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Lin
- grid.13402.340000 0004 1759 700XDepartment of Breast Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jufeng Guo
- grid.13402.340000 0004 1759 700XDepartment of Breast Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shufang Hu
- grid.13402.340000 0004 1759 700XDepartment of Breast Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Rui
- grid.13402.340000 0004 1759 700XDepartment of Breast Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Liu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|