1
|
Zhu M, Xie Y, Li Z, Bao H, Miao D, Guo X, Wang S, Chen K, Chen H, Dai J, Yang N, Yu L, Pei J. Antitumour and anti-angiogenesis efficacy of a multifunctional self-oxygenated active-targeting drug delivery system by encapsulating biological and chemotherapeutic drugs. Colloids Surf B Biointerfaces 2025; 250:114549. [PMID: 39965481 DOI: 10.1016/j.colsurfb.2025.114549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/12/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
The hypoxic tumour microenvironment (TME), resulting from abnormal tumour angiogenesis, is a major factor contributing to treatment failure in breast cancer patients. In this study, we present a ZnO2-based oestrone-conjugated PEGylated liposome (ZnO2@EPL-CDDP/EGCG) that incorporates cisplatin (CDDP) and epigallocatechin-3-gallate (EGCG). ZnO2 remains stable in neutral environments but decomposes under mildly acidic conditions, releasing Zn²⁺ and H₂O₂. These byproducts inhibit the electron transport chain, stimulate the endogenous reactive oxygen species production for chemodynamic therapy (CDT), and generate oxygen at tumour sites to alleviate hypoxia and enhance anti-angiogenic efficacy. EGCG inhibits tumour angiogenesis by down-regulating hypoxia-inducible factor-1α (HIF-1α) and its downstream pathways, while also exhibiting synergistic anti-tumour effects with CDDP. Oestrone-conjugated and polyethylene glycol (PEG) modifications facilitate targeted accumulation at tumour sites. Our findings indicate that ZnO2@EPL-CDDP/EGCG significantly improves the therapeutic efficacy of both EGCG and CDDP, remodels tumour vasculature, and alleviates hypoxia within the TME. This self-oxygenated, actively targeted drug delivery system notably extends the survival of healthy ICR mice without observed toxicity. This novel approach, which co-encapsulates ZnO2, EGCG, and CDDP in an active-targeting liposomal formulation for the first time, represents a promising strategy for effective cancer treatment.
Collapse
Affiliation(s)
- Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Zhiping Li
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Han Bao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Dongfanghui Miao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Xin Guo
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Shanshan Wang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Kejia Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Hongzhu Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Jingwen Dai
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Na Yang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Liangping Yu
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China.
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China.
| |
Collapse
|
2
|
Yan Z, Deng Y, Huang L, Zeng J, Wang D, Tong Z, Fan Q, Tan W, Yan J, Zang X, Chen S. Biopolymer-based bone scaffold for controlled Pt (IV) prodrug release and synergistic photothermal-chemotherapy and immunotherapy in osteosarcoma. J Nanobiotechnology 2025; 23:286. [PMID: 40205459 PMCID: PMC11983740 DOI: 10.1186/s12951-025-03253-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
Achieving bone defect repair while preventing tumor recurrence after osteosarcoma surgery has consistently posed a clinical challenge. Local treatment with 3D-printed scaffolds loaded with chemotherapeutic drugs can exert certain effects in tumor inhibition and bone regeneration. However, the non-specific activation of chemotherapeutic drugs leads to high local toxic side effects and the formation of an immunosuppressive tumor microenvironment, thereby limiting their clinical application and therapeutic efficacy. To address this, we designed a Pt (IV) prodrug with low toxicity and minimal side effects, which releases Pt (II) in response to glutathione. This prodrug was grafted onto polydopamine (PDA) through an amidation reaction, resulting in a composite nanomaterial (PDA@Pt) that possesses both photothermal synergistic chemotherapy and immuno-oncological properties. Subsequently, we innovatively employed selective laser sintering technology to incorporate PDA@Pt into a poly (L-lactic acid)/bioactive glass matrix, successfully constructing a composite scaffold with dual anti-tumor and bone repair capabilities. The study revealed that the composite scaffold significantly inhibited the growth of osteosarcoma cells and activated the cGAS-STING pathway by inducing DNA damage, ultimately converting the 'cold tumor' into a 'hot tumor.' Additionally, the composite scaffold could induce osteogenic differentiation of bone marrow mesenchymal stem cells and exhibited excellent bone repair capabilities in vivo.
Collapse
Affiliation(s)
- Zuyun Yan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Liping Huang
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhaochen Tong
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Qizhi Fan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jinpeng Yan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, 410017, P. R. China
| | - Xiaofang Zang
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China.
| |
Collapse
|
3
|
Huang Y, Xia M, Xu C, Lin Z, Chen M, Shi X, Ding Y, Xiao Y, Zhao C. A GSH-responsive oxidative stress nanoamplifier for self-augmented chemo/chemodynamic therapy to reverse cisplatin resistance. Acta Biomater 2025; 193:440-454. [PMID: 39706539 DOI: 10.1016/j.actbio.2024.12.041] [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: 09/09/2024] [Revised: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Drug resistance and off-target toxicity of cisplatin (CDDP) pose significant challenges in effectively treating non-small cell lung cancer (NSCLC). Recently, chemodynamic therapy (CDT), an emerging reactive oxygen species (ROS)-mediated tumor-specific therapeutic modality, has shown great potential in sensitizing multidrug resistance tumor cells. Herein, a glutathione (GSH)-responsive Pt(IV) prodrug-based oxidative stress nanoamplifier (CuBSO@PtC16) was developed for effective chemo/chemodynamic therapy to reverse CDDP resistance in NSCLC. CuBSO@PtC16, a lipid-coated nanoagent, was constructed by coordinating Cu2+ with l-buthioninesulfoximine (BSO) as the core framework, and Pt(IV) prodrug (PtC16) was concurrently loaded on the outer lipid bilayer. With appropriate particle size (∼35 nm) and good physiological stability, CuBSO@PtC16 efficiently accumulated at tumor tissue. Under high intracellular GSH levels, PtC16 was reduced to generate cytotoxic CDDP that induced cell-killing and boosted intracellular H2O2 levels, and the CuBSO core was disassembled to release Cu ions and BSO simultaneously. The released BSO could efficiently reduce the intracellular GSH content to weaken its detoxification effect on CDDP, leading to more Pt-DNA adduct formation and more severe DNA damage. Meanwhile, Cu ions catalyzed the intracellular elevated H2O2 into highly lethal •OH through Fenton-like reactions, and the reduction of GSH weakened the •OH elimination, which jointly amplified the intracellular oxidative stress levels, finally achieving enhanced chemo/chemodynamic therapeutic effect and reversing CDDP resistance in NSCLC. Therefore, this work offers an inspirational idea for effectively treating drug-resistant cancers. STATEMENT OF SIGNIFICANCE: Cisplatin (CDDP) faces challenges in treating non-small cell lung cancer (NSCLC) due to drug resistance and off-target toxicity. Herein, a GSH-responsive nanoreactor (CuBSO@PtC16) was developed for effective chemo/chemodynamic therapy to address CDDP resistance. CuBSO@PtC16 could efficiently traffic to tumor site and response to high GSH levels in tumor cells to release CDDP, Cu ions and buthioninesulfoximine (BSO) simultaneously. CDDP could induce DNA damage and boost intracellular H2O2 levels, which then served as the substrate of Cu to induce •OH generation through Fenton-like reactions. Meanwhile, the released BSO efficiently reduced the intracellular GSH content to weaken its detoxification effect on CDDP and the elimination of the •OH, leading to amplified intracellular oxidative stress and more severe damage to induce cell death.
Collapse
Affiliation(s)
- Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Meng Xia
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Congjun Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Zijun Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Meixu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xianmin Shi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yaqing Ding
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yan Xiao
- Laboratory Animal Center, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
4
|
Cao Y, Zheng M, Shi J, Si J, Huang G, Ji Y, Hou Y, Ge Z. X-ray-Triggered Activation of Polyprodrugs for Synergistic Radiochemotherapy. Biomacromolecules 2025; 26:579-590. [PMID: 39727263 DOI: 10.1021/acs.biomac.4c01373] [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: 12/28/2024]
Abstract
X-ray-induced photodynamic therapy (XPDT) can penetrate deeply into the tumor tissues to overcome the disadvantage of conventional PDT. However, the therapeutic efficacy of XPDT in cancer therapy is still restricted due to the insufficient reactive oxygen species (ROS) generation at a relatively low irradiation dosage. Herein, we present the tumor pH and ROS-responsive polyprodrug micelles to load the X-ray photosensitizer verteporfin (VP) as an ROS production enhancer. The block copolymer polyprodrug consisting of hydrophilic poly(ethylene glycol) (PEG) as well as the segments of thioketal-linked camptothecin (CPT) methacrylate (CPTKMA) and 2-(pentamethyleneimino)ethyl methacrylate (PEMA) (PEG-b-P(CPTKMA-co-PEMA)) can self-assemble into micelles in aqueous solution and encapsulate VP with a high loading efficiency of 67%. Inside tumor tissues, the zeta potential of the micelles can transform from neutral to positive for promoted cellular internalization under tumor acidity. Followed by X-ray irradiation at the dose of 4 Gy, efficient ROS generation in the presence of VP triggers CPT release. The VP-loaded polyprodrug micelles can finally ablate tumors efficiently via synergistic radiochemotherapy due to deep penetration of X-ray inside tumor tissues, ROS generation enhancement, and triggered CPT release. Consequently, this promising strategy represents a robust therapeutic modality for the enhanced radiochemotherapy of cancers.
Collapse
Affiliation(s)
- Yufei Cao
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Moujiang Zheng
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Jiahong Shi
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Jiale Si
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Guopu Huang
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yuanyuan Ji
- Department of Geriatric General Surgery, Scientific Research Center and Precision Medical Institute, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
5
|
Manturthi S, El-Sahli S, Bo Y, Durocher E, Kirkby M, Popatia A, Mediratta K, Daniel R, Lee SH, Iqbal U, Côté M, Wang L, Gadde S. Nanoparticles Codelivering mRNA and SiRNA for Simultaneous Restoration and Silencing of Gene/Protein Expression In Vitro and In Vivo. ACS NANOSCIENCE AU 2024; 4:416-425. [PMID: 39713729 PMCID: PMC11659891 DOI: 10.1021/acsnanoscienceau.4c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 12/24/2024]
Abstract
RNA-based agents (siRNA, miRNA, and mRNA) can selectively manipulate gene expression/proteins and are set to revolutionize a variety of disease treatments. Nanoparticle (NP) platforms have been developed to deliver functional mRNA or siRNA inside cells to overcome their inherent limitations. Recent studies have focused on siRNA to knock down proteins causing drug resistance or mRNA technology to introduce tumor suppressors. However, cancer needs multitargeted approaches to selectively manipulate multiple gene expressions/proteins. In this proof-of-concept study, we developed NPs containing Luc-mRNA and siRNA-GFP as model agents ((M+S)-NPs) and showed that NPs can simultaneously deliver functional mRNA and siRNA and impact the expression of two genes/proteins in vitro. Additionally, after in vivo administration, (M+S)-NPs successfully knocked down GFP while introducing luciferase into a TNBC mouse model, indicating that our NPs have the potential to develop RNA-based anticancer therapeutics. These studies pave the way to develop RNA-based, multitargeted approaches for complex diseases like cancer.
Collapse
Affiliation(s)
- Shireesha Manturthi
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H
8L6, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Sara El-Sahli
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Yuxia Bo
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H
8L6, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
| | - Emma Durocher
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H
8L6, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Melanie Kirkby
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Alyanna Popatia
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Karan Mediratta
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Redaet Daniel
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Seung-Hwan Lee
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Umar Iqbal
- Human Health
Therapeutics Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | - Marceline Côté
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Lisheng Wang
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Suresh Gadde
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H
8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research Institute, Ottawa, ON K1H
8L6, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa-Carleton
Institute for Biomedical Engineering (OCIBME), Ottawa, ON K1S
5B6, Canada
| |
Collapse
|
6
|
Sun Y, Ying K, Sun J, Qiu L, Wang Y, Ji M, Zhou L, Chen J. Curcumin mediates glutathione depletion via metal-organic framework nanocarriers to enhance cisplatin chemosensitivity on esophageal cancer. DISCOVER NANO 2024; 19:200. [PMID: 39661226 PMCID: PMC11635067 DOI: 10.1186/s11671-024-04168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Cisplatin (CDDP) is the primary drug used in the initial treatment of esophageal cancer (EC). However, its side effects and resistance can limit its effectiveness in clinical therapy. Curcumin (Cur)-mediated glutathione (GSH) depletion can reverse resistance, enhance the chemosensitivity of CDDP, and further improve the efficacy of platinum-containing chemotherapy in the treatment of esophageal cancer. However, it is also faced with problems of poor water solubility and low bioavailability in vivo, which severely hinders cancer treatments. In order to address these issues, we developed a novel nanotherapeutic system called CDCZA, combining Cur/CDDP/Cu/ZIF8@Au to enhance chemotherapy through GSH depletion and chemodynamic therapy through self-produced H2O2. Cu and CDDP were precisely co-loaded into Cu/ZIF8 nanoparticles using a one-pot method, then ultra-small gold nanoparticles mimicking glucose oxidase (Au nanoparticles) were embedded in the outer shell to create the CDCZA nano system. The released Cur could notably decrease intracellular GSH content and thus improve the chemosensitivity of CDDP, resulting in severe cellular apoptosis. And the Au nanoparticles effectively enabled chemodynamic therapy enhancement by accelerating the depletion of β-D-glucose into H2O2. As a result, the CDCZA nanoparticles showed increased tumor accumulation and improved antitumor effectiveness in a model of EC. Taken together, this work provides a new idea for the clinical design of efficient treatment reagents for EC.
Collapse
Affiliation(s)
- Yunhao Sun
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China.
| | - Kaijun Ying
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Jian Sun
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Limin Qiu
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Yao Wang
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Mingming Ji
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Lulu Zhou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, People's Republic of China.
| | - Jinjin Chen
- Department of Oncology, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China.
| |
Collapse
|
7
|
Tang L, Yin Y, Cao Y, Liu H, Qing G, Fu C, Li Z, Zhu Y, Shu W, He S, Gao J, Zhang Y, Wang Z, Bu J, Li X, Zhu M, Liang XJ, Wang W. Bioorthogonal Chemistry-Guided Inhalable Nanoprodrug to Circumvent Cisplatin Resistance in Orthotopic Nonsmall Cell Lung Cancer. ACS NANO 2024; 18:32103-32117. [PMID: 39520399 DOI: 10.1021/acsnano.4c10947] [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: 11/16/2024]
Abstract
Pulmonary delivery of anticancer therapeutics has shown encouraging performance in treating nonsmall cell lung cancer (NSCLC), which is characterized by high aggressiveness and poor prognosis. Cisplatin, a key member of the family of DNA alkylating agents, is extensively employed during NSCLC therapy. However, the development of chemoresistance and the occurrence of side effects severely impede the long-term application of cisplatin-based chemotherapies. Herein, we propose a meaningful strategy to precisely treat cisplatin-resistant NSCLC based on the combination of bioorthogonal chemistry with an inhalation approach. Ethacraplatin (EA-Pt), a platinum prodrug (IV), was synthesized and encapsulated in nitric oxide (NO)-containing micelles to overcome cisplatin chemoresistance. By further modifying bioorthogonal molecules in this nanoplatform (EA-Pt@MDBCO), an improved targeting performance toward pulmonary cancerous regions is achieved after prelabeling with azide via inhalation. Upon entering acidic cancer cells, EA-Pt is swiftly released due to the pH sensitivity of bioorthogonal micelles, which enables its bifunctions to inhibit glutathione S-transferase activity and deplete intracellular glutathione, eventually reversing cisplatin resistance. Moreover, the released NO also improves the overall therapeutic outcome against NSCLC. Consequently, inhalable EA-Pt@MDBCO prelabeled by azide effectively inhibits the progression of cisplatin-resistant orthotopic NSCLC, offering a feasible nanostrategy to expand the treatment options for NSCLC.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yuqi Cao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Guangchao Qing
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
| | - Cong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Zixuan Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yuanbo Zhu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Weijie Shu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jifan Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yi Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Zihan Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jianlan Bu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Xuejing Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Mengliang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing 100049, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
8
|
Bai Y, Wang Z, Liu D, Meng X, Wang H, Yu M, Zhang S, Sun T. Enhancing ovarian cancer treatment with maleimide-modified Pt(IV) prodrug nanoparticles. Mater Today Bio 2024; 27:101131. [PMID: 39050986 PMCID: PMC11267080 DOI: 10.1016/j.mtbio.2024.101131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/06/2024] [Accepted: 06/16/2024] [Indexed: 07/27/2024] Open
Abstract
The limitations of platinum in ovarian cancer therapy, such as poor solubility and significant side effects, often lead to suboptimal therapeutic outcome and mortality. In this study, we have developed a novel approach utilizing biodegradable polymeric nanoparticles as a drug delivery system (NDDS), loaded with advanced platinum (IV) (Pt(IV)) prodrugs. A key feature of our approach is the enhancement of nanoparticles with maleimide, a modification hypothesized to significantly boost tumor tissue accumulation. When tested in mouse models of orthotopic and peritoneal metastasis ovarian cancer, these maleimide-modified nanoparticles are anticipated to show preferential accumulation in tumor tissues, enhancing therapeutic efficiency and minimizing systemic drug exposure. Our findings demonstrate that the maleimide-modified Pt(IV)-loaded NDDSs significantly reduce tumor burden in comparison to traditional cisplatin therapy, while simultaneously reducing adverse side effects. This leads to markedly improved survival rates in models of peritoneal metastasis ovarian cancer, offering a promising new direction in the treatment of this challenging disease.
Collapse
Affiliation(s)
- Yiting Bai
- Department of Obstetrics and Gynaecology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Zhenpeng Wang
- Department of Obstetrics and Gynaecology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Dongzhen Liu
- Department of Obstetrics and Gynaecology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Haorui Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Meiling Yu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Songling Zhang
- Department of Obstetrics and Gynaecology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China
| |
Collapse
|
9
|
Wang Y, Deng T, Liu X, Fang X, Mo Y, Xie N, Nie G, Zhang B, Fan X. Smart Nanoplatforms Responding to the Tumor Microenvironment for Precise Drug Delivery in Cancer Therapy. Int J Nanomedicine 2024; 19:6253-6277. [PMID: 38911497 PMCID: PMC11193972 DOI: 10.2147/ijn.s459710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
The tumor microenvironment (TME) is a complex and dynamic entity, comprising stromal cells, immune cells, blood vessels and extracellular matrix, which is intimately associated with the occurrence and development of cancers, as well as their therapy. Utilizing the shared characteristics of tumors, such as an acidic environment, enzymes and hypoxia, researchers have developed a promising cancer therapy strategy known as responsive release of nano-loaded drugs, specifically targeted at tumor tissues or cells. In this comprehensive review, we provide an in-depth overview of the current fundamentals and state-of-the-art intelligent strategies of TME-responsive nanoplatforms, which include acidic pH, high GSH levels, high-level adenosine triphosphate, overexpressed enzymes, hypoxia and reductive environment. Additionally, we showcase the latest advancements in TME-responsive nanoparticles. In conclusion, we thoroughly examine the immediate challenges and prospects of TME-responsive nanopharmaceuticals, with the expectation that the progress of these targeted nanoformulations will enable the exploitation, overcoming or modulation of the TME, ultimately leading to significantly more effective cancer therapy.
Collapse
Affiliation(s)
- Yujie Wang
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035, People’s Republic of China
| | - Tingting Deng
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035, People’s Republic of China
| | - Xi Liu
- Department of Nephrology, Shenzhen Longgang Central Hospital, Shenzhen, 518116, People’s Republic of China
| | - Xueyang Fang
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035, People’s Republic of China
| | - Yongpan Mo
- Department of Breast Surgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035, People’s Republic of China
| | - Ni Xie
- The Bio-Bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
| | - Guohui Nie
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035, People’s Republic of China
| | - Bin Zhang
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035, People’s Republic of China
| | - Xiaoqin Fan
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, 518035, People’s Republic of China
- The Bio-Bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
| |
Collapse
|
10
|
Chen M, Fu Y, Liu Y, Zhang B, Song X, Chen X, Zhu Z, Gao H, Yang J, Shi X. NIR-Light-Triggered Mild-Temperature Hyperthermia to Overcome the Cascade Cisplatin Resistance for Improved Resistant Tumor Therapy. Adv Healthc Mater 2024; 13:e2303667. [PMID: 38178648 DOI: 10.1002/adhm.202303667] [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: 10/24/2023] [Revised: 12/13/2023] [Indexed: 01/06/2024]
Abstract
Currently, cisplatin resistance has been recognized as a multistep cascade process for its clinical chemotherapy failure. Hitherto, it remains challenging to develop a feasible and promising strategy to overcome the cascade drug resistance (CDR) issue for achieving fundamentally improved chemotherapeutic efficacy. Herein, a novel self-assembled nanoagent is proposed, which is constructed by Pt(IV) prodrug, cyanine dye (cypate), and gadolinium ion (Gd3+), for systematically conquering the cisplatin resistance by employing near-infrared (NIR) light activated mild-temperature hyperthermia in tumor targets. The proposed nanoagents exhibit high photostability, GSH/H+-responsive dissociation, preferable photothermal conversion, and enhanced cellular uptake performance. In particular, upon 785-nm NIR light irradiation, the generated mild temperature of ≈ 43 °C overtly improves the cell membrane permeability and drug uptake, accelerates the disruption of intracellular redox balance, and apparently enhances the formation of Pt-DNA adducts, thereby effectively overcoming the CDR issue and achieves highly improved therapeutic efficacy for cisplatin-resistant tumor ablation.
Collapse
Affiliation(s)
- Mingmao Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Yulei Fu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Yan Liu
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Key Laboratory of Nanomaterials, and State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, China
| | - Baihe Zhang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Xiaorong Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xinchun Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Zhengjia Zhu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Hang Gao
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Key Laboratory of Nanomaterials, and State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, Fuzhou, 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, Fuzhou, 350108, China
| |
Collapse
|
11
|
Bronowicka-Adamska P, Kaczor-Kamińska M, Wróbel M, Bentke-Imiolek A. Differences in nonoxidative sulfur metabolism between normal human breast MCF-12A and adenocarcinoma MCF-7 cell lines. Anal Biochem 2024; 687:115434. [PMID: 38141799 DOI: 10.1016/j.ab.2023.115434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 12/25/2023]
Abstract
Recent studies have revealed the role of endogenous hydrogen sulfide (H2S) in the development of breast cancer. The capacity of cells to generate H2S and the activity and expression of the main enzymes (cystathionine beta synthase; CBS, cystathionase γ-lyase; CGL, 3-mercaptopyruvate sulfurtransferase; MPST and thiosulfate sulfurtransferase; TST) involved in H2S metabolism were analyzed using an in vitro model of a non-tumourigenic breast cell line (MCF-12A) and a human breast adenocarcinoma cell line (MCF-7). In both cell lines, MPST, CGL, and TST expression was confirmed at the mRNA (RT-PCR) and the protein (Western Blot) level, while CBS expression was detected only in MCF-7 cells. Elevated levels of GSH, sulfane sulfur and increased CBS and TST activity were presented in the MCF-7 compared to the MCF-12A cells. It appears that cysteine might be mainly a substrate for GSH synthesis in breast adenocarcinoma. Increased capacity of the cells to generate H2S was shown for MCF-12A compared to MCF-7 cell line. Results suggest an important function of CBS in H2S metabolism in breast adenocarcinoma. The presented work may contribute to further research on new therapeutic possibilities for breast cancer - one of the most frequently diagnosed types of cancer among women.
Collapse
Affiliation(s)
| | - Marta Kaczor-Kamińska
- Jagiellonian University Medical College, Faculty of Medicine, Chair of Medical Biochemistry, Poland(1)
| | - Maria Wróbel
- Jagiellonian University Medical College, Faculty of Medicine, Chair of Medical Biochemistry, Poland(1)
| | - Anna Bentke-Imiolek
- Jagiellonian University Medical College, Faculty of Medicine, Chair of Medical Biochemistry, Poland(1)
| |
Collapse
|
12
|
Liu N, Lin Q, Huang Z, Liu C, Qin J, Yu Y, Chen W, Zhang J, Jiang M, Gao X, Huo S, Zhu X. Mitochondria-Targeted Prodrug Nanoassemblies for Efficient Ferroptosis-Based Therapy via Devastating Ferroptosis Defense Systems. ACS NANO 2024; 18:7945-7958. [PMID: 38452275 DOI: 10.1021/acsnano.3c10133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Ferroptosis is a form of regulated cell death accompanied by lipid reactive oxygen species (ROS) accumulation in an iron-dependent manner. However, the efficiency of tumorous ferroptosis was seriously restricted by intracellular ferroptosis defense systems, the glutathione peroxidase 4 (GPX4) system, and the ubiquinol (CoQH2) system. Inspired by the crucial role of mitochondria in the ferroptosis process, we reported a prodrug nanoassembly capable of unleashing potent mitochondrial lipid peroxidation and ferroptotic cell death. Dihydroorotate dehydrogenase (DHODH) inhibitor (QA) was combined with triphenylphosphonium moiety through a disulfide-containing linker to engineer well-defined nanoassemblies (QSSP) within a single-molecular framework. After being trapped in cancer cells, the acidic condition provoked the structural disassembly of QSSP to liberate free prodrug molecules. The mitochondrial membrane-potential-driven accumulation of the lipophilic cation prodrug was delivered explicitly into the mitochondria. Afterward, the thiol-disulfide exchange would occur accompanied by downregulation of reduced glutathione levels, thus resulting in mitochondria-localized GPX4 inactivation for ferroptosis. Simultaneously, the released QA from the hydrolysis reaction of the adjacent ester bond could further devastate mitochondrial defense and evoke robust ferroptosis via the DHODH-CoQH2 system. This subcellular targeted nanoassembly provides a reference for designing ferroptosis-based strategy for efficient cancer therapy through interfering antiferroptosis systems.
Collapse
Affiliation(s)
- Nian Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Qian Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zhenkun Huang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Chen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Jingbo Qin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Yanlin Yu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Weibin Chen
- School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jingbo Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Min Jiang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xuemin Gao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
13
|
Yang XC, Ding Y, Song SN, Wang WH, Huang S, Pang XY, Li B, Yu YY, Xia YM, Gao WW. Biocompatible N-carbazoleacetic acid decorated Cu xO nanoparticles as self-cascading platforms for synergistic single near-infrared triggered phototherapy treating microbial infections. Biomater Sci 2024; 12:1558-1572. [PMID: 38305728 DOI: 10.1039/d3bm01873c] [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: 02/03/2024]
Abstract
In this work, positively charged N-carbazoleacetic acid decorated CuxO nanoparticles (CuxO-CAA NPs) as novel biocompatible nanozymes have been successfully prepared through a one-step hydrothermal method. CuxO-CAA can serve as a self-cascading platform through effective GSH-OXD-like and POD-like activities, and the former can induce continuous generation of H2O2 through the catalytic oxidation of overexpressed GSH in the bacterial infection microenvironment, which in turn acts as a substrate for the latter to yield ˙OH via Fenton-like reaction, without introducing exogenous H2O2. Upon NIR irradiation, CuxO-CAA NPs possess a high photothermal conversion effect, which can further improve the enzymatic activity for increasing the production rate of H2O2 and ˙OH. Besides, the photodynamic performance of CuxO-CAA NPs can produce 1O2. The generated ROS and hyperthermia have synergetic effects on bacterial mortality. More importantly, CuxO-CAA NPs are more stable and biosafe than Cu2O, and can generate electrostatic adsorption with negatively charged bacterial cell membranes and accelerate bacterial death. Antibacterial results demonstrate that CuxO-CAA NPs are lethal against methicillin-resistant Staphylococcus aureus (MRSA) and ampicillin-resistant Escherichia coli (AREC) through destroying the bacterial membrane and disrupting the bacterial biofilm formation. MRSA-infected animal wound models show that CuxO-CAA NPs can efficiently promote wound healing without causing toxicity to the organism.
Collapse
Affiliation(s)
- Xiao-Chan Yang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Yong Ding
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Sheng-Nan Song
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Wen-Hui Wang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Shan Huang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
- The Third Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Xue-Yao Pang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Bo Li
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Ya-Ya Yu
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Ya-Mu Xia
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Wei-Wei Gao
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
14
|
Yin X, Zhuang Y, Song H, Xu Y, Zhang F, Cui J, Zhao L, Yu Y, Zhang Q, Ye J, Chen Y, Han Y. Antibody-platinum (IV) prodrugs conjugates for targeted treatment of cutaneous squamous cell carcinoma. J Pharm Anal 2024; 14:389-400. [PMID: 38618248 PMCID: PMC11010626 DOI: 10.1016/j.jpha.2023.11.002] [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: 08/05/2023] [Revised: 10/07/2023] [Accepted: 11/01/2023] [Indexed: 04/16/2024] Open
Abstract
Antibody-drug conjugates (ADCs) are a new type of targeting antibodies that conjugate with highly toxic anticancer drugs via chemical linkers to exert high specificity and efficient killing of tumor cells, thereby attracting considerable attention in precise oncology therapy. Cetuximab (Cet) is a typical antibody that offers the benefits of good targeting and safety for individuals with advanced and inoperable cutaneous squamous cell carcinoma (cSCC); however, its anti-tumor activity is limited to a single use. Cisplatin (CisPt) shows good curative effects; however, its adverse effects and non-tumor-targeting ability are major drawbacks. In this study, we designed and developed a new ADC based on a new cytotoxic platinum (IV) prodrug (C8Pt(IV)) and Cet. The so-called antibody-platinum (IV) prodrugs conjugates, named Cet-C8Pt(IV), showed excellent tumor targeting in cSCC. Specifically, it accurately delivered C8Pt(IV) into tumor cells to exert the combined anti-tumor effect of Cet and CisPt. Herein, metabolomic analysis showed that Cet-C8Pt(IV) promoted cellular apoptosis and increased DNA damage in cSCC cells by affecting the vitamin B6 metabolic pathway in tumor cells, thereby further enhancing the tumor-killing ability and providing a new strategy for clinical cancer treatment using antibody-platinum (IV) prodrugs conjugates.
Collapse
Affiliation(s)
- Xiangye Yin
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yingjie Zhuang
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, China
| | - Yujian Xu
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fan Zhang
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianxin Cui
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lei Zhao
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qixu Zhang
- Department of Plastic Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Youbai Chen
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
15
|
Sun S, He Y, Xu J, Leng S, Liu Y, Wan H, Yan L, Xu Y. Enhancing cell pyroptosis with biomimetic nanoparticles for melanoma chemo-immunotherapy. J Control Release 2024; 367:470-485. [PMID: 38290565 DOI: 10.1016/j.jconrel.2024.01.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/11/2024] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
Despite the fact that immunotherapy has significantly improved the prognosis of melanoma patients, the non-response rate of monoimmunotherapy is considerably high due to insufficient tumor immunogenicity. Therefore, it is necessary to develop alternative methods of combination therapy with enhanced antitumor efficiency and less systemic toxicity. In this study, we reported a cancer cell membrane-coated zeolitic imidazole framework-8 (ZIF-8) encapsulating pyroptosis-inducer oxaliplatin (OXA) and immunomodulator imiquimod (R837) for chemoimmunotherapy. With the assistance of DNA methyltransferase inhibitor decitabine (DCT), upregulated Gasdermin E (GSDME) was cleaved by OXA-activated caspase-3, further inducing tumor cell pyroptosis, then localized antitumor immunity was enhanced by immune adjuvant R837, followed by triggering systemic antitumor immune responses. These results provided a proof-of-concept for the use of cell membrane-coated biomimetic nanoparticles as a promising drug carrier of combination therapy and a potential insight for pyroptosis-based melanoma chemo-immunotherapy.
Collapse
Affiliation(s)
- Shiquan Sun
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China; Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Yong He
- R&D Department of 3D printing, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jiaqi Xu
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Shaolong Leng
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yu Liu
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Huanhuan Wan
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China
| | - Leping Yan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China.
| | - Yunsheng Xu
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, PR China.
| |
Collapse
|
16
|
Xu L, Cao Y, Xu Y, Li R, Xu X. Redox-Responsive Polymeric Nanoparticle for Nucleic Acid Delivery and Cancer Therapy: Progress, Opportunities, and Challenges. Macromol Biosci 2024; 24:e2300238. [PMID: 37573033 DOI: 10.1002/mabi.202300238] [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: 05/25/2023] [Revised: 07/25/2023] [Indexed: 08/14/2023]
Abstract
Cancer development and progression of cancer are closely associated with the activation of oncogenes and loss of tumor suppressor genes. Nucleic acid drugs (e.g., siRNA, mRNA, and DNA) are widely used for cancer therapy due to their specific ability to regulate the expression of any cancer-associated genes. However, nucleic acid drugs are negatively charged biomacromolecules that are susceptible to serum nucleases and cannot cross cell membrane. Therefore, specific delivery tools are required to facilitate the intracellular delivery of nucleic acid drugs. In the past few decades, a variety of nanoparticles (NPs) are designed and developed for nucleic acid delivery and cancer therapy. In particular, the polymeric NPs in response to the abnormal redox status in cancer cells have garnered much more attention as their potential in redox-triggered nanostructure dissociation and rapid intracellular release of nucleic acid drugs. In this review, the important genes or signaling pathways regulating the abnormal redox status in cancer cells are briefly introduced and the recent development of redox-responsive NPs for nucleic acid delivery and cancer therapy is systemically summarized. The future development of NPs-mediated nucleic acid delivery and their challenges in clinical translation are also discussed.
Collapse
Affiliation(s)
- Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Ya Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Rong Li
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| |
Collapse
|
17
|
Gan Y, Xie W, Wang M, Wang P, Li Q, Cheng J, Yan M, Xia J, Wu Z, Zhang G. Cancer cell membrane-camouflaged CuPt nanoalloy boosts chemotherapy of cisplatin prodrug to enhance anticancer effect and reverse cisplatin resistance of tumor. Mater Today Bio 2024; 24:100941. [PMID: 38269055 PMCID: PMC10805937 DOI: 10.1016/j.mtbio.2023.100941] [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: 10/04/2023] [Revised: 12/02/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024] Open
Abstract
The biotoxicity and chemotherapeutic resistance of cisplatin (CDDP) pose a challenge for tumor therapy. Practically, the change in the therapeutic response of tumor from resistance to sensitivity are impressive but challenging. To this end, we propose a strategy of "one stone, three birds" by designing a CuPt nanoalloy to simultaneously eliminate GSH, relieve hypoxia, and promote ROS production for effectively reversing the platinum (IV) (Pt(IV), (c,c,t-[Pt(NH3)2Cl2(OOCCH2CH2COOH)2)) resistance. Notably, the CuPt nanoalloy exhibits ternary catalytic capabilities including mimicking GSH oxidase, catalase and peroxidase. With the subsequent disguise of tumor cell membrane, the CuPt nanoalloy is conferred with homologous targeting ability, making it actively recognize tumor cells and then effectively internalized by tumor cells. Upon entering tumor cell, it gives rise to GSH depletion, hypoxia relief, and oxidative stress enhancement by catalyzing the reaction of GSH and H2O2, which mitigates the vicious milieu and ultimately reinforces the tumor response to Pt(IV) treatment. In vivo results prove that combination therapy of mCuPt and Pt(IV) realizes the most significant suppression on A549 cisplatin-resistant tumor. This study provides a potential strategy to design novel nanozyme for conquering resistant tumor.
Collapse
Affiliation(s)
- Yuehao Gan
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, PR China
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Wenteng Xie
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
- Engineering and Materials Science Experiment Center, University of Science and Technology of China, Hefei, 230026, PR China
| | - Miaomiao Wang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, PR China
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Peng Wang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, PR China
| | - Qingdong Li
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, PR China
| | - Junjie Cheng
- Engineering and Materials Science Experiment Center, University of Science and Technology of China, Hefei, 230026, PR China
| | - Miao Yan
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, PR China
| | - Jikai Xia
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, PR China
| | - Zhengyan Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Guilong Zhang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, PR China
| |
Collapse
|
18
|
Li Q, Chen S, Wang X, Cai J, Huang H, Tang S, He D. Cisplatin-Based Combination Therapy for Enhanced Cancer Treatment. Curr Drug Targets 2024; 25:473-491. [PMID: 38591210 DOI: 10.2174/0113894501294182240401060343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Cisplatin, a primary chemotherapeutic drug, is of great value in the realm of tumor treatment. However, its clinical efficacy is strictly hindered by issues, such as drug resistance, relapse, poor prognosis, and toxicity to normal tissue. Cisplatin-based combination therapy has garnered increasing attention in both preclinical and clinical cancer research for its ability to overcome resistance, reduce toxicity, and enhance anticancer effects. This review examines three primary co-administration strategies of cisplatin-based drug combinations and their respective advantages and disadvantages. Additionally, seven types of combination therapies involving cisplatin are discussed, focusing on their main therapeutic effects, mechanisms in preclinical research, and clinical applications. This review also discusses future prospects and challenges, aiming to offer guidance for the development of optimal cisplatin-based combination therapy regimens for improved cancer treatment.
Collapse
Affiliation(s)
- Qi Li
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Siwei Chen
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Xiao Wang
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Jia Cai
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Hongwu Huang
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, China
| | - Dongxiu He
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| |
Collapse
|
19
|
Imran H, Tang Y, Wang S, Yan X, Liu C, Guo L, Wang E, Xu C. Optimized DOX Drug Deliveries via Chitosan-Mediated Nanoparticles and Stimuli Responses in Cancer Chemotherapy: A Review. Molecules 2023; 29:31. [PMID: 38202616 PMCID: PMC10780101 DOI: 10.3390/molecules29010031] [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: 10/15/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Chitosan nanoparticles (NPs) serve as useful multidrug delivery carriers in cancer chemotherapy. Chitosan has considerable potential in drug delivery systems (DDSs) for targeting tumor cells. Doxorubicin (DOX) has limited application due to its resistance and lack of specificity. Chitosan NPs have been used for DOX delivery because of their biocompatibility, biodegradability, drug encapsulation efficiency, and target specificity. In this review, various types of chitosan derivatives are discussed in DDSs to enhance the effectiveness of cancer treatments. Modified chitosan-DOX NP drug deliveries with other compounds also increase the penetration and efficiency of DOX against tumor cells. We also highlight the endogenous stimuli (pH, redox, enzyme) and exogenous stimuli (light, magnetic, ultrasound), and their positive effect on DOX drug delivery via chitosan NPs. Our study sheds light on the importance of chitosan NPs for DOX drug delivery in cancer treatment and may inspire the development of more effective approaches for cancer chemotherapy.
Collapse
Affiliation(s)
- HafizMuhammad Imran
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Yixin Tang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Siyuan Wang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Xiuzhang Yan
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Chang Liu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Lei Guo
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| | - Erlei Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Caina Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.I.); (Y.T.); (S.W.); (X.Y.); (C.L.); (L.G.)
| |
Collapse
|
20
|
Wei D, Sun Y, Zhu H, Fu Q. Stimuli-Responsive Polymer-Based Nanosystems for Cancer Theranostics. ACS NANO 2023; 17:23223-23261. [PMID: 38041800 DOI: 10.1021/acsnano.3c06019] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2023]
Abstract
Stimuli-responsive polymers can respond to internal stimuli, such as reactive oxygen species (ROS), glutathione (GSH), and pH, biological stimuli, such as enzymes, and external stimuli, such as lasers and ultrasound, etc., by changing their hydrophobicity/hydrophilicity, degradability, ionizability, etc., and thus have been widely used in biomedical applications. Due to the characteristics of the tumor microenvironment (TME), stimuli-responsive polymers that cater specifically to the TME have been extensively used to prepare smart nanovehicles for the targeted delivery of therapeutic and diagnostic agents to tumor tissues. Compared to conventional drug delivery nanosystems, TME-responsive nanosystems have many advantages, such as high sensitivity, broad applicability among different tumors, functional versatility, and improved biosafety. In recent years, a great deal of research has been devoted to engineering efficient stimuli-responsive polymeric nanosystems, and significant improvement has been made to both cancer diagnosis and therapy. In this review, we summarize some recent research advances involving the use of stimuli-responsive polymer nanocarriers in drug delivery, tumor imaging, therapy, and theranostics. Various chemical stimuli will be described in the context of stimuli-responsive nanosystems. Accordingly, the functional chemical groups responsible for the responsiveness and the strategies to incorporate these groups into the polymer will be discussed in detail. With the research on this topic expending at a fast pace, some innovative concepts, such as sequential and cascade drug release, NIR-II imaging, and multifunctional formulations, have emerged as popular strategies for enhanced performance, which will also be included here with up-to-date illustrations. We hope that this review will offer valuable insights for the selection and optimization of stimuli-responsive polymers to help accelerate their future applications in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Dengshuai Wei
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Hu Zhu
- Maoming People's Hospital, Guangdong 525000, China
| | - Qinrui Fu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
21
|
Bai Y, Aodeng G, Ga L, Hai W, Ai J. Research Progress of Metal Anticancer Drugs. Pharmaceutics 2023; 15:2750. [PMID: 38140091 PMCID: PMC10747151 DOI: 10.3390/pharmaceutics15122750] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer treatments, including traditional chemotherapy, have failed to cure human malignancies. The main reasons for the failure of these treatments are the inevitable drug resistance and serious side effects. In clinical treatment, only 5 percent of the 50 percent of cancer patients who are able to receive conventional chemotherapy survive. Because of these factors, being able to develop a drug and treatment that can target only cancer cells without affecting normal cells remains a big challenge. Since the special properties of cisplatin in the treatment of malignant tumors were accidentally discovered in the last century, metal anticancer drugs have become a research hotspot. Metal anticancer drugs have unique pharmaceutical properties, such as ruthenium metal drugs with their high selectivity, low toxicity, easy absorption by tumor tissue, excretion, and so on. In recent years, efficient and low-toxicity metal antitumor complexes have been synthesized. In this paper, the scientific literature on platinum (Pt), ruthenium (Ru), iridium (Ir), gold (Au), and other anticancer complexes was reviewed by referring to a large amount of relevant literature at home and abroad.
Collapse
Affiliation(s)
- Yun Bai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Gerile Aodeng
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Lu Ga
- College of Pharmacy, Inner Mongolia Medical University, Jinchuankaifaqu, Hohhot 010110, China;
| | - Wenfeng Hai
- Inner Mongolia Key Laboratory of Carbon Nanomaterials, Nano Innovation Institute (NII), College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Jun Ai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| |
Collapse
|
22
|
Fu Y, Kong Y, Li X, Cheng D, Hou Y, Li Y, Li T, Xiao Y, Zhang Q, Rong R. Novel Pt(IV) prodrug self-assembled nanoparticles with enhanced blood circulation stability and improved antitumor capacity of oxaliplatin for cancer therapy. Drug Deliv 2023; 30:2171158. [PMID: 36744299 PMCID: PMC9904295 DOI: 10.1080/10717544.2023.2171158] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pt(IV) compounds are regarded as prodrugs of active Pt(II) drugs (i.e. cisplatin, carboplatin, and oxaliplatin) and burgeoned as the most ideal candidates to substitute Pt(II) anticancer drugs with severe side effects. Nanoparticle drug delivery systems have been widely introduced to deliver Pt(IV) prodrugs more effectively and safely to tumors, but clinical outcomes were unpredictable owing to limited in vivo pharmacokinetics understanding. Herein, a novel Pt(IV) prodrug of oxaliplatin(OXA) was synthesized and prepared as self-assembled micellar nanoparticles(PEG-OXA NPs). In vitro, PEG-OXA NPs rapidly released biologically active OXA within 5 min in tumor cells while remaining extremely stable in whole blood or plasma. Importantly, the pharmacokinetic results showed that the AUC0-∞, and t1/2 values of PEG-OXA NPs were 1994 ± 117 h·µg/mL and 3.28 ± 0.28 h, respectively, which were much higher than that of free OXA solution (2.03 ± 0.55 h·µg/mL and 0.16 ± 0.07 h), indicating the longer drug circulation of PEG-OXA NPs in vivo. The altered pharmacokinetic behavior of PEG-OXA NPs remarkably contributed to improve antitumor efficacy, decrease systemic toxicity and increase tumor growth inhibition compared to free OXA. These findings establish that PEG-OXA NPs have the potential to offer a desirable self-delivery platform of platinum drugs for anticancer therapeutics.
Collapse
Affiliation(s)
- Yuanlei Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China,Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Ying Kong
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Xiangping Li
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Dongfang Cheng
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Yuqian Hou
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Yan Li
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Tongfang Li
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Yani Xiao
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Qiuyan Zhang
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China,CONTACT Qiuyan Zhang
| | - Rong Rong
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, China,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China,Rong Rong
| |
Collapse
|
23
|
Wang X, Yuan L, Lu B, Lin D, Xu X. Glutathione promotes the synergistic effects of venetoclax and azacytidine against myelodysplastic syndrome‑refractory anemia by regulating the cell cycle. Exp Ther Med 2023; 26:574. [PMID: 38023359 PMCID: PMC10652243 DOI: 10.3892/etm.2023.12274] [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: 05/28/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Azacitidine is a DNA methyltransferase inhibitor that has been used as a singular agent for the treatment of myelodysplastic syndrome-refractory anemia with excess blast-1 and -2 (MDS-RAEB I/II). However, recurrence and overall response rates following this treatment remain unsatisfactory. The combination of azacitidine and venetoclax has been used for the clinical treatment of a variety of hematological diseases due to the synergistic killing effect of the two drugs. Venetoclax is a BCL-2 inhibitor that can inhibit mitochondrial metabolism. In addition, azacitidine has been shown to reduce the levels of myeloid cell leukemia 1 (MCL-1) in acute myeloid leukemia cells. MCL-1 is an anti-apoptotic protein and a potential source of resistance to venetoclax. However, the mechanism underlying the effects of combined venetoclax and azacitidine treatment remains to be fully elucidated. In the present study, the molecular mechanism underlying the impact of venetoclax on the efficacy of azacitidine was investigated by examining its effects on cell cycle progression. SKM-1 cell lines were treated in vitro with 0-2 µM venetoclax and 0-4 µM azacytidine. After 24, 48 and 72 h of treatment, the impact of the drugs on the cell cycle was assessed by flow cytometry. Following drug treatment, changes in cellular glutamine metabolism pathways was analyzed using western blotting (ATF4, CHOP, ASCT2, IDH2 and RB), quantitative PCR (ASCT2 and IDH2), liquid chromatography-mass spectrometry (α-KG, succinate and glutathione) and ELISA (glutamine and glutaminase). Venetoclax was found to inhibit mitochondrial activity though the alanine-serine-cysteine transporter 2 (ASCT2) pathway, which decreased glutamine uptake. Furthermore, venetoclax partially antagonized the action of azacitidine through this ASCT2 pathway, which was reversed by glutathione (GSH) treatment. These results suggest that GSH treatment can potentiate the synergistic therapeutic effects of venetoclax and azacitidine combined treatment on a myelodysplastic syndrome-refractory anemia cell line at lower concentrations.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Lihua Yuan
- Department of Pediatric Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Bo Lu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| |
Collapse
|
24
|
Zheng S, Li G, Shi J, Liu X, Li M, He Z, Tian C, Kamei KI. Emerging platinum(IV) prodrug nanotherapeutics: A new epoch for platinum-based cancer therapy. J Control Release 2023; 361:819-846. [PMID: 37597809 DOI: 10.1016/j.jconrel.2023.08.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Owing to the unique DNA damaging cytotoxicity, platinum (Pt)-based chemotherapy has long been the first-line choice for clinical oncology. Unfortunately, Pt drugs are restricted by the severe dose-dependent toxicity and drug resistance. Correspondingly, Pt(IV) prodrugs are developed with the aim to improve the antitumor performance of Pt drugs. However, as "free" molecules, Pt(IV) prodrugs are still subject to unsatisfactory in vivo destiny and antitumor efficacy. Recently, Pt(IV) prodrug nanotherapeutics, inheriting both the merits of Pt(IV) prodrugs and nanotherapeutics, have emerged and demonstrated the promise to address the underexploited dilemma of Pt-based cancer therapy. Herein, we summarize the latest fronts of emerging Pt(IV) prodrug nanotherapeutics. First, the basic outlines of Pt(IV) prodrug nanotherapeutics are overviewed. Afterwards, how versatile Pt(IV) prodrug nanotherapeutics overcome the multiple biological barriers of antitumor drug delivery is introduced in detail. Moreover, advanced combination therapies based on multimodal Pt(IV) prodrug nanotherapeutics are discussed with special emphasis on the synergistic mechanisms. Finally, prospects and challenges of Pt(IV) prodrug nanotherapeutics for future clinical translation are spotlighted.
Collapse
Affiliation(s)
- Shunzhe Zheng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guanting Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianbin Shi
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinying Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Meng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, Hangzhou 310058, China.
| | - Ken-Ichiro Kamei
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
25
|
Zhou X, An B, Lin Y, Ni Y, Zhao X, Liang X. Molecular mechanisms of ROS-modulated cancer chemoresistance and therapeutic strategies. Biomed Pharmacother 2023; 165:115036. [PMID: 37354814 DOI: 10.1016/j.biopha.2023.115036] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023] Open
Abstract
Drug resistance is the main obstacle to achieving a cure in many cancer patients. Reactive oxygen species (ROS) are master regulators of cancer development that act through complex mechanisms. Remarkably, ROS levels and antioxidant content are typically higher in drug-resistant cancer cells than in non-resistant and normal cells, and have been shown to play a central role in modulating drug resistance. Therefore, determining the underlying functions of ROS in the modulation of drug resistance will contribute to develop therapies that sensitize cancer resistant cells by leveraging ROS modulation. In this review, we summarize the notable literature on the sources and regulation of ROS production and highlight the complex roles of ROS in cancer chemoresistance, encompassing transcription factor-mediated chemoresistance, maintenance of cancer stem cells, and their impact on the tumor microenvironment. We also discuss the potential of ROS-targeted therapies in overcoming tumor therapeutic resistance.
Collapse
Affiliation(s)
- Xiaoting Zhou
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Biao An
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yi Lin
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yanghong Ni
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiao Liang
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
26
|
Ma Y, Feng Q, Han B, Yu R, Jin Z. Elevated HMGB1 promotes the malignant progression and contributes to cisplatin resistance of non-small cell lung cancer. Hereditas 2023; 160:33. [PMID: 37518006 PMCID: PMC10388484 DOI: 10.1186/s41065-023-00294-9] [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: 05/17/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND HMGB1 (high mobility group box B-1) exhibits crucial role in tumor genesis and development, including lung cancer. Whereas, more HMGB1-related details in non-small cell lung cancer (NSCLC) are still largely unclear. METHODS The HMGB1 and inflammatory factors in malignant (MPE) and non-malignant pleural effusion (BPE) were determined by ELISA. Additionally, qRT-PCR, western blot, or immunohistochemistry were used to determine HMGB1, drug-resistant and apoptotic proteins' expressions in NSCLC A549, A549-DDP cell lines, and xenograft model. Cell viability, migration/ invasion, and apoptosis were analyzed using MTT, Transwell, and flow cytometry assays, respectively. RESULTS Inflammatory factors and HMGB1 expressions in MPE were significantly higher than BPE of NSCLC. Compared with preoperative and adjacent tissues, significantly higher HMGB1, drug-resistant protein, and anti-apoptotic protein expressions were observed in recurrent tissues. Overexpressed HMGB1 induced NSCLC cells to exhibit stronger aggressive, proliferative, and drug-resistant features. The related abilities were reversed when HMGB1 was interfered. Overexpressed HMGB1 showed a similar co-localization with drug resistant protein P-gp in cytoplasm in xenograft model, while low HMGB1 expression localized in cell nucleus. CONCLUSIONS HMGB1 overexpression significantly promoted the malignant progression and cisplatin resistance of NSCLC in vitro and in vivo.
Collapse
Affiliation(s)
- Ying Ma
- Department of Thoracic Surgery, Affiliated People's Hospital of Inner Mongolia Medical University, No.1 Tong Dao Bei Road, Hohhot, 010059, China
| | - Qin Feng
- Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, 010020, China
| | - Bateer Han
- Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, 010020, China
| | - Rong Yu
- Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, 010020, China
| | - Zhiyong Jin
- Department of Thoracic Surgery, Affiliated People's Hospital of Inner Mongolia Medical University, No.1 Tong Dao Bei Road, Hohhot, 010059, China.
| |
Collapse
|
27
|
Li C, Chen Y, Gao Y, Wang X, Wang J, Zhang P, Hu X, Li L, Tong W, Ren Z, Yao W. A nanomedicine based on stoichiometric coordination of camptothecin and organoplatinum (II) for synergistic antitumor therapy. Acta Biomater 2023; 164:553-562. [PMID: 37072068 DOI: 10.1016/j.actbio.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/20/2023]
Abstract
Precise combination therapy, involving multiple chemotherapeutics with pharmacologically synergistic antitumor effects, is a promising approach to address the challenge of monotherapy with insufficient activity towards their targets of interest. We employed Pt←pyridine coordination-driven assembly to construct a stoichiometric coordination complex of camptothecin and organoplatinum (II) (Pt-CPT). The Pt-CPT complex exhibited a remarkable synergistic effect toward several tumor cell lines, which is equal to the optimal synergistic effect of (PEt3)2Pt(OTf)2 (Pt) and CPT mixture at various ratios. An amphiphilic polymer with H2O2-responsiveness and glutathione (GSH)-depleting ability (PO) was used to encapsulate Pt-CPT complex to enable the nanomedicine (Pt-CPT@PO) with prolonged blood circulation and elevated tumor accumulation. The Pt-CPT@PO nanomedicine exhibited remarkable synergistic antitumor efficacy and antimetastatic effect on a mice orthotopic breast tumor model. This work demonstrated the potential of stoichiometric coordination-driven assembly of organic therapeutics with metal-based drugs in developing advanced nanomedicine with optimal synergistic antitumor activity. STATEMENT OF SIGNIFICANCE: In this study, for the first time, we employed Pt←pyridine coordination-driven assembly to construct a stoichiometric coordination complex of camptothecin and organoplatinum (II) (Pt-CPT), with an optimal synergistic effect at various ratios. Then it was encapsulated into an amphiphilic polymer with H2O2-responsiveness and glutathione (GSH)-depleting ability (PO) to enable the nanomedicine (Pt-CPT@PO) with prolonged blood circulation and elevated tumor accumulation. The Pt-CPT@PO nanomedicine exhibited remarkable synergistic antitumor efficacy and antimetastatic effect on a mice orthotopic breast tumor model.
Collapse
Affiliation(s)
- Chao Li
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450052 PR China
| | - Yu Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450052 PR China; Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 PR China
| | - Yong Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058 PR China
| | - Xin Wang
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450052 PR China
| | - Jiaqiang Wang
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450052 PR China
| | - Peng Zhang
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450052 PR China
| | - Xiaobo Hu
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 PR China; Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 PR China
| | - Lei Li
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 PR China; Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 PR China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058 PR China.
| | - Zhigang Ren
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 PR China; Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 PR China.
| | - Weitao Yao
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450052 PR China.
| |
Collapse
|
28
|
He P, Xu S, Miao Z, Que Y, Chen Y, Li S, Ma Q, Yang R, Wei W, Zha Z, Hu Y. Anti-Her2 affibody-decorated arsenene nanosheets induce ferroptosis through depleting intracellular GSH to overcome cisplatin resistance. J Nanobiotechnology 2023; 21:203. [PMID: 37370105 DOI: 10.1186/s12951-023-01963-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Ferroptosis, a form of regulated cell death induced by excessive accumulation of reactive oxygen species and lipid peroxidation, has recently attracted extensive attention due to its ability to effectively suppress tumors and overcome drug resistance. Unlike previously reported metal nanomaterials that induce ferroptosis via the Fenton reaction, arsenene nanosheets can effectively deplete intracellular glutathione and then induce ferroptosis by inhibiting glutathione peroxidase 4. In this study, we designed target-modified arsenene nanosheets loaded with cisplatin (Her2-ANs@CDDP), which are capable of selective uptake by tumor cells. Her2-ANs@CDDP promotes both apoptosis and ferroptosis through a reciprocal cascade reaction between cisplatin and the carrier, respectively, and we demonstrate that it can significantly inhibit the activity of drug-resistant cells. Arsenene nanosheets kill drug-resistant tumor cells by inducing ferroptosis and restoring the sensitivity of drug-resistant cells to cisplatin. Cisplatin-loaded arsenene nanosheets can be prepared simply, and exert synergistic effects that overcome drug resistance. They show great potential for applications in the clinical treatment of chemotherapy-insensitive osteosarcoma, expanding the uses of arsenic in the treatment of solid tumors.
Collapse
Affiliation(s)
- Peng He
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Shenglin Xu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Zhaohua Miao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Yukang Que
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Yu Chen
- Department of Pharmacy, Anqing Medical College, Anqing, 246052, Anhui, China
| | - Sheng Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Qiming Ma
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Rui Yang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China.
| | - Yong Hu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
29
|
Liu H, Chen W, Wu G, Zhou J, Liu C, Tang Z, Huang X, Gao J, Xiao Y, Kong N, Joshi N, Cao Y, Abdi R, Tao W. Glutathione-Scavenging Nanoparticle-Mediated PROTACs Delivery for Targeted Protein Degradation and Amplified Antitumor Effects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207439. [PMID: 37066758 PMCID: PMC10238184 DOI: 10.1002/advs.202207439] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/10/2023] [Indexed: 06/04/2023]
Abstract
PROteolysis TArgeting Chimeras (PROTACs) are an emerging class of promising therapeutic modalities that selectively degrade intracellular proteins of interest by hijacking the ubiquitin-proteasome system. However, the lack of techniques to efficiently transport these degraders to targeted cells and consequently the potential toxicity of PROTACs limit their clinical applications. Here, a strategy of nanoengineered PROTACs, that is, Nano-PROTACs, is reported, which improves the bioavailability of PROTACs and maximizes their capacity to therapeutically degrade intracellular oncogenic proteins for tumor therapy. The Nano-PROTACs are developed by encapsulating PROTACs in glutathione (GSH)-responsive poly(disulfide amide) polymeric (PDSA) nanoparticles and show that ARV@PDSA Nano-PROTAC, nanoengineered BRD4 degrader ARV-771, improves BRD4 protein degradation and decreases the downstream oncogene c-Myc expression. Benefiting from the GSH-scavenging ability to amply the c-Myc-related ferroptosis and cell cycle arrest, this ARV@PDSA Nano-PROTACs strategy shows superior anti-tumor efficacy with a low dose administration and good biocompatibility in vivo. The findings reveal the potential of the Nano-PROTACs strategy to treat a broad range of diseases by dismantling associated pathogenic proteins.
Collapse
Affiliation(s)
- Hai‐Jun Liu
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
- Transplantation Research CenterRenal DivisionBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Wei Chen
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Gongwei Wu
- Department of Medical OncologyDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA02215USA
| | - Jun Zhou
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Chuang Liu
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Zhongmin Tang
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Xiangang Huang
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Jingjing Gao
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Yufen Xiao
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Na Kong
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Nitin Joshi
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Yihai Cao
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 77Sweden
| | - Reza Abdi
- Transplantation Research CenterRenal DivisionBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Wei Tao
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| |
Collapse
|
30
|
Liu W, Li X, Wang T, Xiong F, Sun C, Yao X, Huang W. Platinum Drug-Incorporating Polymeric Nanosystems for Precise Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208241. [PMID: 36843317 DOI: 10.1002/smll.202208241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/01/2023] [Indexed: 05/25/2023]
Abstract
Platinum (Pt) drugs are widely used in clinic for cancer therapy, but their therapeutic outcomes are significantly compromised by severe side effects and acquired drug resistance. With the emerging immunotherapy and imaging-guided cancer therapy, precise delivery and release of Pt drugs have drawn great attention these days. The targeting delivery of Pt drugs can greatly increase the accumulation at tumor sites, which ultimately enhances antitumor efficacy. Further, with the combination of Pt drugs and other theranostic agents into one nanosystem, it not only possesses excellent synergistic efficacy but also achieves real-time monitoring. In this review, after the introduction of Pt drugs and their characteristics, the recent progress of polymeric nanosystems for efficient delivery of Pt drugs is summarized with an emphasis on multi-modal synergistic therapy and imaging-guided Pt-based cancer treatment. In the end, the conclusions and future perspectives of Pt-encapsulated nanosystems are given.
Collapse
Affiliation(s)
- Wei Liu
- School of Flexible Electronics (Future Technologies) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Xin Li
- School of Pharmaceutical Science, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Ting Wang
- School of Flexible Electronics (Future Technologies) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Fei Xiong
- School of Flexible Electronics (Future Technologies) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Changrui Sun
- School of Flexible Electronics (Future Technologies) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Xikuang Yao
- School of Flexible Electronics (Future Technologies) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Wei Huang
- School of Flexible Electronics (Future Technologies) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials and Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, P. R. China
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| |
Collapse
|
31
|
He H, Du L, Xue H, An Y, Zeng K, Huang H, He Y, Zhang C, Wu J, Shuai X. Triple Tumor Microenvironment-Responsive Ferroptosis Pathways Induced by Manganese-Based Imageable Nanoenzymes for Enhanced Breast Cancer Theranostics. SMALL METHODS 2023:e2300230. [PMID: 37096886 DOI: 10.1002/smtd.202300230] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Previous studies have found that activated CD8+ T cells secrete elevated levels of interferon-gamma (IFN-γ) to trigger ferroptosis in tumor cells. However, IFN-γ-mediated ferroptosis is induced at low levels in tumor cells because of the limited IFN-γ secreted by CD8+ T cells in the immunosuppressive tumor microenvironment. Recent studies have shown that manganese ion can activate the cyclic guanosine monophosphate-adenosine monophosphate (GMP-AMP) synthase/stimulator of interferon genes (cGAS-STING) pathway and support adaptive immune responses against tumors, which enhances the level of tumor-infiltrating CD8+ T cells. Therefore, tumor microenvironment-responsive Mn-based nanoenzymes (Mn-based NEs) that activated the cGAS-STING pathway are designed to amplify immune-driven ferroptosis. The multifunctional all-in-one nanoplatform is simply and mildly synthesized by the coordination between Mn3+ ions and 3,3'-dithiodipropionic acid. After intracellular delivery, each component of Mn-based NEs exerts its function. That is, glutathione is depleted through disulfide-thiol exchange and redox pair of Mn3+ /Mn2+ , a hydroxyl radical (·OH) is generated via the Fenton-like reaction to cause ferroptosis, and Mn2+ augments cGAS-STING activity to boost immune-driven ferroptosis. In addition, ferroptosis amplifies Mn2+ -induced immunogenic cell death and initiates the antitumor immune "closed loop" along with immune-driven ferroptosis. Notably, this multifunctional nanoplatform is effective in killing both primary and distant tumors.
Collapse
Affiliation(s)
- Haozhe He
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Lihua Du
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Hongman Xue
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yongcheng An
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Kejing Zeng
- Department of Endocrinology, Department of Diabetes and Obesity Reversal Research Centre, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Huaping Huang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jun Wu
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, China
- Department of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
- Nanomedicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| |
Collapse
|
32
|
Hu B, Chen R, Jiang M, Xiong S, Xie A, Liu X, Fu B. MTX-211 Inhibits GSH Synthesis through Keap1/NRF2/GCLM Axis and Exerts Antitumor Effects in Bladder Cancer. Int J Mol Sci 2023; 24:ijms24087608. [PMID: 37108769 PMCID: PMC10142351 DOI: 10.3390/ijms24087608] [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: 02/13/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Globally, bladder cancer (BLCA) is still the leading cause of death in patients with tumors. The function and underlying mechanism of MTX-211, an EFGR and PI3K kinase inhibitor, have not been elucidated. This study examined the function of MTX-211 in BLCA cells using in vitro and in vivo assays. RNA sequencing, quantitative real-time polymerase chain reaction, Western blotting, co-immunoprecipitation, and immunofluorescence were performed to elucidate the underlying mechanism. Our observations revealed that MTX-211 has a time- and concentration-dependent inhibitory effect on bladder cancer cell proliferation. Flow cytometry analysis showed that cell apoptosis and G0/G1 cell cycle arrest were significantly induced by MTX-211. MTX-211 inhibited intracellular glutathione (GSH) metabolism, leading to a decrease in GSH levels and an increase in reactive oxygen species. GSH supplementation partly reversed the inhibitory effects of MTX-211. Further experiments verified that MTX-211 promoted NFR2 protein ubiquitinated degradation via facilitating the binding of Keap1 and NRF2, subsequently resulting in the downregulated expression of GCLM, which plays a vital role in GSH synthesis. This study provided evidence that MTX-211 effectively inhibited BLCA cell proliferation via depleting GSH levels through Keap1/NRF2/GCLM signaling pathway. Thus, MTX-211 could be a promising therapeutic agent for cancer.
Collapse
Affiliation(s)
- Bing Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang 330006, China
- Jiangxi Institute of Urology, Nanchang 430032, China
| | - Ru Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang 330006, China
- Jiangxi Institute of Urology, Nanchang 430032, China
| | - Ming Jiang
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang 330006, China
- Jiangxi Institute of Urology, Nanchang 430032, China
| | - Situ Xiong
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang 330006, China
- Jiangxi Institute of Urology, Nanchang 430032, China
| | - An Xie
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang 330006, China
- Jiangxi Institute of Urology, Nanchang 430032, China
| | - Xiaoqiang Liu
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang 330006, China
- Jiangxi Institute of Urology, Nanchang 430032, China
| | - Bin Fu
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang 330006, China
- Jiangxi Institute of Urology, Nanchang 430032, China
| |
Collapse
|
33
|
Charge-conversional click polyprodrug nanomedicine for targeted and synergistic cancer therapy. J Control Release 2023; 356:567-579. [PMID: 36924894 DOI: 10.1016/j.jconrel.2023.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/03/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Polyprodrug nanomedicines hold great potential for combating tumors. However, the functionalization of polyprodrug nanomedicines to improve therapeutic efficacy is restricted by conventional polymerization methods. Herein, we fabricated a charge-conversional click polyprodrug nanomedicine system by metal-free azide-alkyne cycloaddition click polymerization (AACCP) for targeted and synergistic cancer therapy. Specifically, Pt(IV) prodrug-backboned diazide monomer, DMC prodrug-pendent diazide monomer, dialkyne-terminated PEG monomer and azide-modified folate were click polymerized to obtain the target polyprodrug (P1). P1 could self-assemble into nano-micelles (1-NM), where PEG was the hydrophilic shell with folate on the surface, Pt(IV) and DMC prodrugs as the hydrophobic core. Taking advantage of PEGylation and folate-mediated tumor cell targeting, 1-NM achieved prolonged blood circulation time and high tumor accumulation efficiency. Tumor acidic microenvironment-responsive cleavage and cascade activation of pendant DMC prodrug induced surface charge conversion of 1-NM from negative to positive, which promoted tumor penetration and cellular internalization of the remaining 1-NM. After internalization into tumor cells, the reduction-responsive activation of Pt(IV) prodrug to Pt(II) further showed synergetic effect with DMC for enhanced apoptosis. This first designed charge-conversional click polyprodrug nanomedicine exhibited targeted and synergistic efficacy to suppress tumor proliferation in living mice bearing human ovarian tumor model.
Collapse
|
34
|
Zhu Y, Li Q, Wang C, Hao Y, Yang N, Chen M, Ji J, Feng L, Liu Z. Rational Design of Biomaterials to Potentiate Cancer Thermal Therapy. Chem Rev 2023. [PMID: 36912061 DOI: 10.1021/acs.chemrev.2c00822] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Cancer thermal therapy, also known as hyperthermia therapy, has long been exploited to eradicate mass lesions that are now defined as cancer. With the development of corresponding technologies and equipment, local hyperthermia therapies such as radiofrequency ablation, microwave ablation, and high-intensity focused ultrasound, have has been validated to effectively ablate tumors in modern clinical practice. However, they still face many shortcomings, including nonspecific damages to adjacent normal tissues and incomplete ablation particularly for large tumors, restricting their wide clinical usage. Attributed to their versatile physiochemical properties, biomaterials have been specially designed to potentiate local hyperthermia treatments according to their unique working principles. Meanwhile, biomaterial-based delivery systems are able to bridge hyperthermia therapies with other types of treatment strategies such as chemotherapy, radiotherapy and immunotherapy. Therefore, in this review, we discuss recent progress in the development of functional biomaterials to reinforce local hyperthermia by functioning as thermal sensitizers to endow more efficient tumor-localized thermal ablation and/or as delivery vehicles to synergize with other therapeutic modalities for combined cancer treatments. Thereafter, we provide a critical perspective on the further development of biomaterial-assisted local hyperthermia toward clinical applications.
Collapse
Affiliation(s)
- Yujie Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Quguang Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Chunjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Yu Hao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| |
Collapse
|
35
|
Sun T, Jiang C. Stimuli-responsive drug delivery systems triggered by intracellular or subcellular microenvironments. Adv Drug Deliv Rev 2023; 196:114773. [PMID: 36906230 DOI: 10.1016/j.addr.2023.114773] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
Drug delivery systems (DDS) triggered by local microenvironment represents the state-of-art of nanomedicine design, where the triggering hallmarks at intracellular and subcellular levels could be employed to exquisitely recognize the diseased sites, reduce side effects, and expand the therapeutic window by precisely tailoring the drug-release kinetics. Though with impressive progress, the DDS design functioning at microcosmic levels is fully challenging and underexploited. Here, we provide an overview describing the recent advances on stimuli-responsive DDSs triggered by intracellular or subcellular microenvironments. Instead of focusing on the targeting strategies as listed in previous reviews, we herein mainly highlight the concept, design, preparation and applications of stimuli-responsive systems in intracellular models. Hopefully, this review could give useful hints in developing nanoplatforms proceeding at a cellular level.
Collapse
Affiliation(s)
- Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China.
| |
Collapse
|
36
|
Chen T, Chen J, Zeng T, Huang Q, Chen D, Chen H, Chen J, Zheng B, Wang M, Chen S, Dai J, Sun H, Chen T, Huang Y, Zhao L, Ma S, Liu X. WZ35 inhibits gastric cancer cell metastasis by depleting glutathione to promote cellular metabolic remodeling. Cancer Lett 2023; 555:216044. [PMID: 36574880 DOI: 10.1016/j.canlet.2022.216044] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
This study aimed at elucidating the crosstalk between redox reaction and metabolic remodeling through uncovering the mechanism underlying WZ35-mediated reactive oxygen species (ROS) production and regulation of amino acid metabolism to inhibit gastric cancer (GC) cell metastasis. The activity and biosafety of curcumin analog, WZ35, were verified in vitro and in vivo. The potential molecular mechanism underlying WZ35-mediated enhanced radiotherapeutic sensitivity by reduced Glutathione (GSH) depletion was elucidated by RNA sequencing, single-cell sequencing (scRNA-seq), metabolic mass spectrometry, and other molecular experiments. Compared to curcumin, WZ35 proved more potent anti-proliferative and anti-metastasis properties. Importantly, we demonstrated that WZ35 could consume GSH in multiple ways, including by reduction of raw materials and consumption reserves, inhibition of reformation, and enhanced decomposition. Mechanistically, we identify that WZ35 maintains the GSH depletion phenotype through the ROS-YAP-AXL-ALKBH5-GLS2 loop, further backing the relevance of metabolic remodeling in the tumor microenvironment with tumor metastasis and the role of m6A in tumor metastasis. Collectively, our study identified WZ35 as a novel GSH depletion agent and a previously undiscovered GSH depletion loop mechanism in GC cell metastasis.
Collapse
Affiliation(s)
- Tongke Chen
- Laboratory Animal Centre, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Junbo Chen
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Tianni Zeng
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China
| | - Qianying Huang
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Di Chen
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hong Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiayao Chen
- Wenzhou Medical University Renji College, Wenzhou, 325035, Zhejiang Province, China
| | - Bin Zheng
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengting Wang
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Shinuo Chen
- Laboratory Animal Centre, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jichen Dai
- Second Medical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hanxiao Sun
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tongzuan Chen
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, 325000, China
| | - Yuwen Huang
- Laboratory Animal Centre, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Liqian Zhao
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Shumei Ma
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Xiaodong Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
37
|
Wang W, Wang W, Jin S, Fu F, Huang Z, Huang Y, Wu C, Pan X. Open pocket and tighten holes: Inhalable lung cancer-targeted nanocomposite for enhanced ferroptosis-apoptosis synergetic therapy. CHEMICAL ENGINEERING JOURNAL 2023; 458:141487. [DOI: 10.1016/j.cej.2023.141487] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
|
38
|
Zhang Z, Yan W, Ji Y. A novel manganese dioxide-based drug delivery strategy via in situ coating γ-polyglutamic acid/cisplatin for intelligent anticancer therapy. J Mater Chem B 2023; 11:667-674. [PMID: 36541339 DOI: 10.1039/d2tb01659a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cisplatin (CDDP) is one of the most frequently used chemotherapeutic drugs due to its broad-spectrum and potent antitumor activity. Unfortunately, inactivation due to glutathione (GSH) substances and insufficient cellular uptake of CDDP greatly hinder its clinical applications. Herein, manganese dioxide (MnO2) was reported as an efficient glutathione (GSH) consumption material for promoting the accumulation and preventing premature leakage of CDDP in tumor cells. In this work, γ-polyglutamic acid/cisplatin (PGA/CDDP) conjugates and PGA/CDDP nanoparticles (NPs) were respectively constructed via the ligand exchange reaction and electrostatic interaction. Furthermore, PGA/CDDP NPs were in situ coated with MnO2 (PGA/CDDP@MnO2 NPs) through the redox reaction of the residual carboxyl group (-COOH) and potassium permanganate (KMnO4). As a result, the PGA/CDDP@MnO2 NPs achieved a satisfactory drug-loading efficiency (ca. 37.26%) and multi-responsive controlled drug release. Remarkably, the MnO2 shells exhibited excellent performance for efficient glutathione (GSH) consumption and significantly enhanced the killing effect (ca. 2-3 times) in human lung cancer cells (A549) compared with pure CDDP. Moreover, it was observed that PGA/CDDP@MnO2 NPs could also inhibit the migration and invasion of A549 cells. Overall, these remarkable performances of PGA/CDDP@MnO2 NPs make MnO2 promising for controlled drug release and intelligent anticancer therapy.
Collapse
Affiliation(s)
- Zheng Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Weichen Yan
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
39
|
Xu J, Zhang J, Song J, Liu Y, Li J, Wang X, Tang R. Construction of multifunctional mesoporous silicon nano-drug delivery system and study of dual sensitization of chemo-photodynamic therapy in vitro and in vivo. J Colloid Interface Sci 2022; 628:271-285. [PMID: 35998453 DOI: 10.1016/j.jcis.2022.08.100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022]
Abstract
This study was conducted to construct a multifunctional nanodrug delivery system (NDDS) to deplete glutathione (GSH) in tumor cells and amplify oxidative stress, enhancing the synergistic effect of chemotherapy and photodynamic therapy (PDT). l-Buthionine-sulfoximine (BSO) and chlorin e6 (Ce6) were loaded into mesoporous silicon nanoparticles (MSN), and then MSN were modified with oxidized hyaluronic acid (OHA) as a pore-blocking agent. Cisplatin (Pt(II)) was further loaded by a coordination reaction with carboxyl groups in OHA to yield a multifunctional NDDS (denoted as MSN@OHA-Ce6/BSO/Pt). The physicochemical properties and antitumor activity of the prepared nanoparticles were characterized in detail. In vitro and in vivo experiments demonstrated that OHA was shed from MSN@OHA-Ce6/BSO/Pt under acidic conditions in tumors, resulting in the release of free BSO, Ce6, and Pt(II). The released BSO could reduce intracellular GSH expression by 48.8 %, effectively enhancing the PDT effect of Ce6 and the chemotherapy effect of Pt(II). Finally, the tumor inhibitory rate (vs saline) reached 73.8 % ± 2.5 % for MSN@OHA-Ce6/BSO/Pt in A549/DDP tumor-bearing nude mice. Therefore, the multifunctional NDDS significantly enhanced the synergistic effect of PDT and chemotherapy.
Collapse
Affiliation(s)
- Jiaxi Xu
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Jingwen Zhang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Jinping Song
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Yufei Liu
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Junnan Li
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China.
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China.
| |
Collapse
|
40
|
Tang Z, Tian W, Long H, Jiang S, Zhao J, Zhou J, He Q, Luo X. Subcellular-Targeted Near-Infrared-Responsive Nanomedicine with Synergistic Chemo-photothermal Therapy against Multidrug Resistant Cancer. Mol Pharm 2022; 19:4538-4551. [PMID: 35311257 DOI: 10.1021/acs.molpharmaceut.1c00998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) is a major obstacle to effective cancer treatment. Therefore, developing effective approaches for overcoming the limitation of MDR in cancer therapy is very essential. Chemotherapy combined with photothermal therapy (PTT) is a potential therapeutic option against MDR. Herein, we developed a subcellular-targeted near-infrared (NIR)-responsive nanomedicine (Fe3O4@PDA-TPP/S2-PEG-hyd-DOX, abbreviated as Fe3O4-ATSPD) as a new photothermal agent with improved photothermal stability and efficiency. This system demonstrates high stability in blood circulation and can be accumulated at the tumor site by magnetic targeting enhanced permeability and retention effect (EPR). Near-infrared (NIR) irradiation at the tumor site generates a photothermal effect from the photosensitizer Fe3O4@PDA, leading to a dramatic decrease in mitochondrial membrane potential. Simultaneously, the conjugated drugs released under low pH condition in endosomes or lysosomes cause nucleus DNA damage and cell apoptosis. This subcellular-targeted NIR-responsive nanomedicine with efficient integration of diagnosis and therapy could significantly enhance MDR cancer treatment by combination of chemotherapy and PTT.
Collapse
Affiliation(s)
- Zhaomin Tang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Weijun Tian
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Hongyu Long
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Shuting Jiang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Jianqing Zhao
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Jianren Zhou
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Qian He
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| | - Xia Luo
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, China
| |
Collapse
|
41
|
Zeng Z, Fu C, Sun X, Niu M, Ren X, Tan L, Wu Q, Huang Z, Meng X. Reversing the immunosuppressive microenvironment with reduced redox level by microwave-chemo-immunostimulant Ce-Mn MOF for improved immunotherapy. J Nanobiotechnology 2022; 20:512. [PMID: 36463157 PMCID: PMC9719648 DOI: 10.1186/s12951-022-01699-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 11/05/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUNDS Reversing the immunosuppressive tumor microenvironment (TME) in the tumor is widely deemed to be an effective strategy to improve immune therapy. In particular, the redox balance in TME needs to be well controlled due to its critical role in mediating the functions of various cells, including cancer cells and immune-suppressive cells. RESULTS Here, we propose an efficient strategy to reshape the redox homeostasis to reverse immunosuppressive TME. Specifically, we developed a microwave-chemo-immunostimulant CMMCP to promote the infiltration of the tumor-T cells by simultaneously reducing the reactive oxygen species (ROS) and glutathione (GSH) and improving the oxygen (O2) levels in TME. The CMMCP was designed by loading chemotherapy drugs cisplatin into the bimetallic Ce-Mn MOF nanoparticles coated with polydopamine. The Ce-Mn MOF nanoparticles can effectively improve the catalytic decomposition of ROS into O2 under microwave irradiation, resulting in overcoming hypoxia and limited ROS generation. Besides, the activity of intracellular GSH in TME was reduced by the redox reaction with Ce-Mn MOF nanoparticles. The reprogrammed TME not only boosts the immunogenic cell death (ICD) induced by cisplatin and microwave hyperthermia but also gives rise to the polarization of pro-tumor M2-type macrophages to the anti-tumor M1-type ones. CONCLUSION Our in vivo experimental results demonstrate that the microwave-chemo-immunostimulant CMMCP significantly enhances the T cell infiltration and thus improves the antitumor effect. This study presents an easy, safe, and effective strategy for a whole-body antitumor effect after local treatment.
Collapse
Affiliation(s)
- Zhiheng Zeng
- grid.9227.e0000000119573309Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190 China ,grid.458502.e0000 0004 0644 7196CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Beijing, 100190 China ,grid.13291.380000 0001 0807 1581College of Biomedical Engineering, Sichuan University, Chengdu, 610065 China
| | - Changhui Fu
- grid.9227.e0000000119573309Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190 China ,grid.458502.e0000 0004 0644 7196CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Beijing, 100190 China
| | - Xiaohan Sun
- grid.412636.40000 0004 1757 9485Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang, 110000 China
| | - Meng Niu
- grid.412636.40000 0004 1757 9485Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang, 110000 China
| | - Xiangling Ren
- grid.9227.e0000000119573309Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190 China ,grid.458502.e0000 0004 0644 7196CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Beijing, 100190 China
| | - Longfei Tan
- grid.9227.e0000000119573309Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190 China ,grid.458502.e0000 0004 0644 7196CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Beijing, 100190 China
| | - Qiong Wu
- grid.9227.e0000000119573309Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190 China ,grid.458502.e0000 0004 0644 7196CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Beijing, 100190 China
| | - Zhongbing Huang
- grid.13291.380000 0001 0807 1581College of Biomedical Engineering, Sichuan University, Chengdu, 610065 China
| | - Xianwei Meng
- grid.9227.e0000000119573309Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190 China ,grid.458502.e0000 0004 0644 7196CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Beijing, 100190 China
| |
Collapse
|
42
|
Self-assembled nano-photosensitizer for targeted, activatable, and biosafe cancer phototheranostics. Biomaterials 2022; 291:121916. [PMID: 36410110 DOI: 10.1016/j.biomaterials.2022.121916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/25/2022] [Accepted: 11/13/2022] [Indexed: 11/17/2022]
Abstract
Cancer treatment currently still faces crucial challenges in therapeutic effectiveness, precision, and complexity. Photodynamic therapy (PDT) as a non-invasive tactic has earned widespread popularity for its excellent therapeutic output, flexibility, and restrained toxicity. Nonetheless, drawbacks, including low efficiency, poor cancer specificity, and limited therapeutic depth, remain considerable during the cancer treatment. Although great effort has been made to improve the performance, the overall efficiency and biosafety are still ambiguous and unable to meet urgent clinical needs. Herein, this study integrates merits from previous PDT strategies and develops a cancer-targeting, activatable, biosafe photosensitizer. Owing to excellent self-assembly ability, this photosensitizer can be conveniently prepared as multifunctional nano-photosensitizers, namely MBNPs, and applied to in vivo cancer phototheranostics in "all-in-one" mode. This study successfully verifies the mechanism of MBNPs, then deploys them to cell-based and in vivo cancer PDT. Based on the unique cancer microenvironment, MBNPs achieve precise distribution, accumulation, and activation towards the tumor, releasing methylene blue as a potent photosensitizer for phototherapy. The PDT outcome demonstrates MBNPs' superior cancer specificity, remarkable PDT efficacy, and negligible toxicity. Meanwhile, in vivo NIR fluorescence and photoacoustic imaging have been utilized to guide the PDT treatment synergistically. Additionally, the biosafety of the MBNPs-based PDT treatment is ensured, thus providing potential for future clinical studies.
Collapse
|
43
|
Davoudi M, Jadidi Y, Moayedi K, Farrokhi V, Afrisham R. Ameliorative impacts of polymeric and metallic nanoparticles on cisplatin-induced nephrotoxicity: a 2011-2022 review. J Nanobiotechnology 2022; 20:504. [PMID: 36457031 PMCID: PMC9714065 DOI: 10.1186/s12951-022-01718-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Cisplatin (CDDP) is a well-known platinum-based drug used in the treatment of various malignancies. However, the widespread side effects that this drug leaves on normal tissues make its use limited. Since cisplatin is mainly eliminated from the kidneys, CDDP-induced nephrotoxicity is the most significant dose-limiting complication attributed to cisplatin, which often leads to dose withdrawal. Considering the high efficiency of cisplatin in chemotherapy, finding renoprotective drug delivery systems for this drug is a necessity. In this regard, we can take advantages of different nanoparticle-based approaches to deliver cisplatin into tumors either using passive targeting or using specific receptors. In an effort to find more effective cisplatin-based nano-drugs with less nephrotoxic effect, the current 2011-2022 review study was conducted to investigate some of the nanotechnology-based methods that have successfully been able to mitigate CDDP-induced nephrotoxicity. Accordingly, although cisplatin can cause renal failures through inducing mitochondria dysfunction, oxidative stress, lipid peroxidation and endoplasmic reticulum stress, some CDDP-based nano-carriers have been able to reverse a wide range of these advert effects. Based on the obtained results, it was found that the use of different metallic and polymeric nanoparticles can help renal cells to strengthen their antioxidant systems and stay alive through reducing CDDP-induced ROS generation, inhibiting apoptosis-related pathways and maintaining the integrity of the mitochondrial membrane. For example, nanocurcumin could inhibit oxidative stress and acting as a ROS scavenger. CONPs could reduce lipid peroxidation and pro-inflammatory cytokines. CDDP-loaded silver nanoparticles (AgNPs) could inhibit mitochondria-mediated apoptosis. In addition, tea polyphenol-functionalized SeNPs (Se@TE) NPs could mitigate the increased level of dephosphorylated AKT, phosphorylated p38 MAPK and phosphorylated c-Jun N-terminal kinase (JNK) induced by cisplatin. Moreover, exosomes mitigated cisplatin-induced renal damage through inhibiting Bcl2 and increasing Bim, Bid, Bax, cleaved caspase-9, and cleaved caspase-3. Hence, nanoparticle-based techniques are promising drug delivery systems for cisplatin so that some of them, such as lipoplatins and nanocurcumins, have even reached phases 1-3 trials.
Collapse
Affiliation(s)
- Maryam Davoudi
- grid.411705.60000 0001 0166 0922Department of Clinical Laboratory Sciences, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Jadidi
- grid.411705.60000 0001 0166 0922Department of Clinical Laboratory Sciences, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiana Moayedi
- grid.411705.60000 0001 0166 0922Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vida Farrokhi
- grid.411705.60000 0001 0166 0922Department of Hematology, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Afrisham
- grid.411705.60000 0001 0166 0922Department of Clinical Laboratory Sciences, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
A nuclease-mimetic platinum nanozyme induces concurrent DNA platination and oxidative cleavage to overcome cancer drug resistance. Nat Commun 2022; 13:7361. [PMID: 36450764 PMCID: PMC9712435 DOI: 10.1038/s41467-022-35022-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Platinum (Pt) resistance in cancer almost inevitably occurs during clinical Pt-based chemotherapy. The spontaneous nucleotide-excision repair of cancer cells is a representative process that leads to Pt resistance, which involves the local DNA bending to facilitate the recruitment of nucleotide-excision repair proteins and subsequent elimination of Pt-DNA adducts. By exploiting the structural vulnerability of this process, we herein report a nuclease-mimetic Pt nanozyme that can target cancer cell nuclei and induce concurrent DNA platination and oxidative cleavage to overcome Pt drug resistance. We show that the Pt nanozyme, unlike cisplatin and conventional Pt nanoparticles, specifically induces the nanozyme-catalyzed cleavage of the formed Pt-DNA adducts by generating in situ reactive oxygen species, which impairs the damage recognition factors-induced DNA bending prerequisite for nucleotide-excision repair. The recruitment of downstream effectors of nucleotide-excision repair to DNA lesion sites, including xeroderma pigmentosum groups A and F, is disrupted by the Pt nanozyme in cisplatin-resistant cancer cells, allowing excessive accumulation of the Pt-DNA adducts for highly efficient cancer therapy. Our study highlights the potential benefits of applying enzymatic activities to the use of the Pt nanomedicines, providing a paradigm shift in DNA damaging chemotherapy.
Collapse
|
45
|
A nanodiamond chemotherapeutic folate receptor-targeting prodrug with triggerable drug release. Int J Pharm 2022; 630:122432. [PMID: 36435503 DOI: 10.1016/j.ijpharm.2022.122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/26/2022]
Abstract
Cancer chemotherapy is often accompanied by severe off-target effects that both damage quality of life and can decrease therapeutic compliance. This could be minimized through selective delivery of cytotoxic agents directly to the cancer cells. This would decrease the drug dose, consequently minimizing side effects and cost. With this goal in mind, a dual-gated folate-functionalized nanodiamond drug delivery system (NPFSSD) for doxorubicin with activatable fluorescence and cytotoxicity has been prepared. Both the cytotoxic activity and the fluorescence of doxorubicin (DOX) are quenched when it is covalently immobilized on the nanodiamond. The NPFSSD is preferentially uptaken by cancer cells overexpressing the folate receptor. Then, once inside a cell, the drug is preferentially released within tumor cells due to their high levels of endogenous of glutathione, required for releasing DOX through cleavage of a disulfide linker. Interestingly, once free DOX is loaded onto the nanodiamond, it can also evade resistance mechanisms that use protein pumps to remove drugs from the cytoplasm. This nanodrug, used in an in vivo model with local injection of drugs, effectively inhibits tumor growth with fewer side effects than direct injection of free DOX, providing a potentially powerful platform to improve therapeutic outcomes.
Collapse
|
46
|
Zeng Q, Li X, Xie S, Xing D, Zhang T. Specific disruption of glutathione-defense system with activatable single molecule-assembled nanoprodrug for boosted photodynamic/chemotherapy eradication of drug-resistant tumors. Biomaterials 2022; 290:121867. [DOI: 10.1016/j.biomaterials.2022.121867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/02/2022]
|
47
|
Lin C, Chen Y, Zhu M, Pei J, Zhou Y, Gou M, Ouyang L. A sulfhydryl blocking reagent BT-4 sensitizes cisplatin-based micelle prodrugs for efficient treatment of breast cancer. Int J Pharm 2022; 626:122187. [PMID: 36100145 DOI: 10.1016/j.ijpharm.2022.122187] [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: 05/01/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 10/14/2022]
Abstract
Detoxification of glutathione (GSH) and insufficient cellular uptake of cisplatin (CDDP) severely compromised the therapeutic efficacy of CDDP. Here, a nano-delivery system (BT-4@PtPPNPs) for CDDP prodrug (C16-Pt(Ⅳ)-PEG) based on a novel sulfhydryl blocking reagent methyl 2-(methylsulfonyl) benzothiazole-6-carboxylate (BT-4) was developed. On the one hand, BT-4 can deplete GSH in tumor cells by directly interacting with reactive sulfhydryl group on GSH, thereby increasing the cytotoxicity of CDDP. On the other hand, the CDDP prodrug carrier C16-Pt(IV)-PEG can promote the distribution of CDDP in tumors, reduce the probability of unexpected inactivation of CDDP, and reduce the content of GSH in tumor cells during the conversion to CDDP, thereby making CDDP more effective for treatment. The results showed that the optimized BT-4@PtPPNPs with a small particle size (130 nm) exhibited notable cytotoxicity and apoptosis of 4T1 cells. BT-4@PtPPNPs not only significantly improved the uptake of drugs by tumor cells, but also rapidly targeted and accumulated in the tumors for a long time. Moreover, in vivo efficacy studies showed that BT-4@PtPPNPs could effectively inhibit tumor growth, inhibiting 60.85 % of tumors in a 4T1 breast cancer mice model, showing superior antitumor activity, which can be attributed to GSH-triggered CDDP tolerance reversal. Overall, this study provides an attractive and simple strategy to combine novel sulfhydryl blockers and CDDP prodrugs to potentiate the efficacy of CDDP in breast cancer.
Collapse
Affiliation(s)
- Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Yuxiu Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Mengli Zhu
- Core Facilities of West China Hospital, Chengdu 610041, China.
| | - Junping Pei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Yang Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
48
|
Biodegradable disulfide crosslinked chitosan/stearic acid nanoparticles for dual drug delivery for colorectal cancer. Carbohydr Polym 2022; 294:119833. [PMID: 35868778 DOI: 10.1016/j.carbpol.2022.119833] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/01/2022] [Accepted: 07/02/2022] [Indexed: 01/12/2023]
Abstract
Herein, redox responsive chitosan/stearic acid nanoparticles (CSSA NPs) (≈200 nm) are developed for dual drug delivery. These degradable nanoparticles are prepared based on disulfide (SS) crosslinking chemistry avoiding the use of any external crosslinking agent. CSSA NPs are further loaded with both DOX (hydrophilic) and curcumin (hydrophobic) drugs with ≈86 % and ≈82 % encapsulation efficiency respectively. This approach of combining anticancer therapeutics having different mode of anticancer action allows to develop systems for cancer therapy with enhanced efficacy. In vitro drug release experiments clearly exhibit the low leakage of drug under physiological conditions while ≈98 % DOX and ≈96 % curcumin is released after 136 h under GSH reducing conditions. The cytotoxicity experiments against HCT116 cells demonstrate higher cytotoxicity of dual drug loaded CSSA NPs. In vivo biodistribution experiments with c57bl/6j mice confirms the retention of CSSA NPs in the colon area up to 24 h exhibiting their potential for colorectal cancer therapy.
Collapse
|
49
|
Hu H, Zhang Z, Fang Y, Chen L, Wu J. Therapeutic poly(amino acid)s as drug carriers for cancer therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
50
|
Zhang R, You X, Luo M, Zhang X, Fang Y, Huang H, Kang Y, Wu J. Poly(β-cyclodextrin)/platinum prodrug supramolecular nano system for enhanced cancer therapy: Synthesis and in vivo study. Carbohydr Polym 2022; 292:119695. [PMID: 35725183 DOI: 10.1016/j.carbpol.2022.119695] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/02/2022]
Abstract
The use of cisplatin is restricted by systemic toxicity and drug resistance. Supramolecular nano-drug delivery systems involving drugs as building blocks circumvent these limitations promisingly. Herein, we describe a novel supramolecular system [Pt(IV)-SSNPs] based on poly(β-cyclodextrin), which was synthesized for efficient loading of adamantly-functionalized platinum(IV) prodrug [Pt(IV)-ADA2] via the host-guest interaction between β-cyclodextrin and adamantyl. Pt(IV)-ADA2 can be converted to active cisplatin in reducing environment in cancer cells, which further reduces systemic toxicity. The introduction of the adamantane group-tethered mPEG2k endowed the Pt(IV)-SSNPs with a longer blood circulation time. In vitro assays exhibited that the Pt(IV)-SSNPs could be uptaken by CT26 cells, resulting in cell cycle arrest in the G2/M and S phases, together with apoptosis. Furthermore, the Pt(IV)-SSNPs showed effective tumor accumulation, better antitumor effect, and negligible cytotoxicity to major organs. These results indicate that supramolecular nanoparticles are a promising platform for efficient cisplatin delivery and cancer treatment.
Collapse
Affiliation(s)
- Ruhe Zhang
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China
| | - Xinru You
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Moucheng Luo
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China
| | - Xinyu Zhang
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China
| | - Yifen Fang
- Department of Cardiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jun Wu
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|