1
|
Sato T, Huang Y, Masuda T, Li J, Takai M. Design of enzyme immobilized zwitterionic copolymer nanogels and its size effect on electrochemical reaction. Colloids Surf B Biointerfaces 2025; 246:114370. [PMID: 39551033 DOI: 10.1016/j.colsurfb.2024.114370] [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: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
For enzyme-based electrochemical devices, an improvement in electron transfer between the enzyme and electrode is important. Thus, we developed a nano-scaled hydrogel that includes an electron mediator and enzyme to realize nano-sized effects that enhance the functions. Three different chain lengths (short, medium, and long) of copolymers composed of 2-methacryloyloxyethyl phosphorylcholine (MPC) and methacrylic acid N-hydroxysuccinimide ester (MNHS; poly(MPC-co-MNHS), PMS) were synthesized via reversible addition-fragmentation chain transfer (RAFT) polymerization. The PMS nanogels can bind to the amino ferrocene (AFc) of the electron mediator and glucose oxidase (GOD) as a catalyst. The mono-dispersive PMS nanogels approximately 200-250 nm in size bound with AFc were prepared with different polymer chain lengths and amounts of AFc (PMMFcX_Y%, X= 'degree of polymerization, 50, 75, 100' and Y= 'AFc feeding ratio against the amount of NHS group in the polymer chain, 50 %, 100 %'). The size of PMMFcX_Y% could be controlled by changing degree of polymerization or AFc feeding ratio. After the modification of GOD to PMMFcX_Y%, their size increased slightly from the original size (ca. 200-250 nm) to approximately 250-300 nm. The catalytic activity of nanogel in dispersed system was higher than that of microgel, indicating that nanogels could improve glucose transport in hydrogel layer. Compared to the catalytic reaction of the PMMFc 75_50 %-GOD nanogel-modified electrodes with that of microgel modified electrode, the current response was improved by decreasing the nanogel size, as evaluated by electrochemical measurements. These results revealed that the smaller nanogels could improve both glucose transport and electron transfer via mediator by smaller size, resulting higher efficiency of enzyme immobilized electrode.
Collapse
Affiliation(s)
- Takehiro Sato
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yixuan Huang
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tsukuru Masuda
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Jincai Li
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Madoka Takai
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
2
|
Alqarni SS, Afzal M, Al-Abbasi FA, Moglad E, Bawadood AS, Almalki NAR, Alqurashi MM, Imam F, Syed SM, Kazmi I. Exploring acemannan-loaded nanogel formulation for the treatment of IMQ-induced psoriasis-like inflammation: In vitro characterization and in vivo efficacy assessment. Int Immunopharmacol 2025; 148:114064. [PMID: 39884082 DOI: 10.1016/j.intimp.2025.114064] [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: 11/11/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 02/01/2025]
Abstract
This study aimed to explore a nanogel formulation containing acemannan as a carrier for the treatment of psoriasis-like skin inflammation. Several acemannan concentrations, such as F1 (2.5 %) and F2 (5 %), were used to prepare the nanogel formulation by homogenization. The formulation was then assessed for in-vitro performance. Four groups of animals were randomly assigned to the animals: Cluster I consisted of normal saline control; Cluster II was assigned Imiquimod (IMQ) control (5 %); Cluster III was assigned IMQ + 2.5 % acemannan (F1); and Cluster IV was assigned IMQ + 5 % acemannan (F2). The effectiveness of the gel in the in vivo study was evaluated in terms of body weight, scaly skin, skin redness, inflammation, patches, moisturizing effect, pro-inflammatory cytokines, nitric oxide, and histopathological examination. The prepared nanogel possessed the desired characteristics in terms of in vitro evaluation parameters. The average particle size was around 199.6 nm, with a polydispersibility index (PDI) of 0.338 and a zeta potential of -65.9 mV. The nanogel formulation significantly (P < 0.05) regulated in vivo performance, including redness, scaly skin, inflammation, patches, moisturizing effect, pro-inflammatory cytokines, and nitric oxide. The histopathological findings suggested that acemannan was effective in rejuvenating the affected skin.
Collapse
Affiliation(s)
- Sana Saeed Alqarni
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 11451 Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442 Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589 Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942 Saudi Arabia
| | - Azizah Salim Bawadood
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Naif A R Almalki
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 11451 Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589 Saudi Arabia
| | - May M Alqurashi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589 Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451 Saudi Arabia
| | | | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589 Saudi Arabia.
| |
Collapse
|
3
|
Liu XM, Li Z, Wang XY, Ding BW, Wang JQ, Qiao X, Feng YK, Hao JH, Xu JY. Self-assembled HO-1i-Pt(IV) nanomedicine targeting p38/MAPK and MDR pathways for cancer chemo-immunotherapy. J Control Release 2025; 379:797-813. [PMID: 39848589 DOI: 10.1016/j.jconrel.2025.01.050] [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: 09/05/2024] [Revised: 01/08/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
Platinum(II)-based antitumor drugs are widely used in clinics but limited by severe side effects and resistance. Multi-target Platinum(IV) complexes are emerging as ideal alternatives. Heme oxygenase-1 (HO-1) works as a rate-limiting step in heme degradation and is overexpressed in malignant tumors. Herein, HO-1i-based Platinum(IV) prodrugs are prepared and candidate complex 15 is further developed into self-assembled nanoparticles (15-NPs). 15 and 15-NPs significantly increase cytotoxicity, particularly in HepG2 (74.77- and 96.14-fold increases) and A549cisR (38.6- and 47.24-fold increases), while reducing toxicity towards normal cells compared to cisplatin. In vitro experiments show 15 and 15-NPs activated multiple pathways, including p38/MAPK- and MDR-related proteins, achieving multi-target synergistic chemosensitization and anti-resistance, further verified by RNA-sequencing analysis. In vivo tests demonstrate that 15 and 15-NPs efficiently inhibit tumor growth and systemic toxicity, especially 15-NPs with optimal tumor-inhibition rate and survival (80% and 100%), superior to cisplatin (40% and 50%), attributing to its extra endocytosis, EPR effect, and precisely tumor-targeted release besides the advantage of a free HO-1i-Pt(IV) prodrug. Additionally, 15 and 15-NPs distinctly regulate T-cell and macrophage functions, thereby exhibiting a chemoimmuno-combined action. This study highlights that multi-functional Platinum(IV) prodrug target-delivered to tumors via carrier-free nanoparticles may represent an effective modality for improving cancer therapy.
Collapse
Affiliation(s)
- Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiao-Ya Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Bo-Wen Ding
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jia-Qian Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yu-Kuan Feng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Ji-Hui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
4
|
Michibata J, Kawaguchi Y, Hirose H, Eguchi A, Deguchi S, Takayama K, Xu W, Niidome T, Sasaki Y, Akiyoshi K, Futaki S. Polysaccharide-Based Coacervate Microgel Bearing Cationic Peptides That Achieve Dynamic Cell-Membrane Structure Alteration and Facile Cytosolic Infusion of IgGs. Bioconjug Chem 2024; 35:1888-1899. [PMID: 39500569 DOI: 10.1021/acs.bioconjchem.4c00344] [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: 12/19/2024]
Abstract
Conjugates of the biocompatible polysaccharide pullulan with a cell membrane permeabilizing peptide L17E (PL-L17Es) were prepared with the aim of producing complex coacervates with pronounced intracellular antibody (IgG) delivery activity and stable structures. Coacervates with diameters of a few μm were formed simply by mixing PL-L17Es with IgG labeled with negatively charged fluorescent moieties of Alexa Fluor 488 [IgG(AF488)]. The coacervate resulted in a pronounced cytosolic infusion of IgG(AF488) and IgG binding to the target proteins inside the cell. The droplet structures were maintained even under high salt conditions, and the fluorescence in the droplet was not recovered after photobleaching, suggesting the formation of complex coacervate microgels. Dynamic changes in cell membrane structure to entrap the coacervate microgels were captured by confocal and electron microscopy, resulting in cytosolic IgG infusion. The use of M-lycotoxin instead of L17E resulted in a coacervate microgel with marked IgG delivery activity even in the presence of serum. Successful IgG delivery to primary hepatocytes, undifferentiated induced pluripotent stem (iPS) cells, and iPS cell-derived intestinal epithelial cells was also achieved. The construction of complex coacervate microgels with design flexibility and the validity of intracellular IgG delivery with high salt stability were thus demonstrated.
Collapse
Affiliation(s)
- Junya Michibata
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yoshimasa Kawaguchi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Hisaaki Hirose
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Akiko Eguchi
- Biobank Center, Mie University Hospital and Department of Gastroenterology and Hepatology, School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Wei Xu
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Takuro Niidome
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kazunari Akiyoshi
- Department of Immunology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Kyoto 606-8501, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
5
|
Feng T, Hu J, Wen J, Qian Z, Che G, Zhou Q, Zhu L. Personalized nanovaccines for treating solid cancer metastases. J Hematol Oncol 2024; 17:115. [PMID: 39609851 PMCID: PMC11603676 DOI: 10.1186/s13045-024-01628-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
Cancer vaccines have garnered attention as a potential treatment for cancer metastases. Nevertheless, the clinical response rate to vaccines remains < 30%. Nanoparticles stabilize vaccines and improve antigen recognition and presentation, resulting in high tumor penetration or accumulation, effective co-distribution of drugs to the secondary lymphatic system, and adaptable antigen or adjuvant administration. Such vaccine-like nanomedicines have the ability to eradicate the primary tumors as well as to prevent or eliminate metastases. This review examines state-of-the-art nanocarriers developed to deliver tumor vaccines to metastases, including synthetic, semi-biogenic, and biogenic nanosystems. Moreover, it highlights the physical and pharmacological properties that enhance their anti-metastasis efficiency. This review also addresses the combination of nanovaccines with cancer immunotherapy to target various steps in the metastatic cascade, drawing insights from preclinical and clinical studies. The review concludes with a critical analysis of the challenges and frameworks linked to the clinical translation of cancer nanovaccines.
Collapse
Affiliation(s)
- Tang Feng
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jia Hu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jirui Wen
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Guowei Che
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qinghua Zhou
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Kawasaki R, Miura Y, Kono N, Fujita S, Yamana K, Ikeda A. Boron Agent Delivery Platforms Based on Natural Products for Boron Neutron Capture Therapy. ChemMedChem 2024; 19:e202400323. [PMID: 38830821 DOI: 10.1002/cmdc.202400323] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/05/2024]
Abstract
Boron neutron capture therapy (BNCT) is one of the most promising modalities for cancer treatment due to its minimal invasiveness. Although two types of boron agents are clinically used, several issues persist in their delivery, including poor water solubility, instability in aqueous media, selectivity toward cancer cells, accumulation in cancer cells, retention time in tumor tissue, and efficiency in achieving the boron neutron capture reaction. Addressing these challenges, numerous groups have explored various boron agents to enhance the therapeutic benefits of BNCT. This review summarizes delivery platforms based on natural products for BNCT.
Collapse
Affiliation(s)
- Riku Kawasaki
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Yamato Miura
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Nanami Kono
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Seiya Fujita
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Keita Yamana
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Atsushi Ikeda
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| |
Collapse
|
7
|
Muraoka D, Moi ML, Muto O, Nakatsukasa T, Deng S, Takashima C, Yamaguchi R, Sawada SI, Hayakawa H, Nguyen TTN, Haseda Y, Soga T, Matsushita H, Ikeda H, Akiyoshi K, Harada N. Low-frequency CD8 + T cells induced by SIGN-R1 + macrophage-targeted vaccine confer SARS-CoV-2 clearance in mice. NPJ Vaccines 2024; 9:173. [PMID: 39294173 PMCID: PMC11411095 DOI: 10.1038/s41541-024-00961-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/01/2024] [Indexed: 09/20/2024] Open
Abstract
Vaccine-induced T cells and neutralizing antibodies are essential for protection against SARS-CoV-2. Previously, we demonstrated that an antigen delivery system, pullulan nanogel (PNG), delivers vaccine antigen to lymph node medullary macrophages and thereby enhances the induction of specific CD8+ T cells. In this study, we revealed that medullary macrophage-selective delivery by PNG depends on its binding to a C-type lectin SIGN-R1. In a K18-hACE2 mouse model of SARS-CoV-2 infection, vaccination with a PNG-encapsulated receptor-binding domain of spike protein decreased the viral load and prolonged the survival in the CD8+ T cell- and B cell-dependent manners. T cell receptor repertoire analysis revealed that although the vaccine induced T cells at various frequencies, low-frequency specific T cells mainly promoted virus clearance. Thus, the induction of specific CD8+ T cells that respond quickly to viral infection, even at low frequencies, is important for vaccine efficacy and can be achieved by SIGN-R1+ medullary macrophage-targeted antigen delivery.
Collapse
Affiliation(s)
- Daisuke Muraoka
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan.
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan.
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.
| | - Osamu Muto
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takaaki Nakatsukasa
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Situo Deng
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Chieko Takashima
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shin-Ichi Sawada
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Haruka Hayakawa
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | | | | | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazunari Akiyoshi
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
8
|
Guo J, Liu C, Qi Z, Qiu T, Zhang J, Yang H. Engineering customized nanovaccines for enhanced cancer immunotherapy. Bioact Mater 2024; 36:330-357. [PMID: 38496036 PMCID: PMC10940734 DOI: 10.1016/j.bioactmat.2024.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
Nanovaccines have gathered significant attention for their potential to elicit tumor-specific immunological responses. Despite notable progress in tumor immunotherapy, nanovaccines still encounter considerable challenges such as low delivery efficiency, limited targeting ability, and suboptimal efficacy. With an aim of addressing these issues, engineering customized nanovaccines through modification or functionalization has emerged as a promising approach. These tailored nanovaccines not only enhance antigen presentation, but also effectively modulate immunosuppression within the tumor microenvironment. Specifically, they are distinguished by their diverse sizes, shapes, charges, structures, and unique physicochemical properties, along with targeting ligands. These features of nanovaccines facilitate lymph node accumulation and activation/regulation of immune cells. This overview of bespoke nanovaccines underscores their potential in both prophylactic and therapeutic applications, offering insights into their future development and role in cancer immunotherapy.
Collapse
Affiliation(s)
- Jinyu Guo
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
| | - Changhua Liu
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
| | - Zhaoyang Qi
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| | - Ting Qiu
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
| | - Jin Zhang
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
| | - Huanghao Yang
- 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, 2 Xueyuan Road, Fuzhou, 350108, PR China
| |
Collapse
|
9
|
Katopodi T, Petanidis S, Floros G, Porpodis K, Kosmidis C. Hybrid Nanogel Drug Delivery Systems: Transforming the Tumor Microenvironment through Tumor Tissue Editing. Cells 2024; 13:908. [PMID: 38891040 PMCID: PMC11171955 DOI: 10.3390/cells13110908] [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: 03/26/2024] [Revised: 04/27/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
The future of drug delivery offers immense potential for the creation of nanoplatforms based on nanogels. Nanogels present a significant possibility for pharmaceutical advancements because of their excellent stability and effective drug-loading capability for both hydrophobic and hydrophilic agents. As multifunctional systems, composite nanogels demonstrate the capacity to carry genes, drugs, and diagnostic agents while offering a perfect platform for theranostic multimodal applications. Nanogels can achieve diverse responsiveness and enable the stimuli-responsive release of chemo-/immunotherapy drugs and thus reprogramming cells within the TME in order to inhibit tumor proliferation, progression, and metastasis. In order to achieve active targeting and boost drug accumulation at target sites, particular ligands can be added to nanogels to improve the therapeutic outcomes and enhance the precision of cancer therapy. Modern "immune-specific" nanogels also have extra sophisticated tumor tissue-editing properties. Consequently, the introduction of a multifunctional nanogel-based drug delivery system improves the targeted distribution of immunotherapy drugs and combinational therapeutic treatments, thereby increasing the effectiveness of tumor therapy.
Collapse
Affiliation(s)
- Theodora Katopodi
- Laboratory of Medical Biology and Genetics, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Savvas Petanidis
- Laboratory of Medical Biology and Genetics, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Department of Pulmonology, I.M. Sechenov First Moscow State Medical University, Moscow 119992, Russia
| | - George Floros
- Department of Electrical and Computer Engineering, University of Thessaly, 38334 Volos, Greece;
| | - Konstantinos Porpodis
- Pulmonary Department-Oncology Unit, G. Papanikolaou General Hospital, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece;
| | - Christoforos Kosmidis
- Third Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece;
| |
Collapse
|
10
|
Sela A, Moussa S, Rodov V, Iasur Kruh L, Poverenov E. Carboxymethyl chitosan-N-alkylimine derivatives: Synthesis, characterization and use for preservation of symbiotic biofertilizer bacteria on chickpea seeds. Int J Biol Macromol 2024; 262:130057. [PMID: 38340940 DOI: 10.1016/j.ijbiomac.2024.130057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
A series of carboxymethyl chitosan-N-alkylimine derivatives with side chain length of 4 to 10 carbons (CMCS-n, n = 4, 6, 8, 10) was prepared in a one-step solvent-free synthesis using Schiff base chemistry. The modified polysaccharides were characterized by their spectral, thermal and physical properties. The prepared polymers demonstrated an ability to spontaneous self-assembly with a clear correlation between critical aggregation concentration and the chain length of the alkyl substituent. N-alkylimine-CMCS derivatives were found to deliver hydrophobic (curcumin) and hydrophilic (ascorbic acid) active agents in unfavorable environments of water and oil, respectively. Then, N-alkylimine-CMCS derivatives were used as a platform for the delivery of symbiotic gram-positive bacteria Bacillus subtilis CJ onto chickpea seeds. These bacteria demonstrated a significantly higher survival rate (106 CFU/mL) in dried CMCS-6 derivative film than in other films tested. The seeds treated with N-alkylimine-CMCS coatings that contained B. subtilis CJ demonstrated up to 100-fold increase of this bacterial population on the seedlings in comparison to the pristine CMCS.
Collapse
Affiliation(s)
- Aviad Sela
- Agro-nanotechnology and Advanced Materials Research Center, Department of Food Science, Agriculture Research Organization, The Volcani Institute, Rishon LeZion, Israel; Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Suzana Moussa
- Department of Biotechnology Engineering, Braude College of Engineering, Carmiel, Israel
| | - Victor Rodov
- Department of Postharvest Science, Agricultural Research Organization, The Volcani Institute, Rishon LeZion, Israel
| | - Lilach Iasur Kruh
- Department of Biotechnology Engineering, Braude College of Engineering, Carmiel, Israel
| | - Elena Poverenov
- Agro-nanotechnology and Advanced Materials Research Center, Department of Food Science, Agriculture Research Organization, The Volcani Institute, Rishon LeZion, Israel.
| |
Collapse
|
11
|
Yong J, Shu H, Zhang X, Yang K, Luo G, Yu L, Li J, Huang H. Natural Products-Based Inhaled Formulations for Treating Pulmonary Diseases. Int J Nanomedicine 2024; 19:1723-1748. [PMID: 38414528 PMCID: PMC10898359 DOI: 10.2147/ijn.s451206] [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: 11/22/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
Given the unique physiological and pathological characteristics of the lung, the direct, inhalable route is more conducive to pulmonary drug delivery and disease control than traditional systemic drug delivery, significantly circumventing drug loss, off-target effects, systemic and organ toxicity, etc., and is widely regarded as the preferred regimen for pulmonary drug delivery. However, very few lung diseases are currently treated with the preferred inhaled formulations, such as asthma, chronic obstructive pulmonary disease and pulmonary hypertension. And there is a lack of appropriate inhaled formulations for other critical lung diseases, such as lung cancer and pulmonary fibrosis, due to the fact that the physicochemical properties of the drugs and their pharmacokinetic profiles do not match the physiology of the lung, and conventional inhalation devices are unable to deliver them to the specific parts of the lung. Phytochemicals of natural origin, due to their wide availability and clear safety profile, hold great promise for the preparation of inhalable formulations to improve the current dilemma in the treatment of lung diseases. In particular, the preparation of inhalable formulations based on nano- and microparticulate carriers for drug delivery to deep lung tissues, which overcome the shortcomings of conventional inhalation therapies while targeting the drug activity directly to a specific part of the lung, may be the best approach to change the current dilemma of lung disease treatment. In this review, we discuss recent advances in nano- and micron-carrier-based inhalation formulations for the delivery of natural products for the treatment of pulmonary diseases, which may represent an opportunity for practical clinical translation of natural products.
Collapse
Affiliation(s)
- Jiangyan Yong
- Department of Clinical Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, People’s Republic of China
| | - Hongli Shu
- Department of Clinical Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, People’s Republic of China
| | - Xiao Zhang
- Department of Clinical Laboratory, Chengdu Children Special Hospital, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Kun Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Guining Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Lu Yu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Jiaqi Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Hong Huang
- Department of Clinical Laboratory, the People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, People’s Republic of China
| |
Collapse
|
12
|
Hao J, Ishihara M, Rapenne G, Yasuhara K. Lipid nanodiscs spontaneously formed by an amphiphilic polymethacrylate derivative as an efficient nanocarrier for molecular delivery to intact cells. RSC Adv 2024; 14:6127-6134. [PMID: 38375006 PMCID: PMC10875731 DOI: 10.1039/d3ra07481a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
There is a great demand for the technology of molecular delivery into living cells using nanocarriers to realise molecular therapies such as gene delivery and drug delivery systems. Lipid-based nanocarriers offer several advantages for molecular delivery in biological systems, such as easy preparation, high encapsulation efficiency of water-insoluble drug molecules, and excellent biocompatibility. In this paper, we first report the interaction of lipid nanodiscs spontaneously formed by the complexation of an amphiphilic polymethacrylate derivative and phospholipid with intact cells. We evaluated the internalisation of polymethacrylate-based lipid nanodiscs by intact HeLa cells and applied them to the delivery of paclitaxel (PTX), an anticancer drug. The lipid nanodisc showed excellent uptake efficiency compared to conventional liposomes at a concentration where nanodiscs do not show cytotoxicity. In addition, the nanodisc encapsulating PTX showed significantly higher anticancer activity than PTX-loaded liposomes against HeLa cells, reflecting their excellent activity in delivering payloads to intact cells. This study demonstrated the potential of a polymethacrylate-based lipid nanodisc as a novel nanocarrier for molecular delivery to intact cells.
Collapse
Affiliation(s)
- Jinyu Hao
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST) 8916-5 Takayama-cho Ikoma 630-0192 Japan
| | - Mika Ishihara
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST) 8916-5 Takayama-cho Ikoma 630-0192 Japan
| | - Gwénaël Rapenne
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST) 8916-5 Takayama-cho Ikoma 630-0192 Japan
- CEMES-CNRS, Université de Toulouse, CNRS 29 Rue Marvig F-31055 Toulouse Cedex 4 France
| | - Kazuma Yasuhara
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST) 8916-5 Takayama-cho Ikoma 630-0192 Japan
- Centre for Digital Green-innovation, Nara Institute of Science and Technology (NAIST) 8916-5 Takayama-cho Ikoma 630-0192 Japan
| |
Collapse
|
13
|
Ghafelehbashi R, Salehi M, Kouhi M, AlizadehNaini A, Sajadi-Javan ZS, Nejatidanesh F. Recent progress in cancer immunotherapy: Application of nano-therapeutic systems. J Drug Deliv Sci Technol 2024; 91:105184. [DOI: 10.1016/j.jddst.2023.105184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Wang Z, Fu S, Guo Y, Han Y, Ma C, Li R, Yang X. Classification and design strategies of polysaccharide-based nano-nutrient delivery systems for enhanced bioactivity and targeted delivery: A review. Int J Biol Macromol 2024; 256:128440. [PMID: 38016614 DOI: 10.1016/j.ijbiomac.2023.128440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023]
Abstract
Since many nutrients are highly sensitive, they cannot be absorbed and utilized efficiently by the body. Using nano-delivery systems to encapsulate nutrients is an effective method of solving the problems associated with the application of nutrients at this stage. Polysaccharides, as natural biomaterials, have a unique chemical structure, ideal biocompatibility, biodegradability and low immunogenicity. This makes polysaccharides powerful carriers that can enhance the biological activity of nutrients. However, the true role of polysaccharide-based delivery systems requires an in-depth understanding of the structural and physicochemical characteristics of polysaccharide-based nanodelivery systems, as well as effective modulation of the intestinal delivery mechanism and the latest advances in nano-encapsulation. This review provides an overview of polysaccharide-based nano-delivery systems dependent on different carrier types, emphasizing recent advances in the application of polysaccharides, a biocomposite material designed for nutrient delivery systems. Strategies for polysaccharide-based nano-delivery systems to enhance the bioavailability of orally administered nutrients from the perspective of the intestinal absorption barrier are presented. Characterization methods for polysaccharide-based nano-delivery systems are presented as well as an explanation of the formation mechanisms behind nano-delivery systems from the perspective of molecular forces. Finally, we discussed the challenges currently facing polysaccharide-based nano-delivery systems as well as possible future directions for the future.
Collapse
Affiliation(s)
- Zhili Wang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Shiyao Fu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Yong Guo
- College of Sports and Human Sciences, Harbin Sport University, Harbin 150008, China
| | - Ying Han
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Chao Ma
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Ruiling Li
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Xin Yang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; Chongqing Research Institute, Harbin Institute of Technology, Chongqing 401135, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China.
| |
Collapse
|
15
|
Yang M, Zhang Y, Deng F, Wu X, Chen Y, Ma F, Shi L. Development of self-cooperative nanochaperones with enhanced activity to facilitate protein refolding. MATERIALS HORIZONS 2023; 10:5547-5554. [PMID: 37843027 DOI: 10.1039/d3mh00619k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Regulating protein folding including assisting de novo folding, preventing misfolding and aggregation, and facilitating refolding of proteins are of significant importance for retaining protein's biological activities. Here, we report a mixed shell polymeric micelle (MSPM)-based self-cooperative nanochaperone (self-CO-nChap) with enhanced activity to facilitate protein refolding. This self-CO-nChap was fabricated by introducing Hsp40-mimetic artificial carriers into the traditional nanochaperone to cooperate with the Hsp70-mimetic confined hydrophobic microdomains. The artificial carrier facilitates transfer and immobilization of client proteins into confined hydrophobic microdomains, by which significantly improving self-CO-nChap's capability to inhibit unfolding and aggregation of client proteins, and finally facilitating refolding. Compared to traditional nanochaperones, the self-CO-nChap significantly enhances the thermal stability of horseradish peroxidase (HRP) epicyclically under harsher conditions. Moreover, the self-CO-nChap efficiently protects misfolding-prone proteins, such as immunoglobulin G (IgG) antibody from thermal denaturation, which is hardly achieved using traditional nanochaperones. In addition, a kinetic partitioning mechanism was devised to explain how self-CO-nChap facilitates refolding by regulating the cooperative effect of kinetics between the nanochaperone and client proteins. This work provides a novel strategy for the design of protein folding regulatory materials, including nanochaperones.
Collapse
Affiliation(s)
- Menglin Yang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P.R. China.
| | - Yanli Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P.R. China.
| | - Fei Deng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P.R. China.
| | - Xiaohui Wu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P.R. China.
| | - Yujie Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P.R. China.
| | - Feihe Ma
- State Key Laboratory of Separation Membranes and Membrane Processes and School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P.R. China.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P.R. China.
| |
Collapse
|
16
|
Wang J, Viola M, Migliorini C, Paoletti L, Arpicco S, Di Meo C, Matricardi P. Polysaccharide-Based Nanogels to Overcome Mucus, Skin, Cornea, and Blood-Brain Barriers: A Review. Pharmaceutics 2023; 15:2508. [PMID: 37896268 PMCID: PMC10610445 DOI: 10.3390/pharmaceutics15102508] [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: 10/06/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Nanocarriers have been extensively developed in the biomedical field to enhance the treatment of various diseases. However, to effectively deliver therapeutic agents to desired target tissues and enhance their pharmacological activity, these nanocarriers must overcome biological barriers, such as mucus gel, skin, cornea, and blood-brain barriers. Polysaccharides possess qualities such as excellent biocompatibility, biodegradability, unique biological properties, and good accessibility, making them ideal materials for constructing drug delivery carriers. Nanogels, as a novel drug delivery platform, consist of three-dimensional polymer networks at the nanoscale, offering a promising strategy for encapsulating different pharmaceutical agents, prolonging retention time, and enhancing penetration. These attractive properties offer great potential for the utilization of polysaccharide-based nanogels as drug delivery systems to overcome biological barriers. Hence, this review discusses the properties of various barriers and the associated constraints, followed by summarizing the most recent development of polysaccharide-based nanogels in drug delivery to overcome biological barriers. It is expected to provide inspiration and motivation for better design and development of polysaccharide-based drug delivery systems to enhance bioavailability and efficacy while minimizing side effects.
Collapse
Affiliation(s)
- Ju Wang
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Marco Viola
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Claudia Migliorini
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Luca Paoletti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy;
| | - Chiara Di Meo
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Pietro Matricardi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| |
Collapse
|
17
|
Askari E, Shokrollahi Barough M, Rahmanian M, Mojtabavi N, Sarrami Forooshani R, Seyfoori A, Akbari M. Cancer Immunotherapy Using Bioengineered Micro/Nano Structured Hydrogels. Adv Healthc Mater 2023; 12:e2301174. [PMID: 37612251 PMCID: PMC11468077 DOI: 10.1002/adhm.202301174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/15/2023] [Indexed: 08/25/2023]
Abstract
Hydrogels, a class of materials with a 3D network structure, are widely used in various applications of therapeutic delivery, particularly cancer therapy. Micro and nanogels as miniaturized structures of the bioengineered hydrogels may provide extensive benefits over the common hydrogels in encapsulation and controlled release of small molecular drugs, macromolecular therapeutics, and even cells. Cancer immunotherapy is rapidly developing, and micro/nanostructured hydrogels have gained wide attention regarding their engineered payload release properties that enhance systemic anticancer immunity. Additionally, they are a great candidate due to their local administration properties with a focus on local immune cell manipulation in favor of active and passive immunotherapies. Although applied locally, such micro/nanostructured can also activate systemic antitumor immune responses by releasing nanovaccines safely and effectively inhibiting tumor metastasis and recurrence. However, such hydrogels are mostly used as locally administered carriers to stimulate the immune cells by releasing tumor lysate, drugs, or nanovaccines. In this review, the latest developments in cancer immunotherapy are summarized using micro/nanostructured hydrogels with a particular emphasis on their function depending on the administration route. Moreover, the potential for clinical translation of these hydrogel-based cancer immunotherapies is also discussed.
Collapse
Affiliation(s)
- Esfandyar Askari
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBC V8P 5C2Canada
| | - Mahdieh Shokrollahi Barough
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBC V8P 5C2Canada
- Department of ImmunologySchool of MedicineIran University of Medical SciencesTehran1449614535Iran
- ATMP DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehran1517964311Iran
| | - Mehdi Rahmanian
- Biomaterials and Tissue Engineering DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehran1517964311Iran
| | - Nazanin Mojtabavi
- Department of ImmunologySchool of MedicineIran University of Medical SciencesTehran1449614535Iran
| | | | - Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBC V8P 5C2Canada
- Biomaterials and Tissue Engineering DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehran1517964311Iran
- Center for Advanced Materials and Related TechnologiesUniversity of VictoriaVictoriaBC V8P 5C2Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBC V8P 5C2Canada
- Center for Advanced Materials and Related TechnologiesUniversity of VictoriaVictoriaBC V8P 5C2Canada
- Center for Biomedical ResearchUniversity of VictoriaVictoriaBC V8P 5C2Canada
| |
Collapse
|
18
|
Pavelić K, Pavelić SK, Bulog A, Agaj A, Rojnić B, Čolić M, Trivanović D. Nanoparticles in Medicine: Current Status in Cancer Treatment. Int J Mol Sci 2023; 24:12827. [PMID: 37629007 PMCID: PMC10454499 DOI: 10.3390/ijms241612827] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer is still a leading cause of deaths worldwide, especially due to those cases diagnosed at late stages with metastases that are still considered untreatable and are managed in such a way that a lengthy chronic state is achieved. Nanotechnology has been acknowledged as one possible solution to improve existing cancer treatments, but also as an innovative approach to developing new therapeutic solutions that will lower systemic toxicity and increase targeted action on tumors and metastatic tumor cells. In particular, the nanoparticles studied in the context of cancer treatment include organic and inorganic particles whose role may often be expanded into diagnostic applications. Some of the best studied nanoparticles include metallic gold and silver nanoparticles, quantum dots, polymeric nanoparticles, carbon nanotubes and graphene, with diverse mechanisms of action such as, for example, the increased induction of reactive oxygen species, increased cellular uptake and functionalization properties for improved targeted delivery. Recently, novel nanoparticles for improved cancer cell targeting also include nanobubbles, which have already demonstrated increased localization of anticancer molecules in tumor tissues. In this review, we will accordingly present and discuss state-of-the-art nanoparticles and nano-formulations for cancer treatment and limitations for their application in a clinical setting.
Collapse
Affiliation(s)
- Krešimir Pavelić
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
| | - Sandra Kraljević Pavelić
- Faculty of Health Studies, University of Rijeka, Ulica Viktora Cara Emina 5, 51000 Rijeka, Croatia
| | - Aleksandar Bulog
- Teaching Institute for Public Health of Primorsko-Goranska County, Krešimirova Ulica 52, 51000 Rijeka, Croatia
- Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Andrea Agaj
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
| | - Barbara Rojnić
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
| | - Miroslav Čolić
- Clear Water Technology Inc., 13008 S Western Avenue, Gardena, CA 90429, USA;
| | - Dragan Trivanović
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
- Department of Oncology and Hematology, General Hospital Pula, Santorijeva 24a, 52200 Pula, Croatia
| |
Collapse
|
19
|
Huang M, Zhai BT, Fan Y, Sun J, Shi YJ, Zhang XF, Zou JB, Wang JW, Guo DY. Targeted Drug Delivery Systems for Curcumin in Breast Cancer Therapy. Int J Nanomedicine 2023; 18:4275-4311. [PMID: 37534056 PMCID: PMC10392909 DOI: 10.2147/ijn.s410688] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/19/2023] [Indexed: 08/04/2023] Open
Abstract
Breast cancer (BC) is the most prevalent type of cancer in the world and the main reason women die from cancer. Due to the significant side effects of conventional treatments such as chemotherapy and radiotherapy, the search for supplemental and alternative natural drugs with lower toxicity and side effects is of interest to researchers. Curcumin (CUR) is a natural polyphenol extracted from turmeric. Numerous studies have demonstrated that CUR is an effective anticancer drug that works by modifying different intracellular signaling pathways. CUR's therapeutic utility is severely constrained by its short half-life in vivo, low water solubility, poor stability, quick metabolism, low oral bioavailability, and potential for gastrointestinal discomfort with high oral doses. One of the most practical solutions to the aforementioned issues is the development of targeted drug delivery systems (TDDSs) based on nanomaterials. To improve drug targeting and efficacy and to serve as a reference for the development and use of CUR TDDSs in the clinical setting, this review describes the physicochemical properties and bioavailability of CUR and its mechanism of action on BC, with emphasis on recent studies on TDDSs for BC in combination with CUR, including passive TDDSs, active TDDSs and physicochemical TDDSs.
Collapse
Affiliation(s)
- Mian Huang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Bing-Tao Zhai
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Yu Fan
- School of Basic Medicine, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jing Sun
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Ya-Jun Shi
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Xiao-Fei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jun-Bo Zou
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jia-Wen Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Dong-Yan Guo
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| |
Collapse
|
20
|
Dam P, Celik M, Ustun M, Saha S, Saha C, Kacar EA, Kugu S, Karagulle EN, Tasoglu S, Buyukserin F, Mondal R, Roy P, Macedo MLR, Franco OL, Cardoso MH, Altuntas S, Mandal AK. Wound healing strategies based on nanoparticles incorporated in hydrogel wound patches. RSC Adv 2023; 13:21345-21364. [PMID: 37465579 PMCID: PMC10350660 DOI: 10.1039/d3ra03477a] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
The intricate, tightly controlled mechanism of wound healing that is a vital physiological mechanism is essential to maintaining the skin's natural barrier function. Numerous studies have focused on wound healing as it is a massive burden on the healthcare system. Wound repair is a complicated process with various cell types and microenvironment conditions. In wound healing studies, novel therapeutic approaches have been proposed to deliver an effective treatment. Nanoparticle-based materials are preferred due to their antibacterial activity, biocompatibility, and increased mechanical strength in wound healing. They can be divided into six main groups: metal NPs, ceramic NPs, polymer NPs, self-assembled NPs, composite NPs, and nanoparticle-loaded hydrogels. Each group shows several advantages and disadvantages, and which material will be used depends on the type, depth, and area of the wound. Better wound care/healing techniques are now possible, thanks to the development of wound healing strategies based on these materials, which mimic the extracellular matrix (ECM) microenvironment of the wound. Bearing this in mind, here we reviewed current studies on which NPs have been used in wound healing and how this strategy has become a key biotechnological procedure to treat skin infections and wounds.
Collapse
Affiliation(s)
- Paulami Dam
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Merve Celik
- Biomedical Engineering Graduate Program, TOBB University of Economics and Technology Ankara 06560 Turkey
| | - Merve Ustun
- Graduate School of Sciences and Engineering, Koç University Istanbul 34450 Turkey
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
| | - Sayantan Saha
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Chirantan Saha
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Elif Ayse Kacar
- Graduate Program of Tissue Engineering, Institution of Health Sciences, University of Health Sciences Turkey Istanbul Turkey
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
| | - Senanur Kugu
- Graduate Program of Tissue Engineering, Institution of Health Sciences, University of Health Sciences Turkey Istanbul Turkey
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
| | - Elif Naz Karagulle
- Biomedical Engineering Graduate Program, TOBB University of Economics and Technology Ankara 06560 Turkey
| | - Savaş Tasoglu
- Mechanical Engineering Department, School of Engineering, Koç University Istanbul Turkey
- Koç University Translational Medicine Research Center (KUTTAM), Koç University Istanbul Turkey
| | - Fatih Buyukserin
- Department of Biomedical Engineering, TOBB University of Economics and Technology Ankara 06560 Turkey
| | - Rittick Mondal
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
| | - Priya Roy
- Department of Law, Raiganj University North Dinajpur West Bengal India
| | - Maria L R Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária 79070900 Campo Grande Mato Grosso do Sul 70790160 Brazil
| | - Octávio L Franco
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco Campo Grande 79117900 Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília Brasília DF Brazil
| | - Marlon H Cardoso
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Universidade Federal de Mato Grosso do Sul, Cidade Universitária 79070900 Campo Grande Mato Grosso do Sul 70790160 Brazil
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco Campo Grande 79117900 Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília Brasília DF Brazil
| | - Sevde Altuntas
- Experimental Medicine Research and Application Center, University of Health Sciences Turkey Istanbul 34662 Turkey
- Department of Tissue Engineering, Institution of Health Sciences, University of Health Sciences Turkey Istanbul Turkey
| | - Amit Kumar Mandal
- Chemical Biology Laboratory, Department of Sericulture, Raiganj University North Dinajpur West Bengal India
- Centre for Nanotechnology Sciences (CeNS), Raiganj University North Dinajpur West Bengal India
| |
Collapse
|
21
|
Altuntaş E, Özkan B, Güngör S, Özsoy Y. Biopolymer-Based Nanogel Approach in Drug Delivery: Basic Concept and Current Developments. Pharmaceutics 2023; 15:1644. [PMID: 37376092 DOI: 10.3390/pharmaceutics15061644] [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: 04/27/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Due to their increased surface area, extent of swelling and active substance-loading capacity and flexibility, nanogels made from natural and synthetic polymers have gained significant interest in scientific and industrial areas. In particular, the customized design and implementation of nontoxic, biocompatible, and biodegradable micro/nano carriers makes their usage very feasible for a range of biomedical applications, including drug delivery, tissue engineering, and bioimaging. The design and application methodologies of nanogels are outlined in this review. Additionally, the most recent advancements in nanogel biomedical applications are discussed, with particular emphasis on applications for the delivery of drugs and biomolecules.
Collapse
Affiliation(s)
- Ebru Altuntaş
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| | - Burcu Özkan
- Graduate School of Natural and Applied Science, Yildiz Technical University, 34220 Istanbul, Türkiye
| | - Sevgi Güngör
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| | - Yıldız Özsoy
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Türkiye
| |
Collapse
|
22
|
Wang Z, You T, Cai C, Su Q, Cheng J, Xiao J, Duan X. Biomimetic Gold Nanostructure with a Virus-like Topological Surface for Enhanced Antigen Cross-Presentation and Antitumor Immune Response. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 36897565 DOI: 10.1021/acsami.2c21028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The internalization of antigens by dendritic cells (DCs) is the initial critical step for vaccines to activate the immune response; however, the systemic delivery of antigens into DCs is hampered by various technical challenges. Here we show that a virus-like gold nanostructure (AuNV) can effectively bind to and be internalized by DCs due to its biomimetic topological morphology, thereby significantly promoting the maturation of DCs and the cross-presentation of the model antigen ovalbumin (OVA). In vivo experiments demonstrate that AuNV efficiently delivers OVA to draining lymph nodes and significantly inhibits the growth of MC38-OVA tumors, generating a ∼80% decrease in tumor volume. Mechanistic studies reveal that the AuNV-OVA vaccine induces a remarkable increase in the rate of maturation of DCs, OVA presentation, and CD4+ and CD8+ T lymphocyte populations in both lymph node and tumor and an obvious decrease in myeloid-derived suppressor cells and regulatory T cell populations in spleen. The good biocompatibility, strong adjuvant activity, enhanced uptake of DCs, and improved T cell activation make AuNV a promising antigen delivery platform for vaccine development.
Collapse
Affiliation(s)
- Zhenyu Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tingting You
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chengyuan Cai
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qianyi Su
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jinmei Cheng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jisheng Xiao
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University; Department of Pharmacy, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xiaopin Duan
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
23
|
Zhao C, Xing Z, Wei X, Liao G, Yang D, Liu H, Fan Y. Multibiofunctional TFEB-engineered endothelial progenitor cell-derived extracellular vesicles/hydrogel system for rescuing critical limb ischemia. CHEMICAL ENGINEERING JOURNAL 2023; 460:141730. [DOI: 10.1016/j.cej.2023.141730] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
24
|
Nanotechnology for next-generation cancer immunotherapy: State of the art and future perspectives. J Control Release 2023; 356:14-25. [PMID: 36805873 DOI: 10.1016/j.jconrel.2023.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Over the past decade, immunotherapy aiming to activate an effective antitumor immune response has ushered in a new era of cancer treatment. However, the efficacy of cancer immunotherapy is limited by low response rates and high systemic toxicity. Nanotechnology is an encouraging platform for the development of next-generation cancer immunotherapy to effectively treat advanced cancer. Nanotechnology-enabled immunotherapy has remarkable advantages, ranging from the increased bioavailability and stability of immunotherapeutic agents to the enhanced activation of immune cells and favorable safety profiles. Nanotechnology-enabled immunotherapy can target solid tumors through reprogramming or stimulating immune cells (i.e., nanovaccines); modulating the immunosuppressive tumor microenvironment; or targeting tumor cells and altering their responses to immune cells to generate effective antitumor immunity. In this Oration, I introduce the advanced strategies currently being pursued by our laboratory and other groups to improve the therapeutic efficacy of cancer immunotherapy and discuss the potential challenges and future directions.
Collapse
|
25
|
Zhang Y, Chen J, Shi L, Ma F. Polymeric nanoparticle-based nanovaccines for cancer immunotherapy. MATERIALS HORIZONS 2023; 10:361-392. [PMID: 36541078 DOI: 10.1039/d2mh01358d] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Therapeutic cancer vaccines, which are designed to amplify tumor-specific T cell responses, have been envisioned as one of the most powerful tools for effective cancer immunotherapy. However, increasing the potency, quality and durability of the vaccine response remains a big challenge. In recent years, materials-based delivery systems focusing on the co-delivery of antigens and adjuvants to enhance cancer vaccination therapy have attracted increasing interest. Among various materials, polymeric nanoparticles (NPs) with different physicochemical properties which can incorporate multiple immunological cues are of great interest. In this review, the recent progress in the design and construction of both ex vivo subunit and in situ cancer vaccines using polymeric NPs is summarized. Especially, we will focus on how these NPs improve the adjuvanticity of vaccines. The design principles of polymeric NPs for ex vivo subunit cancer vaccines and in situ cancer vaccination are also discussed. Finally, we want to briefly discuss molecular chaperones in cancer immunity and the applications of our unique self-assembly mixed shell polymeric micelle-based nanochaperones for cancer vaccines.
Collapse
Affiliation(s)
- Yongxin Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Jiajing Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| | - Feihe Ma
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China.
| |
Collapse
|
26
|
Recent Reports on Polysaccharide-Based Materials for Drug Delivery. Polymers (Basel) 2022; 14:polym14194189. [PMID: 36236137 PMCID: PMC9572459 DOI: 10.3390/polym14194189] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Polysaccharides constitute one of the most important families of biopolymers. Natural polysaccharide-based drug delivery systems are of constant interest to the scientific community due to their unique properties: biocompatibility, non-toxicity, biodegradability, and high availability. These promising biomaterials protect sensitive active agents and provide their controlled release in targeted sites. The application of natural polysaccharides as drug delivery systems is also intensively developed by Polish scientists. The present review focuses on case studies from the last few years authored or co-authored by research centers in Poland. A particular emphasis was placed on the diversity of the formulations in terms of the active substance carried, the drug delivery route, the composition of the material, and its preparation method.
Collapse
|
27
|
Zheng J, Song X, Yang Z, Yin C, Luo W, Yin C, Ni Y, Wang Y, Zhang Y. Self-assembly hydrogels of therapeutic agents for local drug delivery. J Control Release 2022; 350:898-921. [PMID: 36089171 DOI: 10.1016/j.jconrel.2022.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
Abstract
Advanced drug delivery systems are of vital importance to enhance therapeutic efficacy. Among various recently developed formulations, self-assembling hydrogels composed of therapeutic agents have shown promising potential for local drug delivery owing to their excellent biocompatibility, high drug-loading efficiency, low systemic toxicity, and sustained drug release behavior. In particular, therapeutic agents self-assembling hydrogels with well-defined nanostructures are beneficial for direct delivery to the target site via injection, not only improving drug availability, but also extending their retention time and promoting cellular uptake. In brief, the self-assembly approach offers better opportunities to improve the precision of pharmaceutical treatment and achieve superior treatment efficacies. In this review, we intend to cover the recent developments in therapeutic agent self-assembling hydrogels. First, the molecular structures, self-assembly mechanisms, and application of self-assembling hydrogels are systematically outlined. Then, we summarize the various self-assembly strategies, including the single therapeutic agent, metal-coordination, enzyme-instruction, and co-assembly of multiple therapeutic agents. Finally, the potential challenges and future perspectives are discussed. We hope that this review will provide useful insights into the design and preparation of therapeutic agent self-assembling hydrogels.
Collapse
Affiliation(s)
- Jun Zheng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Xianwen Song
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Zhaoyu Yang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chao Yin
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Weikang Luo
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunyang Yin
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yaqiong Ni
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yi Zhang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China.
| |
Collapse
|
28
|
Yang M, Olaoba OT, Zhang C, Kimchi ET, Staveley-O’Carroll KF, Li G. Cancer Immunotherapy and Delivery System: An Update. Pharmaceutics 2022; 14:1630. [PMID: 36015256 PMCID: PMC9413869 DOI: 10.3390/pharmaceutics14081630] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 02/06/2023] Open
Abstract
With an understanding of immunity in the tumor microenvironment, immunotherapy turns out to be a powerful tool in the clinic to treat many cancers. The strategies applied in cancer immunotherapy mainly include blockade of immune checkpoints, adoptive transfer of engineered cells, such as T cells, natural killer cells, and macrophages, cytokine therapy, cancer vaccines, and oncolytic virotherapy. Many factors, such as product price, off-target side effects, immunosuppressive tumor microenvironment, and cancer cell heterogeneity, affect the treatment efficacy of immunotherapies against cancers. In addition, some treatments, such as chimeric antigen receptor (CAR) T cell therapy, are more effective in treating patients with lymphoma, leukemia, and multiple myeloma rather than solid tumors. To improve the efficacy of targeted immunotherapy and reduce off-target effects, delivery systems for immunotherapies have been developed in past decades using tools such as nanoparticles, hydrogel matrix, and implantable scaffolds. This review first summarizes the currently common immunotherapies and their limitations. It then synopsizes the relative delivery systems that can be applied to improve treatment efficacy and minimize side effects. The challenges, frontiers, and prospects for applying these delivery systems in cancer immunotherapy are also discussed. Finally, the application of these approaches in clinical trials is reviewed.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
| | - Olamide Tosin Olaoba
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65212, USA
| | - Eric T. Kimchi
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|