1
|
Liu H, Li Y, Wang Y, Zhang L, Liang X, Gao C, Yang Y. Red blood cells-derived components as biomimetic functional materials: Matching versatile delivery strategies based on structure and function. Bioact Mater 2025; 47:481-501. [PMID: 40034412 PMCID: PMC11872572 DOI: 10.1016/j.bioactmat.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
Red blood cells (RBCs), often referred to as "intelligent delivery systems", can serve as biological or hybrid drug carriers due to their inherent advantages and characteristics. This innovative approach has the potential to enhance biocompatibility, pharmacokinetics, and provide targeting properties for drugs. By leveraging the unique structure and contents of RBCs, drug-loading pathways can be meticulously designed to align with these distinctive features. This review article primarily discusses the drug delivery strategies and their applications that are informed by the structural and functional properties of the main components of RBCs, including living RBCs, membranes, hollow RBCs, and hemoglobin. Overall, this review article would assist efforts to make better decisions on optimization and rational utilization of RBCs derivatives-based drug delivery strategies for the future direction in clinical translation.
Collapse
Affiliation(s)
- Hangbing Liu
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Yi Li
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Yuli Wang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Liying Zhang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Xiaoqing Liang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Chunsheng Gao
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| |
Collapse
|
2
|
Fan S, Wang W, Che W, Xu Y, Jin C, Dong L, Xia Q. Nanomedicines Targeting Metabolic Pathways in the Tumor Microenvironment: Future Perspectives and the Role of AI. Metabolites 2025; 15:201. [PMID: 40137165 PMCID: PMC11943624 DOI: 10.3390/metabo15030201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Tumor cells engage in continuous self-replication by utilizing a large number of resources and capabilities, typically within an aberrant metabolic regulatory network to meet their own demands. This metabolic dysregulation leads to the formation of the tumor microenvironment (TME) in most solid tumors. Nanomedicines, due to their unique physicochemical properties, can achieve passive targeting in certain solid tumors through the enhanced permeability and retention (EPR) effect, or active targeting through deliberate design optimization, resulting in accumulation within the TME. The use of nanomedicines to target critical metabolic pathways in tumors holds significant promise. However, the design of nanomedicines requires the careful selection of relevant drugs and materials, taking into account multiple factors. The traditional trial-and-error process is relatively inefficient. Artificial intelligence (AI) can integrate big data to evaluate the accumulation and delivery efficiency of nanomedicines, thereby assisting in the design of nanodrugs. Methods: We have conducted a detailed review of key papers from databases, such as ScienceDirect, Scopus, Wiley, Web of Science, and PubMed, focusing on tumor metabolic reprogramming, the mechanisms of action of nanomedicines, the development of nanomedicines targeting tumor metabolism, and the application of AI in empowering nanomedicines. We have integrated the relevant content to present the current status of research on nanomedicines targeting tumor metabolism and potential future directions in this field. Results: Nanomedicines possess excellent TME targeting properties, which can be utilized to disrupt key metabolic pathways in tumor cells, including glycolysis, lipid metabolism, amino acid metabolism, and nucleotide metabolism. This disruption leads to the selective killing of tumor cells and disturbance of the TME. Extensive research has demonstrated that AI-driven methodologies have revolutionized nanomedicine development, while concurrently enabling the precise identification of critical molecular regulators involved in oncogenic metabolic reprogramming pathways, thereby catalyzing transformative innovations in targeted cancer therapeutics. Conclusions: The development of nanomedicines targeting tumor metabolic pathways holds great promise. Additionally, AI will accelerate the discovery of metabolism-related targets, empower the design and optimization of nanomedicines, and help minimize their toxicity, thereby providing a new paradigm for future nanomedicine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Dong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| | - Qin Xia
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| |
Collapse
|
3
|
Hoveidaei AH, Sadat-Shojai M, Nabavizadeh SS, Niakan R, Shirinezhad A, MosalamiAghili S, Tabaie S. Clinical challenges in bone tissue engineering - A narrative review. Bone 2025; 192:117363. [PMID: 39638083 DOI: 10.1016/j.bone.2024.117363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Bone tissue engineering (BTE) has emerged as a promising approach to address large bone defects caused by trauma, infections, congenital malformations, and tumors. This review focuses on scaffold design, cell sources, growth factors, and vascularization strategies, highlighting their roles in developing effective treatments. We explore the complexities of balancing mechanical properties, porosity, and biocompatibility in scaffold materials, alongside optimizing mesenchymal stem cell delivery methods. The critical role of growth factors in bone regeneration and the need for controlled release systems are discussed. Vascularization remains a significant hurdle, with strategies such as angiogenic factors, co-culture systems, and bioprinting under investigation. Mechanical challenges, tissue responses, and inflammation management are examined, alongside gene therapy's potential for enhancing osteogenesis and angiogenesis via both viral and non-viral delivery methods. The review emphasizes the impact of patient-specific factors on bone healing outcomes and the importance of personalized approaches. Future directions are described, emphasizing the necessity of interdisciplinary cooperation to advance the field of BTE and convert laboratory results into clinically feasible solutions.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Sara S Nabavizadeh
- Otolaryngology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Niakan
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Sean Tabaie
- Department of Orthopaedic Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
4
|
Horta M, Soares P, Leite Pereira C, Lima RT. Emerging Approaches in Glioblastoma Treatment: Modulating the Extracellular Matrix Through Nanotechnology. Pharmaceutics 2025; 17:142. [PMID: 40006509 PMCID: PMC11859630 DOI: 10.3390/pharmaceutics17020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Glioblastoma's (GB) complex tumor microenvironment (TME) promotes its progression and resistance to therapy. A critical component of TME is the extracellular matrix (ECM), which plays a pivotal role in promoting the tumor's invasive behavior and aggressiveness. Nanotechnology holds significant promise for GB treatment, with the potential to address challenges posed by both the blood-brain barrier and the GB ECM. By enabling targeted delivery of therapeutic and diagnostic agents, nanotechnology offers the prospect of improving treatment efficacy and diagnostic accuracy at the tumor site. This review provides a comprehensive exploration of GB, including its epidemiology, classification, and current treatment strategies, alongside the intricacies of its TME. It highlights nanotechnology-based strategies, focusing on nanoparticle formulations such as liposomes, polymeric nanoparticles, and gold nanoparticles, which have shown promise in GB therapy. Furthermore, it explores how different emerging nanotechnology strategies modulate the ECM to overcome the challenges posed by its high density, which restricts drug distribution within GB tumors. By emphasizing the intersection of nanotechnology and GB ECM, this review underscores an innovative approach to advancing GB treatment. It addresses the limitations of current therapies, identifies new research avenues, and emphasizes the potential of nanotechnology to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Horta
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Paula Soares
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Catarina Leite Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Raquel T. Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
5
|
Li B, Zhao Q, Yang H, Wang X, Zhang Z, Gong Y, Wan X. Long-Circulating and Targeted Liposomes Co-loading Cisplatin and Mifamurtide: Formulation and Delivery in Osteosarcoma Cells. AAPS PharmSciTech 2024; 25:272. [PMID: 39592553 DOI: 10.1208/s12249-024-02992-7] [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: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Osteosarcoma (OS) is one of the most common primary bone sarcoma with high malignant degree and poor prognosis, for which there is an urgent need to develop novel therapeutic approaches. Recent research has revealed that mifamurtide significantly improved the outcome of OS patients when combined with adjuvant chemotherapy drugs including cisplatin (DDP). The present study aimed to construct a drug delivery system co-loading DDP and mifamurtide. Long-circulating targeted liposomes co-loading DDP and mifamurtide were constructed with Soy lecithin (SPC), cholesterol (Chol) and 1,2-distearoylglycero-3-phosphoethanolamine-n-[poly(ethyleneglycol)] (DSPE-PEG), modified with MMP14 targeting peptide BCY-B in the surface of liposomes. In addition to characterization, the cellular uptake, endocytosis pathway and inhibition on cell viability, migration, invasion and cell apoptosis of MG-63 cells were explored. The constructed liposomal delivery possessed the basic characteristics of liposomes and showed high affinity to MG-63 cells, resulting in high uptake efficiency in MG-63 cells. The endocytosis might be involved in multiple pathways including caveolae-mediated endocytosis, clathrin-mediated endocytosis and macropinocytosis, dependently on energy. The constructed long-circulating targeted liposomes co-loading DDP and mifamurtide significantly inhibited the cell viability, migration, invasion and cell apoptosis of MG-63 cells, improving the antitumor effect of DDP and mifamurtide in vitro. The constructed liposomal delivery system is suitable for co-loading DDP and mifamurtide to achieve active tumor targeting, supplying a new strategy for the treatment of OS.
Collapse
Affiliation(s)
- Bo Li
- Department of Musculoskeletal Tumor, Gansu Provincial Hospital, Lanzhou, China
| | - Qianhui Zhao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Hanyu Yang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Xueyuying Wang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Zhijun Zhang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, 266042, China.
| | - Xu Wan
- Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [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: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
7
|
Zong L, Xu H, Zhang H, Tu Z, Zhang X, Wang S, Li M, Feng Y, Wang B, Li L, Xie X, He Z, Pu X. A review of matrix metalloproteinase-2-sensitive nanoparticles as a novel drug delivery for tumor therapy. Int J Biol Macromol 2024; 262:130043. [PMID: 38340921 DOI: 10.1016/j.ijbiomac.2024.130043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Matrix metalloproteinase-2 (MMP-2)-responsive nanodrug vehicles have garnered significant attention as antitumor drug delivery systems due to the extensive research on matrix metalloproteinases (MMPs) within the tumor extracellular matrix (ECM). These nanodrug vehicles exhibit stable circulation in the bloodstream and accumulate specifically in tumors through various mechanisms. Upon reaching tumor tissues, their structures are degraded in response to MMP-2 within the ECM, resulting in drug release. This controlled drug release significantly increases drug concentration within tumors, thereby enhancing its antitumor efficacy while minimizing side effects on normal organs. This review provides an overview of MMP-2 characteristics, enzyme-sensitive materials, and current research progress regarding their application as MMP-2-responsive nanodrug delivery system for anti-tumor drugs, as well as considering their future research prospects. In conclusion, MMP-2-sensitive drug delivery carriers have a broad application in all kinds of nanodrug delivery systems and are expected to become one of the main means for the clinical development and application of nanodrug delivery systems in the future.
Collapse
Affiliation(s)
- Lanlan Zong
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China; Huaihe Hospital of Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Hongliang Xu
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Huiqi Zhang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Ziwei Tu
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Xiao Zhang
- Department of Pharmacy, Hebei Provincial Clinical Research Center for Eye Diseases, Hebei Provincial Key Laboratory of Ophthalmology, Hebei Provincial Eye Hospital, Xingtai City, Hebei Province 054001, China
| | - Shumin Wang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Meigui Li
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Yu Feng
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Binke Wang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Luhui Li
- Medical School, Henan Technical Institute, Kaifeng, Henan 475004, China
| | - Xinmei Xie
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China.
| | - Zhonggui He
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China; Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Xiaohui Pu
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China; Huaihe Hospital of Henan University, N. Jinming Ave., Kaifeng 475004, China.
| |
Collapse
|
8
|
Shikalov A, Koman I, Kogan NM. Targeted Glioma Therapy-Clinical Trials and Future Directions. Pharmaceutics 2024; 16:100. [PMID: 38258110 PMCID: PMC10820492 DOI: 10.3390/pharmaceutics16010100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of glioma, with a median survival of 14.6 months post-diagnosis. Understanding the molecular profile of such tumors allowed the development of specific targeted therapies toward GBM, with a major role attributed to tyrosine kinase receptor inhibitors and immune checkpoint inhibitors. Targeted therapeutics are drugs that work by specific binding to GBM-specific or overexpressed markers on the tumor cellular surface and therefore contain a recognition moiety linked to a cytotoxic agent, which produces an antiproliferative effect. In this review, we have summarized the available information on the targeted therapeutics used in clinical trials of GBM and summarized current obstacles and advances in targeted therapy concerning specific targets present in GBM tumor cells, outlined efficacy endpoints for major classes of investigational drugs, and discussed promising strategies towards an increase in drug efficacy in GBM.
Collapse
Affiliation(s)
| | | | - Natalya M. Kogan
- Department of Molecular Biology, Institute of Personalized and Translational Medicine, Ariel University, Ariel 40700, Israel; (A.S.); (I.K.)
| |
Collapse
|
9
|
Girma A, Mebratie G, Alamnie G, Bekele T. Advances With Selected Nanostructured Materials in Health Care. REFERENCE MODULE IN MATERIALS SCIENCE AND MATERIALS ENGINEERING 2024. [DOI: 10.1016/b978-0-323-95486-0.00090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
10
|
Son J, Parveen S, MacPherson D, Marciano Y, Huang RH, Ulijn RV. MMP-responsive nanomaterials. Biomater Sci 2023; 11:6457-6479. [PMID: 37623747 DOI: 10.1039/d3bm00840a] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Matrix metalloproteinases (MMP) are enzymes that degrade the extracellular matrix and regulate essential normal cell behaviors. Inhibition of these enzymes has been a strategy for anti-cancer therapy since the 1990s, but with limited success. A new type of MMP-targeting strategy exploits the innate selective hydrolytic activity and consequent catalytic signal amplification of the proteinases, rather than inhibiting it. Using nanomaterials, the enzymatic chemical reaction can trigger the temporal and spatial activation of the anti-cancer effects, amplify the associated response, and cause mechanical damage or report on cancer cells. We analyzed nearly 60 literature studies that incorporate chemical design strategies that lead to spatial, temporal, and mechanical control of the anti-cancer effect through four modes of action: nanomaterial shrinkage, induced aggregation, formation of cytotoxic nanofibers, and activation by de-PEGylation. From the literature analysis, we derived chemical design guidelines to control and enhance MMP activation of nanomaterials of various chemical compositions (peptide, lipid, polymer, inorganic). Finally, the review includes a guide on how multiple characteristics of the nanomaterial, such as substrate modification, supramolecular structure, and electrostatic charge should be collectively considered for the targeted MMP to result in optimal kinetics of enzyme action on the nanomaterial, which allow access to amplification and additional levels of spatial, temporal, and mechanical control of the response. Although this review focuses on the design strategies of MMP-responsive nanomaterials in cancer applications, these guidelines are expected to be generalizable to systems that target MMP for treatment or detection of cancer and other diseases, as well as other enzyme-responsive nanomaterials.
Collapse
Affiliation(s)
- Jiye Son
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA.
| | - Sadiyah Parveen
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA.
- Department of Biomedical Engineering, The City College of New York, CUNY, 160 Convent Avenue, New York, NY 10031, USA
| | - Douglas MacPherson
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
| | - Yaron Marciano
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA.
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
| | - Richard H Huang
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA.
| | - Rein V Ulijn
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Chemistry, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| |
Collapse
|
11
|
Abstract
Matrix metalloproteinases (MMPs) are a class of endopeptidases that are dependent on zinc and facilitate the degradation of extracellular matrix (ECM) proteins, thereby playing pivotal parts in human physiology and pathology. MMPs regulate normal tissue and cellular functions, including tissue development, remodeling, angiogenesis, bone formation, and wound healing. Several diseases, including cancer, inflammation, cardiovascular diseases, and nervous system disorders, have been linked to dysregulated expression of specific MMP subtypes, which can promote tumor progression, metastasis, and inflammation. Various MMP-responsive drug delivery and release systems have been developed by harnessing cleavage activities and overexpression of MMPs in affected regions. Herein, we review the structure, substrates, and physiological and pathological functions of various MMPs and highlight the strategies for designing MMP-responsive nanoparticles to improve the targeting efficiency, penetration, and protection of therapeutic payloads.
Collapse
Affiliation(s)
- Chenyun Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
12
|
Doron G, Pearson JJ, Guldberg RE, Temenoff JS. Development and characterization of Factor Xa-responsive materials for applications in cell culture and biologics delivery. J Biomed Mater Res A 2023; 111:634-643. [PMID: 36794576 DOI: 10.1002/jbm.a.37513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Stimuli-responsive biomaterials may be used to better control the release of bioactive molecules or cells for applications involving drug delivery and controlled cell release. In this study, we developed a Factor Xa (FXa)-responsive biomaterial capable of controlled release of pharmaceutical agents and cells from in vitro culture. FXa-cleavable substrates were formed as hydrogels that degraded in response to FXa enzyme over several hours. Hydrogels were shown to release both heparin and a model protein in response to FXa. Additionally, RGD-functionalized FXa-degradable hydrogels were used to culture mesenchymal stromal cells (MSCs), enabling FXa-mediated cell dissociation from hydrogels in a manner that preserved multicellular structures. Harvesting MSCs using FXa-mediated dissociation did not influence their differentiation capacity or indoleamine 2,3-dioxygenase (IDO) activity (a measure of immunomodulatory capacity). In all, this FXa-degradable hydrogel is a novel responsive biomaterial system that may be used for on-demand drug delivery, as well as for improving processes for in vitro culture of therapeutic cells.
Collapse
Affiliation(s)
- Gilad Doron
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Joseph J Pearson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Robert E Guldberg
- Knight Campus for Accelerating Scientific Impact, 6231 University of Oregon, Eugene, Oregon, USA
| | - Johnna S Temenoff
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Chountoulesi M, Selianitis D, Pispas S, Pippa N. Recent Advances on PEO-PCL Block and Graft Copolymers as Nanocarriers for Drug Delivery Applications. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2298. [PMID: 36984177 PMCID: PMC10056975 DOI: 10.3390/ma16062298] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
Poly(ethylene oxide)-poly(ε-caprolactone) (PEO-PCL) is a family of block (or graft) copolymers with several biomedical applications. These types of copolymers are well-known for their good biocompatibility and biodegradability properties, being ideal for biomedical applications and for the formation of a variety of nanosystems intended for controlled drug release. The aim of this review is to present the applications and the properties of different nanocarriers derived from PEO-PCL block and graft copolymers. Micelles, polymeric nanoparticles, drug conjugates, nanocapsules, and hybrid polymer-lipid nanoparticles, such as hybrid liposomes, are the main categories of PEO-PCL based nanocarriers loaded with different active ingredients. The advantages and the limitations in preclinical studies are also discussed in depth. PEO-PCL based nanocarriers could be the next generation of delivery systems with fast clinical translation. Finally, current challenges and future perspectives of the PEO-PCL based nanocarriers are highlighted.
Collapse
Affiliation(s)
- Maria Chountoulesi
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Dimitrios Selianitis
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Natassa Pippa
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| |
Collapse
|
14
|
Sachi Das S, Singh SK, Verma PRP, Gahtori R, Sibuh BZ, Kesari KK, Jha NK, Dhanasekaran S, Thakur VK, Wong LS, Djearamane S, Gupta PK. Polyester nanomedicines targeting inflammatory signaling pathways for cancer therapy. Biomed Pharmacother 2022; 154:113654. [PMID: 36067568 DOI: 10.1016/j.biopha.2022.113654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 12/09/2022] Open
Abstract
The growth of cancerous cells and their responses towards substantial therapeutics are primarily controlled by inflammations (acute and chronic) and inflammation-associated products, which either endorse or repress tumor progression. Additionally, major signaling pathways, including NF-κB, STAT3, inflammation-causing factors (cytokines, TNF-α, chemokines), and growth-regulating factors (VEGF, TGF-β), are vital regulators responsible for the instigation and resolution of inflammations. Moreover, the conventional chemotherapeutics have exhibited diverse limitations, including poor pharmacokinetics, unfavorable chemical properties, poor targetability to the disease-specific disease leading to toxicity; thus, their applications are restricted in inflammation-mediated cancer therapy. Furthermore, nanotechnology has demonstrated potential benefits over conventional chemotherapeutics, such as it protected the incorporated drug/bioactive moiety from enzymatic degradation within the systemic circulation, improving the physicochemical properties of poorly aqueous soluble chemotherapeutic agents, and enhancing their targetability in specified carcinogenic cells rather than accumulating in the healthy cells, leading reduced cytotoxicity. Among diverse nanomaterials, polyester-based nanoparticulate delivery systems have been extensively used to target various inflammation-mediated cancers. This review summarizes the therapeutic potentials of various polyester nanomaterials (PLGA, PCL, PLA, PHA, and others)-based delivery systems targeting multiple signaling pathways related to inflammation-mediated cancer.
Collapse
Affiliation(s)
- Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology - Mesra, Ranchi 835215, Jharkhand, India; School of Pharmaceutical and Population Health Informatics, DIT University, Dehradun 248009, Uttarakhand, India
| | - Sandeep Kumar Singh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology - Mesra, Ranchi 835215, Jharkhand, India.
| | - P R P Verma
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology - Mesra, Ranchi 835215, Jharkhand, India
| | - Rekha Gahtori
- Department of Biotechnology, Sir J. C. Bose Technical Campus, Kumaun University, Bhimtal, Nainital 263136, Uttarakhand, India
| | - Belay Zeleke Sibuh
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India
| | - Kavindra Kumar Kesari
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland; Department of Applied Physics, Aalto University, Espoo, Finland
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, Uttarakhand, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India
| | - Sugapriya Dhanasekaran
- Medical Laboratory Sciences Department, College of Applied Medical Sciences, University of Bisha, Bisha 67714, Saudi Arabia
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Centre, SRUC, Edinburgh EH9 3JG, United Kingdom; School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, Uttarakhand, India; Department of Biotechnology, Graphic Era Deemed to be University, Dehradun 248002, Uttarakhand, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia.
| | - Sinouvassane Djearamane
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia.
| | - Piyush Kumar Gupta
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, India; Department of Biotechnology, Graphic Era Deemed to be University, Dehradun 248002, Uttarakhand, India.
| |
Collapse
|
15
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
16
|
Najmi A, Wang S, Huang Y, Seefeldt T, Alqahtani Y, Guan X. 2-(2-Cholesteroxyethoxyl)ethyl-3′-S-glutathionylpropionate (COXP) for brain-targeting liposomes. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
17
|
Xu Y, Xiong Q, Yang Y, Weng N, Li J, Liu J, Yang X, Zeng Z, Zhang Z, Zhu Q. Serum Nardilysin as a Prognostic Biomarker in Pancreatic Ductal Adenocarcinoma. J Clin Med 2022; 11:jcm11113101. [PMID: 35683488 PMCID: PMC9181681 DOI: 10.3390/jcm11113101] [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: 04/18/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Nardilysin, (N-arginine dibasic convertase, NRDC) has been reported to play an important role in cancer progression, and is associated with tumor proliferation signals and inflammatory signals, such as tumor necrosis factor-a (TNF-a) and heparin-binding epidermal growth factor-like growth factor (HB-EGF), through the activation of disintegrin and metalloproteinase (ADAM) proteases. NRDC has recently been revealed to be involved in the tumorigenesis of various types of cancer, including intrahepatic cholangiocarcinoma, malignant cerebral infarction, esophageal squamous cell carcinoma, and gastric cancer. However, the expression profiles and biological relevance of NRDC in pancreatic ductal adenocarcinoma have rarely been reported. Methods: We analyzed the NRDC expression profile in pancreatic ductal adenocarcinoma by enzyme-linked immunosorbent assay (ELISA) and identified NRDC as a circulating biomarker in the serum of 112 pancreatic ductal adenocarcinoma patients. The diagnostic value of NRDC was analyzed by the area under the curve (AUC) and the receiver operating characteristic (ROC) test. Results: Our results demonstrated that the clinical prognosis significance of NRDC with the clinical characteristics in pancreatic ductal adenocarcinoma (PDAC). NRDC was notably decreased in PDAC patient serum compared with the control group (p < 0.001). Furthermore, the present study found that the NRDC expression level was correlated with T grade (p < 0.001), metastasis(p < 0.001), differentiation(p < 0.001), and TNM stage (p = 0.011). Further bioinformatics analysis revealed that NRDC correlated with proliferation and migration pathways; in particular, it mediated cell-matrix adhesion-dependent activation in pancreatic ductal adenocarcinoma. Conclusions: Serum NRDC may play a useful diagnostic biomarker to evaluate the aggressive clinical features in PAAD patients.
Collapse
|
18
|
Singh R, Sharma A, Saji J, Umapathi A, Kumar S, Daima HK. Smart nanomaterials for cancer diagnosis and treatment. NANO CONVERGENCE 2022; 9:21. [PMID: 35569081 PMCID: PMC9108129 DOI: 10.1186/s40580-022-00313-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/26/2022] [Indexed: 05/14/2023]
Abstract
Innovations in nanomedicine has guided the improved outcomes for cancer diagnosis and therapy. However, frequent use of nanomaterials remains challenging due to specific limitations like non-targeted distribution causing low signal-to-noise ratio for diagnostics, complex fabrication, reduced-biocompatibility, decreased photostability, and systemic toxicity of nanomaterials within the body. Thus, better nanomaterial-systems with controlled physicochemical and biological properties, form the need of the hour. In this context, smart nanomaterials serve as promising solution, as they can be activated under specific exogenous or endogenous stimuli such as pH, temperature, enzymes, or a particular biological molecule. The properties of smart nanomaterials make them ideal candidates for various applications like biosensors, controlled drug release, and treatment of various diseases. Recently, smart nanomaterial-based cancer theranostic approaches have been developed, and they are displaying better selectivity and sensitivity with reduced side-effects in comparison to conventional methods. In cancer therapy, the smart nanomaterials-system only activates in response to tumor microenvironment (TME) and remains in deactivated state in normal cells, which further reduces the side-effects and systemic toxicities. Thus, the present review aims to describe the stimulus-based classification of smart nanomaterials, tumor microenvironment-responsive behaviour, and their up-to-date applications in cancer theranostics. Besides, present review addresses the development of various smart nanomaterials and their advantages for diagnosing and treating cancer. Here, we also discuss about the drug targeting and sustained drug release from nanocarriers, and different types of nanomaterials which have been engineered for this intent. Additionally, the present challenges and prospects of nanomaterials in effective cancer diagnosis and therapeutics have been discussed.
Collapse
Affiliation(s)
- Ragini Singh
- College of Agronomy, Liaocheng University, Liaocheng, 252059, Shandong, China.
| | - Ayush Sharma
- Amity Center for Nanobiotechnology and Nanomedicine (ACNN), Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, Rajasthan, India
| | - Joel Saji
- Amity Center for Nanobiotechnology and Nanomedicine (ACNN), Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, Rajasthan, India
| | - Akhela Umapathi
- Amity Center for Nanobiotechnology and Nanomedicine (ACNN), Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, Rajasthan, India
| | - Santosh Kumar
- Shandong Key Laboratory of Optical Communication Science and Technology, School of Physics Science and Information Technology, Liaocheng University, Liaocheng, 252059, Shandong, China
| | - Hemant Kumar Daima
- Amity Center for Nanobiotechnology and Nanomedicine (ACNN), Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303002, Rajasthan, India.
| |
Collapse
|
19
|
Tune BXJ, Sim MS, Poh CL, Guad RM, Woon CK, Hazarika I, Das A, Gopinath SCB, Rajan M, Sekar M, Subramaniyan V, Fuloria NK, Fuloria S, Batumalaie K, Wu YS. Matrix Metalloproteinases in Chemoresistance: Regulatory Roles, Molecular Interactions, and Potential Inhibitors. JOURNAL OF ONCOLOGY 2022; 2022:3249766. [PMID: 35586209 PMCID: PMC9110224 DOI: 10.1155/2022/3249766] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
Cancer is one of the major causes of death worldwide. Its treatments usually fail when the tumor has become malignant and metastasized. Metastasis is a key source of cancer recurrence, which often leads to resistance towards chemotherapeutic agents. Hence, most cancer-related deaths are linked to the occurrence of chemoresistance. Although chemoresistance can emerge through a multitude of mechanisms, chemoresistance and metastasis share a similar pathway, which is an epithelial-to-mesenchymal transition (EMT). Matrix metalloproteinases (MMPs), a class of zinc and calcium-chelated enzymes, are found to be key players in driving cancer migration and metastasis through EMT induction. The aim of this review is to discuss the regulatory roles and associated molecular mechanisms of specific MMPs in regulating chemoresistance, particularly EMT initiation and resistance to apoptosis. A brief presentation on their potential diagnostic and prognostic values was also deciphered. It also aimed to describe existing MMP inhibitors and the potential of utilizing other strategies to inhibit MMPs to reduce chemoresistance, such as upstream inhibition of MMP expressions and MMP-responsive nanomaterials to deliver drugs as well as epigenetic regulations. Hence, manipulation of MMP expression can be a powerful tool to aid in treating patients with chemo-resistant cancers. However, much still needs to be done to bring the solution from bench to bedside.
Collapse
Affiliation(s)
- Bernadette Xin Jie Tune
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Maw Shin Sim
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
| | - Rhanye Mac Guad
- Department of Biomedical Science and Therapeutics, Faculty of Medicine and Health Science, Universiti Malaysia Sabah, Kota Kinabalu, 88400 Sabah, Malaysia
| | - Choy Ker Woon
- Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, 47000 Selangor, Malaysia
| | - Iswar Hazarika
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Guwahati 781017, India
| | - Anju Das
- Department of Pharmacology, Royal School of Pharmacy, Royal Global University, Guwahati 781035, India
| | - Subash C. B. Gopinath
- Faculty of Chemical Engineering Technology, Universiti Malaysia Perlis (UniMAP), Arau, 02600 Perlis, Malaysia
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis, Kangar, 01000 Perlis, Malaysia
| | - Mariappan Rajan
- Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai 625021, India
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Perak, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Pharmacology, School of Medicine, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Selangor 42610, Malaysia
| | | | - Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Semeling, Bedong, Kedah 08100, Malaysia
| | - Kalaivani Batumalaie
- Department of Biomedical Sciences, Faculty of Health Sciences, Asia Metropolitan University, 81750 Johor Bahru, Malaysia
| | - Yuan Seng Wu
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
| |
Collapse
|
20
|
Kaur J, Gulati M, Kapoor B, Jha NK, Gupta PK, Gupta G, Chellappan DK, Devkota HP, Prasher P, Ansari MS, Aba Alkhayl FF, Arshad MF, Morris A, Choonara YE, Adams J, Dua K, Singh SK. Advances in designing of polymeric micelles for biomedical application in brain related diseases. Chem Biol Interact 2022; 361:109960. [PMID: 35533733 DOI: 10.1016/j.cbi.2022.109960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/11/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022]
|
21
|
Yin W, Tian L, Wang S, Zhang D, Guo S, Lang M. Co-delivery systems of paclitaxel prodrug for targeted synergistic therapy of breast cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
22
|
MacPherson DS, McPhee SA, Zeglis BM, Ulijn RV. The Impact of Tyrosine Iodination on the Aggregation and Cleavage Kinetics of MMP-9-Responsive Peptide Sequences. ACS Biomater Sci Eng 2022; 8:579-587. [PMID: 35050574 DOI: 10.1021/acsbiomaterials.1c01488] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Matrix metalloproteinase (MMP) enzymes are over-expressed by some metastatic cancers, in which they are responsible for the degradation and remodeling of the extracellular matrix. In recent years, MMPs have emerged as promising targets for enzyme-responsive diagnostic probes because oligopeptides can be designed to be selectively hydrolyzed by exposure to these enzymes. With the ultimate goal of developing radio-iodinated peptides as supramolecular building blocks for MMP-sensitive tools for nuclear imaging and therapy, we designed three MMP-9-responsive peptides containing either tyrosine or iodotyrosine to assess the impact of iodotyrosine introduction to the peptide structure and cleavage kinetics. We found that the peptides containing iodotyrosine underwent more rapid and more complete hydrolysis by MMP-9. While the peptides under investigation were predominantly disordered, it was found that iodination increased the degree of aromatic residue-driven aggregation of the peptides. We determined that these iodination-related trends stem from the improved overall intramolecular order through H- and halogen bonding, in addition to intermolecular organization of the self-assembled peptides due to steric and electrostatic effects introduced by the halogenated tyrosine. These fundamental observations provide insights for the development of enzyme-triggered peptide aggregation tools for localized radioactive iodine-based tumor imaging.
Collapse
Affiliation(s)
- Douglas S MacPherson
- Department of Chemistry, Hunter College of the City University of New York, New York, New York 10028, United States.,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 St. Nicholas Terrace, New York, New York 10031, United States
| | - Scott A McPhee
- Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 St. Nicholas Terrace, New York, New York 10031, United States
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York, New York, New York 10028, United States.,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Rein V Ulijn
- Department of Chemistry, Hunter College of the City University of New York, New York, New York 10028, United States.,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States.,Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 St. Nicholas Terrace, New York, New York 10031, United States
| |
Collapse
|
23
|
Rabha B, Bharadwaj KK, Pati S, Choudhury BK, Sarkar T, Kari ZA, Edinur HA, Baishya D, Atanase LI. Development of Polymer-Based Nanoformulations for Glioblastoma Brain Cancer Therapy and Diagnosis: An Update. Polymers (Basel) 2021; 13:polym13234114. [PMID: 34883617 PMCID: PMC8659151 DOI: 10.3390/polym13234114] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Brain cancers, mainly high-grade gliomas/glioblastoma, are characterized by uncontrolled proliferation and recurrence with an extremely poor prognosis. Despite various conventional treatment strategies, viz., resection, chemotherapy, and radiotherapy, the outcomes are still inefficient against glioblastoma. The blood–brain barrier is one of the major issues that affect the effective delivery of drugs to the brain for glioblastoma therapy. Various studies have been undergone in order to find novel therapeutic strategies for effective glioblastoma treatment. The advent of nanodiagnostics, i.e., imaging combined with therapies termed as nanotheranostics, can improve the therapeutic efficacy by determining the extent of tumour distribution prior to surgery as well as the response to a treatment regimen after surgery. Polymer nanoparticles gain tremendous attention due to their versatile nature for modification that allows precise targeting, diagnosis, and drug delivery to the brain with minimal adverse side effects. This review addresses the advancements of polymer nanoparticles in drug delivery, diagnosis, and therapy against brain cancer. The mechanisms of drug delivery to the brain of these systems and their future directions are also briefly discussed.
Collapse
Affiliation(s)
- Bijuli Rabha
- Department of Bioengineering & Technology, GUIST, Gauhati University, Guwahati 781014, India; (B.R.); (K.K.B.)
| | - Kaushik Kumar Bharadwaj
- Department of Bioengineering & Technology, GUIST, Gauhati University, Guwahati 781014, India; (B.R.); (K.K.B.)
| | - Siddhartha Pati
- Skills Innovation & Academic Network (SIAN) Institute-Association for Biodiversity Conservation and Research (ABC), Balasore 756001, India;
- NatNov Bioscience Private Limited, Balasore, 756001, India
| | | | - Tanmay Sarkar
- Malda Polytechnic, West Bengal State Council of Technical Education, Govt. of West Bengal, Malda 732102, India;
- Department of Food Technology and Biochemical Engineering, Jadavpur University, Kolkata 700032, India
| | - Zulhisyam Abdul Kari
- Faculty of Agro Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Malaysia;
| | - Hisham Atan Edinur
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia;
| | - Debabrat Baishya
- Department of Bioengineering & Technology, GUIST, Gauhati University, Guwahati 781014, India; (B.R.); (K.K.B.)
- Correspondence: (D.B.); (L.I.A.)
| | - Leonard Ionut Atanase
- Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania
- Correspondence: (D.B.); (L.I.A.)
| |
Collapse
|
24
|
Ravi Kiran AVVV, Kusuma Kumari G, Krishnamurthy PT, Khaydarov RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021; 9:7667-7704. [PMID: 34673853 DOI: 10.1039/d1bm01127h] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over recent years, advancements in nanomedicine have allowed new approaches to diagnose and treat tumors. Nano drug delivery systems exploit the enhanced permeability and retention (EPR) effect and enter the tumor tissue's interstitial space. However, tumor barriers play a crucial role, and cause inefficient EPR or the homing effect. Mounting evidence supports the hypothesis that the components of the tumor microenvironment, such as the extracellular matrix, and cellular and physiological components collectively or cooperatively hinder entry and distribution of drugs, and therefore, limit the theragnostic applications of cancer nanomedicine. This abnormal tumor microenvironment plays a pivotal role in cancer nanomedicine and was recently recognized as a promising target for improving nano-drug delivery and their therapeutic outcomes. Strategies like passive or active targeting, stimuli-triggered nanocarriers, and the modulation of immune components have shown promising results in achieving anticancer efficacy. The present review focuses on the tumor microenvironment and nanoparticle-based strategies (polymeric, inorganic and organic nanoparticles) for intruding the tumor barrier and improving therapeutic effects.
Collapse
Affiliation(s)
- Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Renat R Khaydarov
- Institute of Nuclear Physics, Uzbekistan Academy of Sciences, Tashkent, 100047, Uzbekistan.
| |
Collapse
|
25
|
Stimuli-Responsive Polymeric Nanosystems for Controlled Drug Delivery. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11209541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Biocompatible nanosystems based on polymeric materials are promising drug delivery nanocarrier candidates for antitumor therapy. However, the efficacy is unsatisfying due to nonspecific accumulation and drug release of the nanoparticles in normal tissue. Recently, the nanosystems that can be triggered by tumor-specific stimuli have drawn great interest for drug delivery applications due to their controllable drug release properties. In this review, various polymers and external stimuli that can be employed to develop stimuli-responsive polymeric nanosystems are discussed, and finally, we delineate the challenges in designing this kind of Nanomedicine to improve the therapeutic efficacy.
Collapse
|
26
|
Tao J, Wei Z, Xu M, Xi L, Cheng Y, Lee SMY, Ge W, Zheng Y. Particle Integrity and Size Effect on the Journey of Polymeric Nanocarriers in Zebrafish Model and the Correlation with Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103584. [PMID: 34528394 DOI: 10.1002/smll.202103584] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/15/2021] [Indexed: 05/25/2023]
Abstract
Polymeric nanocarriers have high biocompatibility for potential drug delivery applications. After entering bloodstream, nanocarriers will circulate, interact with proteins, dissociate, or be cleared by reticuloendothelial system. Zebrafish as a visual animal model, can serve as a tool for screening nanomedicines and monitoring nanocarrier behaviors in vivo. However, a comprehensive correlation between zebrafish and rodent models is currently deficient. Here, different-sized poly(caprolactone) nanocarriers (PCL NCs) are fabricated with or without PEGylation to investigate correlation between zebrafish and mice regarding their biofate via Förster resonance energy transfer technique. Results show that PEGylated PCL NCs have higher integrity in both zebrafish and mice. Small PEG-PCL NCs have longer circulation, while large PEG-PCL NCs have dramatically higher macrophage sequestration in zebrafish and mice spleen, leading to poor circulation. PCL NCs dissociate rapidly with less macrophage sequestration. Moreover, in 7 days postfertilization (dpf) zebrafish, polymers are eliminated via hepatobiliary pathway, which is not fully functional at earlier stages of development. The effects of nanocarrier integrity on macrophage sequestration in zebrafish and good correlation with mice spleen are pioneered to be demonstrated. The findings suggest that 7 dpf zebrafish are suitable as an in vivo screening model of nanocarriers and predict their biofate in rodents.
Collapse
Affiliation(s)
- Jinsong Tao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Zhengjie Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Meng Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Yaxin Cheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Wei Ge
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| |
Collapse
|
27
|
Newman H, Shih YV, Varghese S. Resolution of inflammation in bone regeneration: From understandings to therapeutic applications. Biomaterials 2021; 277:121114. [PMID: 34488119 DOI: 10.1016/j.biomaterials.2021.121114] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/10/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022]
Abstract
Impaired bone healing occurs in 5-10% of cases following injury, leading to a significant economic and clinical impact. While an inflammatory response upon injury is necessary to facilitate healing, its resolution is critical for bone tissue repair as elevated acute or chronic inflammation is associated with impaired healing in patients and animal models. This process is governed by important crosstalk between immune cells through mediators that contribute to resolution of inflammation in the local healing environment. Approaches modulating the initial inflammatory phase followed by its resolution leads to a pro-regenerative environment for bone regeneration. In this review, we discuss the role of inflammation in bone repair, the negative impact of dysregulated inflammation on bone tissue regeneration, and how timely resolution of inflammation is necessary to achieve normal healing. We will discuss applications of biomaterials to treat large bone defects with a specific focus on resolution of inflammation to modulate the immune environment following bone injury, and their observed functional benefits. We conclude the review by discussing future strategies that could lead to the realization of anti-inflammatory therapeutics for bone tissue repair.
Collapse
Affiliation(s)
- Hunter Newman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27710, USA
| | - Yuru Vernon Shih
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shyni Varghese
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27710, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
28
|
Prencipe F, Diaferia C, Rossi F, Ronga L, Tesauro D. Forward Precision Medicine: Micelles for Active Targeting Driven by Peptides. Molecules 2021; 26:4049. [PMID: 34279392 PMCID: PMC8271712 DOI: 10.3390/molecules26134049] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Precision medicine is based on innovative administration methods of active principles. Drug delivery on tissue of interest allows improving the therapeutic index and reducing the side effects. Active targeting by means of drug-encapsulated micelles decorated with targeting bioactive moieties represents a new frontier. Between the bioactive moieties, peptides, for their versatility, easy synthesis and immunogenicity, can be selected to direct a drug toward a considerable number of molecular targets overexpressed on both cancer vasculature and cancer cells. Moreover, short peptide sequences can facilitate cellular intake. This review focuses on micelles achieved by self-assembling or mixing peptide-grafted surfactants or peptide-decorated amphiphilic copolymers. Nanovectors loaded with hydrophobic or hydrophilic cytotoxic drugs or with gene silence sequences and externally functionalized with natural or synthetic peptides are described based on their formulation and in vitro and in vivo behaviors.
Collapse
Affiliation(s)
- Filippo Prencipe
- Institute of Crystallography (IC) CNR, Via Amendola 122/o, 70126 Bari, Italy
| | - Carlo Diaferia
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", via Mezzocannone 16, 80134 Naples, Italy
| | - Filomena Rossi
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", via Mezzocannone 16, 80134 Naples, Italy
| | - Luisa Ronga
- Institut des Sciences Analytiques et de Physico-Chimie Pour l'Environnement et les Matériaux, Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
| | - Diego Tesauro
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", via Mezzocannone 16, 80134 Naples, Italy
| |
Collapse
|
29
|
Kostrikov S, Johnsen KB, Braunstein TH, Gudbergsson JM, Fliedner FP, Obara EAA, Hamerlik P, Hansen AE, Kjaer A, Hempel C, Andresen TL. Optical tissue clearing and machine learning can precisely characterize extravasation and blood vessel architecture in brain tumors. Commun Biol 2021; 4:815. [PMID: 34211069 PMCID: PMC8249617 DOI: 10.1038/s42003-021-02275-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Precise methods for quantifying drug accumulation in brain tissue are currently very limited, challenging the development of new therapeutics for brain disorders. Transcardial perfusion is instrumental for removing the intravascular fraction of an injected compound, thereby allowing for ex vivo assessment of extravasation into the brain. However, pathological remodeling of tissue microenvironment can affect the efficiency of transcardial perfusion, which has been largely overlooked. We show that, in contrast to healthy vasculature, transcardial perfusion cannot remove an injected compound from the tumor vasculature to a sufficient extent leading to considerable overestimation of compound extravasation. We demonstrate that 3D deep imaging of optically cleared tumor samples overcomes this limitation. We developed two machine learning-based semi-automated image analysis workflows, which provide detailed quantitative characterization of compound extravasation patterns as well as tumor angioarchitecture in large three-dimensional datasets from optically cleared samples. This methodology provides a precise and comprehensive analysis of extravasation in brain tumors and allows for correlation of extravasation patterns with specific features of the heterogeneous brain tumor vasculature.
Collapse
Affiliation(s)
- Serhii Kostrikov
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Kasper B Johnsen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Thomas H Braunstein
- Core Facility for Integrated Microscopy, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johann M Gudbergsson
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- Laboratory for Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Frederikke P Fliedner
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth A A Obara
- Brain Tumor Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Bispebjerg, Denmark
| | - Petra Hamerlik
- Brain Tumor Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anders E Hansen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Casper Hempel
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark.
| | - Thomas L Andresen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
30
|
Zhao W, Yu X, Peng S, Luo Y, Li J, Lu L. Construction of nanomaterials as contrast agents or probes for glioma imaging. J Nanobiotechnology 2021; 19:125. [PMID: 33941206 PMCID: PMC8091158 DOI: 10.1186/s12951-021-00866-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Malignant glioma remains incurable largely due to the aggressive and infiltrative nature, as well as the existence of blood-brain-barrier (BBB). Precise diagnosis of glioma, which aims to accurately delineate the tumor boundary for guiding surgical resection and provide reliable feedback of the therapeutic outcomes, is the critical step for successful treatment. Numerous imaging modalities have been developed for the efficient diagnosis of tumors from structural or functional aspects. However, the presence of BBB largely hampers the entrance of contrast agents (Cas) or probes into the brain, rendering the imaging performance highly compromised. The development of nanomaterials provides promising strategies for constructing nano-sized Cas or probes for accurate imaging of glioma owing to the BBB crossing ability and other unique advantages of nanomaterials, such as high loading capacity and stimuli-responsive properties. In this review, the recent progress of nanomaterials applied in single modal imaging modality and multimodal imaging for a comprehensive diagnosis is thoroughly summarized. Finally, the prospects and challenges are offered with the hope for its better development.
Collapse
Affiliation(s)
- Wei Zhao
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China
| | - Xiangrong Yu
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China
| | - Shaojun Peng
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China
| | - Yu Luo
- School of Chemical Science and Engineering, Tongji University, 1239 Siping Road, Shanghai, China.
| | - Jingchao Li
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| | - Ligong Lu
- Zhuhai Precision Medical Center, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Affiliated With Jinan University), Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
31
|
Ciobanasu C. Peptides-based therapy and diagnosis. Strategies for non-invasive therapies in cancer. J Drug Target 2021; 29:1063-1079. [PMID: 33775187 DOI: 10.1080/1061186x.2021.1906885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, remarkable progress was registered in the field of cancer research. Though, cancer still represents a major cause of death and cancer metastasis a problem seeking for urgent solutions as it is the main reason for therapeutic failure. Unfortunately, the most common chemotherapeutic agents are non-selective and can damage healthy tissues and cause side effects that affect dramatically the quality of life of the patients. Targeted therapy with molecules that act specifically at the tumour sites interacting with overexpressed cancer receptors is a very promising strategy for achieving the specific delivery of anticancer drugs, radioisotopes or imaging agents. This review aims to give an overview on different strategies for targeting cancer cell receptors localised either at the extracellular matrix or at the cell membrane. Molecules like antibodies, aptamers and peptides targeting the cell surface are presented with advantages and disadvantages, with emphasis on peptides. The most representative peptides are described, including cell penetrating peptides, homing and anticancer peptides with particular consideration on recent discoveries.
Collapse
Affiliation(s)
- Corina Ciobanasu
- Sciences Department, Institute for Interdisciplinary Research, Alexandru I. Cuza University, Iaşi, Romania
| |
Collapse
|
32
|
Najmi A, Wang S, Huang Y, Seefeldt T, Alqahtani Y, Guan X. 2-(2-Cholesteroxyethoxyl)ethyl 3'-S-glutathionylpropionate and its self-assembled micelles for brain delivery: Design, synthesis and evaluation. Int J Pharm 2021; 600:120520. [PMID: 33775725 DOI: 10.1016/j.ijpharm.2021.120520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/07/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) is a barrier that prevents almost all large and most small exogenous molecules from reaching the brain. The barrier is the major cause of treatment failure for most brain diseases. Extensive efforts have been made to facilitate drug molecules to cross the BBB. One of the approaches is to employ an endogenous ligand or ligand analogue that can enter the brain through its transporter or receptor at the BBB as a brain-targeting agent. Glutathione (GSH) transporters are richly expressed at the BBB with limited presence in other tissues except kidneys. 2-(2-Cholesteroxyethoxyl)ethyl 3'-S-glutathionylpropionate (COXP), formed by connecting GSH with cholesterol through a linker, was designed as a GSH transporter-mediated brain targeting molecule. The amphiphilic nature of COXP enables the molecule to self-assemble to form micelles with a CMC value of 3.9 μM. By using DiR as a fluorescence tracking agent and the whole-body fluorescence imaging technique, the brain distribution of DiR delivered by COXP micelles in mice was 20 folds higher when compared with free DiR. Interestingly, the brain targeting effect was further enhanced by co-administration of GSH. The low CMC value and effective brain targeting make COXP micelles a promising drug delivery system to the brain.
Collapse
Affiliation(s)
- Asim Najmi
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Shenggang Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Yue Huang
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Teresa Seefeldt
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Yahya Alqahtani
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Xiangming Guan
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States.
| |
Collapse
|
33
|
Aflori M. Smart Nanomaterials for Biomedical Applications-A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:396. [PMID: 33557177 PMCID: PMC7913901 DOI: 10.3390/nano11020396] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022]
Abstract
Recent advances in nanotechnology have forced the obtaining of new materials with multiple functionalities. Due to their reduced dimensions, nanomaterials exhibit outstanding physio-chemical functionalities: increased absorption and reactivity, higher surface area, molar extinction coefficients, tunable plasmonic properties, quantum effects, and magnetic and photo properties. However, in the biomedical field, it is still difficult to use tools made of nanomaterials for better therapeutics due to their limitations (including non-biocompatible, poor photostabilities, low targeting capacity, rapid renal clearance, side effects on other organs, insufficient cellular uptake, and small blood retention), so other types with controlled abilities must be developed, called "smart" nanomaterials. In this context, the modern scientific community developed a kind of nanomaterial which undergoes large reversible changes in its physical, chemical, or biological properties as a consequence of small environmental variations. This systematic mini-review is intended to provide an overview of the newest research on nanosized materials responding to various stimuli, including their up-to-date application in the biomedical field.
Collapse
Affiliation(s)
- Magdalena Aflori
- Petru Poni Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| |
Collapse
|
34
|
Rudzińska M, Daglioglu C, Savvateeva LV, Kaci FN, Antoine R, Zamyatnin AA. Current Status and Perspectives of Protease Inhibitors and Their Combination with Nanosized Drug Delivery Systems for Targeted Cancer Therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:9-20. [PMID: 33442233 PMCID: PMC7797289 DOI: 10.2147/dddt.s285852] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022]
Abstract
In cancer treatments, many natural and synthetic products have been examined; among them, protease inhibitors are promising candidates for anti-cancer agents. Since dysregulated proteolytic activities can contribute to tumor development and metastasis, antagonization of proteases with tailored inhibitors is an encouraging approach. Although adverse effects of early designs of these inhibitors disappeared after the introduction of next-generation agents, most of the proposed inhibitors did not pass the early stages of clinical trials due to their nonspecific toxicity and lack of pharmacological effects. Therefore, new applications that modulate proteases more specifically and serve their programmed way of administration are highly appreciated. In this context, nanosized drug delivery systems have attracted much attention because preliminary studies have demonstrated that the therapeutic capacity of inhibitors has been improved significantly with encapsulated formulation as compared to their free forms. Here, we address this issue and discuss the current application and future clinical prospects of this potential combination towards targeted protease-based cancer therapy.
Collapse
Affiliation(s)
- Magdalena Rudzińska
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Cenk Daglioglu
- Biotechnology and Bioengineering Application and Research Center, Integrated Research Centers, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Lyudmila V Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Fatma Necmiye Kaci
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Yakutiye, Erzurum 25050, Turkey
| | - Rodolphe Antoine
- CNRS, Institut Lumière Matière, Univ Lyon, Université Claude Bernard Lyon 1, Lyon F-69622, France
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia.,Department of Biotechnology, Sirius University of Science and Technology, Sochi 354340, Russia
| |
Collapse
|
35
|
Li Y, Qi M, Ding F, Lv Y, Ma J, Zhu Y. Tumour targetable and microenvironment-responsive nanoparticles simultaneously disrupt the PD-1/PD-L1 pathway and MAPK/ERK/JNK pathway for efficient treatment of colorectal cancer. J Drug Target 2020; 29:454-465. [PMID: 33233956 DOI: 10.1080/1061186x.2020.1853750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The present study was aimed to develop a novel combination therapeutic strategy of gene therapy and immunotherapy for efficiently treatment of colorectal cancer (CRC). To achieve that goal, the polyethylene glycol-modified poly (2-(N,N-dimethylamino) ethyl methacrylate) (PEG-PDMAEMA)-based nanoparticles loaded with protein tyrosine phosphatase non-receptor type 6 (PTPN6) (NP-PTPN6) was developed first followed by conjugation with anti-PD-L1 monoclonal antibodies (aPD-L1) atezolizumab (aPD-L1NP-PTPN6). Importantly, the aPD-L1 was conjugated on the surface of NP-PTPN6 by the matrix metalloproteinases (MMPs)-cleavable linkage PLGLAG. Therefore, the aPD-L1 would be completely released once the aPD-L1NP-PTPN6 was entrapped into tumour tissues as demonstrated by the release assay. Tumour targeting assay demonstrated the aPD-L1NP-PTPN6 have high affinity to CRC cells and resulted in excellent tumour targeting drug delivery efficacy. Additionally, anti-tumour effect evaluation revealed that the aPD-L1NP-PTPN6 has greater ability to inhibit the growth, invasion and migration of CRC cells and finally led to longer survival time of tumour-bearing mice than other treatments. Further mechanisms studies demonstrated that treatment of CRC cells with aPD-L1NP-PTPN6 contributed to significant suppression of the MAPK/ERK signalling pathway. Besides, it was further demonstrated that treating CRC with aPD-L1NP-PTPN6 resulted in up-regulation of NK cells and T cells percentage within the tumour tissues.
Collapse
Affiliation(s)
- Yan Li
- Department of General Surgery, The First Affiliated Hospital of Jin Zhou Medical University, Jinzhou, China
| | - Ming Qi
- Department of Ultrasound, The First Affiliated Hospital of Jin Zhou Medical University, Jinzhou, China
| | - Feng Ding
- Department of General Surgery, The First Affiliated Hospital of Jin Zhou Medical University, Jinzhou, China
| | - Yong Lv
- Department of General Surgery, The First Affiliated Hospital of Jin Zhou Medical University, Jinzhou, China
| | - Jingyu Ma
- Department of General Surgery, The First Affiliated Hospital of Jin Zhou Medical University, Jinzhou, China
| | - Yufeng Zhu
- Department of General Surgery, The First Affiliated Hospital of Jin Zhou Medical University, Jinzhou, China
| |
Collapse
|
36
|
Anderson AR, Segura T. Injectable biomaterials for treatment of glioblastoma. ADVANCED MATERIALS INTERFACES 2020; 7:2001055. [PMID: 34660174 PMCID: PMC8513688 DOI: 10.1002/admi.202001055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Indexed: 06/13/2023]
Abstract
Despite ongoing advancements in the field of medicine, glioblastoma multiforme (GBM) is presently incurable, making this advanced brain tumor the deadliest tumor type in the central nervous system. The primary treatment strategies for GBM (i.e. surgical resection, radiation therapy, chemotherapy, and newly incorporated targeted therapies) fail to overcome the challenging characteristics of highly aggressive GBM tumors and are presently given with the goal of increasing the quality of life for patients. With the aim of creating effective treatment solutions, research has shifted toward utilizing injectable biomaterial adjuncts to minimize invasiveness of treatment, provide spatiotemporal control of therapeutic delivery, and engage with cells through material-cell interfaces. This review aims to summarize the limitations of the current standard of care for GBM, discuss how these limitations can be addressed by local employment of injectable biomaterial systems, and highlight developments in the field of biomaterials for these applications.
Collapse
Affiliation(s)
- Alexa R. Anderson
- Duke University Department of Biomedical Engineering, 101 Science Drive, Durham, NC 27708, U.S.A
| | - Tatiana Segura
- Duke University Department of Biomedical Engineering, 101 Science Drive, Durham, NC 27708, U.S.A
| |
Collapse
|
37
|
Biodegradable diblock copolymeric PEG-PCL nanoparticles: Synthesis, characterization and applications as anticancer drug delivery agents. POLYMER 2020. [DOI: 10.1016/j.polymer.2020.122901] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Vicente‐Ruiz S, Serrano‐Martí A, Armiñán A, Vicent MJ. Nanomedicine for the Treatment of Advanced Prostate Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sonia Vicente‐Ruiz
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Antoni Serrano‐Martí
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Ana Armiñán
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| |
Collapse
|
39
|
Targeting Glioblastoma: Advances in Drug Delivery and Novel Therapeutic Approaches. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000124] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Wang K, Ye H, Zhang X, Wang X, Yang B, Luo C, Zhao Z, Zhao J, Lu Q, Zhang H, Kan Q, Wang Y, He Z, Sun J. An exosome-like programmable-bioactivating paclitaxel prodrug nanoplatform for enhanced breast cancer metastasis inhibition. Biomaterials 2020; 257:120224. [PMID: 32736255 DOI: 10.1016/j.biomaterials.2020.120224] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 01/16/2023]
Abstract
Metastasis is closely associated with high breast cancer mortality. Although nanotechnology-based anti-metastatic treatments have developed rapidly, the anti-metastasis efficiency is still far from satisfactory, mainly due to the poor recognition of circulating tumor cells (CTCs) in blood. Herein, we developed an exosome-like sequential-bioactivating prodrug nanoplatform (EMPCs) to overcome the obstacle. Specifically, the reactive oxygen species (ROS)-responsive thioether-linked paclitaxel-linoleic acid conjugates (PTX-S-LA) and cucurbitacin B (CuB) are co-encapsulated into polymeric micelles, and the nanoparticles are further decorated with exosome membrane (EM). The resulting EMPCs could specifically capture and neutralize CTCs during blood circulation through the high-affinity interaction between cancer cell membrane and homotypic EM. Following cellular uptake, EMPCs first release CuB, remarkably blocking tumor metastasis via downregulation of the FAK/MMP signaling pathway. Moreover, CuB obviously elevates the intracellular oxidative level to induce a sequential bioactivation of ROS-responsive PTX-S-LA. In vitro and in vivo results demonstrate that EMPCs not only exhibit amplified prodrug bioactivation, prolonged blood circulation, selective targeting of homotypic tumor cells, and enhanced tumor penetration, but also suppress tumor metastasis through CTCs clearance and FAK/MMP signaling pathway regulation. This study proposes an integrated approach for mechanism-based inhibition of tumor metastasis and manifests a promising potential of programmable-bioactivating prodrug nanoplatform for cancer metastasis inhibition.
Collapse
Affiliation(s)
- Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Hao Ye
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Xuanbo Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Xia Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Bin Yang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Zhiqiang Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jian Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Qiming Kan
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Yongjun Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
41
|
Subramaniyan B, Rajaputra P, Nguyen L, Li M, Peer CJ, Kindrick J, Figg WD, Woo S, You Y. Local and Systemic Antitumor Effects of Photo-activatable Paclitaxel Prodrug on Rat Breast Tumor Models. Photochem Photobiol 2020; 96:668-679. [PMID: 31883393 PMCID: PMC8043141 DOI: 10.1111/php.13202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
We demonstrated that a large primary and a small untreated distant breast cancer could be controlled by local treatment with our light-activatable paclitaxel (PTX) prodrug. We hypothesized that the treated tumor would be damaged by the combinational effects of photodynamic therapy (PDT) and locally released PTX and that the distant tumor would be suppressed by systemic antitumor effects. Syngeneic rat breast cancer models (single- and two-tumor models) were established on Fischer 344 rats by subcutaneous injection of MAT B III cells. The rats were injected with PTX prodrug (dose: 1 umole kg-1 , i.v.), and tumors were treated with illumination using a 690-nm laser (75 or 140 mW cm-1 for 30 min, cylindrical light diffuser, drug-light interval [DLI] 9 h). Larger tumors (~16 mm) were effectively ablated (100%) without recurrence for >90 days. All cured rats rejected rechallenged tumor for up to 12 months. In the two-tumor model, the treatment of the local large tumor (~16 mm) also cured the untreated tumor (4-6 mm) through adaptive immune activation. This is our first demonstration that local treatment with our PTX prodrug produces systemic antitumor effects. Further investigations are warranted to understand mechanisms and optimal conditions to achieve clinically translatable systemic antitumor effects.
Collapse
Affiliation(s)
- Bharathiraja Subramaniyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Pallavi Rajaputra
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Luong Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Mengjie Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD
| | - Jessica Kindrick
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| |
Collapse
|
42
|
Ferraris C, Cavalli R, Panciani PP, Battaglia L. Overcoming the Blood-Brain Barrier: Successes and Challenges in Developing Nanoparticle-Mediated Drug Delivery Systems for the Treatment of Brain Tumours. Int J Nanomedicine 2020; 15:2999-3022. [PMID: 32431498 PMCID: PMC7201023 DOI: 10.2147/ijn.s231479] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
High-grade gliomas are still characterized by a poor prognosis, despite recent advances in surgical treatment. Chemotherapy is currently practiced after surgery, but its efficacy is limited by aspecific toxicity on healthy cells, tumour cell chemoresistance, poor selectivity, and especially by the blood–brain barrier (BBB). Thus, despite the large number of potential drug candidates, the choice of effective chemotherapeutics is still limited to few compounds. Malignant gliomas are characterized by high infiltration and neovascularization, and leaky BBB (the so-called blood–brain tumour barrier); surgical resection is often incomplete, leaving residual cells that are able to migrate and proliferate. Nanocarriers can favour delivery of chemotherapeutics to brain tumours owing to different strategies, including chemical stabilization of the drug in the bloodstream; passive targeting (because of the leaky vascularization at the tumour site); inhibition of drug efflux mechanisms in endothelial and cancer cells; and active targeting by exploiting carriers and receptors overexpressed at the blood–brain tumour barrier. Within this concern, a suitable nanomedicine-based therapy for gliomas should not be limited to cytotoxic agents, but also target the most important pathogenetic mechanisms, including cell differentiation pathways and angiogenesis. Moreover, the combinatorial approach of cell therapy plus nanomedicine strategies can open new therapeutical opportunities. The major part of attempted preclinical approaches on animal models involves active targeting with protein ligands, but, despite encouraging results, a few number of nanomedicines reached clinical trials, and most of them include drug-loaded nanocarriers free of targeting ligands, also because of safety and scalability concerns.
Collapse
Affiliation(s)
- Chiara Ferraris
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Pier Paolo Panciani
- Clinic of Neurosurgery, Spedali Civili and University of Brescia, Brescia, Italy
| | - Luigi Battaglia
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| |
Collapse
|
43
|
Fan L, Wang J, Xia C, Zhang Q, Pu Y, Chen L, Chen J, Wang Y. Glutathione-sensitive and folate-targeted nanoparticles loaded with paclitaxel to enhance oral squamous cell carcinoma therapy. J Mater Chem B 2020; 8:3113-3122. [PMID: 32207763 DOI: 10.1039/c9tb02818h] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In this study, a drug delivery system based on glutathione (GSH)-sensitive and folic acid (FA)-targeted nanoparticles loaded with paclitaxel (FA-PEG-S-S-PCL@PTX, FA-NPs) was developed. First, we proved that the FA receptor was significantly expressed in 95 oral squamous cell carcinoma (OSCC) specimens (57.9%). This provided feasibility to release FA-targeted nanoparticles in tumour sites for patients with OSCC. Next, FA-NPs were synthesized and characterized. In vitro, we found enhancement in FA-mediated endocytosis in the HSC3 cells with FA overexpression. Therefore, paclitaxel (PTX) from FA-NPs could be precisely released due to the disulfide bonds that were cleaved by a redox reaction. In vivo, FA-NPs could be accumulated in mice bearing HSC3 cells, where they exhibited effective antitumor effects when compared to the treatments with free PTX and PEG-S-S-PCL@PTX. In summary, this novel drug system has an opportunity to improve OSCC treatment.
Collapse
Affiliation(s)
- Lei Fan
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatology Hospital, Medical School of Nanjing University, Nanjing, China.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020; 10:4557-4588. [PMID: 32292515 PMCID: PMC7150471 DOI: 10.7150/thno.38069] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/24/2020] [Indexed: 02/05/2023] Open
Abstract
In recent years, much progress has been motivated in stimuli-responsive nanocarriers, which could response to the intrinsic physicochemical and pathological factors in diseased regions to increase the specificity of drug delivery. Currently, numerous nanocarriers have been engineered with physicochemical changes in responding to external stimuli, such as ultrasound, thermal, light and magnetic field, as well as internal stimuli, including pH, redox potential, hypoxia and enzyme, etc. Nanocarriers could respond to stimuli in tumor microenvironments or inside cancer cells for on-demanded drug delivery and accumulation, controlled drug release, activation of bioactive compounds, probes and targeting ligands, as well as size, charge and conformation conversion, etc., leading to sensing and signaling, overcoming multidrug resistance, accurate diagnosis and precision therapy. This review has summarized the general strategies of developing stimuli-responsive nanocarriers and recent advances, presented their applications in drug delivery, tumor imaging, therapy and theranostics, illustrated the progress of clinical translation and made prospects.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, 610041, China
| |
Collapse
|
45
|
Darrigues E, Nima ZA, Griffin RJ, Anderson JM, Biris AS, Rodriguez A. 3D cultures for modeling nanomaterial-based photothermal therapy. NANOSCALE HORIZONS 2020; 5:400-430. [PMID: 32118219 DOI: 10.1039/c9nh00628a] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Photothermal therapy (PTT) is one of the most promising techniques for cancer tumor ablation. Nanoparticles are increasingly being investigated for use with PTT and can serve as theranostic agents. Based on the ability of near-infrared nano-photo-absorbers to generate heat under laser irradiation, PTT could prove advantageous in certain situations over more classical cancer therapies. To analyze the efficacy of nanoparticle-based PTT, preclinical in vitro studies typically use 2D cultures, but this method cannot completely mimic the complex tumor organization, bioactivity, and physiology that all control the complex penetration depth, biodistribution, and tissue diffusion parameters of nanomaterials in vivo. To fill this knowledge gap, 3D culture systems have been explored for PTT analysis. These models provide more realistic microenvironments that allow spatiotemporal oxygen gradients and cancer cell adaptations to be considered. This review highlights the work that has been done to advance 3D models for cancer microenvironment modeling, specifically in the context of advanced, functionalized nanoparticle-directed PTT.
Collapse
Affiliation(s)
- Emilie Darrigues
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S University Avenue, Little Rock, AR 72204, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Xia B, Yan X, Fang WW, Chen S, Jiang Z, Wang J, Sun TC, Li Q, Li Z, Lu Y, He T, Cao B, Yang CT. Activatable Cell-Penetrating Peptide Conjugated Polymeric Nanoparticles with Gd-Chelation and Aggregation-Induced Emission for Bimodal MR and Fluorescence Imaging of Tumors. ACS APPLIED BIO MATERIALS 2020; 3:1394-1405. [DOI: 10.1021/acsabm.9b01049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Bin Xia
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - Xu Yan
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - Wei-Wei Fang
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - Sheng Chen
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - ZhiLin Jiang
- Centre for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, People’s Republic of China
| | - JinChen Wang
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - Tian-Ci Sun
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - Qing Li
- The Central Laboratory of Medical Research Centre, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230009, People’s Republic of China
| | - Zhen Li
- Centre for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, People’s Republic of China
| | - Yang Lu
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - Tao He
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People’s Republic of China
| | - BaoQiang Cao
- Department of General Surgery, Anhui No. 2 Provincial People’s Hospital, Hefei, Anhui 230041, People’s Republic of China
| | - Chang-Tong Yang
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608
- Duke-NUS Medical School, 8 College Road, Singapore 169857
| |
Collapse
|
47
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
48
|
Sang M, Luo R, Bai Y, Dou J, Zhang Z, Liu F, Feng F, Liu W. BHQ-Cyanine-Based "Off-On" Long-Circulating Assembly as a Ferroptosis Amplifier for Cancer Treatment: A Lipid-Peroxidation Burst Device. ACS APPLIED MATERIALS & INTERFACES 2019; 11:42873-42884. [PMID: 31553151 DOI: 10.1021/acsami.9b12469] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ferroptosis is an iron-dependent cell death caused by accumulation of lipid peroxidation (LPO), which is a new strategy for cancer treatment. Th current ferroptosis therapy nanodevices have low efficiency and side effects generally. Hence, we developed a Black Hole Quencher (BHQ)-based fluorescence "off-on" nanophotosensitizer complex assembly (CSO-BHQ-IR780-Hex/MIONPs/Sor). CSO-connected BHQ-IR780-Hex and -loaded magnetic iron oxide nanoparticles (MIONPs) and sorafenib (Sor) formed a very concise functionalized delivery system. CSO-BHQ-IR780-Hex disassembled by GSH attack and released IR780-Hex, MIONPs, and sorafenib. IR780-Hex anchored to the mitochondrial membrane, which would contribute to amplifying the efficiency of the photosensitizer. When NIR irradiation was given to CSO-BHQ-IR780-Hex/MIONPs/Sor-treated cells, iron supply increased, the xCT/GSH/GPX-4 system was triggered, and a lot of LPO burst. A malondialdehyde test showed that LPO in complex assembly-treated cells was explosive and increased about 18-fold compared to the control. The accumulation process of particles was monitored by an IR780-Hex photosensitizer, which showed an excellent tumor target ability by magnetic of nanodevice in vivo. Interestingly, the half-life of sorafenib in a nanodevice was increased about 26-fold compared to the control group. Importantly, the complex assembly effectively inhibits tumor growth in the breast tumor mouse model. This work would provide ideas in designing nanomedicines for the ferroptosis treatment of cancer.
Collapse
Affiliation(s)
- Mangmang Sang
- Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing 210009 , China
| | - Renjie Luo
- Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing 210009 , China
| | - Yidan Bai
- Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing 210009 , China
| | - Jun Dou
- Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing 210009 , China
| | - Zhongtao Zhang
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , China
| | | | - Feng Feng
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , China
- Jiangsu Food & Pharmaceutical Science College , 4 Meicheng Donglu , Huaian 223003 , China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing 210009 , China
- Hangzhou Institute of Pharmaceutical Innovation , China Pharmaceutical University , 291 Fucheng Lu , Hangzhou 310018 , China
| |
Collapse
|
49
|
Xiang Y, Duan X, Feng L, Jiang S, Deng L, Shen J, Yang Y, Guo R. tLyp-1-conjugated GSH-sensitive biodegradable micelles mediate enhanced pUNO1-hTRAILa/curcumin co-delivery to gliomas. CHEMICAL ENGINEERING JOURNAL 2019; 374:392-404. [DOI: 10.1016/j.cej.2019.05.186] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
|
50
|
Sang M, Luo R, Bai Y, Dou J, Zhang Z, Liu F, Feng F, Xu J, Liu W. Mitochondrial membrane anchored photosensitive nano-device for lipid hydroperoxides burst and inducing ferroptosis to surmount therapy-resistant cancer. Am J Cancer Res 2019; 9:6209-6223. [PMID: 31534546 PMCID: PMC6735518 DOI: 10.7150/thno.36283] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/23/2019] [Indexed: 01/31/2023] Open
Abstract
Rationale: Ferroptosis is a regulated process of cell death caused by iron-dependent accumulation of lipid hydroperoxides (LPO). It is sensitive to epithelial-to-mesenchymal transition (EMT) cells, a well-known therapy-resistant state of cancer. Previous studies on nanomaterials did not investigate the immense value of ferroptosis therapy (FT) in epithelial cell carcinoma during EMT. Herein, we describe an EMT-specific nanodevice for a comprehensive FT strategy involving LPO burst. Methods: Mitochondrial membrane anchored oxidation/reduction response and Fenton-Reaction-Accelerable magnetic nanophotosensitizer complex self-assemblies loading sorafenib (CSO-SS-Cy7-Hex/SPION/Srfn) were constructed in this study for LPO produced to overcome the therapy-resistant state of cancer. Both in vitro and in vivo experiments were performed using breast cancer cells to investigate the anti-tumor efficacy of the complex self-assemblies. Results: The nano-device enriched the tumor sites by magnetic targeting of enhanced permeability and retention effects (EPR), which were disassembled by the redox response under high levels of ROS and GSH in FT cells. Superparamagnetic iron oxide nanoparticles (SPION) released Fe2+ and Fe3+ in the acidic environment of lysosomes, and the NIR photosensitizer Cy7-Hex anchored to the mitochondrial membrane, combined sorafenib (Srfn) leading to LPO burst, which was accumulated ~18-fold of treatment group in breast cancer cells. In vivo pharmacodynamic test results showed that this nanodevice with small particle size and high cytotoxicity increased Srfn circulation and shortened the period of epithelial cancer treatment. Conclusion: Ferroptosis therapy had a successful effect on EMT cells. These findings have great potential in the treatment of therapy-resistant epithelial cell carcinomas.
Collapse
|