1
|
Wu Y, Lloveras V, Lope-Piedrafita S, Mulero-Acevedo M, Candiota AP, Vidal-Gancedo J. Synthesis and Relaxivity study of amino acid-branched radical dendrimers as MRI contrast agents for potential brain tumor imaging. Acta Biomater 2025; 192:461-472. [PMID: 39647652 DOI: 10.1016/j.actbio.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
This study introduces a series of water-soluble radical dendrimers (G0 to G5) as promising magnetic resonance imaging (MRI) contrast agents that could potentially address clinical safety concerns associated with current gadolinium-based contrast agents. By using a simplified synthetic approach based on a cyclotriphosphazene core and lysine-derived branching units, we successfully developed a G5 dendrimer containing up to 192 units of 2,2,6,6-Tetramethylpiperidinyloxy (TEMPO) radical. This synthesis offers advantages including ease of preparation, purification, and tunable water solubility through the incorporation of glutamic acid anion residues. Comprehensive characterization using 1H NMR, FT-IR, and SEC-HPLC confirmed the dendrimers' structures and purity. Electron paramagnetic resonance (EPR) spectroscopy revealed that TEMPO groups in higher generation dendrimers exhibited decreased mobility and stronger spin exchange in their local environments. In vitro MRI showed that relaxivity (r1) increased with higher dendrimer generations, with G5 exhibiting an exceptionally high r1 of over 24 mM-1s-1. Molecular dynamics simulations provided crucial insights into structure-property relationships, revealing the importance of water accessibility to TEMPO groups for enhancing relaxivity. Vero cell viability assay demonstrated G3 and G3.5 have good biocompatibility. In vivo MRI experiments in mice demonstrated that G3.5 was excreted through the kidneys and selectively accumulated in glioblastoma tumors. STATEMENT OF SIGNIFICANCE: This study explores a class of MRI contrast agents based on organic radical dendrimers as a potential alternative to gadolinium-based agents. We present a simplified synthesis method for water-soluble dendrimers containing up to 192 TEMPO radical units-the highest number achieved to date for this class of compounds-resulting in record-high relaxivity values. Our approach offers easier preparation, purification, and tunable water solubility, representing an improvement over existing methods. Through combined experimental and computational studies, we provide insights into the structure-property relationships governing relaxivity. In vivo experiments demonstrate the dendrimers' potential for glioblastoma imaging, with predominantly renal excretion. This work represents a step towards developing metal-free MRI contrast agents with promising relaxivity and biocompatibility, potentially opening new avenues for diagnostic imaging research.
Collapse
Affiliation(s)
- Yufei Wu
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, E-08193 Bellaterra, Spain
| | - Vega Lloveras
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, E-08193 Bellaterra, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Campus UAB, 08913 Bellaterra, Spain.
| | - Silvia Lope-Piedrafita
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Campus UAB, 08913 Bellaterra, Spain; Departament de Bioquímica i Biologia Molecular, Unitat de Biofísica, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Marta Mulero-Acevedo
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Campus UAB, 08913 Bellaterra, Spain; Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ana Paula Candiota
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Campus UAB, 08913 Bellaterra, Spain; Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - José Vidal-Gancedo
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, E-08193 Bellaterra, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Campus UAB, 08913 Bellaterra, Spain.
| |
Collapse
|
2
|
Wang D, Qiu CJ, Chu Y, Zhang A, Huang R, Pan SJ, Tan L. A Polymeric Vesicle System for Combined Lung Cancer Therapy through Chemotherapy and Vasculature Normalization. Biomater Res 2024; 28:0117. [PMID: 39606153 PMCID: PMC11599482 DOI: 10.34133/bmr.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Lung cancer remains a great threat to human health despite the rapid development of various therapeutic methods. Chemotherapy continues to be the most commonly employed treatment for lung cancer; however, it often suffers from low drug delivery efficiency and severe side effects. To enhance the therapeutic efficacy of chemotherapy, we developed a novel strategy that integrates tumor vasculature normalization with the co-delivery of therapeutic agents. This strategy employs a diblock polymeric vesicle with a reduction-sensitive linkage. Paclitaxel (PTX) is encapsulated in the bilayer, while an acid-sensitive nitric oxide (NO) precursor, DETA NONOate, and zinc oxide nanoparticles (ZnO NPs) are loaded into the central cavity. The resulting nanosystem, (ZnO,NONO)@Ves-PTX, is designed to release NO under the acidic conditions typical of the tumor microenvironment (TME) and intracellular environment. The released NO in the TME inhibits angiogenesis, thereby facilitating the delivery and distribution of therapeutic agents. Upon internalization by tumor cells, (ZnO,NONO)@Ves-PTX decomposes in response to intracellular glutathione (GSH), releasing the loaded agents. DETA NONOate and ZnO NPs generate NO and Zn2+ ions, respectively, at the intracellular pH, which synergistically inhibit tumor growth alongside PTX. This combined therapeutic approach demonstrated remarkable potential in improving the chemotherapeutic efficacy for lung cancer, offering a promising direction for future cancer treatments.
Collapse
Affiliation(s)
- Ding Wang
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
| | - Cheng-Jie Qiu
- Department of Neurosurgery, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yaoqing Chu
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
| | - Anzhuo Zhang
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
| | - Ran Huang
- Academy for Engineering and Technology; Yiwu Research Institute; Zhuhai Fudan Innovation Institute,
Fudan University, Shanghai 200433, China
- Center for Innovation and Entrepreneurship,
Taizhou Institute of Zhejiang University, Taizhou, Zhejiang 318000, China
| | - Si-Jian Pan
- Department of Neurosurgery, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lianjiang Tan
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
- Department of Neurosurgery, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
3
|
Pangua C, Espuelas S, Simón JA, Álvarez S, Martínez-Ohárriz C, Collantes M, Peñuelas I, Calvo A, Irache JM. Enhancing bevacizumab efficacy in a colorectal tumor mice model using dextran-coated albumin nanoparticles. Drug Deliv Transl Res 2024:10.1007/s13346-024-01734-3. [PMID: 39455507 DOI: 10.1007/s13346-024-01734-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Bevacizumab is a monoclonal antibody (mAb) that prevents the growth of new blood vessels and is currently employed in the treatment of colorectal cancer (CRC). However, like other mAb, bevacizumab shows a limited penetration in the tumors, hampering their effectiveness and inducing adverse reactions. The aim of this work was to design and evaluate albumin-based nanoparticles, coated with dextran, as carriers for bevacizumab in order to promote its accumulation in the tumor and, thus, improve its antiangiogenic activity. These nanoparticles (B-NP-DEX50) displayed a mean size of about 250 nm and a payload of about 110 µg/mg. In a CRC mice model, these nanoparticles significantly reduced tumor growth and increased tumor doubling time, tumor necrosis and apoptosis more effectively than free bevacizumab. At the end of study, bevacizumab plasma levels were higher in the free drug group, while tumor levels were higher in the B-NP-DEX50 group (2.5-time higher). In line with this, the biodistribution study revealed that nanoparticles accumulated in the tumor core, potentially improving therapeutic efficacy while reducing systemic exposure. In summary, B-NP-DEX can be an adequate alternative to improve the therapeutic efficiency of biologically active molecules, offering a more specific biodistribution to the site of action.
Collapse
Affiliation(s)
- Cristina Pangua
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | - Socorro Espuelas
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Jon Ander Simón
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
| | - Samuel Álvarez
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | | | - María Collantes
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Iván Peñuelas
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Juan M Irache
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain.
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain.
| |
Collapse
|
4
|
Ifijen IH, Christopher AT, Lekan OK, Aworinde OR, Faderin E, Obembe O, Abdulsalam Akanji TF, Igboanugo JC, Udogu U, Ogidi GO, Iorkula TH, Osayawe OJK. Advancements in tantalum based nanoparticles for integrated imaging and photothermal therapy in cancer management. RSC Adv 2024; 14:33681-33740. [PMID: 39450067 PMCID: PMC11498270 DOI: 10.1039/d4ra05732e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
Tantalum-based nanoparticles (TaNPs) have emerged as promising tools in cancer management, owing to their unique properties that facilitate innovative imaging and photothermal therapy applications. This review provides a comprehensive overview of recent advancements in TaNPs, emphasizing their potential in oncology. Key features include excellent biocompatibility, efficient photothermal conversion, and the ability to integrate multifunctional capabilities, such as targeted drug delivery and enhanced imaging. Despite these advantages, challenges remain in establishing long-term biocompatibility, optimizing therapeutic efficacy through surface modifications, and advancing imaging techniques for real-time monitoring. Strategic approaches to address these challenges include surface modifications like PEGylation to improve biocompatibility, precise control over size and shape for effective photothermal therapy, and the development of biodegradable TaNPs for safe elimination from the body. Furthermore, integrating advanced imaging modalities-such as photoacoustic imaging, magnetic resonance imaging (MRI), and computed tomography (CT)-enable real-time tracking of TaNPs in vivo, which is crucial for clinical applications. Personalized medicine strategies that leverage biomarkers and genetic profiling also hold promise for tailoring TaNP-based therapies to individual patient profiles, thereby enhancing treatment efficacy and minimizing side effects. In conclusion, TaNPs represent a significant advancement in nanomedicine, poised to transform cancer treatment paradigms while expanding into various biomedical applications.
Collapse
Affiliation(s)
- Ikhazuagbe H Ifijen
- Department of Research Outreach, Rubber Research Institute of Nigeria Iyanomo Benin City Nigeria
| | - Awoyemi Taiwo Christopher
- Laboratory Department, Covenant University Medical Centre Canaan land, KM 10, Idiroko Road Ota Ogun State Nigeria
| | - Ogunnaike Korede Lekan
- Department of Chemistry, Wichita State University 1845 Fairmount, Box 150 Wichita KS 67260-0150 USA
| | | | - Emmanuel Faderin
- Department of Pharmaceutical Sciences, Southern Illinois University Edwardsville, 1 Hairpin Drive Edwardsville IL 62026-001 USA
| | | | | | - Juliet C Igboanugo
- Department of Health, Human Performance, and Recreation 155 Stadium Drive Arkansas 72701 USA
| | - Uzochukwu Udogu
- Department of Chemistry, Federal University of Technology Owerri Nigeria
| | | | - Terungwa H Iorkula
- Department of Chemistry and Biochemistry, Brigham Young University Provo Utah USA
| | | |
Collapse
|
5
|
Li Y, He J, Liu J, Um W, Ding J. Challenges and opportunities of poly(amino acid) nanomedicines in cancer therapy. Nanomedicine (Lond) 2024; 19:2495-2504. [PMID: 39381990 PMCID: PMC11520535 DOI: 10.1080/17435889.2024.2402677] [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: 07/15/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
Poly(amino acid) nanomedicines hold significant promise for cancer therapy. However, their clinical translation has not matched the extensive efforts of scientists or the burgeoning body of research. The therapeutic outcomes with most nanomedicines often fall short of the promising results observed in animal experiments. This review explores the challenges faced in cancer therapy using poly(amino acid) nanomedicines, particularly addressing the controversies surrounding the enhanced permeability and retention effect and the lack of methods for controlled and reproducible mass production of poly(amino acid) nanomedicines. Furthermore, this review examines the opportunities emerging in this field due to the rapid advancements in artificial intelligence.
Collapse
Affiliation(s)
- Yuce Li
- College of Life Sciences & Health, Wuhan University of Science & Technology, 2 Huangjiahuxi Road, Wuhan, 430065, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Jing He
- College of Life Sciences & Health, Wuhan University of Science & Technology, 2 Huangjiahuxi Road, Wuhan, 430065, P. R. China
| | - Jixiu Liu
- College of Life Sciences & Health, Wuhan University of Science & Technology, 2 Huangjiahuxi Road, Wuhan, 430065, P. R. China
| | - Wooram Um
- Department of Biotechnology, Pukyong National University, 45 Yongso-ro, Nam-Gu, Busan, 48513, Republic of Korea
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| |
Collapse
|
6
|
Qin S, He G, Yang J. Nanomaterial combined engineered bacteria for intelligent tumor immunotherapy. J Mater Chem B 2024; 12:9795-9820. [PMID: 39225508 DOI: 10.1039/d4tb00741g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cancer remains the leading cause of human death worldwide. Compared to traditional therapies, tumor immunotherapy has received a lot of attention and research focus due to its potential to activate both innate and adaptive immunity, low toxicity to normal tissue, and long-term immune activity. However, its clinical effectiveness and large-scale application are limited due to the immunosuppression microenvironment, lack of spatiotemporal control, expensive cost, and long manufacturing time. Recently, nanomaterial combined engineered bacteria have emerged as a promising solution to the challenges of tumor immunotherapy, which offers spatiotemporal control, reversal of immunosuppression, and scalable production. Therefore, we summarize the latest research on nanomaterial-assisted engineered bacteria for precise tumor immunotherapies, including the cross-talk of nanomaterials and bacteria as well as their application in different immunotherapies. In addition, we further discuss the advantages and challenges of nanomaterial-engineered bacteria and their future prospects, inspiring more novel and intelligent tumor immunotherapy.
Collapse
Affiliation(s)
- Shurong Qin
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
7
|
Nouvel J, Bustos‐Quevedo G, Prinz T, Masood R, Daaboul G, Gainey‐Schleicher T, Wittel U, Chikhladze S, Melykuti B, Helmstaedter M, Winkler K, Nazarenko I, Pütz G. Separation of small extracellular vesicles (sEV) from human blood by Superose 6 size exclusion chromatography. J Extracell Vesicles 2024; 13:e70008. [PMID: 39441012 PMCID: PMC11497763 DOI: 10.1002/jev2.70008] [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: 07/17/2023] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
Extracellular vesicles (EVs) are valuable targets for liquid biopsy. However, attempts to introduce EV-based biomarkers into clinical practice have not been successful to the extent expected. One of the reasons for this failure is the lack of reliable methods for EV baseline purification from complex biofluids, such as cell-free plasma or serum. Because available one-step approaches for EV isolation are insufficient to purify EVs, the majority of studies on clinical samples were performed either on a mixture of EVs and lipoproteins, whilst the real number of EVs and their individual specific biomarker content remained elusive, or on a low number of samples of sufficient volume to allow elaborate 2-step EV separation by size and density, resulting in a high purity but utmost low recovery. Here we introduce Fast Protein Liquid Chromatography (FPLC) using Superose 6 as a matrix to obtain small EVs from biofluids that are almost free of soluble proteins and lipoproteins. Along with the estimation of a realistic number of small EVs in human samples, we show temporal resolution of the effect of the duration of postprandial phase on the proportion of lipoproteins in purified EVs, suggesting acceptable time frames additionally to the recommendation to use fasting samples for human studies. Furthermore, we assessed a potential value of pure EVs for liquid biopsy, exemplarily examining EV- and tumour-biomarkers in pure FPLC-derived fractions isolated from the serum of patients with pancreatic cancer. Consistent among different techniques, showed the presence of diseases-associated biomarkers in pure EVs, supporting the feasibility of using single-vesicle analysis for liquid biopsy.
Collapse
Affiliation(s)
- Jerome Nouvel
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Institute of Clinical Chemistry and Laboratory MedicineFreiburgGermany
| | - Gonzalo Bustos‐Quevedo
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Tony Prinz
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Ramsha Masood
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | | | - Tanja Gainey‐Schleicher
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Uwe Wittel
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Department of General and Visceral SurgeryFreiburgGermany
| | - Sophia Chikhladze
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Department of General and Visceral SurgeryFreiburgGermany
| | - Bence Melykuti
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Martin Helmstaedter
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- IMITATE EM Core FacilityFreiburgGermany
| | - Karl Winkler
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Institute of Clinical Chemistry and Laboratory MedicineFreiburgGermany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Hahn‐SchikardFreiburgGermany
- German Cancer Consortium (DKTK)Partner Site Freiburg and German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Gerhard Pütz
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Institute of Clinical Chemistry and Laboratory MedicineFreiburgGermany
| |
Collapse
|
8
|
Chu K, Liu J, Zhang X, Wang M, Yu W, Chen Y, Xu L, Yang G, Zhang N, Zhao T. Herbal Medicine-Derived Exosome-Like Nanovesicles: A Rising Star in Cancer Therapy. Int J Nanomedicine 2024; 19:7585-7603. [PMID: 39081899 PMCID: PMC11287466 DOI: 10.2147/ijn.s477270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Plant-derived exosome-like nanovesicles (PDNVs) are small nanoscale vesicles containing lipids, RNAs, proteins and some plant natural products secreted by plant cells. Over the last decade, PDNVs have garnered significant interest due to its exceptional therapeutic benefits in the treatment of various diseases. Herbal medicine, as a medicinal plant, plays an important role in the treatment of diseases including cancer. Especially in recent years, the function of herbal medicine derived exosome-like nanovesicles (HMDNVs) in the treatment of cancer has been widely concerned, and has become a research hotspot of nanomedicine. In this review, the biological characteristics, functions and the therapeutic advantages of PDNVs are reviewed, as well as the recent achievements and research progress of HMDNVs in cancer treatment, demonstrating its enormous promise as a cancer therapy, and new insights are provided for future research and development of anti-tumor drugs.
Collapse
Affiliation(s)
- Kaifei Chu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
- College of Life Sciences, Zhejiang Normal University, Jinhua, People’s Republic of China
| | - Jie Liu
- College of Life Sciences, Zhejiang Normal University, Jinhua, People’s Republic of China
| | - Xu Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
- College of Life Sciences, Zhejiang Normal University, Jinhua, People’s Republic of China
| | - Minran Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Wanping Yu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Yuyue Chen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Lingling Xu
- College of Life Sciences, Zhejiang Normal University, Jinhua, People’s Republic of China
| | - Geng Yang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Naru Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Tiejun Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
- College of Life Sciences, Zhejiang Normal University, Jinhua, People’s Republic of China
| |
Collapse
|
9
|
Luo T, Jiang X, Fan Y, Yuan E, Li J, Tillman L, Lin W. STING agonist-conjugated metal-organic framework induces artificial leukocytoid structures and immune hotspots for systemic antitumor responses. Natl Sci Rev 2024; 11:nwae167. [PMID: 38887543 PMCID: PMC11182667 DOI: 10.1093/nsr/nwae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 06/20/2024] Open
Abstract
Radiotherapy is widely used for cancer treatment, but its clinical utility is limited by radioresistance and its inability to target metastases. Nanoscale metal-organic frameworks (MOFs) have shown promise as high-Z nanoradiosensitizers to enhance radiotherapy and induce immunostimulatory regulation of the tumor microenvironment. We hypothesized that MOFs could deliver small-molecule therapeutics to synergize with radiotherapy for enhanced antitumor efficacy. Herein, we develop a robust nanoradiosensitizer, GA-MOF, by conjugating a STING agonist, 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (GA), on MOFs for synergistic radiosensitization and STING activation. GA-MOF demonstrated strong anticancer efficacy by forming immune-cell-rich nodules (artificial leukocytoid structures) and transforming them into immunostimulatory hotspots with radiotherapy. Further combination with an immune checkpoint blockade suppressed distant tumors through systemic immune activation. Our work not only demonstrates the potent radiosensitization of GA-MOF, but also provides detailed mechanisms regarding MOF distribution, immune regulatory pathways and long-term immune effects.
Collapse
Affiliation(s)
- Taokun Luo
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Xiaomin Jiang
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Yingjie Fan
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Eric Yuan
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Jinhong Li
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Langston Tillman
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Wenbin Lin
- Department of Chemistry, University of Chicago, Chicago 60637, USA
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| |
Collapse
|
10
|
Krishnan J, Poomalai P, Ravichandran A, Reddy A, Sureshkumar R. A Concise Review on Effect of PEGylation on the Properties of Lipid-Based Nanoparticles. Assay Drug Dev Technol 2024; 22:246-264. [PMID: 38828531 DOI: 10.1089/adt.2024.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Nanoparticle-based drug delivery systems have emerged as promising platforms for enhancing therapeutic efficacy while minimizing off-target effects. Among various strategies employed to optimize these systems, polyethylene glycol (PEG) modification, known as PEGylation-the covalent attachment of PEG to nanoparticles, has gained considerable attention for its ability to impart stealth properties to nanoparticles while also extending circulation time and improving biocompatibility. PEGylation extends to different drug delivery systems, in specific, nanoparticles for targeting cancer cells, where the concentration of drug in the cancer cells is improved by virtue of PEGylation. The primary challenge linked to PEGylation lies in its confirmation. Numerous research findings provide comprehensive insights into selecting PEG for various PEGylation methods. In this review, we have endeavored to consolidate the outcomes concerning the choice of PEG and diverse PEGylation techniques.
Collapse
Affiliation(s)
- Janesha Krishnan
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Praveena Poomalai
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Ashwin Ravichandran
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Aishwarya Reddy
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Raman Sureshkumar
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| |
Collapse
|
11
|
Wang D, Chu Y, Liu S, Tan L. Chitosan-based hybrid nanospheres for vessel normalization towards enhancing tumor chemotherapy. Int J Biol Macromol 2024; 267:131409. [PMID: 38582478 DOI: 10.1016/j.ijbiomac.2024.131409] [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: 11/09/2023] [Revised: 03/04/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Vessel normalization has proved imperative in tumor growth inhibition. In this work, biopolymer-based hybrid nanospheres capable of normalizing blood vessels were designed to improve the therapeutic effect of chemotherapeutic drugs. Zn0.4Fe2.6O4 nanoparticles (ZFO NPs) were synthesized, and were encapsulated in cross-inked chitosan (CS) along with a nitric oxide (NO) precursor, DETA NONOate, forming hybrid ZFO/NO@CS nanospheres highly stable in physiological environment. The structure, morphology and size of the nanospheres were characterized. The ZFO/NO@CS nanospheres could release NO under acidic conditions typical of intratumoral and intracellular environment. The results of related factors expression, wound healing and tube formation assays demonstrated that both the encapsulated ZFO NPs and the released NO were able to inhibit angiogenesis in tumors. The ZFO/NO@CS nanospheres enhanced the antitumor efficacy of the chemotherapeutic drug DOX by normalizing tumor vessels, as evidenced by in vivo experiments for CT26 tumor-bearing mice. By analyzing the contents of Fe in the tumor and different organs, the nanospheres were found to accumulate primarily at the tumor site. The blood analysis showed little side effect of the nanospheres. The ZFO/NO@CS nanospheres have great potential in improving tumor therapeutic effect when used in combination with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Ding Wang
- School of Materials Science and Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Yaoqing Chu
- School of Materials Science and Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Shuiping Liu
- College of Textile and Clothing, Yancheng Institute of Technology, Yancheng 224051, China
| | - Lianjiang Tan
- School of Materials Science and Engineering, Shanghai Institute of Technology, Shanghai 201418, China.
| |
Collapse
|
12
|
Zhang HF, Yu H, Pan SX, Zhang C, Ma YH, Zhang YF, Zuo LL, Hao CY, Lin XY, Geng H, Wu D, Mu SQ, Yu WL, Shi NQ. Multibarrier-penetrating drug delivery systems for deep tumor therapy based on synergistic penetration strategy. Biomater Sci 2024; 12:2321-2330. [PMID: 38488841 DOI: 10.1039/d3bm01959d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Nanotherapies, valued for their high efficacy and low toxicity, frequently serve as antitumor treatments, but do not readily penetrate deep into tumor tissues and cells. Here we developed an improved tumor-penetrating peptide (TPP)-based drug delivery system. Briefly, the established TPP iNGR was modified to generate a linear NGR peptide capable of transporting nanotherapeutic drugs into tumors through a CendR pathway-dependent, neuropilin-1 receptor-mediated process. Although TPPs have been reported to reach intended tumor targets, they often fail to penetrate cell membranes to deliver tumoricidal drugs to intracellular targets. We addressed this issue by harnessing cell penetrating peptide technology to develop a liposome-based multibarrier-penetrating delivery system (mbPDS) with improved synergistic drug penetration into deep tumor tissues and cells. The system incorporated doxorubicin-loaded liposomes coated with nona-arginine (R9) CPP and cyclic iNGR (CRNGRGPDC) molecules, yielding Lip-mbPDS. Lip-mbPDS tumor-targeting, tumor cell/tissue-penetrating and antitumor capabilities were assessed using CD13-positive human fibrosarcoma-derived cell (HT1080)-based in vitro and in vivo tumor models. Lip-mbPDS evaluation included three-dimensional layer-by-layer confocal laser scanning microscopy, cell internalization/toxicity assays, three-dimensional tumor spheroid-based penetration assays and antitumor efficacy assays conducted in an animal model. Lip-mbPDS provided enhanced synergistic drug penetration of multiple biointerfaces for potentially deep tumor therapeutic outcomes.
Collapse
Affiliation(s)
| | - Huan Yu
- School of Pharmacy, Jilin Medical University, China.
| | | | - Chuang Zhang
- School of Pharmacy, Jilin Medical University, China.
| | - Ying-Hui Ma
- School of Pharmacy, Jilin Medical University, China.
| | - Yan-Fei Zhang
- School of Pharmacy, Jilin Medical University, China.
| | - Li-Li Zuo
- School of public health, Jilin Medical University, China
| | - Cheng-Yi Hao
- School of Pharmacy, Jilin Medical University, China.
| | - Xiao-Ying Lin
- School of Pharmacy, Jilin Medical University, China.
| | - Hao Geng
- School of Pharmacy, Jilin Medical University, China.
| | - Di Wu
- School of Pharmacy, Jilin Medical University, China.
| | | | - Wei-Lun Yu
- School of Bioengineering, Jilin Medical University, China
| | - Nian-Qiu Shi
- School of Pharmacy, Jilin Medical University, China.
- College of Pharmaceutical Sciences, Yanbian University, China
| |
Collapse
|
13
|
Ming‐Kun C, Zi‐Xian C, Mao‐Ping C, Hong C, Zhuang‐Fei C, Shan‐Chao Z. Engineered extracellular vesicles: A new approach for targeted therapy of tumors and overcoming drug resistance. Cancer Commun (Lond) 2024; 44:205-225. [PMID: 38155418 PMCID: PMC10876209 DOI: 10.1002/cac2.12518] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023] Open
Abstract
Targeted delivery of anti-tumor drugs and overcoming drug resistance in malignant tumor cells remain significant clinical challenges. However, there are only few effective methods to address these issues. Extracellular vesicles (EVs), actively secreted by cells, play a crucial role in intercellular information transmission and cargo transportation. Recent studies have demonstrated that engineered EVs can serve as drug delivery carriers and showed promising application prospects. Nevertheless, there is an urgent need for further improvements in the isolation and purification of EVs, surface modification techniques, drug assembly processes, and precise recognition of tumor cells for targeted drug delivery purposes. In this review, we summarize the applications of engineered EVs in cancer treatment and overcoming drug resistance, and current challenges associated with engineered EVs are also discussed. This review aims to provide new insights and potential directions for utilizing engineered EVs as targeted delivery systems for anti-tumor drugs and overcoming drug resistance in the near future.
Collapse
Affiliation(s)
- Chen Ming‐Kun
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Chen Zi‐Xian
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Cai Mao‐Ping
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Chen Hong
- Luoyang Key Laboratory of Organic Functional MoleculesCollege of Food and DrugLuoyang Normal UniversityLuoyangHenanP. R. China
| | - Chen Zhuang‐Fei
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Zhao Shan‐Chao
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| |
Collapse
|
14
|
Yang Q, Li S, Ou H, Zhang Y, Zhu G, Li S, Lei L. Exosome-based delivery strategies for tumor therapy: an update on modification, loading, and clinical application. J Nanobiotechnology 2024; 22:41. [PMID: 38281957 PMCID: PMC10823703 DOI: 10.1186/s12951-024-02298-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Malignancy is a major public health problem and among the leading lethal diseases worldwide. Although the current tumor treatment methods have therapeutic effect to a certain extent, they still have some shortcomings such as poor water solubility, short half-life, local and systemic toxicity. Therefore, how to deliver therapeutic agent so as to realize safe and effective anti-tumor therapy become a problem urgently to be solved in this field. As a medium of information exchange and material transport between cells, exosomes are considered to be a promising drug delivery carrier due to their nano-size, good biocompatibility, natural targeting, and easy modification. In this review, we summarize recent advances in the isolation, identification, drug loading, and modification of exosomes as drug carriers for tumor therapy alongside their application in tumor therapy. Basic knowledge of exosomes, such as their biogenesis, sources, and characterization methods, is also introduced herein. In addition, challenges related to the use of exosomes as drug delivery vehicles are discussed, along with future trends. This review provides a scientific basis for the application of exosome delivery systems in oncological therapy.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Haibo Ou
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yuming Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shaohong Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Lanjie Lei
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
15
|
Tan J, Ding B, Chen H, Meng Q, Li J, Yang C, Zhang W, Li X, Han D, Zheng P, Ma P, Lin J. Effects of Skeleton Structure of Mesoporous Silica Nanoadjuvants on Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305567. [PMID: 37702141 DOI: 10.1002/smll.202305567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/20/2023] [Indexed: 09/14/2023]
Abstract
Mesoporous silica nanoparticles (MSNs) have been widely praised as nanoadjuvants in vaccine/tumor immunotherapy thanks to their excellent biocompatibility, easy-to-modify surface, adjustable particle size, and remarkable immuno-enhancing activity. However, the application of MSNs is still greatly limited by some severe challenges including the unclear and complicated relationships of structure and immune effect. Herein, three commonly used MSNs with different skeletons including MSN with tetrasulfide bonds (TMSN), MSN containing ethoxy framework (EMSN), and pure -Si-O-Si- framework of MSN (MSN) are comprehensively compared to study the impact of chemical construction on immune effect. The results fully demonstrate that the three MSNs have great promise in improving cellular immunity for tumor immunotherapy. Moreover, the TMSN performs better than the other two MSNs in antigen loading, cellular uptake, reactive oxygen species (ROS) generation, lymph node targeting, immune activation, and therapeutic efficiency. The findings provide a new paradigm for revealing the structure-function relationship of mesoporous silica nanoadjuvants, paving the way for their future clinical application.
Collapse
Affiliation(s)
- Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Chunzheng Yang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Wenying Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Xinyang Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Di Han
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Superlight Materials & Surface Technology of Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
16
|
Zheng S, Li G, Shi J, Liu X, Li M, He Z, Tian C, Kamei KI. Emerging platinum(IV) prodrug nanotherapeutics: A new epoch for platinum-based cancer therapy. J Control Release 2023; 361:819-846. [PMID: 37597809 DOI: 10.1016/j.jconrel.2023.08.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Owing to the unique DNA damaging cytotoxicity, platinum (Pt)-based chemotherapy has long been the first-line choice for clinical oncology. Unfortunately, Pt drugs are restricted by the severe dose-dependent toxicity and drug resistance. Correspondingly, Pt(IV) prodrugs are developed with the aim to improve the antitumor performance of Pt drugs. However, as "free" molecules, Pt(IV) prodrugs are still subject to unsatisfactory in vivo destiny and antitumor efficacy. Recently, Pt(IV) prodrug nanotherapeutics, inheriting both the merits of Pt(IV) prodrugs and nanotherapeutics, have emerged and demonstrated the promise to address the underexploited dilemma of Pt-based cancer therapy. Herein, we summarize the latest fronts of emerging Pt(IV) prodrug nanotherapeutics. First, the basic outlines of Pt(IV) prodrug nanotherapeutics are overviewed. Afterwards, how versatile Pt(IV) prodrug nanotherapeutics overcome the multiple biological barriers of antitumor drug delivery is introduced in detail. Moreover, advanced combination therapies based on multimodal Pt(IV) prodrug nanotherapeutics are discussed with special emphasis on the synergistic mechanisms. Finally, prospects and challenges of Pt(IV) prodrug nanotherapeutics for future clinical translation are spotlighted.
Collapse
Affiliation(s)
- Shunzhe Zheng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guanting Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianbin Shi
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinying Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Meng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, Hangzhou 310058, China.
| | - Ken-Ichiro Kamei
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
17
|
Tan KF, In LLA, Vijayaraj Kumar P. Surface Functionalization of Gold Nanoparticles for Targeting the Tumor Microenvironment to Improve Antitumor Efficiency. ACS APPLIED BIO MATERIALS 2023; 6:2944-2981. [PMID: 37435615 DOI: 10.1021/acsabm.3c00202] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Gold nanoparticles (AuNPs) have undergone significant research for their use in the treatment of cancer. Numerous researchers have established their potent antitumor properties, which have greatly impacted the treatment of cancer. AuNPs have been used in four primary anticancer treatment modalities, namely radiation, photothermal therapy, photodynamic therapy, and chemotherapy. However, the ability of AuNPs to destroy cancer is lacking and can even harm healthy cells without the right direction to transport them to the tumor microenvironment. Consequently, a suitable targeting technique is needed. Based on the distinct features of the human tumor microenvironment, this review discusses four different targeting strategies that target the four key features of the tumor microenvironment, including abnormal vasculature, overexpression of specific receptors, an acidic microenvironment, and a hypoxic microenvironment, to direct surface-functionalized AuNPs to the tumor microenvironment and increase antitumor efficacies. In addition, some current completed or ongoing clinical trials of AuNPs will also be discussed below to further reinforce the concept of using AuNPs in anticancer therapy.
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
18
|
Xie Q, Tang J, Guo S, Zhao Q, Li S. Recent Progress of Preparation Strategies in Organic Nanoparticles for Cancer Phototherapeutics. Molecules 2023; 28:6038. [PMID: 37630290 PMCID: PMC10459389 DOI: 10.3390/molecules28166038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/27/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Phototherapy has the advantages of being a highly targeted, less toxic, less invasive, and repeatable treatment, compared with conventional treatment methods such as surgery, chemotherapy, and radiotherapy. The preparation strategies are significant in order to determine the physical and chemical properties of nanoparticles. However, choosing appropriate preparation strategies to meet applications is still challenging. This review summarizes the recent progress of preparation strategies in organic nanoparticles, mainly focusing on the principles, methods, and advantages of nanopreparation strategies. In addition, typical examples of cancer phototherapeutics are introduced in detail to inform the choice of appropriate preparation strategies. The relative future trend and outlook are preliminarily proposed.
Collapse
Affiliation(s)
| | | | | | - Qi Zhao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (Q.X.); (J.T.); (S.G.)
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (Q.X.); (J.T.); (S.G.)
| |
Collapse
|
19
|
Yun WS, Kim J, Lim DK, Kim DH, Jeon SI, Kim K. Recent Studies and Progress in the Intratumoral Administration of Nano-Sized Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2225. [PMID: 37570543 PMCID: PMC10421122 DOI: 10.3390/nano13152225] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
Over the last 30 years, diverse types of nano-sized drug delivery systems (nanoDDSs) have been intensively explored for cancer therapy, exploiting their passive tumor targetability with an enhanced permeability and retention effect. However, their systemic administration has aroused some unavoidable complications, including insufficient tumor-targeting efficiency, side effects due to their undesirable biodistribution, and carrier-associated toxicity. In this review, the recent studies and advancements in intratumoral nanoDDS administration are generally summarized. After identifying the factors to be considered to enhance the therapeutic efficacy of intratumoral nanoDDS administration, the experimental results on the application of intratumoral nanoDDS administration to various types of cancer therapies are discussed. Subsequently, the reports on clinical studies of intratumoral nanoDDS administration are addressed in short. Intratumoral nanoDDS administration is proven with its versatility to enhance the tumor-specific accumulation and retention of therapeutic agents for various therapeutic modalities. Specifically, it can improve the efficacy of therapeutic agents with poor bioavailability by increasing their intratumoral concentration, while minimizing the side effect of highly toxic agents by restricting their delivery to normal tissues. Intratumoral administration of nanoDDS is considered to expand its application area due to its potent ability to improve therapeutic effects and relieve the systemic toxicities of nanoDDSs.
Collapse
Affiliation(s)
- Wan Su Yun
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jeongrae Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Kwon Lim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hwee Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
20
|
Dos Apostolos RCR, Andrada ADS, Oliveira AF, Neto ESF, de Sousa EMB. pH-Sensitive Hybrid System Based on Eu 3+/Gd 3+ Co-Doped Hydroxyapatite and Mesoporous Silica Designed for Theranostic Applications. Polymers (Basel) 2023; 15:2681. [PMID: 37376326 DOI: 10.3390/polym15122681] [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: 05/18/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Nanomaterials such as pH-responsive polymers are promising for targeted drug delivery systems, due to the difference in pH between tumor and healthy regions. However, there is a significant concern about the application of these materials in this field due to their low mechanical resistance, which can be attenuated by combining these polymers with mechanically resistant inorganic materials such as mesoporous silica nanoparticles (MSN) and hydroxyapatite (HA). Mesoporous silica has interesting properties such as high surface area and hydroxyapatite has been widely studied to aid in bone regeneration, providing special properties adding multifunctionality to the system. Furthermore, fields of medicine involving luminescent elements such as rare earth elements are an interesting option in cancer treatment. The present work aims to obtain a pH-sensitive hybrid system based on silica and hydroxyapatite with photoluminescent and magnetic properties. The nanocomposites were characterized by X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FTIR), nitrogen adsorption methods, CHN elemental analysis, Zeta Potential, scanning electron microscopy (SEM), and transmission electron microscopy (TEM), vibrational sample magnetometry (VSM), and photoluminescence analysis. Incorporation and release studies of the antitumor drug doxorubicin were performed to evaluate the potential use of these systems in targeted drug delivery. The results showed the luminescent and magnetic properties of the materials and showed suitable characteristics for application in the release of pH-sensitive drugs.
Collapse
Affiliation(s)
| | - Andreza de Sousa Andrada
- Laboratório Interdisciplinar de Materiais Compósitos e Poliméricos (LIMCOP), Instituto de Engenharias Integradas (IEI) da Universidade Federal de Itajubá, Federal University of Itajubá-UNIFEI, Rua Irmã Ivone Drumond, 200-Campus Itabira, Itabira 35903-087, MG, Brazil
| | - André Felipe Oliveira
- Development Center of Nuclear Technology-CDTN, Avenida Presidente Antônio Carlos, 6.627-Campus UFMG, Belo Horizonte 31270-901, MG, Brazil
| | - Ernesto Soares Freitas Neto
- Laboratório Interdisciplinar de Materiais Compósitos e Poliméricos (LIMCOP), Instituto de Engenharias Integradas (IEI) da Universidade Federal de Itajubá, Federal University of Itajubá-UNIFEI, Rua Irmã Ivone Drumond, 200-Campus Itabira, Itabira 35903-087, MG, Brazil
| | - Edésia Martins Barros de Sousa
- Development Center of Nuclear Technology-CDTN, Avenida Presidente Antônio Carlos, 6.627-Campus UFMG, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
21
|
Pu Y, Ke H, Wu C, Xu S, Xiao Y, Han L, Lyv G, Li S. Superparamagnetic iron oxide nanoparticles target BxPC-3 cells and silence MUC4 for theranostics of pancreatic cancer. Biochim Biophys Acta Gen Subj 2023:130383. [PMID: 37236323 DOI: 10.1016/j.bbagen.2023.130383] [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: 11/20/2022] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Abstract
PURPOSE Superparamagnetic iron oxide nanoparticles (SPION) are excellent magnetic resonance imaging (MRI) contrast agents. Mucin 4 (MUC4) acts as pancreatic cancer (PC) tumor antigen and influences PC progression. Small interfering RNAs (siRNAs) are used as a gene-silencing tool to treat a variety of diseases. METHODS We designed a therapeutic probe based on polyetherimide-superparamagnetic iron oxide nanoparticles (PEI-SPION) combined with siRNA nanoprobes (PEI-SPION-siRNA) to assess the contrast in MRI. The biocompatibility of the nanocomposite, and silencing of MUC4 were characterized and evaluated. RESULTS The prepared molecular probe had a particle size of 61.7 ± 18.5 nmand a surface of 46.7 ± 0.8mVand showed good biocompatibility in vitro and T2 relaxation efficiency. It can also load and protect siRNA. PEI-SPION-siRNA showed a good silencing effect on MUC4. CONCLUSION PEI-SPION-siRNA may be beneficial as a novel theranostic tool for PC.
Collapse
Affiliation(s)
- Yu Pu
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China; Department of Medical Imaging Key Laboratory of Sichuan Province, Affiliated Hospital of North Sichuan Medical College. No. 234, Fujiang Road, Shunqing District, Nanchong City 637000, People's Republic of China; Department of Medicine, Quanzhou Medical College, No. 2 Anji Road, Luojiang District, Quanzhou 362000, People's Republic of China
| | - Helin Ke
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Changqiang Wu
- Department of Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College. No. 55, Dongshun Road, Gaoping District, Nanchong City 637100, People's Republic of China
| | - Shaodan Xu
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Yang Xiao
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Lina Han
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Guorong Lyv
- Department of Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College. No. 55, Dongshun Road, Gaoping District, Nanchong City 637100, People's Republic of China.
| | - Shilin Li
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China.
| |
Collapse
|
22
|
Zhang H, Xue Q, Zhou Z, He N, Li S, Zhao C. Co-delivery of doxorubicin and hydroxychloroquine via chitosan/alginate nanoparticles for blocking autophagy and enhancing chemotherapy in breast cancer therapy. Front Pharmacol 2023; 14:1176232. [PMID: 37229260 PMCID: PMC10203398 DOI: 10.3389/fphar.2023.1176232] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy in women worldwide, and the standard treatment is chemotherapy or radiotherapy after surgery. In order to reduce the side effects of chemotherapy, various nanoparticles (NPs) have been discovered and synthesized, which has become a promising treatment for BC. In this study, a co-delivery nanodelivery drug system (Co-NDDS) was designed and synthesized with 2,3-dimercaptosuccinic acid (DMSA) coated Fe3O4 NPs as core encapsulated into chitosan/alginate nanoparticles (CANPs) shell, doxorubicin (DOX) and hydroxychloroquine (HCQ) as loading drugs. Smaller NPs carrying DOX (FeAC-DOX NPs) were loaded into larger NPs containing HCQ (FeAC-DOX@PC-HCQ NPs) by ionic gelation and emulsifying solvent volatilization methods. The physicochemical properties of this Co-NDDS were characterised, followed by in vitro studies of the anticancer effects and mechanisms using two different BC cell lines, MCF-7 cells and MDA-MB-231 cells. The results indicated that the Co-NDDS showcases exemplary physicochemical qualities and encapsulation capacity, facilitating accurate intracellular release through pH-sensitive attributes. Importantly, NPs can significantly increase the in vitro cytotoxicity of co-administered drugs and effectively inhibit the autophagy level of tumour cells. The Co-NDDS constructed in this study provides a promising strategy for the treatment of BC.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qingwen Xue
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zihan Zhou
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
- Sino Genomics Technology Co., Ltd, Qingdao, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Cheng Zhao
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
23
|
Ji X, Cai Y, Dong X, Wu W, Zhao W. Selection of an aggregation-caused quenching-based fluorescent tracer for imaging studies in nano drug delivery systems. NANOSCALE 2023. [PMID: 37158114 DOI: 10.1039/d3nr01018j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
In order to develop and optimize nano drug delivery systems (NDDSs), it is crucial to understand their in vivo fate. We previously found that P2 (Aza-BODIPY) and P4 (BODIPY) as aggregation-caused quenching (ACQ) probes could be used to unravel the biofate of various nanoparticles owing to their water-sensitive emission. However, previous studies also found that quenched ACQ probe aggregates showed repartition into hydrophobic physiologically relevant constituents, resulting in fluorescence re-illumination. In this paper, we screened various types of fluorophores for ACQ and their re-illumination performance and focused on Aza-BODIPY dyes. BODIPY and Aza-BODIPY dyes were identified to be advantageous over other fluorophores. Some BODIPY and Aza-BODIPY dyes were selected as potential probes with improved performance against re-illumination. The best performing probes were Aza-C7 and Aza-C8. Aza-C7-loaded PMs were found to have decreased fluorescence re-illumination properties over P2 and DiR.
Collapse
Affiliation(s)
- Xin Ji
- Key Laboratory for Special Functional Materials of Ministry of Education, National & Local Joint Engineering Research Center for High-efficiency Display and Lighting Technology, and School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China.
| | - Yifan Cai
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Xiaochun Dong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Weili Zhao
- Key Laboratory for Special Functional Materials of Ministry of Education, National & Local Joint Engineering Research Center for High-efficiency Display and Lighting Technology, and School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China.
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
24
|
Orel VB, Papazoglou ΑS, Tsagkaris C, Moysidis DV, Papadakos S, Galkin OY, Orel VE, Syvak LA. Nanotherapy based on magneto-mechanochemical modulation of tumor redox state. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1868. [PMID: 36289050 DOI: 10.1002/wnan.1868] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/25/2022] [Accepted: 10/10/2022] [Indexed: 05/13/2023]
Abstract
Magnetic nanoparticles (MNs) are typically used as contrast agents for magnetic resonance imaging or as drug carriers with a remotely controlled delivery to the tumor. However, they can also potentiate the action of anticancer drugs under the influence of applied constant magnetic (CMFs) and electromagnetic fields (EMFs). This review demonstrates the role of magneto-mechanochemical effects produced by MNs alone and loaded with anticancer agents (MNCs) in response to CMFs and EMFs for modulation of tumor redox state. The combined treatment is suggested to act by two mechanisms: spin-dependent electron transport propagates free radical chain reactions, while magnetomechanical interactions cause conformational changes in drug molecules loaded onto MNs and generate reactive oxygen species (ROS). By adjusting the parameters of CMFs and EMFs during the magneto-mechanochemical synthesis and subsequent treatment, it is possible to modulate ROS production and switch redox signaling involved in ERK1/2 and NF-κB pathways from initiation of tumor growth to inhibition. Observations of tumor volume in different animal models and treatment combinations reported a 6%-70% reduction as compared with conventional drugs. Despite these results, there is a general lack of research in magnetic nanotheranostics that link redox changes across multiple levels of organization in the tumor-bearing host. Further multidisciplinary studies with more focus on the relationship between the electron transport processes in biomolecules and their effects on the tumor-host interaction should accelerate the clinical translation of magnetic nanotheranostics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Valerii B Orel
- National Cancer Institute, Kyiv, Ukraine
- Faculty of Biomedical Engineering, National Technical University of Ukraine "Igor Sikorsky Kyiv Polytechnic Institute", Kyiv, Ukraine
| | | | - Christos Tsagkaris
- Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
| | - Dimitrios V Moysidis
- Department of Cardiology, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| | | | - Olexander Yu Galkin
- Faculty of Biomedical Engineering, National Technical University of Ukraine "Igor Sikorsky Kyiv Polytechnic Institute", Kyiv, Ukraine
| | - Valerii E Orel
- National Cancer Institute, Kyiv, Ukraine
- Faculty of Biomedical Engineering, National Technical University of Ukraine "Igor Sikorsky Kyiv Polytechnic Institute", Kyiv, Ukraine
| | | |
Collapse
|
25
|
Current understanding of passive and active targeting nanomedicines to enhance tumor accumulation. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
26
|
Shen Q, Du Y. A comprehensive review of advanced drug delivery systems for the treatment of rheumatoid arthritis. Int J Pharm 2023; 635:122698. [PMID: 36754181 DOI: 10.1016/j.ijpharm.2023.122698] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/21/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Rheumatoid arthritis (RA), a chronic autoimmune disease, is characterized by articular pain and swelling, synovial hyperplasia, and cartilage and bone destruction. Conventional treatment strategies for RA involve the use of anti-rheumatic drugs, which warrant high-dose, frequent, and long-term administration, resulting in serious adverse effects and poor patient compliance. To overcome these problems and improve clinical efficacy, drug delivery systems (DDS) have been designed for RA treatment. These systems have shown success in animal models of RA. In this review, representative DDS that target RA through passive or active effects on inflammatory cells are discussed and highlighted using examples. In particular, DDS allowing controlled and targeted drug release based on a variety of stimuli, intra-articular DDS, and transdermal DDS for RA treatment are described. Thus, this review provides an improved understanding of these DDS and paves the way for the development of novel DDS for efficient RA treatment.
Collapse
Affiliation(s)
- Qiying Shen
- School of Pharmacy, Hangzhou Normal University, 2318 Yu-HangTang Road, Hangzhou 311121, China; Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-HangTang Road, Hangzhou 310058, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-HangTang Road, Hangzhou 310058, China.
| |
Collapse
|
27
|
Ernst LM, Mondragón L, Ramis J, Gustà MF, Yudina T, Casals E, Bastús NG, Fernández-Varo G, Casals G, Jiménez W, Puntes V. Exploring the Long-Term Tissue Accumulation and Excretion of 3 nm Cerium Oxide Nanoparticles after Single Dose Administration. Antioxidants (Basel) 2023; 12:765. [PMID: 36979013 PMCID: PMC10045098 DOI: 10.3390/antiox12030765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Nanoparticle (NP) pharmacokinetics significantly differ from traditional small molecule principles. From this emerges the need to create new tools and concepts to harness their full potential and avoid unnecessary risks. Nanoparticle pharmacokinetics strongly depend on size, shape, surface functionalisation, and aggregation state, influencing their biodistribution, accumulation, transformations, and excretion profile, and hence their efficacy and safety. Today, while NP biodistribution and nanoceria biodistribution have been studied often at short times, their long-term accumulation and excretion have rarely been studied. In this work, 3 nm nanoceria at 5.7 mg/kg of body weight was intravenously administrated in a single dose to healthy mice. Biodistribution was measured in the liver, spleen, kidney, lung, brain, lymph nodes, ovary, bone marrow, urine, and faeces at different time points (1, 9, 30, and 100 days). Biodistribution and urinary and faecal excretion were also studied in rats placed in metabolic cages at shorter times. The similarity of results of different NPs in different models is shown as the heterogeneous nanoceria distribution in organs. After the expectable accumulation in the liver and spleen, the concentration of cerium decays exponentially, accounting for about a 50% excretion of cerium from the body in 100 days. Cerium ions, coming from NP dissolution, are most likely excreted via the urinary tract, and ceria nanoparticles accumulated in the liver are most likely excreted via the hepatobiliary route. In addition, nanoceria looks safe and does not damage the target organs. No weight loss or apathy was observed during the course of the experiments.
Collapse
Affiliation(s)
- Lena M. Ernst
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Laura Mondragón
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Joana Ramis
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Muriel F. Gustà
- Institut Català de Nanociència I Nanotecnologia (ICN2), CSIC, The Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Tetyana Yudina
- Institut Català de Nanociència I Nanotecnologia (ICN2), CSIC, The Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Eudald Casals
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Neus G. Bastús
- Institut Català de Nanociència I Nanotecnologia (ICN2), CSIC, The Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Guillermo Fernández-Varo
- Service of Biochemistry and Molecular Genetics, Hospital Clinic, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Gregori Casals
- Service of Biochemistry and Molecular Genetics, Hospital Clinic, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Fundamental Care and Medical-Surgical Nursing, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Wladimiro Jiménez
- Service of Biochemistry and Molecular Genetics, Hospital Clinic, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Departament de Biomedicina, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Victor Puntes
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
- Institut Català de Nanociència I Nanotecnologia (ICN2), CSIC, The Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
28
|
Liu C, Wu H, Duan H, Hou Y, Wang S, Liu Y, Zhang X, Zhao H, Gong L, Wan H, Zeng B, Quan X, Cui M, Chen L, Jin M, Wang Q, Gao Z, Huang W. An EGCG-mediated self-assembled micellar complex acts as a bioactive drug carrier. Food Chem 2023; 418:135939. [PMID: 36948024 DOI: 10.1016/j.foodchem.2023.135939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/17/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023]
Abstract
Epigallocatechin gallate (EGCG) has attracted the increasing attention of many researchers, especially in the field of tumor therapy. However, EGCG has poor fat solubility, low stability, low bioavailability, and a high effective dose in vivo. Traditional drug delivery methods are difficult to deliver the water-soluble EGCG efficiently and in high doses to tumor sites. To address these issues, a new type of strategy has been tried in this study to transform EGCG from a "Bioactive natural ingredient" into a "Bioactive drug carrier". Briefly, the EGCG was modified with a fat-soluble 9-fluorene methoxy carbonyl (Fmoc) motif, and the obtained EGCG-Fmoc showed a considerable improvement in lipid solubility and stability. Interestingly, EGCG-Fmoc obtained the characteristic of self-assembly in water, making it easier to take up by tumor cells. Furthermore, the self-assembled nanocomplex exhibited paclitaxel encapsulation performance and could achieve the dual delivery of EGCG and paclitaxel.
Collapse
Affiliation(s)
- Chao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Hao Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Department of Pharmacy, Yanbian University, Yanji, Jilin 133000, PR China
| | - Hongxia Duan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Yan Hou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Department of Pharmacy, Yanbian University, Yanji, Jilin 133000, PR China
| | - Shuangqing Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Department of Pharmacy, Yanbian University, Yanji, Jilin 133000, PR China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Xintong Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Heming Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Liming Gong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Hongshuang Wan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Department of Gastroenterology, Yanbian University Hospital, Yanji, Jilin 133000, PR China
| | - Bowen Zeng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Department of Gastroenterology, Yanbian University Hospital, Yanji, Jilin 133000, PR China
| | - Xiuquan Quan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Department of Gastroenterology, Yanbian University Hospital, Yanji, Jilin 133000, PR China
| | - Minhu Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Department of Gastroenterology, Yanbian University Hospital, Yanji, Jilin 133000, PR China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Qiming Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China.
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
29
|
Jiang J, Huang Y, Zeng Z, Zhao C. Harnessing Engineered Immune Cells and Bacteria as Drug Carriers for Cancer Immunotherapy. ACS NANO 2023; 17:843-884. [PMID: 36598956 DOI: 10.1021/acsnano.2c07607] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Immunotherapy continues to be in the spotlight of oncology therapy research in the past few years and has been proven to be a promising option to modulate one's innate and adaptive immune systems for cancer treatment. However, the poor delivery efficiency of immune agents, potential off-target toxicity, and nonimmunogenic tumors significantly limit its effectiveness and extensive application. Recently, emerging biomaterial-based drug carriers, including but not limited to immune cells and bacteria, are expected to be potential candidates to break the dilemma of immunotherapy, with their excellent natures of intrinsic tumor tropism and immunomodulatory activity. More than that, the tiny vesicles and physiological components derived from them have similar functions with their source cells due to the inheritance of various surface signal molecules and proteins. Herein, we presented representative examples about the latest advances of biomaterial-based delivery systems employed in cancer immunotherapy, including immune cells, bacteria, and their derivatives. Simultaneously, opportunities and challenges of immune cells and bacteria-based carriers are discussed to provide reference for their future application in cancer immunotherapy.
Collapse
Affiliation(s)
- Jingwen Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Zishan Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| |
Collapse
|
30
|
Winter G, Eberhardt N, Löffler J, Raabe M, Alam MNA, Hao L, Abaei A, Herrmann H, Kuntner C, Glatting G, Solbach C, Jelezko F, Weil T, Beer AJ, Rasche V. Preclinical PET and MR Evaluation of 89Zr- and 68Ga-Labeled Nanodiamonds in Mice over Different Time Scales. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4471. [PMID: 36558325 PMCID: PMC9780863 DOI: 10.3390/nano12244471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Nanodiamonds (NDs) have high potential as a drug carrier and in combination with nitrogen vacancies (NV centers) for highly sensitive MR-imaging after hyperpolarization. However, little remains known about their physiological properties in vivo. PET imaging allows further evaluation due to its quantitative properties and high sensitivity. Thus, we aimed to create a preclinical platform for PET and MR evaluation of surface-modified NDs by radiolabeling with both short- and long-lived radiotracers. Serum albumin coated NDs, functionalized with PEG groups and the chelator deferoxamine, were labeled either with zirconium-89 or gallium-68. Their biodistribution was assessed in two different mouse strains. PET scans were performed at various time points up to 7 d after i.v. injection. Anatomical correlation was provided by additional MRI in a subset of animals. PET results were validated by ex vivo quantification of the excised organs using a gamma counter. Radiolabeled NDs accumulated rapidly in the liver and spleen with a slight increase over time, while rapid washout from the blood pool was observed. Significant differences between the investigated radionuclides were only observed for the spleen (1 h). In summary, we successfully created a preclinical PET and MR imaging platform for the evaluation of the biodistribution of NDs over different time scales.
Collapse
Affiliation(s)
- Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Nina Eberhardt
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jessica Löffler
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
- Department of Internal Medicine II, Experimental Cardiovascular Imaging, Ulm University Medical Center, 89081 Ulm, Germany
| | - Marco Raabe
- Department of Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Md. Noor A. Alam
- Department of Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Li Hao
- Department of Internal Medicine II, Experimental Cardiovascular Imaging, Ulm University Medical Center, 89081 Ulm, Germany
| | - Alireza Abaei
- Department of Internal Medicine II, Experimental Cardiovascular Imaging, Ulm University Medical Center, 89081 Ulm, Germany
| | - Hendrik Herrmann
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Claudia Kuntner
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University Vienna, 1090 Vienna, Austria
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Fedor Jelezko
- Institute for Quantum Optics, Ulm University, 89081 Ulm, Germany
| | - Tanja Weil
- Department of Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Volker Rasche
- Department of Internal Medicine II, Experimental Cardiovascular Imaging, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
31
|
Tefas LR, Toma I, Sesarman A, Banciu M, Jurj A, Berindan-Neagoe I, Rus L, Stiufiuc R, Tomuta I. Co-delivery of gemcitabine and salinomycin in PEGylated liposomes for enhanced anticancer efficacy against colorectal cancer. J Liposome Res 2022:1-17. [PMID: 36472146 DOI: 10.1080/08982104.2022.2153139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Colorectal cancer remains one of the major causes of morbidity and mortality in both developed and emerging countries. Cancer stem cells (CSCs) are a subpopulation of cells within the tumor mass harboring stem cell characteristics, considered responsible for tumor initiation, growth, relapse, and treatment failure. Lately, it has become clear that both CSCs and non-CSCs have to be eliminated for the successful eradication of cancer. Drug delivery systems have been extensively employed to enhance drug efficacy. In this study, salinomycin (SAL), a selective anti-CSC drug, and gemcitabine (GEM), a conventional anticancer drug, were co-loaded in liposomes and tested for optimal therapeutic efficacy. We employed the Design of Experiments approach to develop and optimize a liposomal delivery system for GEM and SAL. The antiproliferative effect of the liposomes was evaluated in SW-620 human colorectal cancer cells. The GEM and SAL-loaded liposomes exhibited adequate size, polydispersity, zeta potential, and drug content. The in vitro release study showed a sustained release of GEM and SAL from the liposomes over 72 h. Moreover, no sign of liposome aggregation was seen over 1 month and in a biological medium (FBS). The in vitro cytotoxic effects of the co-loaded liposomes were superior to that of single GEM either in free or liposomal form. The combination therapy using GEM and SAL co-loaded in liposomes could be a promising strategy for tackling colorectal cancer.
Collapse
Affiliation(s)
- Lucia Ruxandra Tefas
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Ioana Toma
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, “Babes-Bolyai” University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, “Babes-Bolyai” University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Lucia Rus
- Department of Drug Analysis, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Rares Stiufiuc
- Department of Bionanoscopy, MedFuture Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania
| | - Ioan Tomuta
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania
| |
Collapse
|
32
|
Park S, Kim J, Lee C. Injectable rapidly dissolving needle-type gelatin implant capable of delivering high concentrations of H2O2 through intratumoral injection. Biomed Pharmacother 2022; 156:113910. [DOI: 10.1016/j.biopha.2022.113910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/24/2022] Open
|
33
|
Rodrigues CF, Fernandes N, de Melo‐Diogo D, Correia IJ, Moreira AF. Cell-Derived Vesicles for Nanoparticles' Coating: Biomimetic Approaches for Enhanced Blood Circulation and Cancer Therapy. Adv Healthc Mater 2022; 11:e2201214. [PMID: 36121767 PMCID: PMC11481079 DOI: 10.1002/adhm.202201214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/11/2022] [Indexed: 01/28/2023]
Abstract
Cancer nanomedicines are designed to encapsulate different therapeutic agents, prevent their premature release, and deliver them specifically to cancer cells, due to their ability to preferentially accumulate in tumor tissue. However, after intravenous administration, nanoparticles immediately interact with biological components that facilitate their recognition by the immune system, being rapidly removed from circulation. Reports show that less than 1% of the administered nanoparticles effectively reach the tumor site. This suboptimal pharmacokinetic profile is pointed out as one of the main factors for the nanoparticles' suboptimal therapeutic effectiveness and poor translation to the clinic. Therefore, an extended blood circulation time may be crucial to increase the nanoparticles' chances of being accumulated in the tumor and promote a site-specific delivery of therapeutic agents. For that purpose, the understanding of the forces that govern the nanoparticles' interaction with biological components and the impact of the physicochemical properties on the in vivo fate will allow the development of novel and more effective nanomedicines. Therefore, in this review, the nano-bio interactions are summarized. Moreover, the application of cell-derived vesicles for extending the blood circulation time and tumor accumulation is reviewed, focusing on the advantages and shortcomings of each cell source.
Collapse
Affiliation(s)
- Carolina F. Rodrigues
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Natanael Fernandes
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Duarte de Melo‐Diogo
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Ilídio J. Correia
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - André F. Moreira
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
- CPIRN‐UDI/IPG – Center of Potential and Innovation in Natural Resources, Research Unit for Inland DevelopmentInstituto Politécnico da GuardaAvenida Dr. Francisco de Sá CarneiroGuarda6300‐559Portugal
| |
Collapse
|
34
|
Diep YN, Kim TJ, Cho H, Lee LP. Nanomedicine for advanced cancer immunotherapy. J Control Release 2022; 351:1017-1037. [DOI: 10.1016/j.jconrel.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/09/2022]
|
35
|
de Oliveira JV, Oliveira da Rocha MC, de Sousa-Junior AA, Rodrigues MC, Farias GR, da Silva PB, Bao SN, Bakuzis AF, Azevedo RB, Morais PC, Muehlmann LA, Figueiró Longo JP. Tumor vascular heterogeneity and the impact of subtumoral nanoemulsion biodistribution. Nanomedicine (Lond) 2022; 17:2073-2088. [PMID: 36853205 DOI: 10.2217/nnm-2022-0176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Aim: Investigate the heterogeneous tumor tissue organization and examine how this condition can interfere with the passive delivery of a lipid nanoemulsion in two breast cancer preclinical models (4T1 and Ehrlich). Materials & methods: The authors used in vivo image techniques to follow the nanoemulsion biodistribution and microtomography, as well as traditional histopathology and electron microscopy to evaluate the tumor structural characteristics. Results & conclusion: Lipid nanoemulsion was delivered to the tumor, vascular organization depends upon the subtumoral localization and this heterogeneous organization promotes a nanoemulsion biodistribution to the highly vascular peripherical region. Also, the results are presented with a comprehensive mathematical model, describing the differential biodistribution in two different breast cancer models, the 4T1 and Ehrlich models.
Collapse
Affiliation(s)
| | | | | | - Mosar Corrêa Rodrigues
- Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Gabriel Ribeiro Farias
- Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | | | - Sônia Nair Bao
- Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | | | - Ricardo Bentes Azevedo
- Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Paulo César Morais
- Institute of Physics, University of Brasília, Brasília, DF, 70910-900, Brazil
- Biotechnology & Genomic Sciences, Catholic University of Brasília, Brasília, DF, 70790-160, Brazil
| | | | | |
Collapse
|
36
|
Wang T, Lin M, Mao J, Tian L, Gan H, Hu X, Yan L, Long H, Cai J, Zheng X, Xiao Y, Li D, Shuai X, Pang P. Inflammation-Regulated Nanodrug Sensitizes Hepatocellular Carcinoma to Checkpoint Blockade Therapy by Reprogramming the Tumor Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:49542-49554. [PMID: 36314479 DOI: 10.1021/acsami.2c14448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Immune checkpoint blockade (ICB) utilizing programmed death ligand-1 (PD-L1) antibody is a promising treatment strategy in solid tumors. However, in fact, more than half of hepatocellular carcinoma (HCC) patients are unresponsive to PD-L1-based ICB treatment due to multiple immune evasion mechanisms such as the hyperactivation of inflammation pathway, excessive tumor-associated macrophages (TAMs) infiltration, and insufficient infiltration of T cells. Herein, an inflammation-regulated nanodrug was designed to codeliver NF-κB inhibitor curcumin and PD-L1 antibody to reprogram the tumor microenvironment (TME) and activate antitumor immunity. The nanodrug accumulated in TME by an enhanced permeability and retention effect, where it left antibody to block PD-L1 on the membrane of tumor cells and TAMs due to pH-responsiveness. Simultaneously, a new curcumin-encapsulated nanodrug was generated, which was easily absorbed by either tumor cells or TAMs to inhibit the nuclear factor kappa-B (NF-κB) signal and related immunosuppressive genes. The inflammation-regulated nanodrug possessed good biocompatibility. Simultaneously, it reprogrammed TME effectively and exhibited an effective anticancer effect in immunocompetent mice. Overall, this study provided a potent strategy to improve the efficiency of ICB-based treatment for HCC.
Collapse
Affiliation(s)
- TianCheng Wang
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - MinZhao Lin
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - JunJie Mao
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - LiRong Tian
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - HaiRun Gan
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - XinYan Hu
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - LeYe Yan
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - HaoYu Long
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - JianXun Cai
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - XiaoDi Zheng
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - YuDong Xiao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha 410012, China
| | - Dan Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - XinTao Shuai
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - PengFei Pang
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| |
Collapse
|
37
|
Disulfiram enhances chemotherapeutic effects of doxorubicin liposomes against human hepatocellular carcinoma via activating ROS-induced cell stress response pathways. Cancer Chemother Pharmacol 2022; 90:455-465. [PMID: 36251033 DOI: 10.1007/s00280-022-04481-9] [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: 02/17/2022] [Accepted: 10/05/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Increasing evidences have revealed the anti-cancer effect of disulfiram. Current disulfiram-based cancer therapies still have limitations, such as poor tumor-targeting ability and insufficient studies on anti-tumor mechanisms. METHODS In the present study, tumor-targeting liposomes were prepared as drug carriers to increase retention of disulfiram in tumor cells. Then, anti-tumor efficacy of liposomes and the underlying mechanisms were investigated in in vitro, in vivo, and transcriptomic level. RESULTS The results showed that disulfiram enhanced sensitivity of human hepatocellular carcinoma cells to doxorubicin by 15-27-fold, and increased reactive oxygen species (ROS) production as well as caspase-dependent apoptosis. Inhibition of tumor migration and invasion by doxorubicin were further enhanced by disulfiram. In vivo study showed that disulfiram additive doxorubicin liposomes had better performance in suppressing tumor growth than single doxorubicin liposomes. Gene expression profiling found that cellular components destruction, cell stress, check point regulation, and immunoregulation were the main anti-tumor mechanisms of disulfiram. More importantly, disulfiram possessed a great potential to be a protein ubiquitination and murine double minute 4 (MDM4) targeting compound. CONCLUSIONS Due to its low price and good safety, it is worth to repurposing disulfiram as a chemotherapeutic drug. Furthermore, MDM4 may act as a biomarker for observation the clinical effect of disulfiram-based treatment.
Collapse
|
38
|
Yang F, Wang M, Guan X. Exosomes and mimics as novel delivery platform for cancer therapy. Front Pharmacol 2022; 13:1001417. [PMID: 36313380 PMCID: PMC9602403 DOI: 10.3389/fphar.2022.1001417] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/30/2022] [Indexed: 11/15/2022] Open
Abstract
Exosomes are nano-sized biological extracellular vesicles transmitting information between cells and constituting a new intercellular communication mode. Exosomes have many advantages as an ideal drug delivery nanocarrier, including good biocompatibility, permeability, low toxicity, and low immunogenicity. Recently, exosomes have been used to deliver chemotherapeutic agents, natural drugs, nucleic acid drugs, and other antitumor drugs to treat many types of tumors. Due to the limited production of exosomes, synthetic exosome-mimics have been developed as an ideal platform for drug delivery. This review summarizes recent advances in the application of exosomes and exosome-mimics delivering therapeutic drugs in treating cancers.
Collapse
Affiliation(s)
- Fuxu Yang
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
- Key Laboratory of Pharmaceutics and Bioengineering, School of Medical Technology, Beihua University, Jilin, China
| | - Mingyue Wang
- Key Laboratory of Pharmaceutics and Bioengineering, School of Medical Technology, Beihua University, Jilin, China
- Center of Reproductive Medicine and Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Xingang Guan
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
- *Correspondence: Xingang Guan,
| |
Collapse
|
39
|
Qu X, Liu Z, Li N, Ma B, Zhao H, Li Y, Lei B, Du Y. Biodegradable biocompatible MgO/Eu nanodrug with Acid-Base conversion capacity for targeted lung cancer therapy. CHEMICAL ENGINEERING JOURNAL 2022; 446:136323. [DOI: 10.1016/j.cej.2022.136323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
40
|
Wang YF, Zhou Y, Sun J, Wang X, Jia Y, Ge K, Yan Y, Dawson KA, Guo S, Zhang J, Liang XJ. The Yin and Yang of the protein corona on the delivery journey of nanoparticles. NANO RESEARCH 2022; 16:715-734. [PMID: 36156906 PMCID: PMC9483491 DOI: 10.1007/s12274-022-4849-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 06/12/2023]
Abstract
Nanoparticles-based drug delivery systems have attracted significant attention in biomedical fields because they can deliver loaded cargoes to the target site in a controlled manner. However, tremendous challenges must still be overcome to reach the expected targeting and therapeutic efficacy in vivo. These challenges mainly arise because the interaction between nanoparticles and biological systems is complex and dynamic and is influenced by the physicochemical properties of the nanoparticles and the heterogeneity of biological systems. Importantly, once the nanoparticles are injected into the blood, a protein corona will inevitably form on the surface. The protein corona creates a new biological identity which plays a vital role in mediating the bio-nano interaction and determining the ultimate results. Thus, it is essential to understand how the protein corona affects the delivery journey of nanoparticles in vivo and what we can do to exploit the protein corona for better delivery efficiency. In this review, we first summarize the fundamental impact of the protein corona on the delivery journey of nanoparticles. Next, we emphasize the strategies that have been developed for tailoring and exploiting the protein corona to improve the transportation behavior of nanoparticles in vivo. Finally, we highlight what we need to do as a next step towards better understanding and exploitation of the protein corona. We hope these insights into the "Yin and Yang" effect of the protein corona will have profound implications for understanding the role of the protein corona in a wide range of nanoparticles.
Collapse
Affiliation(s)
- Yi-Feng Wang
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260 China
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190 China
| | - Yaxin Zhou
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - JiaBei Sun
- China National Institute of Food and Drug Control, Beijing, 100061 China
| | - Xiaotong Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| | - Yaru Jia
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| | - Kun Ge
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| | - Yan Yan
- Centre for BioNano Interactions, School of Chemistry, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, D04V1W8 Ireland
| | - Kenneth A. Dawson
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260 China
- Centre for BioNano Interactions, School of Chemistry, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, D04V1W8 Ireland
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| | - Xing-Jie Liang
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260 China
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| |
Collapse
|
41
|
Nanoparticles-Based Strategies to Improve the Delivery of Therapeutic Small Interfering RNA in Precision Oncology. Pharmaceutics 2022; 14:pharmaceutics14081586. [PMID: 36015212 PMCID: PMC9415718 DOI: 10.3390/pharmaceutics14081586] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/14/2022] [Accepted: 07/23/2022] [Indexed: 02/07/2023] Open
Abstract
Small interfering RNA (siRNA) can selectively suppress the expression of disease-causing genes, holding great promise in the treatment of human diseases, including malignant cancers. In recent years, with the development of chemical modification and delivery technology, several siRNA-based therapeutic drugs have been approved for the treatment of non-cancerous liver diseases. Nevertheless, the clinical development of siRNA-based cancer therapeutics remains a major translational challenge. The main obstacles of siRNA therapeutics in oncology include both extracellular and intracellular barriers, such as instability under physiological conditions, insufficient tumor targeting and permeability (particularly for extrahepatic tumors), off-target effects, poor cellular uptake, and inefficient endosomal escape. The development of clinically suitable and effective siRNA delivery systems is expected to overcome these challenges. Herein, we mainly discuss recent strategies to improve the delivery and efficacy of therapeutic siRNA in cancer, including the application of non-viral nanoparticle-based carriers, the selection of target genes for therapeutic silencing, and the combination with other therapeutic modalities. In addition, we also provide an outlook on the ongoing challenges and possible future developments of siRNA-based cancer therapeutics during clinical translation.
Collapse
|
42
|
Shakib Z, Mahmoudi A, Moosavian SA, Malaekeh-Nikouei B. PEGylated solid lipid nanoparticles functionalized by aptamer for targeted delivery of docetaxel in mice bearing C26 tumor. Drug Dev Ind Pharm 2022; 48:69-78. [PMID: 35758194 DOI: 10.1080/03639045.2022.2095398] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Colorectal cancer is one of the most deadly cancers in the world. Docetaxel (DTX) is a potentially important chemotherapeutic agent for the treatment of cancer. Many studies have attempted to improve its bioavailability and efficiency using different nanoparticulate drug delivery systems. SIGNIFICANCE In the current study, PEGylated solid lipid nanoparticles (SLNs) containing DTX were prepared and modified with AS1411 anti-nucleolin aptamers to target nucleoin receptors on colorectal cancer cells. METHODS Nanoparticles were characterized and the morphology was evaluated. In vitro studies were investigated on murine colon carcinoma (C26) and Chinese hamster ovary (CHO) cell lines. Then in vivo antitumor efficacy and survival analysis were evaluated in mice bearing the C26 tumor model. RESULTS Results showed 135-140 nm particle size and about 78% DTX entrapment efficiency for actively targeted samples. PEGylated and aptamer-targeted SLNs containing DTX had the lowest IC50 (0.28 and 0.11 nM for 3 and 6 h incubation respectively) and higher cellular uptake values in the C26 cell line. Also in vivo results demonstrated that PEGylated and aptamer-targeted SLNs containing Docetaxel (Apt-PEG-SLN-DTX) improved antitumor activity and inhibited tumor growth in C26 tumor-bearing mice. CONCLUSION These results suggested that PEGylated and aptamer-targeted SLNs containing DTX exhibited efficient characteristics in tumor inhibitory against murine C26 carcinoma model.
Collapse
Affiliation(s)
- Zahra Shakib
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Mahmoudi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Alia Moosavian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bizhan Malaekeh-Nikouei
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
43
|
Polyketal-based nanocarriers: A new class of stimuli-responsive delivery systems for therapeutic applications. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
44
|
Shinde VR, Revi N, Murugappan S, Singh SP, Rengan AK. Enhanced Permeability and Retention Effect: A key facilitator for solid tumor targeting by nanoparticles. Photodiagnosis Photodyn Ther 2022; 39:102915. [PMID: 35597441 DOI: 10.1016/j.pdpdt.2022.102915] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 12/14/2022]
Abstract
Exploring the enhanced permeability and retention (EPR) effect through therapeutic nanoparticles has been a subject of considerable interest in tumor biology. This passive targeting based phenomenon exploits the leaky blood vasculature and the defective lymphatic drainage system of the heterogeneous tumor microenvironment resulting in enhanced preferential accumulation of the nanoparticles within the tumor tissues. This article reviews the fundamental studies to assess how the EPR effect plays an essential role in passive targeting. Further, it summarizes various therapeutic modalities of nanoformulation including chemo-photodynamic therapy, intravascular drug release, and photothermal immunotherapy to combat cancer using enhanced EPR effect in neoplasia region.
Collapse
Affiliation(s)
- Vinod Ravasaheb Shinde
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Neeraja Revi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | | | - Surya Prakash Singh
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India.
| |
Collapse
|
45
|
Wu Y, Wu H, Lu X, Chen Y, Zhang X, Ju J, Zhang D, Zhu B, Huang S. Development and Evaluation of Targeted Optical Imaging Probes for Image‐Guided Surgery in Head and Neck Cancer. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yue Wu
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Xiaoya Lu
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Yi Chen
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Xue Zhang
- University of Jinan Jinan Shandong 250021 China
| | - Jiandong Ju
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Baocun Zhu
- University of Jinan Jinan Shandong 250021 China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| |
Collapse
|
46
|
The Pyrazolo[3,4-d]Pyrimidine Derivative Si306 Encapsulated into Anti-GD2-Immunoliposomes as Therapeutic Treatment of Neuroblastoma. Biomedicines 2022; 10:biomedicines10030659. [PMID: 35327462 PMCID: PMC8945814 DOI: 10.3390/biomedicines10030659] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 12/03/2022] Open
Abstract
Si306, a pyrazolo[3,4-d]pyrimidine derivative recently identified as promising anticancer agent, has shown favorable in vitro and in vivo activity profile against neuroblastoma (NB) models by acting as a competitive inhibitor of c-Src tyrosine kinase. Nevertheless, Si306 antitumor activity is associated with sub-optimal aqueous solubility, which might hinder its further development. Drug delivery systems were here developed with the aim to overcome this limitation, obtaining suitable formulations for more efficacious in vivo use. Si306 was encapsulated in pegylated stealth liposomes, undecorated or decorated with a monoclonal antibody able to specifically recognize and bind to the disialoganglioside GD2 expressed by NB cells (LP[Si306] and GD2-LP[Si306], respectively). Both liposomes possessed excellent morphological and physio-chemical properties, maintained over a period of two weeks. Compared to LP[Si306], GD2-LP[Si306] showed in vitro specific cellular targeting and increased cytotoxic activity against NB cell lines. After intravenous injection in healthy mice, pharmacokinetic profiles showed increased plasma exposure of Si306 when delivered by both liposomal formulations, compared to that obtained when Si306 was administered as free form. In vivo tumor homing and cytotoxic effectiveness of both liposomal formulations were finally tested in an orthotopic animal model of NB. Si306 tumor uptake resulted significantly higher when encapsulated in GD2-LP, compared to Si306, either free or encapsulated into untargeted LP. This, in turn, led to a significant increase in survival of mice treated with GD2-LP[Si306]. These results demonstrate a promising antitumor efficacy of Si306 encapsulated into GD2-targeted liposomes, supporting further therapeutic developments in pre-clinical trials and in the clinic for NB.
Collapse
|
47
|
Saify Nabiabad H, Amini M, Demirdas S. Specific delivering of RNAi using Spike's aptamer-functionalized lipid nanoparticles for targeting SARS-CoV-2: A strong anti-Covid drug in a clinical case study. Chem Biol Drug Des 2022; 99:233-246. [PMID: 34714580 PMCID: PMC8653378 DOI: 10.1111/cbdd.13978] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/24/2021] [Accepted: 10/24/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus (SARS-CoV-2) as a global pandemic has attracted the attention of many scientific centers to find the right treatment. We expressed and purified the recombinant receptor-binding domain (RBD) of the SARS-CoV-2 spike (S) protein, and specific RBD aptamers were designed using SELEX method. RNAi targeting nucleocapsid phosphoprotein was synthesized and human lung cells were inoculated with aptamer-functionalized lipid nanoparticles (LNPs) containing RNAi. The results demonstrated that RBD aptamer having KD values of 0.290 nm possessed good affinity. Based on molecular docking and efficacy prediction analysis, siRNA molecule was showed the best action. LNPs were appropriately functionalized by aptamer and contained RNAi molecules. Antiviral assay using q-PCR and ELISA demonstrated that LNP functionalized with 35 µm Apt and containing 30 nm RNAi/ml of cell culture had the best antiviral activity compared to other concentrations. Applied aptamer in the nanocarrier has two important functions. First, it can deliver the drug (RNAi) to the surface of epithelial cells. Second, by binding to the SARS-CoV-2 spike protein, it inhibits the virus entrance into cells. Our data reveal an interaction between the aptamer and the virus, and RNAi targeted the virus RNA. CT scan and the clinical laboratory tests in a clinical case study, a 36-year old man who presented with severe SARS-CoV-2, demonstrated that inhalation of 10 mg Apt-LNPs-RNAi nebulized/day for six days resulted in an improvement in consolidation and ground-glass opacity in lungs on the sixth day of treatment. Our findings suggest the treatment of SARS-CoV-2 infection through inhalation of Aptamer-LNPs-RNAi.
Collapse
Affiliation(s)
| | - Massoume Amini
- Department of BiotechnologyBu‐Ali Sina UniversityHamadanIran
| | - Serwet Demirdas
- Department of Clinical GeneticsErasmus Medical CentreRotterdamthe Netherlands
| |
Collapse
|
48
|
Koksharov YA, Gubin SP, Taranov IV, Khomutov GB, Gulyaev YV. Magnetic Nanoparticles in Medicine: Progress, Problems, and Advances. JOURNAL OF COMMUNICATIONS TECHNOLOGY AND ELECTRONICS 2022; 67:101-116. [PMCID: PMC8988108 DOI: 10.1134/s1064226922020073] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 10/27/2023]
Abstract
The review presents an analysis of the current state of research related to the design, development, and practical application of methods for biomedical radioelectronics and nanomedicine, including the use of magnetic nanoparticles. The important role of rational scientific physical approaches and experimental methods in the design of efficient and safe magnetic nanoparticle-based agents for therapy, controlled targeted drug delivery, and diagnostics, including spatial imaging, is emphasized. Examples of successful practical application of magnetic nanoparticles in medicine based on these methods are given, and an analysis of the main problems and prospects of this area of science is conducted.
Collapse
Affiliation(s)
- Yu. A. Koksharov
- Moscow State University, 119991 Moscow, Russia
- Kotelnikov Institute of Radioengineering and Electronics, Russian Academy of Sciences, 125009 Moscow, Russia
| | - S. P. Gubin
- Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
| | - I. V. Taranov
- Kotelnikov Institute of Radioengineering and Electronics, Russian Academy of Sciences, 125009 Moscow, Russia
| | - G. B. Khomutov
- Moscow State University, 119991 Moscow, Russia
- Kotelnikov Institute of Radioengineering and Electronics, Russian Academy of Sciences, 125009 Moscow, Russia
| | - Yu. V. Gulyaev
- Kotelnikov Institute of Radioengineering and Electronics, Russian Academy of Sciences, 125009 Moscow, Russia
| |
Collapse
|
49
|
Fobian SF, Cheng Z, ten Hagen TLM. Smart Lipid-Based Nanosystems for Therapeutic Immune Induction against Cancers: Perspectives and Outlooks. Pharmaceutics 2021; 14:26. [PMID: 35056922 PMCID: PMC8779430 DOI: 10.3390/pharmaceutics14010026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy, a promising and widely applied mode of oncotherapy, makes use of immune stimulants and modulators to overcome the immune dysregulation present in cancer, and leverage the host's immune capacity to eliminate tumors. Although some success has been seen in this field, toxicity and weak immune induction remain challenges. Liposomal nanosystems, previously used as targeting agents, are increasingly functioning as immunotherapeutic vehicles, with potential for delivery of contents, immune induction, and synergistic drug packaging. These systems are tailorable, multifunctional, and smart. Liposomes may deliver various immune reagents including cytokines, specific T-cell receptors, antibody fragments, and immune checkpoint inhibitors, and also present a promising platform upon which personalized medicine approaches can be built, especially with preclinical and clinical potentials of liposomes often being frustrated by inter- and intrapatient variation. In this review, we show the potential of liposomes in cancer immunotherapy, as well as the methods for synthesis and in vivo progression thereof. Both preclinical and clinical studies are included to comprehensively illuminate prospects and challenges for future research and application.
Collapse
Affiliation(s)
| | | | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology (LEO), Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (S.-F.F.); (Z.C.)
| |
Collapse
|
50
|
Design and synthesis of targeted star-shaped micelle for guided delivery of camptothecin: In vitro and in vivo evaluation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112529. [PMID: 34857308 DOI: 10.1016/j.msec.2021.112529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
This study aimed to synthesize a star-shaped micelle using 3-azido-2,2-bis(azidomethyl)propan-1-ol (pentaerythritol triazide) core, as an initiator for the synthesis of three-arm polylactic acid (PLA) block. Then, the ends of the PLA arms were converted to PLA triazide followed by conjugation to the three alkyne-PEG-maleamide through click reaction. The maleamide ends were available for coupling with sulfhydryl-modified DNA aptamer against epithelial cell adhesion molecule in order to offer targeted delivery of encapsulated drug, camptothecin to the site of action. The successful synthesis of the star-shaped polymers was confirmed via1HNMR. Hydrophobic anti-cancer drug, camptothecin was encapsulated into the micelles core implementing solvent switching method providing loading content (LC%) and encapsulation efficiency (EE%) of 3.7 ± 0.4 and 73.7 ± 8.2, respectively. The size of both non-targeted and aptamer-targeted micelles was determined to be 154 and 192 nm, respectively with polydispersity index below 0.3. In vitro drug release evaluation at 37 °C, pH 7.4 showed a controlled release pattern for camptothecin during 72 h. In vitro cytotoxicity of the prepared non-targeted and targeted micelles was carried out on human colorectal adenocarcinoma (HT29) and mouse colon carcinoma (C26) as EpCAM positive cell lines and Chinese hamster ovary (CHO) as EpCAM negative cell line. The results verified significantly higher cytotoxicity of the targeted micelles on HT29 and C26 cell lines, while no obvious difference was observed between targeted and non-targeted formulation on CHO cell line. The in vivo therapeutic efficiency investigation on BALB/c C26 tumor-bearing mice showed superior capability of the targeted formulation on tumor suppression and survival rate of the treated mice. The developed platform exhibited excellent characteristics to diminish camptothecin drawbacks and its adverse effects while considerably increasing its therapeutic index.
Collapse
|