1
|
Soman S, Kulkarni S, Kulkarni J, Dhas N, Roy AA, Pokale R, Mukharya A, Mutalik S. Metal-organic frameworks: a biomimetic odyssey in cancer theranostics. NANOSCALE 2025; 17:12620-12647. [PMID: 40336407 DOI: 10.1039/d5nr00794a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
This review comprehensively explores biomimetic metal-organic frameworks (MOFs) and their significant applications in cancer theranostics. Although MOFs have promising features such as adjustable porosity, improved surface area, and multifunctionality, they are limited by factors like low biocompatibility and specificity. Biomimetic strategies involving biological membranes and materials are proposed to address these challenges. The review begins by examining the unique characteristics and preparation methods of biomimetic carriers used in MOF-based nanoplatforms, with a comparative analysis of each method. It then delves into the various biomedical applications of biomimetic MOFs, including biosensing, bioimaging, immunotherapy, gene therapy, and multimodal therapies. The review also discusses the bio-interaction of these nanoplatforms, including their immunogenicity and interactions with fluids and tissues. Toxicity perspectives are also critically assessed. Overall, the article emphasizes the need for continued research into biomimetic MOFs, highlighting their potential to overcome current obstacles and provide safe, effective, and targeted therapeutic options.
Collapse
Affiliation(s)
- Soji Soman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, Tamil Nadu, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Jahnavi Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Amrita Arup Roy
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Rahul Pokale
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Anoushka Mukharya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
2
|
Song K, Ming J, Tao B, Zhao F, Huang S, Wu W, Jiang C, Li X. Emerging glucose oxidase-delivering nanomedicines for enhanced tumor therapy. J Control Release 2025; 381:113580. [PMID: 40024341 DOI: 10.1016/j.jconrel.2025.02.076] [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: 08/30/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Abnormalities in glucose metabolism have been shown to characterize malignant tumors. Glucose depletion by glucose oxidase (GOD) has shown great potential in tumor therapy by causing tumor starvation. Since 2017, nanomedicines have been designed and utilized to deliver GOD for more precise and effective glucose modulation, which can overcome intrinsic limitations of different cancer therapeutic modalities by remodeling the tumor microenvironment to enhance antitumor therapy. To date, the topic of GOD-delivering nanomedicines for enhancing tumor therapy has not been comprehensively summarized. Herein, this review aims to provide an overview and discuss in detail recent advances in GOD delivery and directly involved starvation therapy strategies, GOD-sensitized various tumor therapy strategies, and GOD-mediated multimodal antitumor strategies. Finally, the challenges and outlooks for the future progress of the emerging tumor therapeutic nanomedicines are discussed. This review provides intuitive and specific insights to a broad audience in the fields of nanomedicines, biomaterials, and cancer therapy.
Collapse
Affiliation(s)
- Kaiyue Song
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jiang Ming
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Feng Zhao
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China.
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Cong Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200092, China.
| | - Xianglong Li
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
3
|
Alavinia S, Ghorbani-Vaghei R, Haddadi R, Sanemar K, Uroomiye SS, Nourian A, Emami N. Anticancer effects of silymarin-loaded ACA-HA/sulfonamide IR-MOF nanoplatform in CT-26 xenograft model. Int J Biol Macromol 2025; 309:142651. [PMID: 40158557 DOI: 10.1016/j.ijbiomac.2025.142651] [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: 12/17/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Silymarin, a herbal compound with antioxidant and anti-inflammatory properties, has shown potential in inhibiting colon cancer cell growth. However, its clinical application is hindered by poor bioavailability. This study introduces a novel metal-organic framework, Sul-IRMOF, modified with 6-aminocaproic acid (ACA) and hyaluronic acid (HA) to create Sul-IRMOF-ACA-HA, which enables targeted drug delivery via HA receptors. Histological staining, biochemical analyses, and flow cytometry revealed that Sul-IRMOF-ACA-HA@Sily significantly enhances cytotoxicity against CT-26 cells compared to pure silymarin. According to oxidative stress markers, Sul-IRMOF-ACA-HA@Sily significantly reduced total thiol and glutathione (GSH) content compared to the control (p < 0.001) and silymarin (p < 0.001). Catalase (CAT) and superoxide dismutase (SOD) activities were significantly reduced in all treatment groups (p < 0.001), with Sul-IRMOF-ACA-HA@Sily showing the most pronounced effects (p < 0.001 vs. silymarin). Additionally, malondialdehyde (MDA) and nitric oxide (NO) levels were significantly elevated in the silymarin and Sul-IRMOF-ACA-HA@Sily groups compared to the control (p < 0.001), with Sul-IRMOF-ACA-HA@Sily further increasing both markers compared to silymarin (p < 0.001). The synergistic design of ACA, HA, and Sul-IRMOF offers a promising strategy to overcome silymarin's bioavailability limitations, positioning Sul-IRMOF-ACA-HA@Sily as a novel therapeutic candidate for colorectal cancer.
Collapse
Affiliation(s)
- Sedigheh Alavinia
- Department of Organic Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, 6517838683 Hamadan, Iran
| | - Ramin Ghorbani-Vaghei
- Department of Organic Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, 6517838683 Hamadan, Iran; Department of Organic Chemistry, Faculty of Chemistry, University of Guilan, Rasht, Iran.
| | - Rasool Haddadi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Hamadan University of Medical Sciences, 6517838678, Iran.
| | - Khashayar Sanemar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Hamadan University of Medical Sciences, 6517838678, Iran
| | - Seyed Sepehr Uroomiye
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Hamadan University of Medical Sciences, 6517838678, Iran
| | - Alireza Nourian
- Department of Pathobiology, School of Veterinary Science, Bu-Ali Sina University, Hamadan 6517658978, Iran
| | - Nafiseh Emami
- Department of Mechanical Engineering, Khomeinishahr Branch, Islamic Azad University, Khomeinishahr, Iran
| |
Collapse
|
4
|
Kostevšek N. Erythrocyte membrane vesicles as drug delivery systems: A systematic review of preclinical studies on biodistribution and pharmacokinetics. BIOMATERIALS ADVANCES 2025; 170:214234. [PMID: 39961269 DOI: 10.1016/j.bioadv.2025.214234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
This systematic review aims to summarize the development of erythrocyte membrane vesicles (EMVs) as drug delivery carriers, with a focus on elucidating their fate in terms of biodistribution and pharmacokinetics in preclinical studies. The PubMed database was systematically reviewed to search for original peer-reviewed published studies on the use of EMVs for drug delivery to summarize the preclinical findings, following the PRISMA guidelines. A total of 142 articles matched the selection criteria and were included in the review. For each study, the following parameters were extracted: type of active pharmaceutical ingredient (API) encapsulated into EMVs, EMVs-API formulation method and final particle size, EMVs surface modifications for active targeting, cell lines and animal models used in the study, crucial treatment data, biodistribution data and finally, where applicable, data about the EMVs circulation time and blood half-life. EMVs size did not vary significantly among the different formulation methods. A complete list of cell lines and animal models used is provided. Circulation times and data for blood half-life were grouped per animal type. For the most commonly used animal type, BALB/c mice, the average half-life of EMV-API was calculated to be 10.4 h, and in all cases, up to a 10-fold increase was observed compared with that of free API. Surface modifications did not drastically change the circulation time but did improve target tissue accumulation. The most critical weaknesses in the analysed studies were identified. Key points for future studies are provided to fill the current knowledge gaps and improve the quality of publications.
Collapse
Affiliation(s)
- Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia.
| |
Collapse
|
5
|
Fu Y, Sun J, Yang C, Li W, Wang Y. Diversified nanocarrier design to optimize glucose oxidase-mediated anti-tumor therapy: Strategy and progress. Int J Biol Macromol 2025; 306:141581. [PMID: 40023419 DOI: 10.1016/j.ijbiomac.2025.141581] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Given the inherent complexity and heterogeneity of tumors, current therapeutic approaches often fall short in meeting prognostic requirements. Starvation therapy (ST) utilizing glucose oxidase (GOx) has emerged as a promising strategy, specifically targeting tumor glucose consumption to disrupt nutrient supply. However, the therapeutic potential of GOx is significantly hampered by its inherent limitations as a protein, particularly its poor stability and short in vivo half-life. In recent years, the development of nanocarriors has provided an effective platform for intravenous and local tumor delivery of GOx. This review systematically examines three key strategies in GOx delivery: stimulus-response, biofilm modification, and local delivery. The progress in various carrier systems for GOx-mediated tumor therapy is comprehensively summarized, providing valuable insights for nanocarrier design. Furthermore, the existing challenges and future directions to advance the development of GOx-based tumor therapies are critically analyzed.
Collapse
Affiliation(s)
- Yuhan Fu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jialin Sun
- Department of medicine, Heilongjiang Minzu College, Harbin, Heilongjiang Province, China
| | - Chunyu Yang
- Department of Pathology, Harbin 242 Hospital, Harbin, Heilongjiang Province, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| |
Collapse
|
6
|
Premji TP, Dash BS, Lu YJ, Govindaraju DT, Chen JP. Reduced graphene oxide quantum dots/manganese dioxide/glucose oxidase nanoparticles for cascade catalytic cancer treatment in multimodal starvation therapy-augmented chemodynamic/photothermal therapy. Colloids Surf B Biointerfaces 2025; 253:114713. [PMID: 40279818 DOI: 10.1016/j.colsurfb.2025.114713] [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: 01/29/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Combination cancer therapy can boost the overall treatment efficacy using functional nanomaterials that specifically target cancer cells. Furthermore, the treatment outcome can be improved by focusing on specific characteristics in the tumor microenvironment (TME). In this study, tumor-targeting multifunctional nanoparticles were constructed from reduced graphene oxide quantum dots (rGOQD), manganese dioxide (MnO2), glucose oxidase (GOx), and cell-penetrating peptide (CPP). The rGOQD/MnO2/GOx/CPP nanoparticles can treat tumors by strengthening chemodynamic/photothermal therapy (CDT/PTT) with starvation therapy (ST). The MnO2 reacts with high concentrations of endogenous H2O2 in an acidic TME to produce reactive oxygen species (ROSs) from Mn2+. The highly cytotoxic hydroxyl radical (•OH) kills cancer cells and initiates CDT. The MnO2 can also consume the •OH scavenger glutathione (GSH) in cancer cells and eliminate their antioxidant defense. The GOx oxidizes glucose to cause cancer cell glucose starvation for ST, which produces H2O2 to boost the efficacy of CDT. By consuming glucose, ST mediated by GOx leads to reduced ATP production in the glycolysis pathway. This downregulates the expression of ATP-dependent heat shock proteins that provide cancer cell thermal resistance when the photothermal agent rGOQD is irradiated with near-infrared (NIR) light for PTT. Therefore, we prepare different rGOQD-based nanoparticles and characterize their physicochemical and biological properties. The nanoparticles were studied in vitro against U87 glioblastoma cells for targeted cancer therapy. Using nude mice bearing subcutaneous U87 tumors, the in vivo study indicates rGOQD/MnO2/GOx/CPP plus NIR irradiation can substantially inhibit the tumor growth rate without causing adverse effects from CPP-mediated trimodal ST/CDT/PTT.
Collapse
Affiliation(s)
- Thejas P Premji
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, School of Medicine, Chang Gung University, Kwei-San, Taoyuan 33305, Taiwan
| | | | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, School of Medicine, Chang Gung University, Kwei-San, Taoyuan 33305, Taiwan; Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33302, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
7
|
Han M, Zhou S, Liao Z, Zishan C, Yi X, Wu C, Zhang D, He Y, Leong KW, Zhong Y. Bimetallic peroxide-based nanotherapeutics for immunometabolic intervention and induction of immunogenic cell death to augment cancer immunotherapy. Biomaterials 2025; 315:122934. [PMID: 39509856 DOI: 10.1016/j.biomaterials.2024.122934] [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: 07/19/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
Immunotherapy has transformed cancer treatment, but its efficacy is often limited by the immunosuppressive characteristics of the tumor microenvironment (TME), which are predominantly influenced by the metabolism of cancer cells. Among these metabolic pathways, the indoleamine 2,3-dioxygenase (IDO) pathway is particularly crucial, as it significantly contributes to TME suppression and influences immune cell activity. Additionally, inducing immunogenic cell death (ICD) in tumor cells can reverse the immunosuppressive TME, thereby enhancing the efficacy of immunotherapy. Herein, we develop CGDMRR, a novel bimetallic peroxide-based nanodrug based on copper-cerium peroxide nanoparticles. These nanotherapeutics are engineered to mitigate tumor hypoxia and deliver therapeutics such as 1-methyltryptophan (1MT), glucose oxidase (GOx), and doxorubicin (Dox) in a targeted manner. The design aims to alleviate tumor hypoxia, reduce the immunosuppressive effects of the IDO pathway, and promote ICD. CGDMRR effectively inhibits the growth of 4T1 tumors and elicits antitumor immune responses by leveraging immunometabolic interventions and therapies that induce ICD. Furthermore, when CGDMRR is combined with a clinically certified anti-PD-L1 antibody, its efficacy in inhibiting tumor growth is enhanced. This improved efficacy extends beyond unilateral tumor models, also affecting bilateral tumors and lung metastases, due to the activation of systemic antitumor immunity. This study underscores CGDMRR's potential to augment the efficacy of PD-L1 blockade in breast cancer immunotherapy.
Collapse
Affiliation(s)
- Min Han
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Shiying Zhou
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Zunde Liao
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Chen Zishan
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Xiangting Yi
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China
| | - Chuanbin Wu
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Dongmei Zhang
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Yao He
- Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, United States.
| | - Yiling Zhong
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China; Department of Biomedical Engineering, Columbia University, New York, NY, 10027, United States.
| |
Collapse
|
8
|
Migliavacca M, Cedrún-Morales M, Ceballos M, Soprano E, Polo E, Pelaz B, Pino PD. Engineered cell membrane-cloaked metal-organic framework nanocrystals for intracellular cargo delivery. J Colloid Interface Sci 2025; 682:31-40. [PMID: 39612761 DOI: 10.1016/j.jcis.2024.11.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/07/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
This investigation demonstrates the development and functionality of cell membrane-cloaked UiO-67 nanosized metal-organic frameworks (NMOFs), which are engineered for precise intracellular delivery of encapsulated cargoes. Utilizing the robust and porous nature of UiO-67, we enveloped these NMOFs with fusogenic cell membrane-derived nanovesicles (FCSMs) sourced from adenocarcinomic human alveolar basal epithelial (A549) cells. This biomimetic coating enhances biocompatibility and leverages the homotypic targeting capabilities of the cell-derived coatings, facilitating direct cytoplasmic delivery and avoiding endolysosomal entrapment. Our nanoplatform exhibited high efficiency in loading and releasing two model cargoes: cresyl violet (CV), a staining agent, and carboplatin (CbPt), a chemotherapeutic agent. This approach demonstrated substantial potential in drug delivery in both 2D cell cultures and 3D spheroids. This study underscores the enhanced cellular uptake facilitated by the engineered biomimetic shell and highlights the system's capacity for sustained release, offering a promising strategy for targeted drug delivery.
Collapse
Affiliation(s)
- Martina Migliavacca
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Manuela Cedrún-Morales
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Manuel Ceballos
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Enrica Soprano
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Ester Polo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Bioquímica y Biología Molecular, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Pablo Del Pino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain.
| |
Collapse
|
9
|
Guo Z, Xiao Y, Wu W, Zhe M, Yu P, Shakya S, Li Z, Xing F. Metal-organic framework-based smart stimuli-responsive drug delivery systems for cancer therapy: advances, challenges, and future perspectives. J Nanobiotechnology 2025; 23:157. [PMID: 40022098 PMCID: PMC11871784 DOI: 10.1186/s12951-025-03252-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
Cancer treatment is currently one of the most critical healthcare issues globally. A well-designed drug delivery system can precisely target tumor tissues, improve efficacy, and reduce damage to normal tissues. Stimuli-responsive drug delivery systems (SRDDSs) have shown promising application prospects. Intelligent nano drug delivery systems responsive to endogenous stimuli such as weak acidity, complex redox characteristics, hypoxia, active energy metabolism, as well as exogenous stimuli like high temperature, light, pressure, and magnetic fields are increasingly being applied in chemotherapy, radiotherapy, photothermal therapy, photodynamic therapy, and various other anticancer approaches. Metal-organic frameworks (MOFs) have become promising candidate materials for constructing SRDDSs due to their large surface area, tunable porosity and structure, ease of synthesis and modification, and good biocompatibility. This paper reviews the application of MOF-based SRDDSs in various modes of cancer therapy. It summarizes the key aspects, including the classification, synthesis, modifications, drug loading modes, stimuli-responsive mechanisms, and their roles in different cancer treatment modalities. Furthermore, we address the current challenges and summarize the potential applications of artificial intelligence in MOF synthesis. Finally, we propose strategies to enhance the efficacy and safety of MOF-based SRDDSs, ultimately aiming at facilitating their clinical translation.
Collapse
Affiliation(s)
- Ziliang Guo
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuzhen Xiao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China
| | - Wenting Wu
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiyun Yu
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Sujan Shakya
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhihui Li
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Fei Xing
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Lu Y, Shan B, Li L, Jiang R, Li M. Tumor-Homing Biomimetic Near-Infrared II SERS Probes for Targeted Intraoperative Resection Guidance of Orthotopic Glioblastoma. NANO LETTERS 2025; 25:2325-2333. [PMID: 39884958 DOI: 10.1021/acs.nanolett.4c05622] [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: 02/01/2025]
Abstract
In vivo optical imaging holds great potential for surgical guidance with the ability to intraoperatively identify tumor lesions in a surgical bed and navigate their surgical excision in real time. Nevertheless, its full potential remains underexploited, mainly due to the dearth of high-performance optical probes. Herein, hybrid cell membrane-biomimetic near-infrared II surface-enhanced Raman spectroscopy (NIR-II SERS) probes are reported for intraoperative resection guidance of orthotopic glioblastoma. A novel class of plasmonic Au nanorod (AuNR)@Au-Ag frames is developed with remarkable plasmonic properties tunable beyond 1700 nm. We demonstrate the exceptional NIR-II SERS performance both in vitro and in vivo of the biomimetic NIR-II SERS probes created with AuNR@Au-Ag frames and hybrid cell membranes. The biomimetic NIR-II SERS probes are successfully applied in an orthotopic glioblastoma mouse model for intraoperative resection guidance with complete tumor removal and improved surgical outcomes. This study presents a promising strategy for precise NIR-II SERS surgical navigation.
Collapse
Affiliation(s)
- Yaxuan Lu
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Beibei Shan
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Linhu Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Renting Jiang
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
11
|
Mao L, Ma Y, Wen X, Luo Z, Zhu H, Kong J, Liu S, Fan X, Wang J, He C, Wu YL. Iron-glucose oxidase nanogel assembly for amplified starvation-ferroptosis anti-tumor therapy. Int J Biol Macromol 2025; 289:138804. [PMID: 39689793 DOI: 10.1016/j.ijbiomac.2024.138804] [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: 08/15/2024] [Revised: 12/14/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Developing advanced and effective enzyme-drug systems for cancer treatment is of significant interest. Herein, a novel approach is reported to create a highly active and robust enzyme-drug system. Glucose oxidase nanogels (nGOx) are first synthesized by polymerization on the surface of GOx using vinylimidazole as comonomers. Fe3+ are utilized to induce self-assembly of nGOx through the imidazole-metal coordination interaction to form GOx nanogel clusters (Fe@nGOx), enhancing the permeability and retention of nGOx into tumor cells by EPR effect. nGOx can deplete glucose in the presence of oxygen and generate H2O2, which is converted to highly cytotoxic hydroxyl radical (·OH) by Fe3+ and GSH, and the proximity between Fe3+ and GOx act in tandem for enhanced tumor therapy. The FeIII/FeII redox cycle reacts with GSH and H2O2, enabling continuous generation of ·OH within tumor cells, thus facilitating the anticancer effect. Moreover, the generation of H2O2 and ·OH can further promote the repolarization of tumor-associated macrophages from an M2 phenotype towards an M1 phenotype polarization, thus enhancing immune response. The cascade reaction between GOx and Fe3+/Fe2+ endows Fe@nGOx with excellent anti-tumor efficacy in mice models, highlighting its potential as a promising anticancer drug for clinical applications. This work establishes a new platform for utilizing enzyme/protein and metal ion complexes in versatile applications, advancing the field of enzyme-based cancer therapies.
Collapse
Affiliation(s)
- Liuzhou Mao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China
| | - Yedong Ma
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| | - Xiaoqing Wen
- Drug clinical trial institution, The first affiliated hospital of Xiamen university, Xiamen, Fujian, PR China
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China
| | - Houjuan Zhu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore
| | - Junhua Kong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore
| | - Siqi Liu
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| | - Xiaotong Fan
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore.
| | - Jinling Wang
- Department of Emergency and Critical Care Center, The Second Affiliated Hospital of Guangdong Medical University, No. 12 Minyou Road, Xiashan, Zhanjiang, Guangdong 524003, PR China.
| | - Chaobin He
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore; Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore.
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
12
|
Wang Y, Tang Y, Guo L, Yang X, Wu S, Yue Y, Xu C. Recent advances in zeolitic imidazolate frameworks as drug delivery systems for cancer therapy. Asian J Pharm Sci 2025; 20:101017. [PMID: 39931355 PMCID: PMC11808527 DOI: 10.1016/j.ajps.2025.101017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 04/16/2024] [Accepted: 11/24/2024] [Indexed: 02/13/2025] Open
Abstract
Biological nanotechnologies based on functional nanoplatforms have synergistically catalyzed the emergence of cancer therapies. As a subtype of metal-organic frameworks (MOFs), zeolitic imidazolate frameworks (ZIFs) have exploded in popularity in the field of biomaterials as excellent protective materials with the advantages of conformational flexibility, thermal and chemical stability, and functional controllability. With these superior properties, the applications of ZIF-based materials in combination with various therapies for cancer treatment have grown rapidly in recent years, showing remarkable achievements and great potential. This review elucidates the recent advancements in the use of ZIFs as drug delivery agents for cancer therapy. The structures, synthesis methods, properties, and various modifiers of ZIFs used in oncotherapy are presented. Recent advances in the application of ZIF-based nanoparticles as single or combination tumor treatments are reviewed. Furthermore, the future prospects, potential limitations, and challenges of the application of ZIF-based nanomaterials in cancer treatment are discussed. We except to fully explore the potential of ZIF-based materials to present a clear outline for their application as an effective cancer treatment to help them achieve early clinical application.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yixin Tang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Lei Guo
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xi Yang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Shanli Wu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Caina Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
13
|
Jiang S, Gurram B, Zhu J, Lei S, Zhang Y, He T, Tagit O, Fang H, Huang P, Lin J. Self-Boosting Programmable Release of Multiple Therapeutic Agents by Activatable Heterodimeric Prodrug-Enzyme Assembly for Antitumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409960. [PMID: 39569709 PMCID: PMC11727268 DOI: 10.1002/advs.202409960] [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: 10/08/2024] [Indexed: 11/22/2024]
Abstract
Endogenous stimuli-responsive prodrugs, due to their disease lesion specificity and reduced systemic toxicity, have been widely explored for antitumor therapy. However, reactive oxygen species (ROS) as classical endogenous stimuli in the tumor microenvironment (TME) are not enough to achieve the expected drug release. Herein, a ROS-activatable heterodimeric prodrug-loaded enzyme assembly is developed for self-boosting programmable release of multiple therapeutic agents. The heterodimeric prodrug NBS-TK-PTX (namely NTP) is composed of 5-(ethylamino)-9-diethylaminobenzo[a]phenothiazinium chloride analog (NBS), paclitaxel (PTX) and ROS-responsive thioketal (TK) linker, which shows a strong binding affinity with glucose oxidase (GOx), thus obtaining NTP@GOx assembly. Notably, the enzymatic activity of GOx in NTP@GOx is inhibited by NTP. The programmable release is achieved by following steps: i) NTP@GOx is partially dissociated in acidic TME, thus releasing a small segment of NTP and GOx. Thereupon, the enzymatic activity of GOx is recovered; ii) GOx-triggered pH reduction further facilitates the dissociation of NTP@GOx, thus accelerating a large amount of NTP and GOx release; iii) The TK linker of prodrug NTP is cleaved by hydrogen peroxide generated by GOx catalysis, thus expediting the release of NBS for Type-I photodynamic therapy and PTX for chemotherapy, respectively. The NTP@GOx shows great potential for multimodal synergistic cancer therapy.
Collapse
Affiliation(s)
- Shanshan Jiang
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
- Nanophotonics Research CenterShenzhen Key Laboratory of Micro‐Scale Optical Information TechnologyInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060China
| | - Bhaskar Gurram
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
- Department of BioInterfacesInstitute for Chemistry and BioanalyticsSchool of Life SciencesFHNW University of Applied Sciences and Arts Northwestern SwitzerlandMuttenz4132Switzerland
| | - Junfei Zhu
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
| | - Shan Lei
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
| | - Yifan Zhang
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
| | - Ting He
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
| | - Oya Tagit
- Department of BioInterfacesInstitute for Chemistry and BioanalyticsSchool of Life SciencesFHNW University of Applied Sciences and Arts Northwestern SwitzerlandMuttenz4132Switzerland
| | - Hui Fang
- Nanophotonics Research CenterShenzhen Key Laboratory of Micro‐Scale Optical Information TechnologyInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060China
| | - Peng Huang
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
| | - Jing Lin
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary TheranosticsSchool of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhen518055China
| |
Collapse
|
14
|
Agrawal S, Singh GK, Tiwari S. Focused starvation of tumor cells using glucose oxidase: A comprehensive review. Int J Biol Macromol 2024; 281:136444. [PMID: 39389487 DOI: 10.1016/j.ijbiomac.2024.136444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Starvation therapy targets the high metabolic demand of tumor cells. It primarily leans over the consumption of intracellular glucose and simultaneous blockade of alternative metabolic pathways. The strategy involves the use of glucose oxidase (GOx) for catalyzing the conversion of glucose into gluconic acid and hydrogen peroxide. Under these conditions, metabolic re-programming of tumor cells enables the utilization of substrates such as amino acids, fatty acids and lipids. This can be overcome by co-administration of chemo-, photo- and immuno-therapeutics together with glucose oxidase. Targeted delivery of glucose oxidase at tumor site can be enabled with the use of nanoformulations. In this review, we highlight that the outcomes of starvation therapy can be improved using rationally developed nano-formulations. It is possible to load synergistically acting bioactives in these formulations and deliver in site-specific manner and hence achieve the elimination of tumors cells with greater efficacy.
Collapse
Affiliation(s)
- Shivanshu Agrawal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Gireesh K Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya 824236, India
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India.
| |
Collapse
|
15
|
Wei P, Wang Y, Feng H, Zhang F, Ji Z, Zhang K, Zhang Q, Jiang L, Qian Y, Fu Y. Gene-Engineered Cerium-Exosomes Mediate Atherosclerosis Therapy Through Remodeling of the Inflammatory Microenvironment and DNA Damage Repair. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404463. [PMID: 39235409 DOI: 10.1002/smll.202404463] [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: 06/02/2024] [Revised: 07/10/2024] [Indexed: 09/06/2024]
Abstract
The pro-inflammatory immune microenvironment in the localized lesion areas and the absence of DNA damage repair mechanisms in endothelial cells serve as essential accelerating factors in the development of atherosclerosis. The lack of targeted therapeutic strategies represents a significant limitation in the efficacy of therapeutic agents for atherosclerosis. In this study, Genetically engineered SNHG12-loaded cerium-macrophage exosomes (Ce-Exo) are designed as atherosclerosis-targeting agents. In vivo studies demonstrated that Ce-Exo exhibited multivalent targeting properties for macrophages, with a 4.1-fold higher atherosclerotic plaque-aggregation ability than that of the control drugs. This suggests that Ce-Exo has a higher homing capacity and deeper penetration into the atherosclerotic plaque. In apolipoprotein E-deficient mice, Ce-Exo found to effectively remodel the immune microenvironment in the lesion area, repair endothelial cell damage, and inhibit the development of atherosclerosis. This study provides a novel approach to the treatment of atherosclerosis and demonstrates the potential of cell-derived drug carriers in biomedicine.
Collapse
Affiliation(s)
- Peng Wei
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yifan Wang
- Department of Emergency Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Haiyan Feng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
| | - Fan Zhang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, P. R. China
| | - Zhenyan Ji
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Kai Zhang
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Qiang Zhang
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Lixian Jiang
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yuxuan Qian
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yimu Fu
- Department of Emergency Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| |
Collapse
|
16
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [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: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
17
|
Zheng H, Huang L, An G, Guo L, Wang N, Yang W, Zhu Y. A Nanoreactor Based on Metal-Organic Frameworks With Triple Synergistic Therapy for Hepatocellular Carcinoma. Adv Healthc Mater 2024; 13:e2401743. [PMID: 39015058 DOI: 10.1002/adhm.202401743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/17/2024] [Indexed: 07/18/2024]
Abstract
The transformation of monotherapy into multimodal combined targeted therapy to fully exploit synergistic efficacy is of increasing interest in tumor treatment. In this work, a novel nanodrug-carrying platform based on iron-based MOFs, which is loaded with doxorubicin hydrochloride (DOX), dihydroartemisinin (DHA), and glucose oxidase (GOx), and concurrently covalently linked to the photosensitizer 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) in polydopamine (PDA)-encapsulated MIL-101(Fe) (denoted as MIL-101(Fe)-DOX-DHA@TCPP/GOx@PDA, MDDTG@P), is successfully developed. Upon entering the tumor microenvironment, MDDTG@P catalyzes the hydrogen peroxide (H2O2) into hydroxyl radicals (·OH) and depletes glutathione (GSH); thus, exerting the role of chemodynamic therapy (CDT). The reduced Fe2+ can also activate DHA, further expanding CDT and promoting tumor cell apoptosis. The introduced GOx will rapidly consume glucose and oxygen (O2) in the tumor; while, replenishing H2O2 for Fenton reaction, starving the cancer cells; and thus, realizing starvation and chemodynamic therapy. In addition, the covalent linkage of TCPP endows MDDTG@P with good photodynamic therapeutic (PDT) properties. Therefore, this study develops a nanocarrier platform for triple synergistic chemodynamic/photodynamic/starvation therapy, which has promising applications in the efficient treatment of tumors.
Collapse
Affiliation(s)
- Heming Zheng
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Lei Huang
- School of Stomatology, Minzhu Clinic of Stomatology Hospital Affiliated to Guangxi Medical University, Guangxi, 530007, China
| | - Guanghui An
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Lianshan Guo
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Wenhui Yang
- Department of Medical Laboratory, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QF, UK
| |
Collapse
|
18
|
Prakash O, Verma D, Singh PC. Exploring enzyme-immobilized MOFs and their application potential: biosensing, biocatalysis, targeted drug delivery and cancer therapy. J Mater Chem B 2024; 12:10198-10214. [PMID: 39283204 DOI: 10.1039/d4tb01556h] [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: 10/18/2024]
Abstract
Enzymes are indispensable in several applications including biosensing and degradation of pollutants and in the drug industry. However, adverse conditions restrict enzymes' utility in biocatalysis due to their inherent limitations. Metal-organic frameworks (MOFs), with their robust structure, offer an innovative avenue for enzyme immobilization, enhancing their resilience against harsh solvents and temperatures. This advancement is pivotal for application in bio-sensing, bio-catalysis, and specifically, targeted drug delivery in cancer therapy, where enzyme-MOF composites enable precise therapeutic localization, minimizing the side effects of traditional treatment. The adaptable nature of MOFs enhances drug biocompatibility and availability, significantly improving therapeutic outcomes. Moreover, the integration of enzyme-immobilized MOFs into bio-sensing represents a leap forward in the rapid and accurate identification of biomarkers, facilitating early diagnosis and disease monitoring. In bio-catalysis, this synergy promotes efficient and environmentally safe chemical synthesis, enhancing reaction rates and yields and broadening the scope of enzyme application in pharmaceutical and bio-fuel production. This review article explores the immobilization techniques and their biomedical applications, specifically focusing on drug delivery in cancer therapy and bio-sensing. Additionally, it addresses the challenges faced in this expanding field.
Collapse
Affiliation(s)
- Om Prakash
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226 007, India.
| | - Deepika Verma
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226 007, India.
| | - Poonam C Singh
- Division of Microbial Technology, CSIR-NBRI, Lucknow 226001, India
| |
Collapse
|
19
|
Cai J, Xu Y, Liao F. Advances in multifunctional metal-organic framework (MOF)-based nanoplatforms for cancer starvation therapy. Expert Rev Mol Med 2024; 26:e27. [PMID: 39397711 PMCID: PMC11488333 DOI: 10.1017/erm.2024.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/28/2024] [Accepted: 07/23/2024] [Indexed: 10/15/2024]
Abstract
Cancer remains a significant threat to human health today. Even though starvation therapy and other treatment methods have recently advanced to a new level of rapid development in tumour treatment, their limited therapeutic effectiveness and unexpected side effects prevent them from becoming the first option in clinical treatment. With rapid advancement in nanotechnology, the utilization of nanomaterials in therapeutics offers the potential to address the shortcomings in cancer treatment. Notably, multifunctional metal-organic framework (MOF) has been widely employed in cancer therapy due to their customizable shape, adjustable diameter, high porosity, diverse compositions, large specific surface area, high degree of functionalization and strong biocompatibility. This paper reviews the current progress and success of MOF-based multifunctional nanoplatforms for cancer starvation therapy, as well as the prospects and potential barriers for the application of MOF nanoplatforms in cancer starvation therapy.
Collapse
Affiliation(s)
- Jinghan Cai
- Renmin Hospital of Wuhan University, Wuhan University, Wuhan, P. R. China
| | - Yan Xu
- University Hospital, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| |
Collapse
|
20
|
Leitão MIPS, Morais TS. Tailored Metal-Based Catalysts: A New Platform for Targeted Anticancer Therapies. J Med Chem 2024; 67:16967-16990. [PMID: 39348603 DOI: 10.1021/acs.jmedchem.4c01680] [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: 10/02/2024]
Abstract
Innovative strategies for targeted anticancer therapies have gained significant momentum, with metal complexes emerging as tunable catalysts for more effective and safer treatments. Rational design and engineering of metal complexes enable the development of tailored molecular structures optimized for precision oncology. The strategic incorporation of metal complex catalysts within combinatorial therapies amplifies their anticancer properties. This perspective highlights the advancements in synthetic strategies and rational design since 2019, showing how tailored metal catalysts are optimized by designing structures to release or in situ synthesize active drugs, leveraging the target-specific characteristics to develop more precise cancer therapies. This review explores metal-based catalysts, including those conjugated with biomolecules, nanostructures, and metal-organic frameworks (MOFs), highlighting their catalytic activity in biological environments and their in vitro/in vivo performance. To sum up, the potential of metal complexes as catalysts to reshape the landscape of anticancer therapies and foster novel avenues for therapeutic advancement is emphasized.
Collapse
Affiliation(s)
- Maria Inês P S Leitão
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Tânia S Morais
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| |
Collapse
|
21
|
Deng Y, Huang S, Jiang G, Zhou L, Nezamzadeh-Ejhieh A, Liu J, Zhou Z. Current status and prospects of MOFs loaded with H 2O 2-related substances for ferroptosis therapy. RSC Med Chem 2024; 15:2996-3016. [PMID: 39309362 PMCID: PMC11411616 DOI: 10.1039/d4md00261j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024] Open
Abstract
Ferroptosis is a programmed cell death mechanism characterized by the accumulation of iron (Fe)-dependent lipid peroxides within cells. Ferroptosis holds excellent promise in tumor therapy. Metal-organic frameworks (MOFs) offer unique advantages in tumor ferroptosis treatment due to their high porosity, excellent stability, high biocompatibility, and targeting capabilities. Inducing ferroptosis in tumor cells primarily involves the production of reactive oxygen species (ROS), like hydroxyl radicals (˙OH), through iron-mediated Fenton reactions. However, the intrinsic H2O2 levels in tumor cells are often insufficient to sustain prolonged consumption, limiting therapeutic efficacy if ˙OH production is inadequate. Therefore, catalyzing or supplementing the intracellular H2O2 levels in tumor cells is essential for inducing ferroptosis by nanoscale metal-organic frameworks. This article reviews the biological characteristics and molecular mechanisms of ferroptosis, introduces H2O2-related substances, and reviews MOF-based nanoscale strategies for enhancing intracellular H2O2 levels in tumor cells. Finally, the challenges and prospects of this approach are discussed, aiming to provide insights into improving the effectiveness of ferroptosis induced by MOFs.
Collapse
Affiliation(s)
- Yu Deng
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Sida Huang
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Guanming Jiang
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital) 78 Wandao Road South Dongguan 523059 Guangdong China
| | - Luyi Zhou
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | | | - Jianqiang Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Zhikun Zhou
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| |
Collapse
|
22
|
Wang KN, Li ZZ, Zhou K, Liu B, Rao L, Bu LL. Cell Membrane-Coated Nanoparticles for Dental, Oral, and Craniofacial Diseases. RESEARCH (WASHINGTON, D.C.) 2024; 7:0478. [PMID: 39296987 PMCID: PMC11409001 DOI: 10.34133/research.0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024]
Abstract
Dental, oral, and craniofacial diseases can substantially impact the quality of human life, thereby posing a serious public health concern. Although conventional therapies such as surgery have solved these problems largely, the prognosis of patients is not always satisfactory. Cell membrane-coated nanoparticles (CMCNPs) carry nanodrugs with the help of natural cell membranes, therefore utilizing their remarkable ability to interface and interact with their surrounding environment. These nanoparticles have demonstrated substantial advantages in drug targeting, prolonging blood circulation time, penetrating biofilms, and immune escape. With the assistance of CMCNPs, the therapeutic effects of dental, oral, and craniofacial diseases can reach a higher level. CMCNPs have been applied for dental, oral, and craniofacial diseases for various conditions such as head and neck cancer, periodontal disease, and oral biosignal detection. For the therapies of head and neck cancer, CMCNPs have been widely utilized as a tool of chemotherapy, phototherapy, and immunotherapy, while yet to be exploited in imaging technique. In the end, we summarized the challenges and prospectives of CMCNPs for dental, oral, and craniofacial diseases: large-scale production with uniform standards and high quantity, extensive application directions in dental, oral, and craniofacial regions (implant, endodontics), and the promotion of its clinical application.
Collapse
Affiliation(s)
- Kang-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral & Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral & Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
23
|
Zhang W, Li L, Wu Y, Li C, Xu Z, Zhang N, Wang X, Zhao Y, Zu T, He Q, Jiao J, Zheng R. Biomimetic Iron-Based Nanoparticles Remodel Immunosuppressive Tumor Microenvironment for Metabolic Immunotherapy. Int J Nanomedicine 2024; 19:9333-9349. [PMID: 39286354 PMCID: PMC11403131 DOI: 10.2147/ijn.s473463] [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: 06/11/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Immunotherapy has led to a paradigm shift in reinvigorating treatment of cancer. Nevertheless, tumor associated macrophages (TAMs) experience functional polarization on account of the generation of suppressive metabolites, contributing to impaired antitumor immune responses. Methods Hence, metabolic reprogramming of tumor microenvironment (TME) can synergistically improve the efficacy of anti-tumor immunotherapy. Herein, we engineered an iron-based nanoplatform termed ERFe3O4 NPs. This platform features hollow Fe3O4 nanoparticles loaded with the natural product emodin, the outer layer is coated with red blood cell membrane (mRBCs) inserted with DSPE-PEG2000-galactose. This effectively modulates lactate production, thereby reversing the tumor immune suppressive microenvironment (TIME). Results The ERFe3O4 NPs actively targeted TAMs on account of their ability to bind to M2-like TAMs with high expression of galectin (Mgl). ERFe3O4 NPs achieved efficient ability to reverse TIME via the production of reducing lactate and prompting enrichment iron of high concentrations. Furthermore, ERFe3O4 NPs resulted in heightened expression of CD16/32 and enhanced TNF-α release, indicating promotion of M1 TAMs polarization. In vitro and in vivo experiments revealed that ERFe3O4 NPs induced significant apoptosis of tumor cells and antitumor immune response. Discussion This study combines Traditional Chinese Medicine (TCM) with nanomaterials to synergistically reprogram TAMs and reverse TIME, opening up new ideas for improving anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Wenyu Zhang
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Linquan Li
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Yaguang Wu
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Chengzhilin Li
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Zi'ang Xu
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Nianlei Zhang
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Xinyu Wang
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Yingchun Zhao
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Tingjian Zu
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Qingbin He
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Runxiao Zheng
- School of Clinical and Basic Medical Sciences, Medical Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| |
Collapse
|
24
|
Meng L, Tang L, Gao F, Zhu L, Liu X, Zhang J, Chang Y, Ma X, Guo Y. Hollow CeO 2-Based Nanozyme with Self-Accelerated Cascade Reactions for Combined Tumor Therapy. Chemistry 2024; 30:e202401640. [PMID: 38935332 DOI: 10.1002/chem.202401640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 06/28/2024]
Abstract
Nanozymes have obvious advantages in improving the efficiency of cancer treatment. However, due to the lack of tissue specificity, low catalytic efficiency, and so on, their clinical applications are limited. Herein, the nanoplatform CeO2@ICG@GOx@HA (CIGH) with self-accelerated cascade reactions is constructed. The as-prepared nanozyme shows the superior oxidase (OXD)-like, superoxide dismutase (SOD)-like, catalase (CAT)-like and peroxidase (POD)-like activities. At the same time, under 808 nm near-infrared (NIR) irradiation, the photodynamic and photothermal capabilities are also significantly enhanced due to the presence of indocyanine green (ICG). We demonstrate that the nanozyme CIGH can efficiently accumulate in the tumor and exhibit amplified cascade antitumor effects with negligible systemic toxicity through the combination of photodynamic therapy (PDT), photothermal therapy (PTT), chemodynamic therapy (CDT) and starvation therapy. The nanozyme prepared in this study provides a promising candidate for catalytic nanomedicines for efficient tumor therapy.
Collapse
Affiliation(s)
- Lili Meng
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Lingxue Tang
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Fangli Gao
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Liang Zhu
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xinhe Liu
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Jie Zhang
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Yi Chang
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xiaoming Ma
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Yuming Guo
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| |
Collapse
|
25
|
Manoharan D, Wang LC, Chen YC, Li WP, Yeh CS. Catalytic Nanoparticles in Biomedical Applications: Exploiting Advanced Nanozymes for Therapeutics and Diagnostics. Adv Healthc Mater 2024; 13:e2400746. [PMID: 38683107 DOI: 10.1002/adhm.202400746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Catalytic nanoparticles (CNPs) as heterogeneous catalyst reveals superior activity due to their physio-chemical features, such as high surface-to-volume ratio and unique optical, electric, and magnetic properties. The CNPs, based on their physio-chemical nature, can either increase the reactive oxygen species (ROS) level for tumor and antibacterial therapy or eliminate the ROS for cytoprotection, anti-inflammation, and anti-aging. In addition, the catalytic activity of nanozymes can specifically trigger a specific reaction accompanied by the optical feature change, presenting the feasibility of biosensor and bioimaging applications. Undoubtedly, CNPs play a pivotal role in pushing the evolution of technologies in medical and clinical fields, and advanced strategies and nanomaterials rely on the input of chemical experts to develop. Herein, a systematic and comprehensive review of the challenges and recent development of CNPs for biomedical applications is presented from the viewpoint of advanced nanomaterial with unique catalytic activity and additional functions. Furthermore, the biosafety issue of applying biodegradable and non-biodegradable nanozymes and future perspectives are critically discussed to guide a promising direction in developing span-new nanozymes and more intelligent strategies for overcoming the current clinical limitations.
Collapse
Affiliation(s)
- Divinah Manoharan
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Liu-Chun Wang
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wei-Peng Li
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
26
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
27
|
Torres-Herrero B, Armenia I, Ortiz C, de la Fuente JM, Betancor L, Grazú V. Opportunities for nanomaterials in enzyme therapy. J Control Release 2024; 372:619-647. [PMID: 38909702 DOI: 10.1016/j.jconrel.2024.06.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
In recent years, enzyme therapy strategies have rapidly evolved to catalyze essential biochemical reactions with therapeutic potential. These approaches hold particular promise in addressing rare genetic disorders, cancer treatment, neurodegenerative conditions, wound healing, inflammation management, and infectious disease control, among others. There are several primary reasons for the utilization of enzymes as therapeutics: their substrate specificity, their biological compatibility, and their ability to generate a high number of product molecules per enzyme unit. These features have encouraged their application in enzyme replacement therapy where the enzyme serves as the therapeutic agent to rectify abnormal metabolic and physiological processes, enzyme prodrug therapy where the enzyme initiates a clinical effect by activating prodrugs, and enzyme dynamic or starving therapy where the enzyme acts upon host substrate molecules. Currently, there are >20 commercialized products based on therapeutic enzymes, but approval rates are considerably lower than other biologicals. This has stimulated nanobiotechnology in the last years to develop nanoparticle-based solutions that integrate therapeutic enzymes. This approach aims to enhance stability, prevent rapid clearance, reduce immunogenicity, and even enable spatio-temporal activation of the therapeutic catalyst. This comprehensive review delves into emerging trends in the application of therapeutic enzymes, with a particular emphasis on the synergistic opportunities presented by incorporating enzymes into nanomaterials. Such integration holds the promise of enhancing existing therapies or even paving the way for innovative nanotherapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Torres-Herrero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Cecilia Ortiz
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Jesús Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Lorena Betancor
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
28
|
Li J, Luo P, Liu S, Fu M, Lin A, Liu Y, He Z, Qiao K, Fang Y, Qu L, Yang K, Wang K, Wang L, Jiang A. Effective strategies to enhance the diagnosis and treatment of RCC: The application of biocompatible materials. Mater Today Bio 2024; 27:101149. [PMID: 39100279 PMCID: PMC11296058 DOI: 10.1016/j.mtbio.2024.101149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 08/06/2024] Open
Abstract
Renal cell carcinoma (RCC) is recognized as one of the three primary malignant tumors affecting the urinary system, posing a significant risk to human health and life. Despite advancements in understanding RCC, challenges persist in its diagnosis and treatment, particularly in early detection and diagnosis due to issues of low specificity and sensitivity. Consequently, there is an urgent need for the development of effective strategies to enhance diagnostic accuracy and treatment outcomes for RCC. In recent years, with the extensive research on materials for applications in the biomedical field, some materials have been identified as promising for clinical applications, e.g., in the diagnosis and treatment of many tumors, including RCC. Herein, we summarize the latest materials that are being studied and have been applied in the early diagnosis and treatment of RCC. While focusing on their adjuvant effects, we also discuss their technical principles and safety, thus highlighting the value and potential of their application. In addition, we also discuss the limitations of the application of these materials and possible future directions, providing new insights for improving RCC diagnosis and treatment.
Collapse
Affiliation(s)
- Jinxin Li
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Shiyang Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Meiling Fu
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361101, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Ziwei He
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Kun Qiao
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Le Qu
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 East Zhongshan Road, Nanjing, 210000, China
| | - Kaidi Yang
- Department of Oncology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan, 572000, China
- Department of Oncology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Kunpeng Wang
- Department of Urology, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, 222061, China
- Department of Urology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The first People's Hospital of Lianyungang, 222061, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| |
Collapse
|
29
|
Huang QF, Li YH, Huang ZJ, Wu QX, Mei J, Wang W, Gui P, Cheng F, Wang GH. High-performance self-cascade nanoreactors for combined ferroptosis, photothermal therapy, and starving therapy. Eur J Pharm Biopharm 2024; 201:114367. [PMID: 38876360 DOI: 10.1016/j.ejpb.2024.114367] [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: 04/11/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Despite the great potential of starving therapy caused by nanoreactor based on glucose oxidase (GOX) in tumor therapy, efficiency and uncontrolled reaction rates in vivo lead to inevitable toxicity to normal tissues, which seriously hindering their clinical conversion. Herein, a cascade nanoreactor (GOX/Mn/MPDA) was constructed by coating mesoporous polydopamine nanoparticles (MPDA) with MnO2 shell and then depositing GOX into honeycomb-shaped manganese oxide nanostructures to achieve a combination of ferroptosis, photothermal therapy and starving therapy. Upon uptake of nanodrugs to cancer cells, the MnO2 shell would deplete glutathione (GSH) and produce Mn2+, while a large amount of H2O2 generated from the catalytic oxidation of glucose by GOX would accelerate the Fenton-like reaction mediated by Mn2+, producing high toxic •OH. More importantly, the cascade reaction between GOX and MnO2 would be further strengthened by localized hyperthermia caused by irradiated by near-infrared laser (NIR), inducing significant anti-tumor effects in vitro and in vivo. Regarding the effectiveness of tumor treatment in vivo, the tumor inhibition rate achieved an impressive 64.33%. This study provided a new strategy for anti-tumor therapeutic by designing a photothermal-enhanced cascade catalytic nanoreactor.
Collapse
Affiliation(s)
- Qun-Fa Huang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China
| | - Yan-Hong Li
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China
| | - Zeng-Jin Huang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China
| | - Quan-Xin Wu
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China
| | - Jun Mei
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China
| | - Wei Wang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China
| | - Ping Gui
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China
| | - Fan Cheng
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China.
| | - Guan-Hai Wang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, 523710 Dongguan, China.
| |
Collapse
|
30
|
Alshehri AM, Wilson OC. Biomimetic Hydrogel Strategies for Cancer Therapy. Gels 2024; 10:437. [PMID: 39057460 PMCID: PMC11275631 DOI: 10.3390/gels10070437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Recent developments in biomimetic hydrogel research have expanded the scope of biomedical technologies that can be used to model, diagnose, and treat a wide range of medical conditions. Cancer presents one of the most intractable challenges in this arena due to the surreptitious mechanisms that it employs to evade detection and treatment. In order to address these challenges, biomimetic design principles can be adapted to beat cancer at its own game. Biomimetic design strategies are inspired by natural biological systems and offer promising opportunities for developing life-changing methods to model, detect, diagnose, treat, and cure various types of static and metastatic cancers. In particular, focusing on the cellular and subcellular phenomena that serve as fundamental drivers for the peculiar behavioral traits of cancer can provide rich insights into eradicating cancer in all of its manifestations. This review highlights promising developments in biomimetic nanocomposite hydrogels that contribute to cancer therapies via enhanced drug delivery strategies and modeling cancer mechanobiology phenomena in relation to metastasis and synergistic sensing systems. Creative efforts to amplify biomimetic design research to advance the development of more effective cancer therapies will be discussed in alignment with international collaborative goals to cure cancer.
Collapse
Affiliation(s)
- Awatef M. Alshehri
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
- Department of Nanomedicine, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdelaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia;
| | - Otto C. Wilson
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
31
|
Pang ASR, Dinesh T, Pang NYL, Dinesh V, Pang KYL, Yong CL, Lee SJJ, Yip GW, Bay BH, Srinivasan DK. Nanoparticles as Drug Delivery Systems for the Targeted Treatment of Atherosclerosis. Molecules 2024; 29:2873. [PMID: 38930939 PMCID: PMC11206617 DOI: 10.3390/molecules29122873] [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: 04/11/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis continues to be a leading cause of morbidity and mortality globally. The precise evaluation of the extent of an atherosclerotic plaque is essential for forecasting its likelihood of causing health concerns and tracking treatment outcomes. When compared to conventional methods used, nanoparticles offer clear benefits and excellent development opportunities for the detection and characterisation of susceptible atherosclerotic plaques. In this review, we analyse the recent advancements of nanoparticles as theranostics in the management of atherosclerosis, with an emphasis on applications in drug delivery. Furthermore, the main issues that must be resolved in order to advance clinical utility and future developments of NP research are discussed. It is anticipated that medical NPs will develop into complex and advanced next-generation nanobotics that can carry out a variety of functions in the bloodstream.
Collapse
Affiliation(s)
- Alexander Shao-Rong Pang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Tarini Dinesh
- Department of Medicine, Government Kilpauk Medical College, Chennai 600010, Tamilnadu, India;
| | - Natalie Yan-Lin Pang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Vishalli Dinesh
- Department of Pathology, Dhanalakshmi Srinivasan Medical College Hospital, Perambalur 621113, Tamilnadu, India;
| | - Kimberley Yun-Lin Pang
- Division of Medicine, South Australia Health, Northern Adelaide Local Health Network, Adelaide, SA 5112, Australia; (K.Y.-L.P.); (S.J.J.L.)
| | - Cai Ling Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Shawn Jia Jun Lee
- Division of Medicine, South Australia Health, Northern Adelaide Local Health Network, Adelaide, SA 5112, Australia; (K.Y.-L.P.); (S.J.J.L.)
| | - George W. Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| | - Dinesh Kumar Srinivasan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| |
Collapse
|
32
|
Pandya I, Kumar S, Aswal VK, El Seoud O, Assiri MA, Malek N. Metal organic framework-based polymeric hydrogel: A promising drug delivery vehicle for the treatment of breast cancer. Int J Pharm 2024; 658:124206. [PMID: 38734276 DOI: 10.1016/j.ijpharm.2024.124206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/04/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
The constraints associated with current cancer therapies have inspired scientists to develop advanced, precise, and safe drug delivery methods. These delivery systems boost treatment effectiveness, minimize harm to healthy cells, and combat cancer recurrence. To design advanced drug delivery vehicle with these character, in the present manuscript, we have designed a self-healing and injectable hybrid hydrogel through synergistically interacting metal organic framework, CuBTC with the poly(vinyl alcohol) (PVA). This hybrid hydrogel acts as a localized drug delivery system and was used to encapsulate and release the anticancer drug 5-Fluorouracil selectively at the targeted site in response to the physiological pH. The hydrogel was formed through transforming the gaussian coil like matrix of PVA-CuBTC into a three-dimensional network of hydrogel upon the addition of crosslinker; borax. The biocompatible character of the hydrogel was confirmed through cell viability test. The biocompatible hybrid hydrogel then was used to encapsulate and studied for the pH responsive release behavior of the anti-cancer drug, 5-FU. The in vitro cytotoxicity of the drug-loaded hydrogel was evaluated against MCF-7 and HeLa cells. The study confirms that the hybrid hydrogel is effective for targeted and sustained release of anticancer drugs at cancer sites.
Collapse
Affiliation(s)
- Ishani Pandya
- Ionic Liquids Research Laboratory, Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India
| | - Sugam Kumar
- Solid State Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Vinod K Aswal
- Solid State Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Omar El Seoud
- Institute of Chemistry, University of São Paulo, 05508-000 São Paulo, SP, Brazil
| | - Mohammed A Assiri
- Chemistry Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Naved Malek
- Ionic Liquids Research Laboratory, Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India; Institute of Chemistry, University of São Paulo, 05508-000 São Paulo, SP, Brazil.
| |
Collapse
|
33
|
Li M, Liu Z, Tang J, Cheng L, Xue Y, Liu Y, Liu J. Facile Synthesis of a Multifunctional Porous Organic Polymer Nanosonosensitizer (mHM@HMME) for Enhanced Cancer Sonodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28104-28117. [PMID: 38769350 DOI: 10.1021/acsami.4c02651] [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: 05/22/2024]
Abstract
Sonodynamic therapy (SDT), which involves the activation of sonosensitizers to generate cytotoxic reactive oxygen species under ultrasound irradiation, is a promising noninvasive modality for cancer treatment. However, the clinical translational application of SDT is impeded by the lack of efficient sonosensitizers, the inefficient accumulation of sonosensitizers at tumor sites, and the complicated immunosuppressive tumor microenvironment. Herein, we developed a facilely synthesized multifunctional porous organic polymer nanosonosensitizer (mHM@HMME) for enhanced SDT. Specifically, mHM@HMME nanosonosensitizers were prepared by incorporating chemotherapeutic mitoxantrone into the one-step synthesis process of disulfide bond containing porous organic polymers, followed by loading with organic sonosensitizer (HMME) and camouflaging with a cancer cell membrane. Due to the cancer cell membrane camouflage, this multifunctional mHM@HMME nanosonosensitizer showed prolonged blood circulation and tumor targeting aggregation. Under ultrasound irradiation, the mHM@HMME nanosonosensitizer exhibited a satisfactory SDT performance both in vitro and in vivo. Moreover, the potent SDT combined with glutathione-responsive drug release in tumor cells induced robust immunogenic cell death to enhance the antitumor effect of SDT in turn. Overall, this facilely synthesized multifunctional mHM@HMME nanosonosensitizer shows great potential application in enhanced SDT.
Collapse
Affiliation(s)
- Meiting Li
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Zhuoyin Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Junjie Tang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - LiLi Cheng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Yifan Xue
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Yadong Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, People's Republic of China
| |
Collapse
|
34
|
Liu Y, Jiang Z, Yang X, Wang Y, Yang B, Fu Q. Engineering Nanoplatforms for Theranostics of Atherosclerotic Plaques. Adv Healthc Mater 2024; 13:e2303612. [PMID: 38564883 DOI: 10.1002/adhm.202303612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Atherosclerotic plaque formation is considered the primary pathological mechanism underlying atherosclerotic cardiovascular diseases, leading to severe cardiovascular events such as stroke, acute coronary syndromes, and even sudden cardiac death. Early detection and timely intervention of plaques are challenging due to the lack of typical symptoms in the initial stages. Therefore, precise early detection and intervention play a crucial role in risk stratification of atherosclerotic plaques and achieving favorable post-interventional outcomes. The continuously advancing nanoplatforms have demonstrated numerous advantages including high signal-to-noise ratio, enhanced bioavailability, and specific targeting capabilities for imaging agents and therapeutic drugs, enabling effective visualization and management of atherosclerotic plaques. Motivated by these superior properties, various noninvasive imaging modalities for early recognition of plaques in the preliminary stage of atherosclerosis are comprehensively summarized. Additionally, several therapeutic strategies are proposed to enhance the efficacy of treating atherosclerotic plaques. Finally, existing challenges and promising prospects for accelerating clinical translation of nanoplatform-based molecular imaging and therapy for atherosclerotic plaques are discussed. In conclusion, this review provides an insightful perspective on the diagnosis and therapy of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
35
|
Wang Z, Wang R, Geng Z, Luo X, Jia J, Pang S, Fan X, Bilal M, Cui J. Enzyme hybrid nanoflowers and enzyme@metal-organic frameworks composites: fascinating hybrid nanobiocatalysts. Crit Rev Biotechnol 2024; 44:674-697. [PMID: 37032548 DOI: 10.1080/07388551.2023.2189548] [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: 08/24/2022] [Accepted: 02/06/2023] [Indexed: 04/11/2023]
Abstract
Hybrid nanomaterials have recently emerged as a new interface of nanobiocatalysis, serving as a host platform for enzyme immobilization. Enzyme immobilization in inorganic crystal nanoflowers and metal-organic frameworks (MOFs) has sparked the bulk of scientific interest due to their superior performances. Many breakthroughs have been achieved recently in the preparation of various types of enzyme@MOF and enzyme-hybrid nanoflower composites. However, it is unfortunate that there are few reviews in the literature related to enzyme@MOF and enzyme-hybrid nanoflower composites and their improved synthesis strategies and their applications in biotechnology. In this review, innovative synthetic strategies for enzyme@MOF composites and enzyme-hybrid nanoflower composites are discussed. Enzyme@MOF composites and enzyme-hybrid nanoflower composites are reviewed in terms of biotechnological applications and potential research directions. We are convinced that a fundamental study and application of enzyme@MOF composites and enzyme-hybrid nanoflower composites will be understood by the reader as a result of this work. The summary of different synthetic strategies for enzyme@MOF composites and enzyme-hybrid nanoflower composites and the improvement of their synthetic strategies will also benefit the readers and provide ideas and thoughts in the future research process.
Collapse
Affiliation(s)
- Zichen Wang
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin Economic and Technological Development Area (TEDA), Tianjin, China
| | - Ruirui Wang
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin Economic and Technological Development Area (TEDA), Tianjin, China
| | - Zixin Geng
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin Economic and Technological Development Area (TEDA), Tianjin, China
| | - Xiuyan Luo
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin Economic and Technological Development Area (TEDA), Tianjin, China
| | - Jiahui Jia
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin Economic and Technological Development Area (TEDA), Tianjin, China
| | - Saizhao Pang
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin Economic and Technological Development Area (TEDA), Tianjin, China
| | - Xianwei Fan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guang Xi University, Nanning, China
| | - Muhammad Bilal
- Institute of Chemical Technology and Engineering, Poznan University of Technology, Poznan, Poland
| | - Jiandong Cui
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin Economic and Technological Development Area (TEDA), Tianjin, China
| |
Collapse
|
36
|
Han J, Dong H, Zhu T, Wei Q, Wang Y, Wang Y, Lv Y, Mu H, Huang S, Zeng K, Xu J, Ding J. Biochemical hallmarks-targeting antineoplastic nanotherapeutics. Bioact Mater 2024; 36:427-454. [PMID: 39044728 PMCID: PMC11263727 DOI: 10.1016/j.bioactmat.2024.05.042] [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: 02/20/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 07/25/2024] Open
Abstract
Tumor microenvironments (TMEs) have received increasing attention in recent years as they play pivotal roles in tumorigenesis, progression, metastases, and resistance to the traditional modalities of cancer therapy like chemotherapy. With the rapid development of nanotechnology, effective antineoplastic nanotherapeutics targeting the aberrant hallmarks of TMEs have been proposed. The appropriate design and fabrication endow nanomedicines with the abilities for active targeting, TMEs-responsiveness, and optimization of physicochemical properties of tumors, thereby overcoming transport barriers and significantly improving antineoplastic therapeutic benefits. This review begins with the origins and characteristics of TMEs and discusses the latest strategies for modulating the TMEs by focusing on the regulation of biochemical microenvironments, such as tumor acidosis, hypoxia, and dysregulated metabolism. Finally, this review summarizes the challenges in the development of smart anti-cancer nanotherapeutics for TME modulation and examines the promising strategies for combination therapies with traditional treatments for further clinical translation.
Collapse
Affiliation(s)
- Jing Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - He Dong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Tianyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Yongheng Wang
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Yun Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Yu Lv
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Shandeng Huang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Ke Zeng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jing Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
37
|
Chen M, Qin Y, Peng Y, Mai R, Teng H, Qi Z, Mo J. Advancing stroke therapy: the potential of MOF-based nanozymes in biomedical applications. Front Bioeng Biotechnol 2024; 12:1363227. [PMID: 38798955 PMCID: PMC11119330 DOI: 10.3389/fbioe.2024.1363227] [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: 12/30/2023] [Accepted: 04/12/2024] [Indexed: 05/29/2024] Open
Abstract
In this study, we explored the growing use of metal-organic framework (MOF)-based Nanozymes in biomedical research, with a specific emphasis on their applications in stroke therapy. We have discussed the complex nature of stroke pathophysiology, highlighting the crucial role of reactive oxygen species (ROS), and acknowledging the limitations of natural enzymes in addressing these challenges. We have also discussed the role of nanozymes, particularly those based on MOFs, their structural similarities to natural enzymes, and their potential to improve reactivity in various biomedical applications. The categorization of MOF nanozymes based on enzyme-mimicking activities is discussed, and their applications in stroke therapy are explored. We have reported the potential of MOF in treating stroke by regulating ROS levels, alleviation inflammation, and reducing neuron apoptosis. Additionally, we have addressed the challenges in developing efficient antioxidant nanozyme systems for stroke treatment. The review concludes with the promise of addressing these challenges and highlights the promising future of MOF nanozymes in diverse medical applications, particularly in the field of stroke treatment.
Collapse
Affiliation(s)
- Meirong Chen
- The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Medical College of Guangxi University, Nanning, China
| | - Yang Qin
- Department of Graduate and Postgraduate Education Management, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yongmei Peng
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Ruyu Mai
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Huanyao Teng
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Zhongquan Qi
- Medical College of Guangxi University, Nanning, China
| | - Jingxin Mo
- The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Lab of Neurology, The Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
38
|
Tao T, Rehman SU, Xu S, Zhang J, Xia H, Guo Z, Li Z, Ma K, Wang J. A biomimetic camouflaged metal organic framework for enhanced siRNA delivery in the tumor environment. J Mater Chem B 2024; 12:4080-4096. [PMID: 38577851 DOI: 10.1039/d3tb02827e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Gene silencing through RNA interference (RNAi), particularly using small double-stranded RNA (siRNA), has been identified as a potent strategy for targeted cancer treatment. Yet, its application faces challenges such as nuclease degradation, inefficient cellular uptake, endosomal entrapment, off-target effects, and immune responses, which have hindered its effective delivery. In the past few years, these challenges have been addressed significantly by using camouflaged metal-organic framework (MOF) nanocarriers. These nanocarriers protect siRNA from degradation, enhance cellular uptake, and reduce unintended side effects by effectively targeting desired cells while evading immune detection. By combining the properties of biomimetic membranes and MOFs, these nanocarriers offer superior benefits such as extended circulation times, enhanced stability, and reduced immune responses. Moreover, through ligand-receptor interactions, biomimetic membrane-coated MOFs achieve homologous targeting, minimizing off-target adverse effects. The MOFs, acting as the core, efficiently encapsulate and protect siRNA molecules, while the biomimetic membrane-coated surface provides homologous targeting, further increasing the precision of siRNA delivery to cancer cells. In particular, the biomimetic membranes help to shield the MOFs from the immune system, avoiding unwanted immune responses and improving their biocompatibility. The combination of siRNA with innovative nanocarriers, such as camouflaged-MOFs, presents a significant advancement in cancer therapy. The ability to deliver siRNA with precision and effectiveness using these camouflaged nanocarriers holds great promise for achieving more personalized and efficient cancer treatments in the future. This review article discusses the significant progress made in the development of siRNA therapeutics for cancer, focusing on their effective delivery through novel nanocarriers, with a particular emphasis on the role of metal-organic frameworks (MOFs) as camouflaged nanocarriers.
Collapse
Affiliation(s)
- Tongxiang Tao
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- University of Science and Technology of China, Hefei 230036, Anhui, P. R. China
| | - Sajid Ur Rehman
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
| | - Shuai Xu
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Jing Zhang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Haining Xia
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Zeyong Guo
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Zehua Li
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Kun Ma
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
| | - Junfeng Wang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- University of Science and Technology of China, Hefei 230036, Anhui, P. R. China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, Anhui, P. R. China
| |
Collapse
|
39
|
Weng Y, Chen R, Hui Y, Chen D, Zhao CX. Boosting Enzyme Activity in Enzyme Metal-Organic Framework Composites. CHEM & BIO ENGINEERING 2024; 1:99-112. [PMID: 38566967 PMCID: PMC10983012 DOI: 10.1021/cbe.3c00091] [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: 11/16/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024]
Abstract
Enzymes, as highly efficient biocatalysts, excel in catalyzing diverse reactions with exceptional activity and selective properties under mild conditions. Nonetheless, their broad applications are hindered by their inherent fragility, including low thermal stability, limited pH tolerance, and sensitivity to organic solvents and denaturants. Encapsulating enzymes within metal-organic frameworks (MOFs) can protect them from denaturation in these harsh environments. However, this often leads to a compromised enzyme activity. In recent years, extensive research efforts have been dedicated to enhancing enzymatic activity within MOFs, leading to the development of new enzyme-MOF composites that not only preserve their catalytic potential but also outperform their free counterparts. This Review provides a comprehensive review on recent developments in enzyme-MOF composites with a specific emphasis on their enhanced enzymatic activity compared to free enzymes.
Collapse
Affiliation(s)
- Yilun Weng
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Rui Chen
- School
of Chemical Engineering, The University
of Adelaide, Adelaide, SA 5005, Australia
| | - Yue Hui
- School
of Chemical Engineering, The University
of Adelaide, Adelaide, SA 5005, Australia
| | - Dong Chen
- State
Key Laboratory of Clean Energy Utilization, College of Energy Engineering, Zhejiang University, Hangzhou 310003, China
| | - Chun-Xia Zhao
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- School
of Chemical Engineering, The University
of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
40
|
Li X, Sun T, Jiang C. Intelligent Delivery Systems in Tumor Metabolism Regulation: Exploring the Path Ahead. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309582. [PMID: 38105387 DOI: 10.1002/adma.202309582] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/07/2023] [Indexed: 12/19/2023]
Abstract
Cancer metabolism plays multifaceted roles in the initiation and progression of tumors, and interventions in metabolism are considered fundamental approaches for cancer control. Within the vast metabolic networks of tumors, there exist numerous potential therapeutic targets, intricately interconnected with each other and with signaling networks related to immunity, metastasis, drug resistance, and more. Based on the characteristics of the tumor microenvironment, constructing drug delivery systems for multi-level modulation of the tumor microenvironment is proven as an effective strategy for achieving multidimensional control of cancer. Consequently, this article summarizes several features of tumor metabolism to provide insights into recent advancements in intelligent drug delivery systems for achieving multi-level regulation of the metabolic microenvironment in cancer, with the aim of offering a novel paradigm for cancer treatment.
Collapse
Affiliation(s)
- Xuwen Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tao Sun
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| |
Collapse
|
41
|
Li B, Ashrafizadeh M, Jiao T. Biomedical application of metal-organic frameworks (MOFs) in cancer therapy: Stimuli-responsive and biomimetic nanocomposites in targeted delivery, phototherapy and diagnosis. Int J Biol Macromol 2024; 260:129391. [PMID: 38242413 DOI: 10.1016/j.ijbiomac.2024.129391] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
The nanotechnology is an interdisciplinary field that has become a hot topic in cancer therapy. Metal-organic frameworks (MOFs) are porous materials and hybrid composites consisted of organic linkers and metal cations. Despite the wide application of MOFs in other fields, the potential of MOFs for purpose of cancer therapy has been revealed by the recent studies. High surface area and porosity, significant drug loading and encapsulation efficiency are among the benefits of using MOFs in drug delivery. MOFs can deliver genes/drugs with selective targeting of tumor cells that can be achieved through functionalization with ligands. The photosensitizers and photo-responsive nanostructures including carbon dots and gold nanoparticles can be loaded in/on MOFs to cause phototherapy-mediated tumor ablation. The immunogenic cell death induction and increased infiltration of cytotoxic CD8+ and CD4+ T cells can be accelerated by MOF platforms in providing immunotherapy of tumor cells. The stimuli-responsive MOF platforms responsive to pH, redox, enzyme and ion can accelerate release of therapeutics in tumor site. Moreover, MOF nanocomposites can be modified ligands and green polymers to improve their selectivity and biocompatibility for cancer therapy. The application of MOFs for the detection of cancer-related biomarkers can participate in the early diagnosis of patients.
Collapse
Affiliation(s)
- Beixu Li
- School of Policing Studies, Shanghai University of Political Science and Law, Shanghai 201701, China; Shanghai Fenglin Forensic Center, Shanghai 200231, China; State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Department of Pathology, University of Maryland, Baltimore, MD 21201, USA
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.
| | - Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang 110001, China.
| |
Collapse
|
42
|
Liu Z, Liu X, Zhang W, Gao R, Wei H, Yu CY. Current advances in modulating tumor hypoxia for enhanced therapeutic efficacy. Acta Biomater 2024; 176:1-27. [PMID: 38232912 DOI: 10.1016/j.actbio.2024.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Hypoxia is a common feature of most solid tumors, which promotes the proliferation, invasion, metastasis, and therapeutic resistance of tumors. Researchers have been developing advanced strategies and nanoplatforms to modulate tumor hypoxia to enhance therapeutic effects. A timely review of this rapidly developing research topic is therefore highly desirable. For this purpose, this review first introduces the impact of hypoxia on tumor development and therapeutic resistance in detail. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are also systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We provide a detailed discussion of the rationale and research progress of these strategies. Through a review of current trends, it is hoped that this comprehensive overview can provide new prospects for clinical application in tumor treatment. STATEMENT OF SIGNIFICANCE: As a common feature of most solid tumors, hypoxia significantly promotes tumor progression. Advanced nanoplatforms have been developed to modulate tumor hypoxia to enhanced therapeutic effects. In this review, we first introduce the impact of hypoxia on tumor progression. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We discuss the rationale and research progress of the above strategies in detail, and finally introduce future challenges for treatment of hypoxic tumors. By reviewing the current trends, this comprehensive overview can provide new prospects for clinical translatable tumor therapy.
Collapse
Affiliation(s)
- Zihan Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xinping Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Wei Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ruijie Gao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
43
|
Xing F, Xu J, Zhou Y, Yu P, Zhe M, Xiang Z, Duan X, Ritz U. Recent advances in metal-organic frameworks for stimuli-responsive drug delivery. NANOSCALE 2024; 16:4434-4483. [PMID: 38305732 DOI: 10.1039/d3nr05776c] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
After entering the human body, drugs for treating diseases, which are prone to delivery and release in an uncontrolled manner, are affected by various factors. Based on this, many researchers utilize various microenvironmental changes encountered during drug delivery to trigger drug release and have proposed stimuli-responsive drug delivery systems. In recent years, metal-organic frameworks (MOFs) have become promising stimuli-responsive agents to release the loaded therapeutic agents at the target site to achieve more precise drug delivery due to their high drug loading, excellent biocompatibility, and high stimuli-responsiveness. The MOF-based stimuli-responsive systems can respond to various stimuli under pathological conditions at the site of the lesion, releasing the loaded therapeutic agent in a controlled manner, and improving the accuracy and safety of drug delivery. Due to the changes in different physical and chemical factors in the pathological process of diseases, the construction of stimuli-responsive systems based on MOFs has become a new direction in drug delivery and controlled release. Based on the background of the rapidly increasing attention to MOFs applied in drug delivery, we aim to review various MOF-based stimuli-responsive drug delivery systems and their response mechanisms to various stimuli. In addition, the current challenges and future perspectives of MOF-based stimuli-responsive drug delivery systems are also discussed in this review.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiawei Xu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Germany
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhou Xiang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Xin Duan
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedic Surgery, The Fifth People's Hospital of Sichuan Province, Chengdu, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
44
|
Sadiq S, Khan S, Khan I, Khan A, Humayun M, Wu P, Usman M, Khan A, Alanazi AF, Bououdina M. A critical review on metal-organic frameworks (MOFs) based nanomaterials for biomedical applications: Designing, recent trends, challenges, and prospects. Heliyon 2024; 10:e25521. [PMID: 38356588 PMCID: PMC10864983 DOI: 10.1016/j.heliyon.2024.e25521] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Nanomaterials (NMs) have garnered significant attention in recent decades due to their versatile applications in a wide range of fields. Thanks to their tiny size, enhanced surface modifications, impressive volume-to-surface area ratio, magnetic properties, and customized optical dispersion. NMs experienced an incredible upsurge in biomedical applications including diagnostics, therapeutics, and drug delivery. This minireview will focus on notable examples of NMs that tackle important issues, demonstrating various aspects such as their design, synthesis, morphology, classification, and use in cutting-edge applications. Furthermore, we have classified and outlined the distinctive characteristics of the advanced NMs as nanoscale particles and hybrid NMs. Meanwhile, we emphasize the incredible potential of metal-organic frameworks (MOFs), a highly versatile group of NMs. These MOFs have gained recognition as promising candidates for a wide range of bio-applications, including bioimaging, biosensing, antiviral therapy, anticancer therapy, nanomedicines, theranostics, immunotherapy, photodynamic therapy, photothermal therapy, gene therapy, and drug delivery. Although advanced NMs have shown great potential in the biomedical field, their use in clinical applications is still limited by issues such as stability, cytotoxicity, biocompatibility, and health concerns. This review article provides a thorough analysis offering valuable insights for researchers investigating to explore new design, development, and expansion opportunities. Remarkably, we ponder the prospects of NMs and nanocomposites in conjunction with current technology.
Collapse
Affiliation(s)
- Samreen Sadiq
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
| | - Shoaib Khan
- College of Horticulture and Landscape Architecture, Northeast Agricultural University, Harbin, 150030, China
| | - Iltaf Khan
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management (IRC-HTCM), King Fahd University of Petroleum and Minerals, Dhahran, 31261, Saudi Arabia
| | - Aftab Khan
- Department of Physics, School of Science, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212100, China
| | - Muhammad Humayun
- Energy, Water and Environment Lab, College of Humanities and Sciences, Prince Sultan University, Riyadh, 11586, Saudi Arabia
| | - Ping Wu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
| | - Muhammad Usman
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management (IRC-HTCM), King Fahd University of Petroleum and Minerals, Dhahran, 31261, Saudi Arabia
| | - Abbas Khan
- Energy, Water and Environment Lab, College of Humanities and Sciences, Prince Sultan University, Riyadh, 11586, Saudi Arabia
- Department of Chemistry, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Amal Faleh Alanazi
- Energy, Water and Environment Lab, College of Humanities and Sciences, Prince Sultan University, Riyadh, 11586, Saudi Arabia
| | - Mohamed Bououdina
- Energy, Water and Environment Lab, College of Humanities and Sciences, Prince Sultan University, Riyadh, 11586, Saudi Arabia
| |
Collapse
|
45
|
Chen S, Luo Y, He Y, Li M, Liu Y, Zhou X, Hou J, Zhou S. In-situ-sprayed therapeutic hydrogel for oxygen-actuated Janus regulation of postsurgical tumor recurrence/metastasis and wound healing. Nat Commun 2024; 15:814. [PMID: 38280861 PMCID: PMC10821930 DOI: 10.1038/s41467-024-45072-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
Surgery is the mainstay of treatment modality for malignant melanoma. However, the deteriorative hypoxic microenvironment after surgery is recognized as a stemming cause for tumor recurrence/metastasis and delayed wound healing. Here we design and construct a sprayable therapeutic hydrogel (HIL@Z/P/H) encapsulating tumor-targeted nanodrug and photosynthetic cyanobacteria (PCC 7942) to prevent tumor recurrence/metastasis while promote wound healing. In a postsurgical B16F10 melanoma model in female mice, the nanodrug can disrupt cellular redox homeostasis via the photodynamic therapy-induced cascade reactions within tumor cells. Besides, the photosynthetically generated O2 by PCC 7942 can not only potentiate the oxidative stress-triggered cell death to prevent local recurrence of residual tumor cells, but also block the signaling pathway of hypoxia-inducible factor 1α to inhibit their distant metastasis. Furthermore, the long-lasting O2 supply and PCC 7942-secreted extracellular vesicles can jointly promote angiogenesis and accelerate the wound healing process. Taken together, the developed HIL@Z/P/H capable of preventing tumor recurrence/metastasis while promoting wound healing shows great application potential for postsurgical cancer therapy.
Collapse
Affiliation(s)
- Shuiling Chen
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yang Luo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yang He
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Ming Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yongjian Liu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xishen Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jianwen Hou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
46
|
Han X, Gong C, Yang Q, Zheng K, Wang Z, Zhang W. Biomimetic Nano-Drug Delivery System: An Emerging Platform for Promoting Tumor Treatment. Int J Nanomedicine 2024; 19:571-608. [PMID: 38260239 PMCID: PMC10802790 DOI: 10.2147/ijn.s442877] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
With the development of nanotechnology, nanoparticles (NPs) have shown broad prospects as drug delivery vehicles. However, they exhibit certain limitations, including low biocompatibility, poor physiological stability, rapid clearance from the body, and nonspecific targeting, which have hampered their clinical application. Therefore, the development of novel drug delivery systems with improved biocompatibility and high target specificity remains a major challenge. In recent years, biofilm mediated biomimetic nano-drug delivery system (BNDDS) has become a research hotspot focus in the field of life sciences. This new biomimetic platform uses bio-nanotechnology to encapsulate synthetic NPswithin biomimetic membrane, organically integrating the low immunogenicity, low toxicity, high tumor targeting, good biocompatibility of the biofilm with the adjustability and versatility of the nanocarrier, and shows promising applications in the field of precision tumor therapy. In this review, we systematically summarize the new progress in BNDDS used for optimizing drug delivery, providing a theoretical reference for optimizing drug delivery and designing safe and efficient treatment strategies to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Xiujuan Han
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Qingru Yang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Kaile Zheng
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
| | - Zhuo Wang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Wei Zhang
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
47
|
Guo X, Han L, Chen W, He H, Zhang W, Huang C, Wang X. Hypoxia and Singlet Oxygen Dual-Responsive Micelles for Photodynamic and Chemotherapy Therapy Featured with Enhanced Cellular Uptake and Triggered Cargo Delivery. Int J Nanomedicine 2024; 19:247-261. [PMID: 38229704 PMCID: PMC10790668 DOI: 10.2147/ijn.s432407] [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: 07/26/2023] [Accepted: 12/13/2023] [Indexed: 01/18/2024] Open
Abstract
Introduction Combination therapy provides better outcomes than a single therapy and becomes an efficient strategy for cancer treatment. In this study, we designed a hypoxia- and singlet oxygen-responsive polymeric micelles which contain azo and nitroimidazole groups for enhanced cellular uptake, repaid cargo release, and codelivery of photosensitizer Ce6 and hypoxia-activated prodrug tirapazamine TPZ (DHM-Ce6@TPZ), which could be used for combining Ce6-mediated photodynamic therapy (PDT) and PDT-activated chemotherapy to enhance the therapy effect of cancer. Methods The hypoxia- and singlet oxygen-responsive polymeric micelles DHM-Ce6@TPZ were prepared by film hydration method. The morphology, physicochemical properties, stimuli responsiveness, in vitro singlet oxygen production, cellular uptake, and cell viability were evaluated. In addition, the in vivo therapeutic effects of the micelles were verified using a tumor xenograft mice model. Results The resulting dual-responsive micelles not only increased the concentration of intracellular photosensitizer and TPZ, but also facilitated photosensitizer and TPZ release for enhanced integration of photodynamic and chemotherapy therapy. As a photosensitizer, Ce6 induced PDT by generating toxic singlet reactive oxygen species (ROS), resulting in a hypoxic tumor environment to activate the prodrug TPZ to achieve efficient chemotherapy, thereby evoking a synergistic photodynamic and chemotherapy therapeutic effect. The cascade synergistic therapeutic effect of DHM-Ce6@TPZ was effectively evaluated both in vitro and in vivo to inhibit tumor growth in a breast cancer mice model. Conclusion The designed multifunctional micellar nano platform could be a convenient and powerful vehicle for the efficient co-delivery of photosensitizers and chemical drugs for enhanced synergistic photodynamic and chemotherapy therapeutic effect of cancer.
Collapse
Affiliation(s)
- Xuliang Guo
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Lefei Han
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Wenyu Chen
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Huixin He
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Weijin Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Chaoqi Huang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Xiu Wang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| |
Collapse
|
48
|
Premji TP, Dash BS, Das S, Chen JP. Functionalized Nanomaterials for Inhibiting ATP-Dependent Heat Shock Proteins in Cancer Photothermal/Photodynamic Therapy and Combination Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:112. [PMID: 38202567 PMCID: PMC10780407 DOI: 10.3390/nano14010112] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024]
Abstract
Phototherapies induced by photoactive nanomaterials have inspired and accentuated the importance of nanomedicine in cancer therapy in recent years. During these light-activated cancer therapies, a nanoagent can produce heat and cytotoxic reactive oxygen species by absorption of light energy for photothermal therapy (PTT) and photodynamic therapy (PDT). However, PTT is limited by the self-protective nature of cells, with upregulated production of heat shock proteins (HSP) under mild hyperthermia, which also influences PDT. To reduce HSP production in cancer cells and to enhance PTT/PDT, small HSP inhibitors that can competitively bind at the ATP-binding site of an HSP could be employed. Alternatively, reducing intracellular glucose concentration can also decrease ATP production from the metabolic pathways and downregulate HSP production from glucose deprivation. Other than reversing the thermal resistance of cancer cells for mild-temperature PTT, an HSP inhibitor can also be integrated into functionalized nanomaterials to alleviate tumor hypoxia and enhance the efficacy of PDT. Furthermore, the co-delivery of a small-molecule drug for direct HSP inhibition and a chemotherapeutic drug can integrate enhanced PTT/PDT with chemotherapy (CT). On the other hand, delivering a glucose-deprivation agent like glucose oxidase (GOx) can indirectly inhibit HSP and boost the efficacy of PTT/PDT while combining these therapies with cancer starvation therapy (ST). In this review, we intend to discuss different nanomaterial-based approaches that can inhibit HSP production via ATP regulation and their uses in PTT/PDT and cancer combination therapy such as CT and ST.
Collapse
Affiliation(s)
- Thejas P. Premji
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
| | - Suprava Das
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (T.P.P.); (B.S.D.); (S.D.)
- Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
49
|
Zhang Y, Lu Y, Li Y, Xu Y, Song W. Poly(Glutamic Acid)-Engineered Nanoplatforms for Enhanced Cancer Phototherapy. Curr Drug Deliv 2024; 21:326-338. [PMID: 36650626 DOI: 10.2174/1567201820666230116164511] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 01/19/2023]
Abstract
Phototherapies, including photothermal therapy and photodynamic therapy, have gained booming development over the past several decades for their attractive non-invasiveness nature, negligible adverse effects, minimal systemic toxicity, and high spatial selectivity. Phototherapy usually requires three components: light irradiation, photosensitizers, and molecular oxygen. Photosensitizers can convert light energy into heat or reactive oxygen species, which can be used in the tumor-killing process. The direct application of photosensitizers in tumor therapy is restricted by their poor water solubility, fast clearance, severe toxicity, and low cellular uptake. The encapsulation of photosensitizers into nanostructures is an attractive strategy to overcome these critical limitations. Poly(glutamic acid) (PGA) is a kind of poly(amino acid)s containing the repeating units of glutamic acid. PGA has superiority for cancer treatment because of its good biocompatibility, low immunogenicity, and modulated pH responsiveness. The hydrophilicity nature of PGA allows the physical entrapment of photosensitizers and anticancer drugs via the construction of amphiphilic polymers. Moreover, the pendent carboxyl groups of PGA enable chemical conjugation with therapeutic agents. In this mini-review, we highlight the stateof- the-art design and fabrication of PGA-based nanoplatforms for phototherapy. We also discuss the potential challenges and future perspectives of phototherapy, and clinical translation of PGA-based nanomedicines.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Yiming Lu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Yicong Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai-200093, P. R. China
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Wenliang Song
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai-200093, P. R. China
| |
Collapse
|
50
|
Yang N, Li H, Cao C, Zhao L, Song X, Wang W, Xu W, Zhang Y, Chen P, Dong X. Tumor microenvironment-activated theranostic nanoreactor for NIR-II Photoacoustic imaging-guided tumor-specific photothermal therapy. FUNDAMENTAL RESEARCH 2024; 4:178-187. [PMID: 38933846 PMCID: PMC11197737 DOI: 10.1016/j.fmre.2022.04.021] [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: 12/05/2021] [Revised: 04/08/2022] [Accepted: 04/20/2022] [Indexed: 11/21/2022] Open
Abstract
Theranostic agents that can be sensitively and specifically activated by the tumor microenvironment (TME) have recently attracted considerable attention. In this study, TME-activatable 3,3',5,5'-tetramethylbenzidine (TMB)-copper peroxide (CuO2)@poly(lactic-co-glycolic acid) (PLGA)@red blood cell membrane (RBCM) (TCPR) nanoparticles (NPs) for second near-infrared photoacoustic imaging-guided tumor-specific photothermal therapy were developed by co-loading CuO2 NPs and TMB into PLGA camouflaged by RBCMs. As an efficient H2O2 supplier, once exposed to a proton-rich TME, CuO2 NPs can generate H2O2 and Cu2+, which are further reduced to Cu+ by endogenous glutathione. Subsequently, the Cu+-mediated Fenton-like reaction produces cytotoxic ·OH to kill the cancer cells and induce TMB-mediated photoacoustic and photothermal effects. Combined with the RBCM modification-prolonged blood circulation, TCPR NPs display excellent specificity and efficiency in suppressing tumor growth, paving the way for more accurate, safe, and efficient cancer theranostics.
Collapse
Affiliation(s)
- Nan Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Hui Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Changyu Cao
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Lei Zhao
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Wenjun Wang
- School of Physical Science and Information Technology, Liaocheng University, Liaocheng 252059, China
| | - Wenjing Xu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Yewei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Peng Chen
- School of Chemical and Biomedical Engineering, Lee Kong Chian School of Medicine, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| |
Collapse
|