1
|
Sindeeva OA, Kozyreva ZV, Abdurashitov AS, Sukhorukov GB. Engineering colloidal systems for cell manipulation, delivery, and tracking. Adv Colloid Interface Sci 2025; 340:103462. [PMID: 40037017 DOI: 10.1016/j.cis.2025.103462] [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: 10/14/2024] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
Men-made colloidal systems are widely presented across various aspects of biomedical science. There is a strong demand for engineering colloids to tailor their functions and properties to meet the requirements of biological and medical tasks. These requirements are not only related to size, shape, capacity to carry bioactive compounds as drug delivery systems, and the ability to navigate via chemical and physical targeting. Today, the more challenging aspects of colloid design are how the colloidal particles interact with biological cells, undergo internalization by cells, how they reside in the cell interior, and whether we can explore cells with colloids, intervene with biochemical processes, and alter cell functionality. Cell tracking, exploitation of cells as natural transporters of internalized colloidal carriers loaded with drugs, and exploring physical methods as external triggers of cell functions are ongoing topics in the research agenda. In this review, we summarize recent advances in these areas, focusing on how colloidal particles interact and are taken up by mesenchymal stem cells, dendritic cells, neurons, macrophages, neutrophils and lymphocytes, red blood cells, and platelets. The engineering of colloidal vesicles with cell membrane fragments and exosomes facilitates their application. The perspectives of different approaches in colloid design, their limitations, and obstacles on the biological side are discussed.
Collapse
Affiliation(s)
- Olga A Sindeeva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| | - Zhanna V Kozyreva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Arkady S Abdurashitov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia; Life Improvement by Future Technologies (LIFT) Center, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Gleb B Sukhorukov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| |
Collapse
|
2
|
Dong H, Fa J, Yan M, Tan Y, Cheng W, Huang J, Ling Z. Development and analysis of chitin/cellulose reinforced galactomannan fluidic hydrogel for drug delivery application. J Colloid Interface Sci 2025; 686:701-710. [PMID: 39919515 DOI: 10.1016/j.jcis.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/23/2025] [Accepted: 02/01/2025] [Indexed: 02/09/2025]
Abstract
Rising demand for advanced materials in biomedical applications has made the development of effective hydrogels a critical focus, especially for drug delivery. Herein, a novel fluidic hydrogel with high drug-loading (>95 %) capacity was developed for controlled release of acetylsalicylic acid. Nature-derived galactomannan (GM) was served as the hydrogel matrix, and chitin nanocrystals (ChNC) and cellulose nanocrystals (CNC) were used to enhance structure and antimicrobial properties. Physical crosslinking of the components were tuned to optimize the porosity, flowability, and mechanical strength of the materials, as well as improving rheology and enabling rapid self-healing within 60 s. Co-culturing experiments demonstrate excellent biocompatibility of both ChNC@GM and CNC@GM hydrogels. Moreover, ChNC@GM shows superior 82 % antimicrobial activity, and the hydrogel exhibits pH-responsive drug release. Therefore, the proposed fluidic hydrogel may act as promising material for applications fields of wound healing, drug delivery, and biomedical engineering, etc.
Collapse
Affiliation(s)
- Hanqi Dong
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Jingjing Fa
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengxing Yan
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Yang Tan
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Wenbo Cheng
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianfeng Huang
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhe Ling
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China.
| |
Collapse
|
3
|
Eskandari A, Safavi SN, Sahrayi H, Alizadegan D, Eskandarisani M, Javanmard A, Tajik M, Sadeghi Z, Toutounch A, Yeganeh FE, Noorbazargan H. Antimicrobial and antibiofilm activity of prepared thymol@UIO-66 and thymol/ZnONPs@UIO-66 nanoparticles against Methicillin-resistant Staphylococcus aureus: A synergistic approach. Colloids Surf B Biointerfaces 2025; 249:114529. [PMID: 39879671 DOI: 10.1016/j.colsurfb.2025.114529] [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: 10/10/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/31/2025]
Abstract
This study introduces a novel approach to enhance the antibacterial properties of UIO-66 by incorporating both Thymol and ZnO nanoparticles within its framework which represents a significant advancement like exhibiting a synergistic antibacterial effect, providing a prolonged and controlled release, and mitigating cytotoxicity associated with the release of free ZnO nanoparticles by combining these two antimicrobial agents within a single, well-defined metal-organic framework. UIO-66 frameworks are investigated as carriers for the natural antimicrobial agent, Thymol, and ZnONPs offering a novel drug delivery system for antibacterial applications. Results demonstrated 132, 90, 184, and 223 nm sizes for UIO-66, ZnONPs, UIO-66 encapsulated Thymol, and UIO-66 encapsulated both Thymol and ZnONPs, respectively. Successful encapsulation of the antibacterial drug with a high entrapment efficiency of 64 % for Thymol was approved, and 49 % in-vitro release of Thymol was achieved for 72 hours. In-vitro antibacterial assays revealed promising results, with the drug-loaded nanoparticles exhibiting significantly lower MIC values and enhanced bactericidal activity against S. Aureus bacterial strains compared to the free drug, as demonstrated by agar disk diffusion and time-kill assays. MIC values reduced from a range of 31.25-250 µg/ml for free Thymol and 12.5-100 µg/ml for free ZnONPs to 3.9-62.5 µg/ml for Thymol@UIO-66 and 1.95-15.63 µg/ml for Thymol/ZnONPs@UIO-66. According to the results, the mixture of both Thymol and ZnONPs had 41 % and 16 % more antibiofilm activities in comparison with free Thymol and free ZnONPs, respectively. Furthermore, Thymol@UIO-66 had 25 % higher antibiofilm activities relative to not-encapsulated Thymol and ZnONPs, and this improvement was even 46 % more in Thymol/ZnONPs@UIO-66 in comparison with Thymol@UIO-66. Overall, this study demonstrates the potential of Thymol/ZnONPs@UIO-66 frameworks as a promising drug delivery platform for effective antibacterial therapy. This approach to overcome antibiotic resistance and improve treatment efficacy potentially.
Collapse
Affiliation(s)
- Alireza Eskandari
- CTERC, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Nooshin Safavi
- Department of Polymer Engineering, Faculty of Engineering, Qom University of Technology, Qom, Iran
| | - Hamidreza Sahrayi
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Dorsa Alizadegan
- Faculty of Pharmacy, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | | | - Alireza Javanmard
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA 16802-1503, United States
| | - Mohammadreza Tajik
- Biomedical Engineering Department, Carnegie Mellon University, Pittsburgh, PA 15219, United States
| | - Zohre Sadeghi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Disease, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Arvin Toutounch
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Hassan Noorbazargan
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Mousavi R, Soltani M, Souri M. Microneedle patch capable of dual drug release for drug delivery to brain tumors. Drug Deliv Transl Res 2025; 15:1567-1594. [PMID: 39186235 DOI: 10.1007/s13346-024-01696-6] [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] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
Primary brain tumors are mostly managed using surgical resection procedures. Nevertheless, in certain cases, a thin layer of tumors may remain outside of the resection process due to the possibility of permanent injury; these residual tumors expose patients to the risk of tumor recurrence. This study has introduced the use of microneedle patches implanted after surgery with a dual-release mechanism for the administration of doxorubicin. The proposed patches possess the capability to administer drugs directly to the residual tumors and initiate chemotherapy immediately following surgical procedures. Three-dimensional simulation of drug delivery to residual tumors in the brain has been performed based on a finite element method. The impact of four important parameters on drug delivery has been investigated, involving the fraction of drug released in the burst phase, the density of microneedles on the patch, the length of microneedles, and the microvascular density of the tumor. The simulation findings indicate that lowering the fraction of drug released in the initial burst phase reduces the maximum average concentration, but the sustained release that continues for a longer period, increasing the bioavailability of free drug. However, the area under curve (AUC) for different release rates remains unchanged due to the fact that an identical dose of drug is supplied in each instance. By increasing the density of microneedles on the patch, concentration accumulation is provided over an extensive region of tumor, which in turn induces more cancer cell death. A comparative analysis of various lengths reveals that longer microneedles facilitate profound penetration into the tumor layers and present better therapeutic response due to extensive area of the tumor which is exposure to chemotherapeutic drugs. Furthermore, high microvascular density, as a characteristic of the tumor microenvironment, is shown to have a significant impact on the blood microvessels drainage of drugs and consequently lower therapeutic response outcome. Our approach offers a computational framework for creating localized drug delivery systems and addressing the challenges related to residual brain tumors.
Collapse
Affiliation(s)
- Robab Mousavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada.
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada.
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada.
- Centre for Sustainable Business, International Business University, Toronto, Canada.
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
5
|
Huang D, Yu Q, Yang K, Li X, Huang C, Yang X, Wu C, Cao C, Zhang L, Zhu D, Li J. Nonfouling Core-Shell Microneedle for Sequential and Sustained Drug Release: Enhancing Synergistic Photothermal Chemotherapy in Melanoma Treatment. ACS APPLIED BIO MATERIALS 2025; 8:3356-3374. [PMID: 40180602 DOI: 10.1021/acsabm.5c00120] [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: 04/05/2025]
Abstract
Melanoma is a highly aggressive and metastatic malignancy, where current treatment methods often result in damage to healthy tissues, suboptimal therapeutic outcomes, and immune-related side effects. Microneedles, as a drug delivery system, offer advantages such as localized administration, minimal invasiveness, and high delivery efficiency. In this study, we first synthesized tetradecyl-thiol-grafted PAMAM dendrimers, which significantly enhanced cellular uptake and enabled sustained release of doxorubicin (DOX), improving cumulative drug release efficiency. Based on this, we developed a core-shell structured zwitterionic polymer-based microneedle delivery system. The outer shell, loaded with the photothermal agent indocyanine green (ICG), achieved precise photothermal therapy under near-infrared irradiation, effectively targeting melanoma tissues. The inner core, composed of a zwitterionic polymer matrix, encapsulated DOX-loaded dendrimers, enabling controlled and prolonged drug release through gradual polymer swelling and dendrimer expansion. Experiments show that the microneedle drug delivery system based on PAMAM dendrimer grafted with tetradecyl mercaptan and zwitterionic polymer has excellent anti protein adsorption properties, and it can minimize the cytotoxicity of carrier and improve the efficiency of drug delivery. This system effectively inhibited tumor growth through synergistic photothermal-chemotherapy, reducing systemic toxicity and improving drug bioavailability. This microneedle platform provides a promising strategy for targeted and synergistic melanoma therapy, offering a high-efficiency and low-toxicity treatment alternative.
Collapse
Affiliation(s)
- Dongyu Huang
- Department of Polymer Science, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Qingyu Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Kaiyue Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xiuqiang Li
- Department of Polymer Science, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Chenlu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xinyu Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Chaoxiong Wu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Cheng Cao
- Department of Polymer Science, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Linhua Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Junjie Li
- Department of Polymer Science, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300350, China
| |
Collapse
|
6
|
Liu N, Wang X, Wang Z, Kan Y, Fang Y, Gao J, Kong X, Wang J. Nanomaterials-driven in situ vaccination: a novel frontier in tumor immunotherapy. J Hematol Oncol 2025; 18:45. [PMID: 40247328 PMCID: PMC12007348 DOI: 10.1186/s13045-025-01692-4] [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: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
In situ vaccination (ISV) has emerged as a promising strategy in cancer immunotherapy, offering a targeted approach that uses the tumor microenvironment (TME) to stimulate an immune response directly at the tumor site. This method minimizes systemic exposure while maintaining therapeutic efficacy and enhancing safety. Recent advances in nanotechnology have enabled new approaches to ISV by utilizing nanomaterials with unique properties, including enhanced permeability, retention, and controlled drug release. ISV employing nanomaterials can induce immunogenic cell death and reverse the immunosuppressive and hypoxic TME, thereby converting a "cold" tumor into a "hot" tumor and facilitating a more robust immune response. This review examines the mechanisms through which nanomaterials-based ISV enhances anti-tumor immunity, summarizes clinical applications of these strategies, and evaluates its capacity to serve as a neoadjuvant therapy for eliminating micrometastases in early-stage cancer patients. Challenges associated with the clinical translation of nanomaterials-based ISV, including nanomaterial metabolism, optimization of treatment protocols, and integration with other therapies such as radiotherapy, chemotherapy, and photothermal therapy, are also discussed. Advances in nanotechnology and immunotherapy continue to expand the possible applications of ISV, potentially leading to improved outcomes across a broad range of cancer types.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yonemori Kan
- Department of Medical Oncology, National Cancer Center Hospital (NCCH), 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518127, China.
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
7
|
Kumar K, Saini M, Rani V, Mishra M, Priya, Jatin, Tiwari S, Mishra B, Chawla R. Crafting a Molecular Trojan Horse: Sialic Acid-Modified PLGA Nanoparticles for Targeted Lung Cancer Therapy. Mol Pharm 2025; 22:1816-1830. [PMID: 40021483 DOI: 10.1021/acs.molpharmaceut.4c00957] [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: 03/03/2025]
Abstract
The glycan receptors prominently expressed on the surface of lung cancer cells offers promising targets for drug delivery. The prepared gemcitabine (GB)-loaded PLGA-NPs and sialic acid (Siac)-modified PLGA-NPs exhibited a uniform polydispersity index (PDI) value below 0.3, a particle size under 200 nm, and negative zeta potentials ranging from -17.45 to -21.45 mV. Entrapment efficiency (% EE) and drug loading values exceeded 70% and 8%, respectively. SEM and TEM showed that the particles were uniformly dispersed with a spherical shape. FTIR, XRD, TGA, and DSC analyses indicated the physiochemical stability of the drug within the nanoformulations. Controlled (26.92 to 31.64% within 24 h at pH 7.4) and pH-sensitive (36.80 to 40.25% within 24 h at pH 5.5) GB release were observed for the different formulations of PLGA-NPs. The MTT cytotoxicity assay revealed IC50 values for the GB control, GB-PLGA-NPs, and GB-PLGA-Siac-NPs as 13.65 ± 1.20, 8.14 ± 1.24, and 4.16 ± 1.05 μg/mL, respectively. The Co6-GB-PLGA-Siac-NPs showed significantly higher cellular uptake than that of the Co6-GB control (p < 0.001) and Co6-GB-PLGA-NPs (p < 0.01) respectively. Pharmacokinetic profiles indicated higher AUC values (ng·h/mL) for GB-PLGA-Siac-NPs (8355.07 ± 2006.45) compared to GB-PLGA-NPs (6145.58 ± 969.25) and the GB control (1510.72 ± 81.08), resulting in higher bioavailability of GB-PLGA-Siac-NPs. Biodistribution studies confirmed superior localization of DiD-GB-PLGA-Siac-NPs, indicated by radiant efficiency signal on B[a]P induced lung cancerous tissues relative to DiD-GB-PLGA-NPs after 1 h (p < 0.001), 4 h (p < 0.01), and 12 h (p < 0.001), which could be attributed to their ability to target glycans. In vivo anticancer efficacy in a B[a]P-induced lung cancer mice model depicted that GB-PLGA-Siac-NPs effectively inhibited lung cancer cells and reduced systemic toxicity, as evidenced by the average number of lung cancer cells, body weight values, survival analysis, biochemical parameters associated with organs (such as the liver and kidney), and histopathological analysis. Therefore, GB-loaded Siac-coated PLGA nanoparticles could serve as an efficient vehicle for GB delivery via targeting glycan receptors in lung cancer therapy.
Collapse
Affiliation(s)
- Krishan Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Manjit Saini
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Varsha Rani
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
- Jichi Medical University Shimostuke, Shimotsuke, Tochigi Pefecture 329-0431, Japan
| | - Mohini Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Priya
- Department of Pharmacy, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Jatin
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Siddharth Tiwari
- School of Pharmaceutical Sciences, CSJM University, Kanpur, Uttar Pradesh 208024, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Ruchi Chawla
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
8
|
Razavi ZS, Razavi FS, Alizadeh SS. Inorganic nanoparticles and blood-brain barrier modulation: Advancing targeted neurological therapies. Eur J Med Chem 2025; 287:117357. [PMID: 39947054 DOI: 10.1016/j.ejmech.2025.117357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
The blood-brain barrier (BBB) is a protective barrier that complicates the treatment of neurological disorders. Pharmaceutical compounds encounter significant challenges in crossing the central nervous system (CNS). Nanoparticles (NPs) are promising candidates for treating neurological conditions as they help facilitate drug delivery. This review explores the diverse characteristics and mechanisms of inorganic NPs (INPs), including metal-based, ferric-oxide, and carbon-based nanoparticles, which facilitate their passage through the BBB. Emphasis is placed on the physicochemical properties of NPs such as size, shape, surface charge, and surface modifications and their role in enhancing drug delivery efficacy, reducing immune clearance, and improving BBB permeability. Specific synthesis approaches are demonstrated, with an emphasis on the influence of each one on NP property, biological activity and the capability of an NP for its intended application. As for the advances in the field, the review emphasizes those characterized the NP formulation and surface chemistry that conquered the BBB and tested the need for its alteration. Current findings indicate that NP therapy can in the future enable effective targeting of specific brain disorders and eventually evolve this drug delivery system, which would allow for lower doses with less side effects.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
| | - Fateme Sadat Razavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | |
Collapse
|
9
|
Sokač K, Vrban L, Liović M, Škorić I, Vianello R, Bregović N, Žižek K. Controlled release of dasatinib from cyclodextrin-based inclusion complexes by mechanochemistry: A computational and experimental study. Int J Pharm 2025; 675:125552. [PMID: 40187706 DOI: 10.1016/j.ijpharm.2025.125552] [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: 01/23/2025] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Dasatinib, a potent tyrosine kinase inhibitor, is widely used to treat chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. However, its poor aqueous solubility and high first-pass metabolism result in limited oral bioavailability and potentially severe side effects, such as cardiotoxicity, hepatotoxicity, and pulmonary complications, which are intensified by rapid concentration peaks in the bloodstream. To address these challenges, this study examines the development of a controlled-release formulation of dasatinib using cyclodextrins as macrocyclic receptors to form inclusion complexes. Cyclodextrins, known for their ability to form host-guest complexes, enhance drug solubility and stability while enabling controlled drug release and aligning with green chemistry principles when synthesized mechanochemically. Different solid-state and solution-based characterization methods confirmed successful complexation and drug amorphization. Additionally, molecular dynamics simulations provided valuable insights into the binding interactions between dasatinib and cyclodextrins in both gas-phase and aqueous medium, simulating experimental conditions in the absence of a solvent and a physiological environment. Formulated tablets exhibited enhanced solubility and improved in vitro release profiles, suggesting a potential reduction in adverse side effects and improved patient compliance. The results demonstrate the efficacy of cyclodextrins as carriers for dasatinib, highlighting their potential to improve the drug's therapeutic profile in leukemia treatment by facilitating a steady, controlled release and minimizing toxicity.
Collapse
Affiliation(s)
- Katarina Sokač
- University of Zagreb, Faculty of Chemical Engineering and Technology, Department of Mechanical and Thermal Process Engineering, Trg Marka Marulića 19, 10 000 Zagreb, Croatia
| | - Lucija Vrban
- Laboratory for the Computational Design and Synthesis of Functional Materials, Ruđer Bošković Institute, Bijenička cesta 54, 10 000 Zagreb, Croatia
| | - Marin Liović
- University of Zagreb, Faculty of Science, Department of Chemistry, Horvatovac 102a, 10 000 Zagreb, Croatia
| | - Irena Škorić
- University of Zagreb, Faculty of Chemical Engineering and Technology, Department of Organic Chemistry, Trg Marka Marulića 19, 10 000 Zagreb, Croatia
| | - Robert Vianello
- Laboratory for the Computational Design and Synthesis of Functional Materials, Ruđer Bošković Institute, Bijenička cesta 54, 10 000 Zagreb, Croatia
| | - Nikola Bregović
- University of Zagreb, Faculty of Science, Department of Chemistry, Horvatovac 102a, 10 000 Zagreb, Croatia
| | - Krunoslav Žižek
- University of Zagreb, Faculty of Chemical Engineering and Technology, Department of Mechanical and Thermal Process Engineering, Trg Marka Marulića 19, 10 000 Zagreb, Croatia.
| |
Collapse
|
10
|
Luo S, Zhao C, Wang R, Wu D. Sequential drug release nanocomposites for synergistic therapy in disease treatment. J Mater Chem B 2025; 13:4313-4329. [PMID: 40104923 DOI: 10.1039/d5tb00026b] [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: 03/20/2025]
Abstract
Time-sequenced drug release, or sequential drug release, represents a pivotal strategy in the synergistic treatment of diseases using nanocomposites. Achieving this requires the rational integration of multiple therapeutic agents within a single nanocomposite, coupled with precise time-controlled release mechanisms. These nanocomposites offer many advantages, including enhanced therapeutic synergy, reduced side effects, attenuated adverse interactions, improved stability and optimized drug utilization. Consequently, research in the field of drug delivery and synergistic therapy has become increasingly important. Currently, sequential drug release research is still in the data collection and basic research stages, and its potential has not yet been fully explored. Although prior studies have explored the sequential drug release strategy in various contexts, a comprehensive review of the underlying mechanisms and their applications in nanocomposites remains scarce. This review categorizes different types of sequential drug release strategies and summarizes diverse nanocomposites, focusing on both physical approaches driven by structural variations and chemical methods based on stimulus-responsive mechanisms. Furthermore, we highlight the major applications of sequential drug release strategies in the treatment of various diseases and detail their therapeutic efficacy. Finally, emerging trends and challenges in advancing sequential drug release strategies based on nanocomposites for disease treatment are also discussed.
Collapse
Affiliation(s)
- Siyuan Luo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| | - Chenyu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| | - Rong Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| |
Collapse
|
11
|
Zhou K, Liu Y, Tang C, Zhu H. Pancreatic Cancer: Pathogenesis and Clinical Studies. MedComm (Beijing) 2025; 6:e70162. [PMID: 40182139 PMCID: PMC11965705 DOI: 10.1002/mco2.70162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy, with pancreatic ductal adenocarcinoma (PDAC) being the most common and aggressive subtype, characterized by late diagnosis, aggressive progression, and resistance to conventional therapies. Despite advances in understanding its pathogenesis, including the identification of common genetic mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and dysregulated signaling pathways (e.g., KRAS-MAPK, PI3K-AKT, and TGF-β pathways), effective therapeutic strategies remain limited. Current treatment modalities including chemotherapy, targeted therapy, immunotherapy, radiotherapy, and emerging therapies such as antibody-drug conjugates (ADCs), chimeric antigen receptor T (CAR-T) cells, oncolytic viruses (OVs), cancer vaccines, and bispecific antibodies (BsAbs), face significant challenges. This review comprehensively summarizes these treatment approaches, emphasizing their mechanisms, limitations, and potential solutions, to overcome these bottlenecks. By integrating recent advancements and outlining critical challenges, this review aims to provide insights into future directions and guide the development of more effective treatment strategies for PC, with a specific focus on PDAC. Our work underscores the urgency of addressing the unmet needs in PDAC therapy and highlights promising areas for innovation in this field.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yingping Liu
- Department of RadiotherapyCancer HospitalChinese Academy of Medical SciencesBeijingChina
| | - Chuanyun Tang
- The First Clinical Medical College of Nanchang UniversityNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
12
|
Huang R, Yu J, Zhang B, Li X, Liu H, Wang Y. Emerging COX-2 inhibitors-based nanotherapeutics for cancer diagnosis and treatment. Biomaterials 2025; 315:122954. [PMID: 39549439 DOI: 10.1016/j.biomaterials.2024.122954] [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: 07/28/2024] [Revised: 10/27/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Increasing evidence has showed that tumorigenesis is closely linked to inflammation, regulated by multiple signaling pathways. Among these, the cyclooxygenase-2/prostaglandin E2 (COX-2/PGE2) axis plays a crucial role in the progression of both inflammation and cancer. Inhibiting the activity of COX-2 can reduce PGE2 secretion, thereby suppressing tumor growth. Therefore, COX-2 inhibitors are considered potential therapeutic agents for cancers. However, their clinical applications are greatly hindered by poor physicochemical properties and serious adverse effects. Fortunately, the advent of nanotechnology offers solutions to these limitations, enhancing drug delivery efficiency and mitigating adverse effects. Given the considerable progress in this area, it is timely to review emerging COX-2 inhibitors-based nanotherapeutics for cancer diagnosis and therapy. In this review, we first outline the various antineoplastic mechanisms of COX-2 inhibitors, then comprehensively summarize COX-2 inhibitors-based nanotherapeutics for cancer monotherapy, combination therapy, and diagnosis. Finally, we highlight and discuss future perspectives and challenges in the development of COX-2 inhibitors-based nanomedicine.
Collapse
Affiliation(s)
- Ruiping Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Baoyue Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Xin Li
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
13
|
Madineh H, Mansourinia F, Zarrintaj P, Poostchi M, Gnatowski P, Kucinska-Lipka J, Ghaffari M, Hasanin MS, Chapi S, Yazdi MK, Ashrafizadeh M, Bączek T, Saeb MR, Wang G. Stimuli-responsive delivery systems using carbohydrate polymers: A review. Int J Biol Macromol 2025; 310:142648. [PMID: 40174846 DOI: 10.1016/j.ijbiomac.2025.142648] [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: 09/15/2024] [Revised: 03/08/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Carbohydrate polymers, including Chitosan, Cellulose, Starch, Dextran, Pectin, Alginate, and Hyaluronic Acid, have been considered as stimuli-responsive biopolymers demonstrating significant potential for drug delivery approaches. Relying on the specific design and fabrication, such biopolymers are able to respond to fluctuations in pH, temperature, or enzymatic activity. This review investigates stimuli-responsive biopolymers, known as carbohydrate polymers, mainly chitosan, cellulose, and alginate, utilized as drug delivery approaches, emphasizing that these stimuli-responsive biopolymers accelerate controlled drug release. The pH-responsive delivery systems selectively target acidic tumor microenvironments, while temperature-responsive materials provide precise control for drug release produced by hyperthermia. Light-responsive biopolymers provide spatial and temporal control, providing appropriate for targeted therapy. Redox-responsive structures are especially efficient in responding to elevated glutathione (GSH) in tumor microenvironment, facilitating targeted drug release. Electro- and magnetic-responsive systems provide remote control functionalities, improving the accuracy of drug administration. The incorporation of multi-stimuli-responsive mechanisms implies a remarkable progression in drug delivery, providing a more versatile and adaptable framework for therapeutic applications. Accordingly, the future research on carbohydrate polymer-based stimuli-responsive delivery systems should focus on improving the responsiveness and targeting efficacy through complicated optimization of features and performance of carbohydrate polymers, where the integration of multifunctional moieties facilitates transformation of targeted drugs for broader biological functions.
Collapse
Affiliation(s)
- Hossein Madineh
- Polymer Engineering Department, Chemical Engineering Faculty, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Mansourinia
- Polymer Engineering Department, Chemical Engineering Faculty, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Poostchi
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Przemysław Gnatowski
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland; Department of Environmental Toxicology, Faculty of Health Sciences with the Institute of Maritime and Tropical Medicine, Medical University of Gdańsk, Dębowa 23A, Gdańsk 80-204, Poland.
| | - Justyna Kucinska-Lipka
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Mehdi Ghaffari
- Polymer Group, Faculty of Technical and Engineering, Golestan University, P. O. Box 155, Gorgan, Golestan, Iran
| | - Mohamed S Hasanin
- Cellulose and Paper Department, National Research Centre, 33 El Bohouth St., Cairo 12622, Egypt; Department of Polymer and Biomaterials Science, West Pomeranian University of Technology in Szczecin, Al. Piastow 45, 70-311 Szczecin, Poland
| | - Sharanappa Chapi
- Department of Physics, B.M.S. College of Engineering, Basavanagudi - 560019, Bengaluru, Karnataka, India
| | - Mohsen Khodadadi Yazdi
- Division of Electrochemistry and Surface Physical Chemistry, Faculty of Applied Physics and Mathematics, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdańsk, Poland; Advanced Materials Center, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland
| | - Mohammad Reza Saeb
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland.
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China.
| |
Collapse
|
14
|
Kamrani S, Naseramini R, Khani P, Razavi ZS, Afkhami H, Atashzar MR, Nasri F, Alavimanesh S, Saeidi F, Ronaghi H. Mesenchymal stromal cells in bone marrow niche of patients with multiple myeloma: a double-edged sword. Cancer Cell Int 2025; 25:117. [PMID: 40140850 PMCID: PMC11948648 DOI: 10.1186/s12935-025-03741-x] [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: 10/24/2024] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy defined by the abnormal proliferation and accumulation of plasma cells (PC) within the bone marrow (BM). While multiple myeloma impacts the bone, it is not classified as a primary bone cancer. The bone marrow microenvironment significantly influences the progression of myeloma and its treatment response. Mesenchymal stromal cells (MSCs) in this environment engage with myeloma cells and other bone marrow components via direct contact and the secretion of soluble factors. This review examines the established roles of MSCs in multiple facets of MM pathology, encompassing their pro-inflammatory functions, contributions to tumor epigenetics, effects on immune checkpoint inhibitors (ICIs), influence on reprogramming, chemotherapy resistance, and senescence. This review investigates the role of MSCs in the development and progression of MM.
Collapse
Affiliation(s)
- Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Naseramini
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Mohammad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hossein Ronaghi
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
15
|
You T, Zhang S. Recent advances in PLGA polymer nanocarriers for ovarian cancer therapy. Front Oncol 2025; 15:1526718. [PMID: 40196734 PMCID: PMC11973302 DOI: 10.3389/fonc.2025.1526718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecologic malignancy worldwide, and early diagnosis and effective treatment have been the focus of research in this field. It is because of its late diagnosis, acquired resistance mechanisms, and systemic toxicity of chemotherapeutic agents that the treatment of ovarian cancer is challenging. Combination chemotherapy can potentially improve therapeutic efficacy by activating multiple downstream pathways to overcome resistance and reduce the required dose. In recent years, PLGA-lipid hybrid nanoparticles have demonstrated their potential as an emerging drug delivery system for treating ovarian cancer. PLGA (poly (lactic-co-glycolic acid) has become a highly sought-after biomaterial for the clinical translation of adjustable drug delivery regimens due to its biodegradability, biocompatibility, and multifunctionality, coupled with controlled drug release, which can effectively overcome multidrug resistance and improve the efficiency of chemotherapy. Combination therapies are gradually becoming an ideal alternative to traditional drug formulations. The application of nanoparticles not only improves the therapeutic effect but also reduces the side effects, which provides strong support for personalized precision medicine. We review polymeric nanoparticle carriers for drug combinations used in the treatment of ovarian cancer, particularly the combination of paclitaxel analogs (commonly used first-line therapy for ovarian cancer) with other small molecule therapeutic agents and cavitation combination therapy under ultrasound targeting (Figure 1). The elucidation of these issues will provide a theoretical basis for future exploration of novel NNDDS targeting GRPR for anti-OC therapy. This review presents research on recent advances in PLGA polymer nanoparticles in ovarian cancer, focusing on the use of PLGA degradable microspheres for loading chemotherapeutic agents and ultrasound combination therapy.
Collapse
Affiliation(s)
| | - Shengmin Zhang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
16
|
Su D, Lai X, Lin Z, Xu Y, Fu Z, Chen J, Wu X. Innovative nanodelivery systems for targeted breast cancer therapy: overcoming drug delivery challenges and exploring future perspectives. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04039-5. [PMID: 40095056 DOI: 10.1007/s00210-025-04039-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Cancer chemotherapy is often limited by insufficient drug targeting, which can cause severe adverse effects on healthy tissues. The advent of nanodelivery systems offers a potential solution to this issue. It intricately enhances drug accumulation in tumor regions, while optimizing drug solubility, ensuring colloidal stability, and promoting cellular uptake, significantly improving the precision and efficacy of treatment, opening a gentler yet more effective new path for cancer therapy. This article begins with the pathogenesis of breast cancer and extends to the current treatment methods and their shortcomings, exploring in-depth the targeting therapeutic effects of five innovative nanodelivery technologies used in the treatment of breast cancer in recent years. Finally, it discusses the potential opportunities and challenges that nanodelivery systems may face in future development.
Collapse
Affiliation(s)
- Dandan Su
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China
| | - Xiaolin Lai
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China
| | - Zhizhe Lin
- Shanghai Wei Er Lab, Shanghai, 201707, China
| | - Youfa Xu
- Shanghai Wei Er Lab, Shanghai, 201707, China
| | - Zhiqin Fu
- Shanghai Wei Er Lab, Shanghai, 201707, China
| | - Jianming Chen
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China.
| | - Xin Wu
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China.
- Shanghai Wei Er Lab, Shanghai, 201707, China.
| |
Collapse
|
17
|
Zhang J, Yang Z, Liu Y, Liu Y, Qu J, Pan X. Recent Advances in Smart Linkage Strategies for Developing Drug Conjugates for Targeted Delivery. Top Curr Chem (Cham) 2025; 383:13. [PMID: 40080285 DOI: 10.1007/s41061-025-00497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Targeted drug delivery systems effectively solve the problem of off-target toxicity of chemotherapeutic drugs by combining chemotherapeutic drugs with antibodies or peptides, thereby promoting drug targeting to the tumor site and bringing further hope for cancer treatment. The development of stimulus-responsive smart linkage technologies has led to the emergence of drug conjugates. Linkage technologies play a crucial role in the design, synthesis, and in vivo circulation of drug conjugates, as they determine the release of cytotoxic drugs from the conjugates and their subsequent therapeutic efficacy. This article reviews some of the smart linkage strategies used in designing drug conjugates, with a focus on the tumor microenvironment and exogenous stimuli as conditions influencing controlled drug release. This review introduces linker classifications and cleavage mechanisms, discusses modular linkers that promote the efficient synthesis of conjugates, and discusses the differences between linkage strategies. Furthermore, this article focuses on the implementation of self-assembly in drug conjugates, which is currently of great interest. Related concepts are introduced and relevant examples of their applications are provided. Furthermore, a comprehensive discourse is presented on the challenges that may arise in the research and clinical implementation of diverse linkage strategies, along with the associated enhancement measures. Finally, the factors that should be considered when designing linkage strategies for drug conjugates are summarized, offering strategies and ideas for scientists involved in drug conjugate research. It is particularly noteworthy that appropriate linkage strategies allow for the intracellular release of drugs after internalization of the conjugates, thereby maximizing their tumor cell-killing effect.
Collapse
Affiliation(s)
- Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zeyu Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yu Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jingkun Qu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
18
|
Salazar Sandoval S, Díaz-Saldívar P, Araya I, Celis F, Cortés-Arriagada D, Riveros A, Rojas-Romo C, Jullian C, Silva N, Yutronic N, Kogan MJ, Jara P. Controlled Release of the Anticancer Drug Cyclophosphamide from a Superparamagnetic β-Cyclodextrin Nanosponge by Local Hyperthermia Generated by an Alternating Magnetic Field. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13001-13017. [PMID: 38640460 DOI: 10.1021/acsami.3c18038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
A β-cyclodextrin (β-CD) nanosponge (NS) was synthesized using diphenyl carbonate (DPC) as a cross-linker to encapsulate the antitumor drug cyclophosphamide (CYC), thus obtaining the NSs-CYC system. The formulation was then associated with magnetite nanoparticles (MNPs) to develop the MNPs-NSs-CYC ternary system. The formulations mentioned above were characterized to confirm the deposition of the MNPs onto the organic matrix and that the superparamagnetic nature of the MNPs was preserved upon association. The association of the MNPs with the NSs-drug complex was confirmed through field emission scanning electron microscopy, energy dispersive spectroscopy, transmission electron microscopy, X-ray photoelectron spectroscopy, dynamic light scattering, ζ-potential, atomic absorption spectroscopy, X-ray powder diffraction, selected area electron diffraction, and vibrating-sample magnetometer. The superparamagnetic properties of the ternary system allowed the release of CYC by utilizing magnetic hyperthermia upon the exposure of an alternating magnetic field (AMF). The drug release experiments were carried out at different frequencies and intensities of the magnetic field, complying with the "Atkinson-Brezovich criterion". The assays in AMF showed the feasibility of release by controlling hyperthermia of the drug, finding that the most efficient conditions were F = 280 kHz, H = 15 mT, and a concentration of MNPs of 5 mg/mL. CYC release was temperature-dependent, facilitated by local heat generation through magnetic hyperthermia. This phenomenon was confirmed by DFT calculations. Furthermore, the ternary systems outperformed the formulations without MNPs regarding the amount of released drug. The MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assays demonstrated that including CYC within the magnetic NS cavities reduced the effects on mitochondrial activity compared to those observed with the free drug. Finally, the magnetic hyperthermia assays showed that the tertiary system allows the generation of apoptosis in HeLa cells, demonstrating that the MNPs embedded maintain their properties to generate hyperthermia. These results suggest that using NSs associated with MNPs could be a potential tool for a controlled drug delivery in tumor therapy since the materials are efficient and potentially nontoxic.
Collapse
Affiliation(s)
- Sebastián Salazar Sandoval
- Departamento de Química, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile
- Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, Sergio Livingstone 1007, Santiago 8380492, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
- Facultad de Diseño, Universidad del Desarrollo, Avenida Plaza 680, Las Condes, Santiago 7610658, Chile
| | - Patricia Díaz-Saldívar
- Laboratorio de Nanomedicina y Biosensores, Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile
| | - Ingrid Araya
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomás, Santiago 8370003, Chile
| | - Freddy Celis
- Laboratorio de Procesos Fotónicos y Electroquímicos, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Valparaíso 2360002, Chile
| | - Diego Cortés-Arriagada
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, Ignacio Valdivieso 2409, San Joaquín, Santiago 8940577, Chile
| | - Ana Riveros
- Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, Sergio Livingstone 1007, Santiago 8380492, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Carlos Rojas-Romo
- Departamento de Química, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile
| | - Carolina Jullian
- Departamento de Química Orgánica y Fisicoquímica, Universidad de Chile, Sergio Livingstone 1007, Santiago 8380492, Chile
| | - Nataly Silva
- Facultad de Diseño, Universidad del Desarrollo, Avenida Plaza 680, Las Condes, Santiago 7610658, Chile
| | - Nicolás Yutronic
- Departamento de Química, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile
| | - Marcelo J Kogan
- Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, Sergio Livingstone 1007, Santiago 8380492, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Paul Jara
- Departamento de Química, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile
| |
Collapse
|
19
|
Mazzotta E, Romeo M, Sacco G, De Benedittis S, Qualtieri A, Perrotta ID, Muzzalupo R. The Impact of Hyaluronic Acid Coating on the Cationic Niosomal Surface for Doxorubicin Delivery. Molecules 2025; 30:1148. [PMID: 40076371 PMCID: PMC11901725 DOI: 10.3390/molecules30051148] [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: 01/14/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
This study was designed to develop cationic vesicles for doxorubicin (DOX) delivery and to compare anticancer efficacy of these systems uncoated and coated with hyaluronic acid. Cationic nanoformulation was first optimized using various amounts of Span80, DODAB, and cholesterol. The optimized niosomal formulation (CTN4) in terms of vesicle size, surface zeta potential, and colloidal stability was coated with hyaluronic acid and the in vitro therapeutic effectiveness in uterine cervix cancer cells of vesicles loaded with DOX was tested. In vitro studies revealed significantly superior cytotoxicity against Hela cells of niosomes coated with HA compared to uncoated formulations. Moreover, cytotoxicity was also evaluated on normal fibroblast murine cell line, NIH-3T3 cells, and the results obtained demonstrated that HA-coated vesicles exhibited lower cytotoxicity to NIH-3T3 cells compared to uncoated nanovesicles. These findings highlighted how the surface coating influences the effectiveness of niosomes developed as a target drug delivery system and the selectivity and the antitumour efficacy of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Elisabetta Mazzotta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.M.); (M.R.); (G.S.)
| | - Martina Romeo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.M.); (M.R.); (G.S.)
| | - Giuseppina Sacco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.M.); (M.R.); (G.S.)
| | - Selene De Benedittis
- Institute for the Research and the Biomedical Innovation (IRIB)-CNR-Mangone (CS), 00185 Rome, Italy; (S.D.B.); (A.Q.)
| | - Antonio Qualtieri
- Institute for the Research and the Biomedical Innovation (IRIB)-CNR-Mangone (CS), 00185 Rome, Italy; (S.D.B.); (A.Q.)
| | - Ida Daniela Perrotta
- Centre for Microscopy and Microanalysis (CM2), Department of Biology Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy;
| | - Rita Muzzalupo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.M.); (M.R.); (G.S.)
| |
Collapse
|
20
|
Sivakumar PM, Zarepour A, Akhter S, Perumal G, Khosravi A, Balasekar P, Zarrabi A. Anionic polysaccharides as delivery carriers for cancer therapy and theranostics: An overview of significance. Int J Biol Macromol 2025; 294:139211. [PMID: 39732249 DOI: 10.1016/j.ijbiomac.2024.139211] [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: 07/24/2024] [Revised: 11/13/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Recently, cancer therapy has witnessed remarkable advancements with a growing focus on precision medicine and targeted drug delivery strategies. The application of anionic polysaccharides has gained traction in various drug delivery systems. Anionic polysaccharides have emerged as promising delivery carriers in cancer therapy and theranostics, offering numerous advantages such as biocompatibility, low toxicity, and the ability to encapsulate and deliver therapeutic agents to tumor sites with high specificity. This review underscores the significance of anionic polysaccharides as essential components of the evolving landscape of cancer therapy and theranostics. These polymers can be tailored to carry a wide range of therapeutic cargo, including chemotherapeutic agents, nucleic acids, and imaging agents. Their negative charge enables electrostatic interactions with positively charged drugs and facilitates the formation of stable nanoparticles, liposomes, or hydrogels for controlled drug release. Additionally, their hydrophilic nature aids in prolonging circulation time, reducing drug degradation, and minimizing off-target effects. Besides, some of them could act as targeting agents or therapeutic compounds that lead to improved therapeutic performance. This review offers valuable information for researchers, clinicians, and biomedical engineers. It provides insights into the recent progress in the applications of anionic polysaccharide-based delivery platforms in cancer theranostics to transform patient outcomes.
Collapse
Affiliation(s)
- Ponnurengam Malliappan Sivakumar
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam; School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India.
| | - Sohail Akhter
- New Product Development, Global R&D, Sterile ops, TEVA Pharmaceutical Industries Ltd., Aston Ln N, Halton, Preston Brook, Runcorn WA7 3FA, UK.
| | - Govindaraj Perumal
- Department of Biomedical Engineering, School of Dental Medicine, University of Connecticut (UConn) Health, Farmington, CT 06030, USA.
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye
| | - Premkumar Balasekar
- Department of Pharmacology, K.K. College of Pharmacy, Affiliated to The Tamilnadu Dr. M.G.R. Medical University, Gerugambakkam 600128, India
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
21
|
Moradi Kashkooli F, Mirala F, H H Tehrani M, Alirahimi M, Souri M, Golzaryan A, Kar S, Soltani M. Mechanical Forces in Tumor Growth and Treatment: Perspectives From Biology, Physics, Engineering, and Mathematical Modeling. WIREs Mech Dis 2025; 17:e70000. [PMID: 40170456 DOI: 10.1002/wsbm.70000] [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: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 04/03/2025]
Abstract
The progression of tumors is influenced by mechanical forces and biological elements, such as hypoxia and angiogenesis. Mechanical factors, including stress, pressure, interstitial fluid pressure, and cellular traction forces, compromise normal tissue architecture, augmenting stiffness and thus promoting tumor growth and invasion. The selective elimination of specific tumor components can reduce growth-induced mechanical stress, thereby improving therapeutic efficacy. Furthermore, stress-relief drugs have the potential in enhancing chemotherapy outcomes. In this setting, computational modeling functions as an essential tool for quantitatively elucidating the mechanical principles underlying tumor formation. These models can precisely replicate the impact of mechanical pressures on solid tumors, offering insight into the regulation of tumor behavior by these forces. Tumor growth produces mechanical forces, including compression, displacement, and deformation, leading to irregular stress patterns, expedited tumor advancement, and reduced treatment efficacy. This review analyzes the impact of mechanical forces on carcinogenesis and solid tumor proliferation, emphasizing the significance of stress alleviation in regulating tumor growth. Furthermore, we investigate the influence of mechanical forces on tumor dissemination and emphasize the promise of integrating computational modeling with force-targeted cancer therapies to improve treatment efficacy by tackling the fundamental mechanics of tumor proliferation.
Collapse
Affiliation(s)
| | - Fatemeh Mirala
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Masoud H H Tehrani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mahvash Alirahimi
- Department of Obstetrics & Gynecology, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Aryan Golzaryan
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Egaila, Kuwait
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
22
|
Sadat Razavi Z, Sina Alizadeh S, Sadat Razavi F, Souri M, Soltani M. Advancing neurological disorders therapies: Organic nanoparticles as a key to blood-brain barrier penetration. Int J Pharm 2025; 670:125186. [PMID: 39788400 DOI: 10.1016/j.ijpharm.2025.125186] [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: 09/04/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier (BBB) plays a vital role in protecting the central nervous system (CNS) by preventing the entry of harmful pathogens from the bloodstream. However, this barrier also presents a significant obstacle when it comes to delivering drugs for the treatment of neurodegenerative diseases and brain cancer. Recent breakthroughs in nanotechnology have paved the way for the creation of a wide range of nanoparticles (NPs) that can serve as carriers for diagnosis and therapy. Regarding their promising properties, organic NPs have the potential to be used as effective carriers for drug delivery across the BBB based on recent advancements. These remarkable NPs have the ability to penetrate the BBB using various mechanisms. This review offers a comprehensive examination of the intricate structure and distinct properties of the BBB, emphasizing its crucial function in preserving brain balance and regulating the transport of ions and molecules. The disruption of the BBB in conditions such as stroke, Alzheimer's disease, and Parkinson's disease highlights the importance of developing creative approaches for delivering drugs. Through the encapsulation of therapeutic molecules and the precise targeting of transport processes in the brain vasculature, organic NP formulations present a hopeful strategy to improve drug transport across the BBB. We explore the changes in properties of the BBB in various pathological conditions and investigate the factors that affect the successful delivery of organic NPs into the brain. In addition, we explore the most promising delivery systems associated with NPs that have shown positive results in treating neurodegenerative and ischemic disorders. This review opens up new possibilities for nanotechnology-based therapies in cerebral diseases.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | | - Fateme Sadat Razavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada; Centre for Sustainable Business, International Business University, Toronto, Canada.
| |
Collapse
|
23
|
Mubarak N, Waqar MA, Khan AM, Asif Z, Alvi AS, Virk AA, Amir S. A comprehensive insight of innovations and recent advancements in nanocarriers for nose-to-brain drug targeting. Des Monomers Polym 2025; 28:7-29. [PMID: 39935823 PMCID: PMC11812116 DOI: 10.1080/15685551.2025.2464132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Central Nervous System (CNS) disorders are the leading cause of illness and affect the everyday lives of people all around the globe and are predicted to increase tremendously in the upcoming decades. Traditional methods of delivering drugs to the CNS face considerable limitations. Nose-to-brain targeting offers a promising alternative that bypasses the blood-brain barrier (BBB), enabling targeted drug administration to the central nervous system (CNS). Nanotechnology has brought forward innovative solutions to the challenges of drug delivery in CNS disorders. Nanocarriers such as liposomes, nanoparticles, nanoemulsions and dendrimers can enhance drug stability, bioavailability, and targeted delivery to the brain. These nanocarriers are designed to overcome physiological barriers and provide controlled and sustained drug release directly to the CNS. Nanocarrier technology has made significant strides in recent years, enabling more effective and targeted delivery of drugs to the brain. With recent advancements, intranasal delivery coupled with nanocarriers seems to be a promising combination that can provide better clinical profiles, pharmacokinetics, and pharmacodynamics for neurodegenerative disorders. This study focuses on exploring the nose-to-brain drug delivery system, emphasizing the use of various nanocarriers designed for this purpose. Additionally, the study encompasses recent advancements in nanocarrier technology tailored specifically to improve the efficiency of drug administration through the nasal route to the brain.
Collapse
Affiliation(s)
- Naeem Mubarak
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Lahore University of Biological and Applied Sciences, Lahore, Pakistan
| | - Muhammad Ahsan Waqar
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological and Applied Sciences, Lahore, Pakistan
| | - Asad Majeed Khan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological and Applied Sciences, Lahore, Pakistan
| | - Zainab Asif
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological and Applied Sciences, Lahore, Pakistan
| | - Aima Subia Alvi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological and Applied Sciences, Lahore, Pakistan
| | - Aqsa Arshad Virk
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological and Applied Sciences, Lahore, Pakistan
| | - Sakeena Amir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological and Applied Sciences, Lahore, Pakistan
| |
Collapse
|
24
|
Dong Y, Li J, Dai Y, Zhang X, Jiang X, Wang T, Zhao B, Liu W, Sun H, Du P, Qin L, Jiao Z. A novel nanocarrier based on natural polyphenols enhancing gemcitabine sensitization ability for improved pancreatic cancer therapy efficiency. Mater Today Bio 2025; 30:101463. [PMID: 39866791 PMCID: PMC11764724 DOI: 10.1016/j.mtbio.2025.101463] [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: 11/07/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/28/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy with rapid progression and poor prognosis. Despite the widespread use of gemcitabine (Gem)-based chemotherapy as the first-line treatment for PC, its efficacy is often compromised by significant drug resistance. 1,2,3,4,6-Pentagaloyl glucose (PGG), a natural polyphenol, has demonstrated potential in sensitizing PC cells to Gem. However, its clinical application is limited by poor water solubility and bioavailability. In this study, we developed a novel PGG-based nanocarrier (FP) using a straightforward, one-step self-assembly method with Pluronic F127 and PGG. Our results showed that FP induced DNA damage and immunogenic cell death (ICD) in both in vitro cell experiments and patient-derived organoid models, exhibiting potent anti-tumor effects. Furthermore, in mouse KPC and PDX models, FP, when combined with Gem, showed enhanced Gem sensitization compared to pure PGG, largely due to increased DNA damage and ICD induction. These findings demonstrate the potential of FP to improve the stability and utilization of PGG as effective Gem sensitizers in the treatment of pancreatic cancer, providing a promising pathway for clinical application and translational research.
Collapse
Affiliation(s)
- Yuman Dong
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Jieru Li
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Yiwei Dai
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Xinyu Zhang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Xiangyan Jiang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Tao Wang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Bin Zhao
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Wenbo Liu
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Haonan Sun
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Pengcheng Du
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Long Qin
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zuoyi Jiao
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| |
Collapse
|
25
|
Ma R, Zhang Y, Ji H, Fu H, Gu S, Su Q, Lin Y, Deng Q, Xue W, Yang Y. Glucose oxidase-driven self-accelerating drug release nanosystem based on metal-phenolic networks orchestrates tumor chemotherapy and ferroptosis-based therapy. Int J Biol Macromol 2025; 290:139103. [PMID: 39716697 DOI: 10.1016/j.ijbiomac.2024.139103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
Nanocarriers responding to tumor microenvironment have been extensively exploited to improve the antitumor outcome of chemotherapeutic drugs. However, selectively and completely releasing drugs within the tumor remains a challenge, thereby limiting the therapeutic effect of drug delivery nanosystem. To tackle this challenge, a metal-phenolic networks (MPNs)-based nanosystem (F-MGD) showing the capability of self-accelerating drug release was originally fabricated in this study. Glucose oxidase (GOx) encapsulated in F-MGD could conduct the glucose transformation in tumor to cause the oxygen consumption and the production of gluconic acid and H2O2. Therefore, F-MGD with acid and hypoxia sensitivities thoroughly disintegrated under the aggravated acidity and hypoxia to achieve a more complete drug release. Besides, the product of H2O2 was readily decomposed into hydroxyl radicals via the iron ion-mediated Fenton reaction, which markedly augmented the oxidative stress in tumor cells and promoted ferroptosis. The results of both in vitro and in vivo assays demonstrated the significant antitumor efficacy of F-MGD. Collectively, this study proposes a strategy to expedite drug release in tumor and improve the tumor treatment effect by combining ferroptosis-based therapy and chemotherapy.
Collapse
Affiliation(s)
- Rongying Ma
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Yufei Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Hongting Ji
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Huiling Fu
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Shuzhen Gu
- Department of Gynecology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Qianhong Su
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Yumian Lin
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Qingchun Deng
- Department of Gynecology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yong Yang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|
26
|
He X, Liu S, Tong J, Qiao Y, Li X, Dong K, Yu D, Gu Y. Pathogen-targeting bismuth nanocluster via photothermally-mediated NDM-1 inactivation and bacterial membrane destabilization combat NDM-1-producing bacteria. Process Biochem 2025; 149:158-168. [DOI: 10.1016/j.procbio.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
27
|
Yoo H, Kang SB, Kim J, Cho W, Ha H, Oh S, Jeong SH, Lee S, Lee H, Park CS, Lee DY, Chung TD, Lee KM, Sun JY. Ionic Diode-Based Drug Delivery System. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412377. [PMID: 39718239 DOI: 10.1002/adma.202412377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/29/2024] [Indexed: 12/25/2024]
Abstract
Drug delivery systems hold promise for delivering cytotoxic drugs by controlling the timing and location of the drug release. However, conventional delivery mechanisms often fall short of achieving spatiotemporally controlled yet sustained release, which is crucial for ensuring drug efficacy and minimizing impact on surrounding tissues. Here, an ionic diode-based drug delivery system is reported that is controlled by an electric potential and capable of releasing drugs at scales ranging from nanogram to microgram. The migrated drug is slowly but continuously diffused to the lesion through the hydrogel at the desired rate. The ionic diode provides flow-free drug delivery while minimizing unintended drug leakage over prolonged periods. Implanted in a freely moving tumor-bearing mouse model, the system filled with doxorubicin demonstrated superior anti-tumor efficacy and minimal off-target immune toxicity compared to the intratumoral injection of free doxorubicin. With its mechanically compliant and biocompatible components, the system offers a safe and readily translatable approach to patients with surgically unresectable tumors.
Collapse
Affiliation(s)
- Hyunjae Yoo
- Department of Material Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Soon-Bo Kang
- Department of Material Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeongsoo Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Wonkyung Cho
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyojeong Ha
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Seyoung Oh
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Seol-Ha Jeong
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sihwan Lee
- Department of Material Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyemin Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Chang Seo Park
- Department of Material Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong-Yup Lee
- Department of Material Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taek Dong Chung
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyung-Mi Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jeong-Yun Sun
- Department of Material Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute of Advanced Materials (RIAM), Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
28
|
Abdurashtov AS, Proshin PI, Sukhorukov GB. The pursuit of linear dosage in pharmacy: reservoir-based drug delivery systems from macro to micro scale. Expert Opin Drug Deliv 2025; 22:219-238. [PMID: 39764701 DOI: 10.1080/17425247.2024.2448026] [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: 10/16/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION The pursuit of linear dosage in pharmacy is essential for achieving consistent therapeutic release and enhancing patient compliance. This review provides a comprehensive summary of zero-order drug delivery systems, with a particular focus on reservoir-based systems emanated from different microfabrication technologies. AREAS COVERED The consideration of recent advances in drug delivery systems is given to encompass the key areas including the importance of achieving a constant drug release rate for therapeutic applications. Detailed examination of reservoir-based systems, their design, mechanisms of action and materials used are highlighted. By addressing these areas, the discussion aims to provide a thorough understanding of most recent zero-order drug delivery systems, their performance advantages and methods of their manufacturing. To ensure the complete coverage of the explored research area, modern AI-assistant tools were used to find not only the most relevant, but also connected and similar articles. EXPERT OPINION Future developments in reservoir-based drug delivery systems are expected to significantly enhance therapeutic effectiveness and patient outcomes through the integration of innovative materials and technologies. The fabrication of intelligent drug delivery systems that utilize sensors and feedback mechanisms can enable real-time monitoring of drug release and patient reactions.
Collapse
Affiliation(s)
- Arkady S Abdurashtov
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Pavel I Proshin
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Gleb B Sukhorukov
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| |
Collapse
|
29
|
Paranandi KS, Amar-Lewis E, Mirkin CA, Artzi N. Nomadic Nanomedicines: Medicines Enabled by the Paracrine Transfer Effect. ACS NANO 2025; 19:21-30. [PMID: 39746105 DOI: 10.1021/acsnano.4c15052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
In nanomedicine, the cellular export of nanomaterials has been less explored than uptake. Traditionally viewed in a negative light, recent findings highlight the potential of nanomedicine export to enhance therapeutic effects. This Perspective examines key pathways for export and how nanomaterial design affects removal rates. We present the idea of the "paracrine transfer effect" (PTE), where nanomaterials are first internalized by a "waypoint" cell and then exported to a "destination" cell, influencing both in potentially exploitable ways. Essential characteristics for nanomedicines to leverage the PTE are discussed, along with two case studies: STING-stimulating polymeric nanoparticles and TLR9-stimulating liposomal spherical nucleic acids. We propose future research directions to better understand and utilize the PTE in developing more effective nanomedicines.
Collapse
Affiliation(s)
- Krishna S Paranandi
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Eliz Amar-Lewis
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| | - Chad A Mirkin
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| |
Collapse
|
30
|
Zhang S, Wang H. Targeting the lung tumour stroma: harnessing nanoparticles for effective therapeutic interventions. J Drug Target 2025; 33:60-86. [PMID: 39356091 DOI: 10.1080/1061186x.2024.2410462] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Lung cancer remains an influential global health concern, necessitating the development of innovative therapeutic strategies. The tumour stroma, which is known as tumour microenvironment (TME) has a central impact on tumour expansion and treatment resistance. The stroma of lung tumours consists of numerous cells and molecules that shape an environment for tumour expansion. This environment not only protects tumoral cells against immune system attacks but also enables tumour stroma to attenuate the action of antitumor drugs. This stroma consists of stromal cells like cancer-associated fibroblasts (CAFs), suppressive immune cells, and cytotoxic immune cells. Additionally, the presence of stem cells, endothelial cells and pericytes can facilitate tumour volume expansion. Nanoparticles are hopeful tools for targeted drug delivery because of their extraordinary properties and their capacity to devastate biological obstacles. This review article provides a comprehensive overview of contemporary advancements in targeting the lung tumour stroma using nanoparticles. Various nanoparticle-based approaches, including passive and active targeting, and stimuli-responsive systems, highlighting their potential to improve drug delivery efficiency. Additionally, the role of nanotechnology in modulating the tumour stroma by targeting key components such as immune cells, extracellular matrix (ECM), hypoxia, and suppressive elements in the lung tumour stroma.
Collapse
Affiliation(s)
- Shushu Zhang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Hui Wang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
31
|
Cordeiro BM, Leite Fontes CF, Meyer-Fernandes JR. Molecular Basis of Na, K-ATPase Regulation of Diseases: Hormone and FXYD2 Interactions. Int J Mol Sci 2024; 25:13398. [PMID: 39769162 PMCID: PMC11678576 DOI: 10.3390/ijms252413398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The Na, K-ATPase generates an asymmetric ion gradient that supports multiple cellular functions, including the control of cellular volume, neuronal excitability, secondary ionic transport, and the movement of molecules like amino acids and glucose. The intracellular and extracellular levels of Na+ and K+ ions are the classical local regulators of the enzyme's activity. Additionally, the regulation of Na, K-ATPase is a complex process that occurs at multiple levels, encompassing its total cellular content, subcellular distribution, and intrinsic activity. In this context, the enzyme serves as a regulatory target for hormones, either through direct actions or via signaling cascades triggered by hormone receptors. Notably, FXYDs small transmembrane proteins regulators of Na, K-ATPase serve as intermediaries linking hormonal signaling to enzymatic regulation at various levels. Specifically, members of the FXYD family, particularly FXYD1 and FXYD2, are that undergo phosphorylation by kinases activated through hormone receptor signaling, which subsequently influences their modulation of Na, K-ATPase activity. This review describes the effects of FXYD2, cardiotonic steroid signaling, and hormones such as angiotensin II, dopamine, insulin, and catecholamines on the regulation of Na, K-ATPase. Furthermore, this review highlights the implications of Na, K-ATPase in diseases such as hypertension, renal hypomagnesemia, and cancer.
Collapse
Affiliation(s)
- Bárbara Martins Cordeiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Carlos Frederico Leite Fontes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| |
Collapse
|
32
|
Rananaware P, Pandit P, Brahmkhatri V. Gold nanoparticle encapsulated hybrid MOF: synthesis, characterization, and co-drug delivery of 5-fluorouracil and curcumin. DISCOVER NANO 2024; 19:201. [PMID: 39661211 PMCID: PMC11635076 DOI: 10.1186/s11671-024-04152-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
The unique features of Metal-Organic Frameworks (MOFs), including structural flexibility, high surface area, and variable pore size, have drawn attention in cancer therapy. However, despite advances in surface functionalization, engineering structural features, and porosity, achieving controlled release, stability, scalability, and toxicity remains a challenge. The current study reports gold nanoparticle (AuNP) encapsulated dual metal-organic frameworks (MOFs) comprising zeolitic imidazolate (ZIF8) and cobalt-imidazole (ZIF67) by a simple precipitation method for dual drug delivery applications. This combination associates the advantages of AuNPs and MOFs, creating a potent platform for cancer theranostics that combines diagnosis and treatment into one unit. The synthesized composite (AuNPs@ZIF-8/ZIF-67) is functionalized with Folic acid (FA) and loaded with the anticancer agents Curcumin (C) and 5-fluorouracil (5-FU) for co-drug delivery The synthesized composites, namely Au/ZIF8, Au/ZIF8/ZIF67/FA, Au/ZIF8/ZIF67/FA/5-FU, and Au/ZIF8/ZIF67/FA/5-FU/C were characterized using diverse analytical techniques such as FESEM, XRD, FTIR, TEM, and BET. The characterization methods showed that the hybrid MOF structure was stable and intact after AuNP encapsulation and drug loading. The dual MOF composite exhibits a better affinity for loading C and 5-FU with 60% and 40% drug loading capacity, respectively. The simultaneous drug release studies suggest that AuNPs@ZIF-8/ZIF-67 are more responsive to the acidic pH and show a higher cumulative drug release of 5FU and C at the lower value of pH 5. For further validation, the release kinetics data were fitted into the Korsmeyer-Peppas model in the current study. The observed value of n which is less than 0.5 suggests the pseudo-Fickian diffusion mechanism for drug release, demonstrating long-term release of 5FU and C from Au/ZIF8/ZIF67/FA/5-FU/C. The targeted drug delivery system is anticipated to display synergistic therapeutic efficacy from the combined effect of the two anticancer agents and the pH-responsive nature of ZIF systems.
Collapse
Affiliation(s)
- Pranita Rananaware
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bengaluru, Karnataka, 562112, India
| | - Parimal Pandit
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bengaluru, Karnataka, 562112, India
| | - Varsha Brahmkhatri
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bengaluru, Karnataka, 562112, India.
- Department of Chemistry, Centre of Excellence in Materials and Sensors, CMR Institute of Technology, Bengaluru, 560037, India.
| |
Collapse
|
33
|
Taheri-Ledari R, Ghafori-Gorab M, Ramezanpour S, Mahdavi M, Safavi M, Akbarzadeh AR, Maleki A. MIL-101 magnetic nanocarrier for solid-phase delivery of doxorubicin to breast and lung cancer cells. Int J Biol Macromol 2024; 283:137615. [PMID: 39551314 DOI: 10.1016/j.ijbiomac.2024.137615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
An efficient strategy for passive delivery of doxorubicin (DOX) to the breast (MDA-MB-231) and lung (A-549) cancer cells is presented and compared with MCF-10A normal breast cells. Two versions of a peptide structure (linear and cyclic) have been designed and assessed. The molecular dynamic simulations in Material Studio2017 exhibited a higher adsorption capacity for L2 (cyclic version) compared with the adsorption capacity of L1 (linear version) on the PG surface by electrostatic interactions between guanidine of arginine and -OH of PG. The prepared final product based on iron oxide nanoparticles and MIL-101(Fe) (formulated as DOX@Fe3O4/MIL-101-(C,L)C[RW]3) is characterized and the drug content has been estimated. The release profiles revealed an ultra-fast stimulus-sensitive model in acidic media, which corroborates a pH-triggered release. The in vitro assessments disclosed that aggregation of nanocargo around the cancer cells and resulted toxicity are more than the neat DOX in the same dosage as DOX@Fe3O4/MIL-101-CC[RW]3. The obtained distinguished features lie in ability to utilize a biocompatible nanocargo structure to release an appropriate dose of DOX in a controlled manner in the cancer cell environment. Moreover, the functionalization of MIL-101 using cyclic and linear peptides and their comparison is one of the important features of this project.
Collapse
Affiliation(s)
- Reza Taheri-Ledari
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Mostafa Ghafori-Gorab
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Sorour Ramezanpour
- Department of Chemistry, K. N. Toosi University of Technology, P.O. Box 15875-4416, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), P. O. Box 3353-5111, Tehran, Iran.
| | - Ali Reza Akbarzadeh
- Department of Chemistry, Iran University of Science and Technology, PO Box: 16846-13114, Tehran, Iran.
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran.
| |
Collapse
|
34
|
Nowak P, Ilnicka A, Ziegler-Borowska M. Hydrazidomethyl starch as a pH-sensitive coating for magnetic core in tailored magnetic nanoparticles with selective doxorubicin release. Int J Biol Macromol 2024; 283:137716. [PMID: 39579836 DOI: 10.1016/j.ijbiomac.2024.137716] [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: 07/07/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
The work aimed to use and modify starch as a biodegradable and biocompatible polysaccharide to create a modern pH-sensitive anticancer drug carrier based on a hydrazone bond. The multi-step reaction created a material that can bind to the carbonyl group of anticancer drugs. Additionally, polysaccharide was used to coat magnetic nanoparticles to increase the applicability of the carrier system. At each synthesis stage, the material was characterized in detail by performing FTIR-ATR spectra, thermal analysis, XRD, and SEM photos. In the next step, doxorubicin was loaded with a maximum of 19 % drug loading to the carrier via hydrazone bond. In the last research stage, the carrier-hydrazone bond-drug system was tested in solutions with different pH values, imitating the environments of a cancer cell, a healthy cell, and their subcellular elements regarding drug release from the carrier. The obtained release results indicate a >4-fold increase in the amount of drug released from the carrier in conditions of a slightly lower pH environment (70 %), compared to neutral pH (15 %). This represents a promising potential for using the material as an intelligent drug delivery system (DDS).
Collapse
Affiliation(s)
- Paweł Nowak
- Doctoral School of Exact and Natural Sciences "Academia Scientiarum Thoruniensis", Grudziadzka 5, 87-100 Torun, Poland; Nicolaus Copernicus University in Torun, Faculty of Chemistry, Department of Biomedical Chemistry and Polymers, Medicinal Chemistry Research Group, Gagarina 7, 87-100 Torun, Poland
| | - Anna Ilnicka
- Nicolaus Copernicus University in Torun, Faculty of Chemistry, Department of Chemistry of Materials, Adsorption and Catalysis, Gagarina 7, 87-100 Torun, Poland
| | - Marta Ziegler-Borowska
- Nicolaus Copernicus University in Torun, Faculty of Chemistry, Department of Biomedical Chemistry and Polymers, Medicinal Chemistry Research Group, Gagarina 7, 87-100 Torun, Poland.
| |
Collapse
|
35
|
Yun D, Fagan E, Shin D, Back W, Lee S, Kim MS, Park H, Park JH, Kim YC. pH and Redox Dual-Responsive Nanoparticle with Enhanced Dendritic Cell Maturation for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64592-64608. [PMID: 39538128 DOI: 10.1021/acsami.4c15342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Type I interferons (IFNs) are essential for activating dendritic cells (DCs) and presenting tumor-associated antigens to T cells. IFNs are primarily produced from DCs among immune cells. A combination of chemotherapy and metalloimmunotherapy induces IFN production by activating the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. However, chemotherapeutic agents deplete DC populations, suppressing immunostimulatory activities, despite their potent anticancer activities. Furthermore, an optimal ratio between chemotherapeutic agents and metal for activating DCs at the highest level has not been reported, and evidence for ensuring DC survival is lacking. In this study, we hypothesized that there is an optimal ratio to yield the highest DC maturation and anticancer activity with minimal DC depletion. To demonstrate it, we have designed a pH and redox dual-responsive nanoparticle, MnO2@BSA@DOX (MD), to prevent DCs from depleting and activate the cGAS-STING pathway both in cancer cells and DCs, inducing considerable levels of IFNs and maturation. MD consists of a core-layer structure, a manganese dioxide (MnO2) core, and a cross-linked layer with bovine serum albumin (BSA) and doxorubicin (DOX), with a specific ratio of DOX to manganese. MD exhibits structure-based selectivity between cancer cells and DCs by targeting the extracellular pH of the tumor microenvironment and intracellular redox reactions in cancer cells. Among various formulations, the 1:1 ratio shows the highest maturation with no significant depletion. Moreover, it induces distinct cytotoxicity in cancer cells through apoptosis and cGAS-STING activation, leading to increased calreticulin expression and enhanced DC phagocytosis. Consequently, it results in superior tumor suppression and prolonged survival with the high accumulation of MD in the tumor and no observed systemic toxicities, highlighting its potential as a therapeutic agent in cancer treatments.
Collapse
Affiliation(s)
- Dohyun Yun
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Erinn Fagan
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Dongik Shin
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Woojin Back
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Susam Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Mun Sik Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Heewon Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| |
Collapse
|
36
|
Gulati S, Ansari N, Moriya Y, Joshi K, Prasad D, Sajwan G, Shukla S, Kumar S, Varma RS. Nanobiopolymers in cancer therapeutics: advancing targeted drug delivery through sustainable and controlled release mechanisms. J Mater Chem B 2024; 12:11887-11915. [PMID: 39502076 DOI: 10.1039/d4tb00599f] [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: 11/28/2024]
Abstract
Nanobiopolymers have emerged as a transformative frontier in cancer treatment, leveraging nanotechnology to transform drug delivery. This review provides a comprehensive exploration of the multifaceted landscape of nano-based biopolymers, emphasizing their diverse sources, synthesis methods, and classifications. Natural, synthetic, and microbial nanobiopolymers are scrutinized, along with elucidation of their underlying mechanisms and impact on cancer drug delivery; the latest findings on their deployment as targeted drug delivery agents for cancer treatment are discussed. A detailed analysis of nanobiopolymer sources, including polysaccharides, peptides, and nucleic acids, highlights critical attributes like biodegradability, renewability, and sustainability essential for therapeutic applications. The classification of nanobiopolymers based on their origin and differentiation among natural, synthetic, and microbial sources are thoroughly examined for inherent advantages, challenges, and suitability for cancer therapeutics. The importance of targeted drug release at tumour sites, crucial for minimizing adverse effects on normal tissues, is discussed, encompassing various mechanisms. The role of polymer membrane coatings as a pivotal barrier for facilitating controlled drug release through diffusion is elucidated, providing further insight into efficient methods for cancer treatment and thus consolidating the current knowledge base for researchers and practitioners in the field of nanobiopolymers and cancer therapeutics.
Collapse
Affiliation(s)
- Shikha Gulati
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Nabeela Ansari
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Yamini Moriya
- Department of Life Sciences, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Kumud Joshi
- Department of Life Sciences, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Disha Prasad
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Gargi Sajwan
- Department of Biological Sciences, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Shefali Shukla
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Sanjay Kumar
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Rajender S Varma
- Centre of Excellence for Research in Sustainable Chemistry, Department of Chemistry, Federal University of São Carlos, 13565-905 São Carlos - SP, Brazil.
| |
Collapse
|
37
|
Wang Z, Wang X, He Y, Wu H, Mao R, Wang H, Qiu L. Exploring Framework Nucleic Acids: A Perspective on Their Cellular Applications. JACS AU 2024; 4:4110-4128. [PMID: 39610738 PMCID: PMC11600171 DOI: 10.1021/jacsau.4c00776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/30/2024]
Abstract
Cells are fundamental units of life. The coordination of cellular functions and behaviors relies on a cascade of molecular networks. Technologies that enable exploration and manipulation of specific molecular events in living cells with high spatiotemporal precision would be critical for pathological study, disease diagnosis, and treatment. Framework nucleic acids (FNAs) represent a novel class of nucleic acid materials characterized by their monodisperse and rigid nanostructure. Leveraging their exceptional programmability, convenient modification property, and predictable atomic-level architecture, FNAs have attracted significant attention in diverse cellular applications such as cell recognition, imaging, manipulation, and therapeutic interventions. In this perspective, we will discuss the utilization of FNAs in living cell systems while critically assessing the opportunities and challenges presented in this burgeoning field.
Collapse
Affiliation(s)
- Zhaoyang Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Xin Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
- The
Key Laboratory of Zhejiang Province for Aptamers and Theranostics,
Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yao He
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Hui Wu
- The
Key Laboratory of Zhejiang Province for Aptamers and Theranostics,
Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Rui Mao
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Haiyuan Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Liping Qiu
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
- The
Key Laboratory of Zhejiang Province for Aptamers and Theranostics,
Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
38
|
Souri M, Elahi S, Moradi Kashkooli F, Kohandel M, Soltani M. Enhancing localized chemotherapy with anti-angiogenesis and nanomedicine synergy for improved tumor penetration in well-vascularized tumors. NPJ Syst Biol Appl 2024; 10:136. [PMID: 39567549 PMCID: PMC11579323 DOI: 10.1038/s41540-024-00467-w] [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: 04/13/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
Intratumoral delivery and localized chemotherapy have demonstrated promise in tumor treatment; however, the rapid drainage of therapeutic agents from well-vascularized tumors limits their ability to achieve maximum therapeutic efficacy. Therefore, innovative approaches are needed to enhance treatment efficacy in such tumors. This study utilizes a mathematical modeling platform to assess the efficacy of combination therapy using anti-angiogenic drugs and drug-loaded nanoparticles. Anti-angiogenic drugs are included to reduce blood microvascular density and facilitate drug retention in the extracellular space. In addition, incorporating negatively charged nanoparticles aims to enhance diffusion and distribution of therapeutic agents within well-vascularized tumors. The findings indicate that, in the case of direct injection of free drugs, using compounds with lower drainage rates and higher diffusion coefficients is beneficial for achieving broader diffusion. Otherwise, drugs tend to accumulate primarily around the injection site. For instance, the drug doxorubicin, known for its rapid drainage, requires the prior direct injection of an anti-angiogenic drug with a high diffusion rate to reduce microvascular density and facilitate broader distribution, enhancing penetration depth by 200%. Moreover, the results demonstrate that negatively charged nanoparticles effectively disperse throughout the tissue due to their high diffusion coefficient. In addition, a faster drug release rate from nanoparticles further enhance treatment efficacy, achieving the necessary concentration for complete eradication of tumor compared to slower drug release rates. This study demonstrates the potential of utilizing negatively charged nanoparticles loaded with chemotherapy drugs exhibiting high release rates for localized chemotherapy through intratumoral injection in well-vascularized tumors.
Collapse
Affiliation(s)
- Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Sohail Elahi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON, Canada
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada.
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada.
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
- Centre for Sustainable Business, International Business University, Toronto, ON, Canada.
| |
Collapse
|
39
|
Rezaei Aghdam H, Peymani M, Salehzadeh A, Rouhi L, Zarepour A, Zarrabi A. Precision Nanomedicine: Lapatinib-Loaded Chitosan-Gold Nanoparticles Targeting LINC01615 for Lung Cancer Therapy. AAPS J 2024; 27:4. [PMID: 39562465 DOI: 10.1208/s12248-024-00990-y] [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: 08/29/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) play essential roles as oncogenic factors in cancer progression by influencing cell proliferation, apoptosis, and metastasis pathways. This study aims to investigate the expression changes of LINC01615 in prevalent cancers, explore its correlation with patient mortality rates, and introduce a novel therapeutic approach to reduce LINC01615 expression. Using The Cancer Genome Atlas (TCGA) data, the expression changes of LINC01615 in various cancers were analyzed, and its relationship with patient survival rates through Cox regression analysis weas assessed. Co-expressed pathways related to LINC01615 were identified via network analysis. Potential drugs to decrease LINC01615 expression were identified using the GSE38376 study. Besides, chitosan-coated nanoparticles were fabricated and functionalized with the identified drug, Lapatinib, to examine their effect on lung cancer cell lines and changes in LINC01615 expression. Our results indicated elevated LINC01615 expression in various common cancers, particularly in lung cancer, which was associated with poor prognosis in lung, breast, and kidney cancers. Co-expression network analysis suggested links to metastasis-related genes. Lapatinib, identified through GEO data, was found to modulate LINC01615 expression effectively. Chitosan-gold nanoparticles conjugated with Lapatinib significantly reduced LINC01615 expression in lung cancer cell lines while enhancing apoptosis rates. Therefore, these nanoparticles could be considered a promising therapeutic candidate for treating cancers with overexpression of LINC01615.
Collapse
Affiliation(s)
- Hadi Rezaei Aghdam
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Leila Rouhi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, 600 077, India
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Sariyer, Istanbul, Türkiye.
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan.
| |
Collapse
|
40
|
Farajollahi A, Baharvand M. Advancements in photoacoustic imaging for cancer diagnosis and treatment. Int J Pharm 2024; 665:124736. [PMID: 39326479 DOI: 10.1016/j.ijpharm.2024.124736] [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: 06/24/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Photoacoustic imaging provides in vivo morphological and functional information about tumors within surrounding tissue. By integrating ultrasound guidance, this technique enables precise localization and characterization of tumors. Moreover, the introduction of targeted contrast agents has further expanded the capabilities of photoacoustic imaging in the realm of in vivo molecular imaging. These contrast agents facilitate enhanced molecular and cellular characterization of cancer, enabling detailed insights into the disease. This review aims to provide a concise summary of the extensive research conducted in the field of Photoacoustic imaging for cancer management. It encompasses the development of the technology, its applications in clinical settings, and the advancements made in molecular imaging. By consolidating and synthesizing the existing knowledge, this review contributes to a better understanding of the potential of photoacoustic imaging in cancer care. In conclusion, photoacoustic imaging has emerged as a non-ionizing and noninvasive modality with the ability to visualize tissue's optical absorption properties while maintaining ultrasound's spatial resolution. Its integration with targeted contrast agents has enhanced molecular and cellular characterization of cancer. This review serves as a succinct overview of the extensive research conducted in the field, shedding light on the potential of photoacoustic imaging in the management of cancer.
Collapse
Affiliation(s)
| | - Mohammad Baharvand
- Department of Mechanical Engineering, Islamic Azad University, Tehran, Iran
| |
Collapse
|
41
|
Wang H, Wang X, Wang L, Wang H, Zhang Y. Exploiting lignin-based nanomaterials for enhanced anticancer therapy: A comprehensive review and future direction. Int J Biol Macromol 2024; 281:136266. [PMID: 39366596 DOI: 10.1016/j.ijbiomac.2024.136266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Lignin, a renewable and abundant natural polymer, has emerged as a promising candidate for anticancer therapy due to its unique properties and biocompatibility. This review provides a comprehensive overview of recent advancements in the utilization of lignin-based nanomaterials for enhancing anticancer drug delivery and therapeutic outcomes. A detailed examination of the literature reveals several synthesis methods, including nanoprecipitation, microemulsion, and solvent exchange, which produce lignin nanoparticles with improved drug solubility and bioavailability. The anticancer mechanisms of lignin nanoparticles, such as the generation of reactive oxygen species (ROS), induction of apoptosis, and enhanced cellular uptake, are also explored. Lignin nanoparticles loaded with drugs like curcumin, doxorubicin, camptothecin, and resveratrol have demonstrated the ability to improve drug efficacy, selectively target cancer cells, overcome multidrug resistance, and minimize toxicity in both in vitro and in vivo studies. These nanoparticles have shown significant potential in suppressing tumor growth, inducing cell death through apoptotic pathways, and enhancing the synergistic effects of combination therapies, such as chemo-phototherapy. Future research directions include optimizing lignin nanoparticle formulations for clinical applications, refining targeted delivery mechanisms to cancer cells, and conducting thorough biocompatibility and toxicity assessments. Overall, this review highlights the significant progress made in utilizing lignin-based nanomaterials for cancer therapy and outlines promising areas for further exploration in this rapidly evolving field.
Collapse
Affiliation(s)
- Haoyu Wang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China; Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoyang Wang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Long Wang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China
| | - Haifan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yuxing Zhang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China.
| |
Collapse
|
42
|
Zhang H, Li Y, Huang J, Shen L, Xiong Y. Precise targeting of lipid metabolism in the era of immuno-oncology and the latest advances in nano-based drug delivery systems for cancer therapy. Acta Pharm Sin B 2024; 14:4717-4737. [PMID: 39664426 PMCID: PMC11628863 DOI: 10.1016/j.apsb.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 12/13/2024] Open
Abstract
Over the past decade, research has increasingly identified unique dysregulations in lipid metabolism within the tumor microenvironment (TME). Lipids, diverse biomolecules, not only constitute biological membranes but also function as signaling molecules and energy sources. Enhanced synthesis or uptake of lipids in the TME significantly promotes tumorigenesis and proliferation. Moreover, lipids secreted into the TME influence tumor-resident immune cells (TRICs), thereby aiding tumor survival against chemotherapy and immunotherapy. This review aims to highlight recent advancements in understanding lipid metabolism in both tumor cells and TRICs, with a particular emphasis on exogenous lipid uptake and endogenous lipid de novo synthesis. Targeting lipid metabolism for intervention in anticancer therapies offers a promising therapeutic avenue for cancer treatment. Nano-drug delivery systems (NDDSs) have emerged as a means to maximize anti-tumor effects by rewiring tumor metabolism. This review provides a comprehensive overview of recent literature on the development of NDDSs targeting tumor lipid metabolism, particularly in the context of tumor immunotherapy. It covers four key aspects: reprogramming lipid uptake, reprogramming lipolysis, reshaping fatty acid oxidation (FAO), and reshuffling lipid composition on the cell membrane. The review concludes with a discussion of future prospects and challenges in this burgeoning field of research.
Collapse
Affiliation(s)
- Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yujie Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jingyi Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Limei Shen
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
43
|
Li Q, Yang X, Xia X, Xia XX, Yan D. Affibody-Functionalized Elastin-like Peptide-Drug Conjugate Nanomicelle for Targeted Ovarian Cancer Therapy. Biomacromolecules 2024; 25:6474-6484. [PMID: 39235966 DOI: 10.1021/acs.biomac.4c00640] [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: 09/07/2024]
Abstract
Recombinant elastin-like polypeptides (ELPs) have emerged as an attractive nanoplatform for drug delivery due to their tunable genetically encoded sequence, biocompatibility, and stimuli-responsive self-assembly behaviors. Here, we designed and biosynthesized an HER2 (human epidermal growth factor receptor 2)-targeted affibody-ELP fusion protein (Z-ELP), which was subsequently conjugated with monomethyl auristatin E (MMAE) to build a protein-drug conjugate (Z-ELP-M). Due to its thermal response, Z-ELP-M can immediately self-assemble into a nanomicelle at physiological temperature. Benefiting from its active targeting and nanomorphology, Z-ELP-M exhibits enhanced cellular internalization and deep tumor penetration in vitro. Moreover, Z-ELP-M shows excellent tumor targeting and superior antitumor efficacy in HER2-positive ovarian cancer, demonstrating a relative tumor growth inhibition of 104.6%. These findings suggest that an affibody-functionalized elastin-like peptide-drug conjugate nanomicelle is an efficient strategy to improve antitumor efficacy and biosafety in cancer therapy.
Collapse
Affiliation(s)
- Qingrong Li
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiaoyuan Yang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xuelin Xia
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiao-Xia Xia
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| |
Collapse
|
44
|
Chen Y, Tang Y, Li Y, Rui Y, Zhang P. Enhancing the Efficacy of Active Pharmaceutical Ingredients in Medicinal Plants through Nanoformulations: A Promising Field. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1598. [PMID: 39404324 PMCID: PMC11478102 DOI: 10.3390/nano14191598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
This article explores the emerging field of nanomedicine as a drug delivery system, aimed at enhancing the therapeutic efficacy of active pharmaceutical ingredients in medicinal plants. The traditional methods of applying medicinal plants present several limitations, such as low bioavailability, poor solubility, challenges in accurately controlling drug dosage, and inadequate targeting. Nanoformulations represent an innovative approach in drug preparation that employs nanotechnology to produce nanoscale particles or carriers, which are designed to overcome these limitations. Nanoformulations offer distinct advantages, significantly enhancing the solubility and bioavailability of drugs, particularly for the poorly soluble components of medicinal plants. These formulations effectively enhance solubility, thereby facilitating better absorption and utilization by the human body, which in turn improves drug efficacy. Furthermore, nanomedicine enables targeted drug delivery, ensuring precise administration to the lesion site and minimizing side effects on healthy tissues. Additionally, nanoformulations can regulate drug release rates, extend the duration of therapeutic action, and enhance the stability of treatment effects. However, nanoformulations present certain limitations and potential risks; their stability and safety require further investigation, particularly regarding the potential toxicity with long-term use. Nevertheless, nanomaterials demonstrate substantial potential in augmenting the efficacy of active pharmaceutical ingredients in medicinal plants, offering novel approaches and methodologies for their development and application.
Collapse
Affiliation(s)
- Yuhao Chen
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuying Tang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuanbo Li
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yukui Rui
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
- Tangshan Jinhai New Material Co., Ltd., Tangshan 063000, China
- Faculty of Resources and Environment, China Agricultural University, Shanghe County Baiqiao Town Science and Technology Courtyard, Jinan 250100, China
| | - Peng Zhang
- Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
45
|
Cao Z, Liu J, Yang X. Deformable nanocarriers for enhanced drug delivery and cancer therapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230037. [PMID: 39439489 PMCID: PMC11491306 DOI: 10.1002/exp.20230037] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/28/2024] [Indexed: 10/25/2024]
Abstract
Recently, the field of nanomedicine has witnessed substantial advancements in the development of nanocarriers for targeted drug delivery, emerges as promising platforms to enhance therapeutic efficacy and minimize adverse effects associated with conventional chemotherapy. Notably, deformable nanocarriers have garnered considerable attention due to their unique capabilities of size changeable, tumor-specific aggregation, stimuli-triggered disintegration, and morphological transformations. These deformable nanocarriers present significant opportunities for revolutionizing drug delivery strategies, by responding to specific stimuli or environmental cues, enabling achieved various functions at the tumor site, including size-shrinkage nanocarriers enhance drug penetration, aggregative nanocarriers enhance retention effect, disintegrating nanocarriers enable controlled drug release, and shape-changing nanocarriers improve cellular uptake, allowing for personalized treatment approaches and combination therapies. This review provides an overview of recent developments and applications of deformable nanocarriers for enhancing tumor therapy, underscores the diverse design strategies employed to create deformable nanocarriers and elucidates their remarkable potential in targeted tumor therapy.
Collapse
Affiliation(s)
- Ziyang Cao
- Department of General SurgeryGuangzhou First People's Hospitalthe Second Affiliated HospitalSouth China University of TechnologyGuangzhouPeople's Republic of China
- Center for Medical Research on Innovation and TranslationInstitute of Clinical MedicineSchool of MedicineGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouPeople's Republic of China
| | - Jing Liu
- School of ChemistryChemical Engineering and Biotechnology Nanyang Technological UniversitySingaporeSingapore
| | - Xianzhu Yang
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdongPeople's Republic of China
| |
Collapse
|
46
|
Chen Y, Liu F, Pal S, Hu Q. Proteolysis-targeting drug delivery system (ProDDS): integrating targeted protein degradation concepts into formulation design. Chem Soc Rev 2024; 53:9582-9608. [PMID: 39171633 DOI: 10.1039/d4cs00411f] [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: 08/23/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a revolutionary paradigm in drug discovery and development, offering a promising avenue to tackle challenging therapeutic targets. Unlike traditional drug discovery approaches that focus on inhibiting protein function, TPD aims to eliminate proteins of interest (POIs) using modular chimeric structures. This is achieved through the utilization of proteolysis-targeting chimeras (PROTACs), which redirect POIs to E3 ubiquitin ligases, rendering them for degradation by the cellular ubiquitin-proteasome system (UPS). Additionally, other TPD technologies such as lysosome-targeting chimeras (LYTACs) and autophagy-based protein degraders facilitate the transportation of proteins to endo-lysosomal or autophagy-lysosomal pathways for degradation, respectively. Despite significant growth in preclinical TPD research, many chimeras fail to progress beyond this stage in the drug development. Various factors contribute to the limited success of TPD agents, including a significant hurdle of inadequate delivery to the target site. Integrating TPD into delivery platforms could surmount the challenges of in vivo applications of TPD strategies by reshaping their pharmacokinetics and pharmacodynamic profiles. These proteolysis-targeting drug delivery systems (ProDDSs) exhibit superior delivery performance, enhanced targetability, and reduced off-tissue side effects. In this review, we will survey the latest progress in TPD-inspired drug delivery systems, highlight the importance of introducing delivery ideas or technologies to the development of protein degraders, outline design principles of protein degrader-inspired delivery systems, discuss the current challenges, and provide an outlook on future opportunities in this field.
Collapse
Affiliation(s)
- Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fengyuan Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Samira Pal
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
47
|
Cao S, Wei Y, Yue Y, Wang D, Yang J, Xiong A, Zeng H. Mapping the evolution and research landscape of ferroptosis-targeted nanomedicine: insights from a scientometric analysis. Front Pharmacol 2024; 15:1477938. [PMID: 39386034 PMCID: PMC11461269 DOI: 10.3389/fphar.2024.1477938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Objective Notable progress has been made in "ferroptosis-based nano drug delivery systems (NDDSs)" over the past 11 years. Despite the ongoing absence of a comprehensive scientometric overview and up-to-date scientific mapping research, especially regarding the evolution, critical research pathways, current research landscape, central investigative themes, and future directions. Methods Data ranging from 1 January 2012, to 30 November 2023, were obtained from the Web of Science database. A variety of advanced analytical tools were employed for detailed scientometric and visual analyses. Results The results show that China significantly led the field, contributing 82.09% of the total publications, thereby largely shaping the research domain. Chen Yu emerged as the most productive author in this field. Notably, the journal ACS Nano had the greatest number of relevant publications. The study identified liver neoplasms, pancreatic neoplasms, gliomas, neoplasm metastases, and melanomas as the top five crucial disorders in this research area. Conclusion This research provides a comprehensive scientometric assessment, enhancing our understanding of NDDSs focused on ferroptosis. Consequently, it enables rapid access to essential information and facilitates the extraction of novel ideas in the field of ferroptotic nanomedicine for both experienced and emerging researchers.
Collapse
Affiliation(s)
- Siyang Cao
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yihao Wei
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, Hong Kong, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, China
| | - Yaohang Yue
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Deli Wang
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Ao Xiong
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hui Zeng
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Orthopedics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
48
|
Guo L, Zhao Q, Wang M. Core-Shell Microspheres with Encapsulated Gold Nanoparticle Carriers for Controlled Release of Anti-Cancer Drugs. J Funct Biomater 2024; 15:277. [PMID: 39452576 PMCID: PMC11509066 DOI: 10.3390/jfb15100277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Cancer is one of the major threats to human health and lives. However, effective cancer treatments remain a great challenge in clinical medicine. As a common approach for cancer treatment, chemotherapy has saved the life of millions of people; however, patients who have gone through chemotherapy often suffer from severe side effects owing to the inherent cytotoxicity of anti-cancer drugs. Stabilizing the blood concentration of an anti-cancer drug will reduce the occurrence or severity of side effects, and relies on using an appropriate drug delivery system (DDS) for achieving sustained or even on-demand drug delivery. However, this is still an unmet clinical challenge since the mainstay of anti-cancer drugs is small molecules, which tend to be diffused rapidly in the body, and conventional DDSs exhibit the burst release phenomenon. Here, we establish a class of DDSs based on biodegradable core-shell microspheres with encapsulated doxorubicin hydrochloride-loaded gold nanoparticles (DOX@Au@MSs), with the core-shell microspheres being made of poly(lactic-co-glycolic acid) in the current study. By harnessing the physical barrier of the biodegradable shell of core-shell microspheres, DOX@Au@MSs can provide a sustained release of the anti-cancer drug in the test duration (which is 21 days in the current study). Thanks to the photothermal properties of the encapsulated gold nanoparticle carriers, the core-shell biodegradable microspheres can be ruptured through remotely controlled near-infrared (NIR) light, thereby achieving an NIR-controlled triggered release of the anti-cancer drug. Furthermore, the route of the DOX-Au@MS-enabled controlled release of the anti-cancer drug can provide durable cancer cell ablation for the long period of 72 h.
Collapse
Affiliation(s)
- Lin Guo
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China;
| | - Qilong Zhao
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China
| | - Min Wang
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China;
| |
Collapse
|
49
|
Fawzy MP, Hassan HAFM, Sedky NK, Nafie MS, Youness RA, Fahmy SA. Revolutionizing cancer therapy: nanoformulation of miRNA-34 - enhancing delivery and efficacy for various cancer immunotherapies: a review. NANOSCALE ADVANCES 2024:d4na00488d. [PMID: 39309515 PMCID: PMC11414826 DOI: 10.1039/d4na00488d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
Despite recent advancements in cancer therapies, challenges such as severe toxic effects, non-selective targeting, resistance to chemotherapy and radiotherapy, and recurrence of metastatic tumors persist. Consequently, there has been considerable effort to explore innovative anticancer compounds, particularly in immunotherapy, which offer the potential for enhanced biosafety and efficacy in cancer prevention and treatment. One such avenue of exploration involves the miRNA-34 (miR-34) family, known for its ability to inhibit tumorigenesis across various cancers. Dysregulation of miR-34 has been observed in several human cancers, and it is recognized as a tumor suppressor microRNA due to its synergistic interaction with the well-established tumor suppressor p53. However, challenges have arisen with the therapeutic application of miR-34a. These include its susceptibility to degradation by RNase in serum, limiting its ability to penetrate capillary endothelium and reach target cells, as well as reports of immunoreactive adverse reactions. Furthermore, unexpected side effects may occur, such as the accumulation of therapeutic miRNAs in healthy tissues due to interactions with serum proteins on nano-vector surfaces, nanoparticle breakdown in the bloodstream due to shearing stress, and unsuccessful extravasation of nanocarriers to target cells owing to interstitial fluid pressure. Despite these challenges, miR-34a remains a promising candidate for cancer therapy, and other members of the miR-34 family have also shown potential in inhibiting tumor cell proliferation. While the in vivo applications of miR-34b/c are limited, they warrant further exploration for oncotherapy. Recently, procedures utilizing nanoparticles have been developed to address the challenges associated with the clinical use of miR-34, demonstrating efficacy both in vitro and in vivo. This review highlights emerging trends in nanodelivery systems for miR-34 targeting cancer cells, offering insights into novel nanoformulations designed to enhance the anticancer therapeutic activity and targeting precision of miR-34. As far as current knowledge extends, no similar recent review comprehensively addresses the diverse nanoformulations aimed at optimizing the therapeutic potential of miR-34 in anticancer strategies.
Collapse
Affiliation(s)
- Marola Paula Fawzy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt
| | - Hatem A F M Hassan
- Medway School of Pharmacy, University of Kent Central Avenue, Chatham Maritime Canterbury ME44TB UK
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University 11562 Cairo Egypt
| | - Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo 11835 Egypt
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah (P.O. 27272) Sharjah United Arab Emirates (UAE)
- Chemistry Department, Faculty of Science, Suez Canal University (P.O. 41522) Ismailia Egypt
| | - Rana A Youness
- Molecular Genetics and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| |
Collapse
|
50
|
Xu X, Lu W, Zhang H, Wang X, Huang C, Huang Q, Xu W, Xu W. Hepatoma-Targeting and ROS-Responsive Polymeric Micelle-Based Chemotherapy Combined with Photodynamic Therapy for Hepatoma Treatment. Int J Nanomedicine 2024; 19:9613-9635. [PMID: 39309184 PMCID: PMC11414760 DOI: 10.2147/ijn.s475531] [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: 05/01/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Background The combination of nanoplatform-based chemotherapy and photodynamic therapy (PDT) is a promising way to treat cancer. Celastrol (Cela) exhibits highly effective anti-hepatoma activity with low water solubility, poor bioavailability, non-tumor targeting, and toxic side effects. The combination of Cela-based chemotherapy and PDT via hepatoma-targeting and reactive oxygen species (ROS)-responsive polymeric micelles (PMs) could solve the application problem of Cela and further enhance antitumor efficacy. Methods In this study, Cela and photosensitizer chlorin e6 (Ce6) co-loaded glycyrrhetinic acid-modified carboxymethyl chitosan-thioketal-rhein (GCTR) PMs (Cela/Ce6/GCTR PMs) were prepared and characterized. The safety, ROS-sensitive drug release, and intracellular ROS production were evaluated. Furthermore, the in vitro anti-hepatoma effect and cellular uptaken in HepG2 and BEL-7402 cells, and in vivo pharmacokinetic, tissue distribution, and antitumor efficacy of Cela/Ce6/GCTR PMs in H22 tumor-bearing mice were then investigated. Results Cela/Ce6/GCTR PMs were successfully prepared with nanometer-scale particle size, favorable drug loading capacity, and encapsulation efficiency. Cela/Ce6/GCTR PMs exhibited a strong safety profile and better hemocompatibility, exhibiting less damage to normal tissues. Compared with Cela-loaded GCTR PMs, the ROS-responsiveness of Cela/Ce6/GCTR PMs was increased, and the release of Cela was accelerated after combination with PDT. Cela/Ce6/GCTR PMs can efficiently target liver tumor cells by uptake and have a high cell-killing effect in response to ROS. The combination of GCTR PM-based chemotherapy and PDT resulted in increased bioavailability of Cela and Ce6, improved liver tumor targeting, and better anti-hepatoma effects in vivo. Conclusion Hepatoma-targeting and ROS-responsive GCTR PMs co-loaded with Cela and Ce6 combined with PDT exhibited improved primary hepatic carcinoma therapeutic effects with lower toxicity to normal tissues, overcoming the limitations of monotherapy and providing new strategies for tumor treatment.
Collapse
Affiliation(s)
- Xueya Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| | - Weili Lu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| | - Hua Zhang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| | - Xiaoying Wang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| | - Caixia Huang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| | - Qiuping Huang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| | - Wen Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| | - Wei Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People’s Republic of China
| |
Collapse
|