1
|
He X, Tian Y, Dong J, Yuan Y, Zhang S, Jing H. RNA-Seq Reveals the Mechanism of Pyroptosis Induced by Oxygen-Enriched IR780 Nanobubbles-Mediated Sono-Photodynamic Therapy. Int J Nanomedicine 2024; 19:13029-13045. [PMID: 39654803 PMCID: PMC11625641 DOI: 10.2147/ijn.s487412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Background Sono-photodynamic therapy (SPDT), the combination of sonodynamic therapy (SDT) and photodynamic therapy (PDT), is a promising tumor treatment method. However, the hypoxic tumor microenvironment greatly compromises the efficacy of SPDT. Pyroptosis, a new type of programmed cell death, is mainly induced by some chemotherapeutic drugs in the current research, and rarely by SPDT. RNA sequencing (RNA-seq) is a high-throughput sequencing technique that comprehensively profiles the transcriptome, revealing the full spectrum of RNA molecules in a cell. Here, we constructed IR780@O2 nanobubbles (NBs) with photoacoustic dual response and hypoxia improvement properties to fight triple negative breast cancer (TNBC), and demonstrated that SPDT could kill TNBC cells through pyroptosis pathway. RNA-seq further revealed potential mechanisms and related differentially expressed genes. Methods Thin-film hydration and mechanical vibration method were utilized to synthesize IR780@O2 NBs. Subsequently, we characterized IR780@O2 NBs and examined the cytotoxicity as well as ROS production ability. A series of experiments were conducted to verify that SPDT killed TNBC cells through pyroptosis. Results IR780@O2 NBs were successfully prepared and had certain stability. Compared with SDT alone, SPDT increased therapeutic effect by 1.67 times by generating more ROS, and the introduction of NBs and O2 NBs (2.23 times and 2.93 times compared with SDT alone) could further promote this process. Other experiments proved that TNBC cells died by pyroptosis pathway. Moreover, the in-depth mechanism revealed that colony stimulating factor (CSF) and C-X-C motif chemokine ligand (CXCL) could be potential targets for the occurrence of pyroptosis in TNBC cells. Conclusion The IR780@O2 NBs prepared in this study increased the degree of TNBC cell pyroptosis through SPDT effect and alleviation of hypoxia, and cellular senescence might be a biological process closely related to pyroptosis in TNBC.
Collapse
Affiliation(s)
- Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Jialin Dong
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Shijie Zhang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
2
|
Eker F, Akdaşçi E, Duman H, Bechelany M, Karav S. Gold Nanoparticles in Nanomedicine: Unique Properties and Therapeutic Potential. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1854. [PMID: 39591094 PMCID: PMC11597456 DOI: 10.3390/nano14221854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
Gold nanoparticles (NPs) have demonstrated significance in several important fields, including drug delivery and anticancer research, due to their unique properties. Gold NPs possess significant optical characteristics that enhance their application in biosensor development for diagnosis, in photothermal and photodynamic therapies for anticancer treatment, and in targeted drug delivery and bioimaging. The broad surface modification possibilities of gold NPs have been utilized in the delivery of various molecules, including nucleic acids, drugs, and proteins. Moreover, gold NPs possess strong localized surface plasmon resonance (LSPR) properties, facilitating their use in surface-enhanced Raman scattering for precise and efficient biomolecule detection. These optical properties are extensively utilized in anticancer research. Both photothermal and photodynamic therapies show significant results in anticancer treatments using gold NPs. Additionally, the properties of gold NPs demonstrate potential in other biological areas, particularly in antimicrobial activity. In addition to delivering antigens, peptides, and antibiotics to enhance antimicrobial activity, gold NPs can penetrate cell membranes and induce apoptosis through various intracellular mechanisms. Among other types of metal NPs, gold NPs show more tolerable toxicity capacity, supporting their application in wide-ranging areas. Gold NPs hold a special position in nanomaterial research, offering limited toxicity and unique properties. This review aims to address recently highlighted applications and the current status of gold NP research and to discuss their future in nanomedicine.
Collapse
Affiliation(s)
- Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey; (F.E.); (E.A.); (H.D.)
| | - Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey; (F.E.); (E.A.); (H.D.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey; (F.E.); (E.A.); (H.D.)
| | - Mikhael Bechelany
- Institut Européen des Membranes (IEM), UMR 5635, University Montpellier, ENSCM, CNRS, F-34095 Montpellier, France
- Functional Materials Group, Gulf University for Science and Technology (GUST), Masjid Al Aqsa Street, Mubarak Al-Abdullah 32093, Kuwait
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey; (F.E.); (E.A.); (H.D.)
| |
Collapse
|
3
|
Zhao L, Liu Y, Jin F, Hu K, Lv M, Zhou Y, Zhao W, Hu Y, Wu J, Yang Y, Wang W. Multifunctional nanoparticles potentiate in-situ tumor vaccines via reversing insufficient Photothermal therapy by disrupting tumor vasculature. J Control Release 2024; 376:842-860. [PMID: 39401677 DOI: 10.1016/j.jconrel.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
Photothermal therapy can trigger immunogenic cell death and release personalized in-situ tumor vaccine, activating immune responses to eliminate systemic tumors beyond the irradiated zone. However, the immune response of the in-situ tumor vaccines is often undermined by the residual tumor cells and their induced immunosuppressive tumor microenvironment (TME), which is attributed to insufficient photothermal effects stemming from the limited accumulation of photosensitizers. To overcome these limitations, we developed multi-functional nanoparticles (VI@Gd-NPs) that integrate a tumor vasculature-specific disrupting agent (Vadimezan, Phase III clinical drug), a photosensitizer (Indocyanine Green, ICG), and a magnetic resonance imaging contrast agent (Gadolinium, Gd) through chemical self-assembly. By selectively disrupting the tumor vasculature, these nanoparticles enhance the intratumoral delivery of photosensitizers (ICG and blood cells), and Gd. With the guidance of Gd-enhanced MRI, the improved delivery facilitates comprehensive photothermal ablation and regulates the TME, further initiating the in-situ tumor vaccine. Notably, this approach significantly enhances anti-tumor immune responses, improves survival rates, and reduces tumor recurrence and metastasis in various animal models. Moreover, depleting CD8+ T cells reverses these therapeutic benefits, highlighting the critical role of adaptive T cell immunity. Therefore, the VI@Gd-NPs treatment holds great potential for reigniting the in-situ tumor vaccine of photothermal therapy.
Collapse
Affiliation(s)
- Lili Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiran Liu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Fangfei Jin
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Kaiyuan Hu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Miao Lv
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yuehua Zhou
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Weijun Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yong Yang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Wenguang Wang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
4
|
Iqbal S, Chen X, Sohail M, Wu F, Fang S, Ma J, Wang H, Zhao Z, Shu G, Chen M, Du YZ, Ji J. Self-targeted smart polyester nanoparticles for simultaneous Delivery of photothermal and chemotherapeutic agents for efficient treatment of HCC. Biomater Sci 2024. [PMID: 39494574 DOI: 10.1039/d4bm01120a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Advances in nanotechnology offer promising strategies to overcome the limitations of single-drug therapies in hepatocellular carcinoma (HCC) and other cancers such as multidrug resistance and variable drug tolerances. This study proposes a targeted nanoparticle system based on a poly(β-aminoester) (PβAE) core and a hyaluronic acid (HA) shell, designed for the codelivery of doxorubicin (DOX) and indocyanine green (ICG) to effectively treat HCC. These nanoparticles demonstrated remarkable physicochemical and colloidal stability, pH- and temperature-responsive release, enhanced cellular uptake, and drug retention within tumors. Upon near-infrared (NIR) irradiation, the photothermal conversion of ICG elevated local tumor temperatures up to 53.6 °C, enhancing apoptotic cell death significantly compared to chemotherapy alone (p < 0.05). Furthermore, the dual delivery system significantly enhanced therapeutic efficacy, as evidenced by a marked decrease in tumor growth in vivo compared to controls (p < 0.01). These findings illustrate that the HA/PβAE/DOX/ICG nanoparticles are not only able to precisely target tumor cells but also overcome the limitations associated with traditional chemotherapies and photothermal treatments, suggesting a promising avenue for clinical translation of cancer therapy.
Collapse
Affiliation(s)
- Sajid Iqbal
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Xiaoxiao Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Muhammad Sohail
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Ji Ma
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Haiyong Wang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Gaofeng Shu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, China
| |
Collapse
|
5
|
Javid H, Oryani MA, Rezagholinejad N, Hashemzadeh A, Karimi-Shahri M. Unlocking the potential of RGD-conjugated gold nanoparticles: a new frontier in targeted cancer therapy, imaging, and metastasis inhibition. J Mater Chem B 2024; 12:10786-10817. [PMID: 39351647 DOI: 10.1039/d4tb00281d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
In the rapidly evolving field of cancer therapeutics, the potential of gold nanoparticles (AuNPs) conjugated with RGD peptides has emerged as a promising avenue for targeted therapy and imaging. Despite numerous studies demonstrating the effectiveness of RGD-conjugated AuNPs in specifically targeting tumor cells and enhancing radiation therapy (RT), a comprehensive review of these advancements is currently lacking. This review aims to fill this critical gap in the literature. Our analysis reveals that RGD-conjugated AuNPs have shown significant promise in improving the diagnosis and treatment of various types of cancer, including breast cancer. However, the full potential of this technology is yet to be realized. The development of multifunctional nanoplatforms incorporating AuNPs has opened new horizons for targeted therapy, dual-mode imaging, and inhibition of tumor growth and metastasis. This review is of paramount importance as it provides a comprehensive overview of the current state of research in this area, and highlights the areas where further research is needed. It is hoped that this review will inspire further investigations into this promising nanotechnology, ultimately leading to improved cancer diagnosis and therapy. Therefore, the findings presented in this review underscore the potential of AuNPs conjugated with RGD peptides as a revolutionary approach in cancer therapeutics. It is our fervent hope that this review will serve as a catalyst for further research in this exciting field.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
6
|
You W, Cai Z, Xiao F, Zhao J, Yu X, Wang W, Chen Z, Hu W, Sun G, Wang Z. Local delivery of MoS2/FeS2 heterojunction by biomolecular microneedles for multimodal therapy of infected wounds. CHEMICAL ENGINEERING JOURNAL 2024; 498:155722. [DOI: 10.1016/j.cej.2024.155722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
|
7
|
Fan W, He Y, Hu P, Liu L, Yang X, Ge T, Jin K, Mou X, Cai Y. A novel acceptor-donor-acceptor structured molecule-based nanosystem for tumor mild photothermal therapy. J Colloid Interface Sci 2024; 670:762-773. [PMID: 38788443 DOI: 10.1016/j.jcis.2024.05.143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Although photothermal therapy (PTT) is effective at killing tumor cells, it can inadvertently damage healthy tissues surrounding the tumor. Nevertheless, lowering the treatment temperature will reduce the therapeutic effectiveness. In this study, we employed 2,2'-((2Z,2'Z)-((4,4,9,9-Tetrahexyl-4,9-dihydro-s-indaceno[1,2-b:5,6-b']dithiophene-2,7-diyl)bis(methanylylidene))bis(3-oxo-2,3-dihydro-1H-indene-2,1-diylidene)) dimalononitrile (IDIC), a molecule possessing a conventional acceptor-donor-acceptor (A-D-A) structure, as a photothermal agent (PTA) to facilitate effective mild photothermal therapy (mPTT). IDIC promotes intramolecular charge transfer under laser irradiation, making it a promising candidate for mPTT. To enhance the therapeutic potential of IDIC, we incorporated quercetin (Qu) into IDIC to form IDIC-Qu nanoparticles (NPs), which can inhibit heat shock protein (HSP) activity during the process of mPTT. Moreover, IDIC-Qu NPs exhibited exceptional water dispersibility and passive targeting abilities towards tumor tissues, attributed to its enhanced permeation and retention (EPR) effect. These advantageous properties position IDIC-Qu NPs as a promising candidate for targeted tumor treatment. Importantly, the IDIC-Qu NPs demonstrated controllable photothermal effects, leading to outstanding in vitro cytotoxicity against cancer cells and effective in vivo tumor ablation through mPTT. IDIC-Qu NPs nano-system enriches the family of organic PTAs and holds significant promise for future clinical applications of mPTT.
Collapse
Affiliation(s)
- Weijiao Fan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Center for Rehabilitation Medicine, Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Yichen He
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Peiyang Hu
- Department of Traumatology, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Hangzhou Medical College, Taizhou 317200, China
| | - Longcai Liu
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Xue Yang
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Tong Ge
- Department of Traumatology, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Hangzhou Medical College, Taizhou 317200, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, China.
| | - Xiaozhou Mou
- Center for Rehabilitation Medicine, Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China.
| | - Yu Cai
- Center for Rehabilitation Medicine, Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China.
| |
Collapse
|
8
|
Xie J, Li D, Niu S, Sheng Y, Shen R, He Y, Xu C, Zhang Y, Wang T, Xue Y. Nano-Titanium Oxide-Coated Carbon Nanotubes for Photothermal Therapy in the Treatment of Colorectal Cancer. Adv Healthc Mater 2024; 13:e2401009. [PMID: 38885692 DOI: 10.1002/adhm.202401009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Carbon nanotubes (CNTs) display good potential in tumor photothermal therapy (PTT). In this study, it is aimed to investigate the therapeutic potential of nano-titanium oxide-coated multi-walled carbon nanotubes (MCNTs) against colorectal cancer (CRC). First, TiO2 nanosheets are modified on the surface of MCNTs to obtain nano-TiO2-coated MCNTs. Next, cell compatibility validation is conducted on nano-TiO2-coated MCNTs, and it is found that nano-TiO2-coated MCNTs are safe within a certain concentration range (0-200 µg mL⁻1). Interestingly, nano-TiO2-coated MCNTs display a good killing effect in CRC cells under near-infrared (NIR) laser irradiation. Subsequently, nano-TiO2-coated MCNTs markedly promote the proapoptotic effects of NIR laser irradiation and significantly inhibit the expression of cell cycle proteins CCNA1 and CCND1 in CRC cells under NIR laser irradiation, which indicates that nano-TiO2-coated MCNTs exert anti-CRC effects under NIR laser irradiation by regulating cell apoptosis and cell cycle. Furthermore, nano-TiO2-coated MCNTs accelerate inhibitory effects on the AKT signaling pathway under NIR laser irradiation. Finally, a cell line-derived xenograft model is established, and the results showed that nano-TiO2-coated MCNTs significantly exhibit superior tumor-killing ability under NIR laser irradiation in vivo. Collectively, these results demonstrate that nano-TiO2-coated MCNTs with NIR laser irradiation may serve as an effective strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Jun Xie
- Department of Pediatric Internal Medicine, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, Jiangsu, 214023, China
| | - Da Li
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Sen Niu
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yufan Sheng
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Renhui Shen
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yiding He
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Chenhao Xu
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Ye Zhang
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Tong Wang
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
9
|
Yu X, Xu C, Sun J, Xu H, Huang H, Gan Z, George A, Ouyang S, Liu F. Recent developments in two-dimensional molybdenum disulfide-based multimodal cancer theranostics. J Nanobiotechnology 2024; 22:515. [PMID: 39198894 PMCID: PMC11351052 DOI: 10.1186/s12951-024-02785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
Recent advancements in cancer research have led to the generation of innovative nanomaterials for improved diagnostic and therapeutic strategies. Despite the proven potential of two-dimensional (2D) molybdenum disulfide (MoS2) as a versatile platform in biomedical applications, few review articles have focused on MoS2-based platforms for cancer theranostics. This review aims to fill this gap by providing a comprehensive overview of the latest developments in 2D MoS2 cancer theranostics and emerging strategies in this field. This review highlights the potential applications of 2D MoS2 in single-model imaging and therapy, including fluorescence imaging, photoacoustic imaging, photothermal therapy, and catalytic therapy. This review further classifies the potential of 2D MoS2 in multimodal imaging for diagnostic and synergistic theranostic platforms. In particular, this review underscores the progress of 2D MoS2 as an integrated drug delivery system, covering a broad spectrum of therapeutic strategies from chemotherapy and gene therapy to immunotherapy and photodynamic therapy. Finally, this review discusses the current challenges and future perspectives in meeting the diverse demands of advanced cancer diagnostic and theranostic applications.
Collapse
Affiliation(s)
- Xinbo Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Chen Xu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jingxu Sun
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
| | - Hainan Xu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Hanwei Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ziyang Gan
- Institute of Physical Chemistry, Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany
| | - Antony George
- Institute of Physical Chemistry, Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany
| | - Sihui Ouyang
- College of Materials Science and Engineering, Chongqing University, National Engineering Research Center for Magnesium Alloys, Chongqing University, Chongqing, 400044, China.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China.
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
10
|
Shang J, Xia Q, Sun Y, Wang H, Chen J, Li Y, Gao F, Yin P, Yuan Z. Bufalin-Loaded Multifunctional Photothermal Nanoparticles Inhibit the Anaerobic Glycolysis by Targeting SRC-3/HIF-1α Pathway for Improved Mild Photothermal Therapy in CRC. Int J Nanomedicine 2024; 19:7831-7850. [PMID: 39105099 PMCID: PMC11299722 DOI: 10.2147/ijn.s470005] [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: 03/21/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Purpose Compared with traditional photothermal therapy (PTT, >50°C), mild PTT (≤45°C) is a promising strategy for tumor therapy with fewer adverse effects. Unfortunately, its anti-tumor efficacy is hampered by thermoresistance induced by overexpression of heat shock proteins (HSPs). In our previous study, we found bufalin (BU) is a glycolysis inhibitor that depletes HSPs, which is expected to overcome thermotolerance of tumor cells. In this study, BU-loaded multifunctional nanoparticles (NPs) were developed for enhancing the mild PTT of colorectal cancer (CRC). Methods Fe3O4 NPs coated with the polydopamine (PDA) shell modified with polyethylene glycol (PEG) and cyclic arginine-glycyl-aspartic peptide (cRGD) for loading BU (Fe3O4@PDA-PEG-cRGD/BU NPs) were developed. The thermal variations in Fe3O4@PDA-PEG-cRGD/BU NPs solution under different conditions were measured. Glycolysis inhibition was evaluated by measuring the glucose uptake, extracellular lactate, and intracellular adenosine triphosphate (ATP) levels. The cellular cytotoxicity of Fe3O4@PDA-PEG-cRGD/BU NPs was analyzed using a cell counting kit-8 assay, Calcein-AM/PI double staining, and flow cytometry in HCT116 cells. The magnetic resonance imaging (MRI) performance and anti-tumor therapeutic efficacy of Fe3O4@PDA-PEG-cRGD/BU NPs were evaluated in HCT116-tumor bearing mice. Results Fe3O4@PDA-PEG-cRGD/BU NPs had an average diameter of 260.4±3.5 nm, the zeta potential of -23.8±1.6 mV, the drug loading rate of 1.1%, which had good thermal stability, photothermal conversion efficiencies and MRI performance. In addition, the released BU not only killed tumor cells but also interfered with glycolysis by targeting the steroid receptor coactivator 3 (SRC-3)/HIF-1α pathway, preventing intracellular ATP synthesis, and combating HSP-dependent tumor thermoresistance, ultimately strengthening the thermal sensitivity toward mild PTT both in vitro and in vivo. Conclusion This study provides a highly effective strategy for enhancing the therapeutic effects of mild PTT toward tumors.
Collapse
Affiliation(s)
- Jing Shang
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
| | - Qi Xia
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
| | - Yuji Sun
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, People’s Republic of China
| | - Hongtao Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, People’s Republic of China
| | - Jia Chen
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
| | - Yue Li
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
| | - Feng Gao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, People’s Republic of China
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Shanghai, 200062, People’s Republic of China
- The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Zeting Yuan
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, People’s Republic of China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Shanghai, 200062, People’s Republic of China
- The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, 230032, People’s Republic of China
| |
Collapse
|
11
|
Lai J, Shi Q, Xie Y, Zhu Y, Liang S, Chen Y, Yuan J, Liu L. Self-Delivery Nanomedicines Reverse Thermal Resistance to Enhance Tumor Mild-Temperature Photothermal Therapy. Mol Pharm 2024; 21:1526-1536. [PMID: 38379524 DOI: 10.1021/acs.molpharmaceut.3c01217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Tumoral thermal defense mechanisms considerably attenuate the therapeutic outcomes of mild-temperature photothermal therapy (PTT). Thus, developing a simple, efficient, and universal therapeutic strategy to sensitize mild-temperature PTT is desirable. Herein, we report self-delivery nanomedicines ACy NPs comprising a near-infrared (NIR) photothermal agent (Cypate), mitochondrial oxidative phosphorylation inhibitor (ATO), and distearoylphosphatidylethanolamine-polyethylene glycol 2000 (DSPE-PEG2000), which have a high drug-loading efficiency that can reverse tumoral thermal resistance, thereby increasing mild-temperature PTT efficacy. ACy NPs achieved targeted tumor accumulation and performed NIR fluorescence imaging capability in vivo to guide tumor PTT for optimized therapeutic outcomes. The released ATO reduced intracellular ATP levels to downregulate multiple heat shock proteins (including HSP70 and HSP90) before PTT, which reversed the thermal resistance of tumor cells, contributing to the excellent results of mild-temperature PTT in vitro and in vivo. Therefore, this study provides a simple, biosafe, advanced, and universal heat shock protein-blocking strategy for tumor PTT.
Collapse
Affiliation(s)
- Jinmei Lai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| | - Qunying Shi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| | - Yongqi Xie
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| | - Yinyin Zhu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| | - Shiyu Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| | - Yi Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| | - Jiali Yuan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| | - Lihan Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong510515, P. R. China
| |
Collapse
|
12
|
Tao N, Jiao L, Li H, Deng L, Wang W, Zhao S, Chen W, Chen L, Zhu C, Liu YN. A Mild Hyperthermia Hollow Carbon Nanozyme as Pyroptosis Inducer for Boosted Antitumor Immunity. ACS NANO 2023; 17:22844-22858. [PMID: 37942890 DOI: 10.1021/acsnano.3c07601] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
The immune checkpoint blockade (ICB) antibody immunotherapy has demonstrated clinical benefits for multiple cancers. However, the efficacy of immunotherapy in tumors is suppressed by deficient tumor immunogenicity and immunosuppressive tumor microenvironments. Pyroptosis, a form of programmed cell death, can release tumor antigens, activate effective tumor immunogenicity, and improve the efficiency of ICB, but efficient pyroptosis for tumor treatment is currently limited. Herein, we show a mild hyperthermia-enhanced pyroptosis-mediated immunotherapy based on hollow carbon nanozyme, which can specifically amplify oxidative stress-triggered pyroptosis and synchronously magnify pyroptosis-mediated anticancer responses in the tumor microenvironment. The hollow carbon sphere modified with iron and copper atoms (HCS-FeCu) with multiple enzyme-mimicking activities has been engineered to induce cell pyroptosis via the radical oxygen species (ROS)-Tom20-Bax-Caspase 3-gasdermin E (GSDME) signaling pathway under light activation. Both in vitro and in vivo antineoplastic results confirm the superiority of HCS-FeCu nanozyme-induced pyroptosis. Moreover, the mild photothermal-activated pyroptosis combining anti-PD-1 can enhance antitumor immunotherapy. Theoretical calculations further indicate that the mild photothermal stimulation generates high-energy electrons and enhances the interaction between the HCS-FeCu surface and adsorbed oxygen, facilitating molecular oxygen activation, which improves the ROS production efficiency. This work presents an approach that effectively transforms immunologically "cold" tumors into "hot" ones, with significant implications for clinical immunotherapy.
Collapse
Affiliation(s)
- Na Tao
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, PR China
| | - Lei Jiao
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410083, PR China
| | - Liu Deng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, PR China
| | - Wei Wang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, PR China
| | - Senfeng Zhao
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, PR China
| | - Wansong Chen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, PR China
| | - Limiao Chen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, PR China
| | - Chengzhou Zhu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - You-Nian Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, PR China
| |
Collapse
|
13
|
Ta N, Jiang X, Zhang Y, Wang H. Ferroptosis as a promising therapeutic strategy for melanoma. Front Pharmacol 2023; 14:1252567. [PMID: 37795022 PMCID: PMC10546212 DOI: 10.3389/fphar.2023.1252567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
Malignant melanoma (MM) is the most common and deadliest type of skin cancer and is associated with high mortality rates across all races and ethnicities. Although present treatment options combined with surgery provide short-term clinical benefit in patients and early diagnosis of non-metastatic MM significantly increases the probability of survival, no efficacious treatments are available for MM. The etiology and pathogenesis of MM are complex. Acquired drug resistance is associated with a pool prognosis in patients with advanced-stage MM. Thus, these patients require new therapeutic strategies to improve their treatment response and prognosis. Multiple studies have revealed that ferroptosis, a non-apoptotic form of regulated cell death (RCD) characterized by iron dependant lipid peroxidation, can prevent the development of MM. Recent studies have indicated that targeting ferroptosis is a promising treatment strategy for MM. This review article summarizes the core mechanisms underlying the development of ferroptosis in MM cells and its potential role as a therapeutic target in MM. We emphasize the emerging types of small molecules inducing ferroptosis pathways by boosting the antitumor activity of BRAFi and immunotherapy and uncover their beneficial effects to treat MM. We also summarize the application of nanosensitizer-mediated unique dynamic therapeutic strategies and ferroptosis-based nanodrug targeting strategies as therapeutic options for MM. This review suggests that pharmacological induction of ferroptosis may be a potential therapeutic target for MM.
Collapse
Affiliation(s)
- Na Ta
- Department of Neurosurgery, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Xiaodong Jiang
- Department of Anatomy, College of Basic Medicine, Chifeng University Health Science Center, Chifeng, China
| | - Yongchun Zhang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Hongquan Wang
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
14
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
15
|
Mohamed AA, Caussat T, Kelly S, Johansen PM, Lucke-Wold B. Choroid plexus tumors: A spectrum from benign to malignant. TUMOR DISCOVERY 2023; 2:1057. [PMID: 37799733 PMCID: PMC10552314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Choroid plexus tumors (CPT) are believed to originate from outgrowths of the choroid plexus. Despite their broad spectrum of symptoms, invasive nature, and prognosis, most CPTs typically exhibit similar presentations due to their relationship with the cerebral ventricles, as well as the mechanical obstruction and mass effect associated with their growth. In addition, these tumors mainly affect the pediatric population, further complicating the differentiation between benign and malignant subtypes. The World Health Organization classifies CPTs into three grades, namely, grades I, II, or III, based on their mitotic activity, which determine the benign or malignant nature of the tumors. CPTs classified by the World Health Organization (WHO) include choroid plexus papillomas (CPP), atypical CPPs (aCPP), and malignant choroid plexus carcinomas (CPC). Choroid plexus adenomas represent an additional category of benign CPTs not officially classified by the WHO. Despite the variations in histology, immunohistochemistry, imaging, treatment, and prognosis, CPTs cannot be reliably distinguished based solely on clinical presentation. Therefore, in this review, we aim to provide a comprehensive overview of each tumor subtype, along with the current management approach and emerging treatments.
Collapse
Affiliation(s)
- Ali A. Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Thomas Caussat
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Sophie Kelly
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Phillip M. Johansen
- Department of Neurosurgery, University of South Florida, Orlando, Florida, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
16
|
Yu Y, Wang T, Meng X, Jiang T, Zhao X. Chitosan Thermosensitive Hydrogel Based on DNA Damage Repair Inhibition and Mild Photothermal Therapy for Enhanced Antitumor Treatment. Biomacromolecules 2023; 24:3755-3766. [PMID: 37506051 DOI: 10.1021/acs.biomac.3c00430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
The DNA damage repair of tumor cells limits the effect of photothermal therapy (PTT), and high temperatures induced by PTT can damage adjacent normal tissues. To overcome these limitations, we developed a novel composite hydrogel (OLA-Au-Gel) based on chitosan (CS) and β-glycerophosphate (β-GP), which encapsulated olaparib-liposomes (OLA-lips) and CS-capped gold nanoparticles (CS-AuNPs). OLA-Au-Gel achieved the combination of mild PTT (mPTT) by CS-AuNPs and tumor DNA damage repair inhibition by OLA. The hydrogel showed good biocompatibility, injectability, and photothermal response. Under near-infrared laser irradiation, OLA-Au-Gel inhibited the proliferation of tumor cells, induced the generation of reactive oxygen species in vitro, and effectively inhibited the growth of breast tumors in vivo. OLA-Au-Gel shows a promising application prospect for inhibiting tumor development and improving the antitumor effect. Collectively, we propose a novel strategy for enhanced antitumor therapy based on the combination of mPTT and DNA damage repair inhibition.
Collapse
Affiliation(s)
- Yang Yu
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Teng Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xin Meng
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Tianze Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Xia Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|