1
|
Chen KC, Yang SJ, Yang SH, Pai JA, Shieh MJ. Hyaluronan-coated gold nanoshells for enhanced synergistic effect and immunogenic cell response of chemo-photothermal therapy on lung cancer. Int J Biol Macromol 2025; 300:140114. [PMID: 39837437 DOI: 10.1016/j.ijbiomac.2025.140114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Lung cancer (LC) is the predominant cause of cancer-related fatalities globally, with the highest death rates in both genders, primarily attributed to smoking. The non-kinase transmembrane cell surface glycoprotein, CD44, enhances LC cell migration and invasion, leading to drug resistance and an unfavorable prognosis. This research formulated a cisplatin-loaded gold nanoshell (HCP@GNS) integrated with hyaluronan (HCP@GNS@HA) to enhance targeting capability and realize a synergistic effect of chemo-photothermal therapy (chemo-PTT) against LC. The coating of hyaluronic acid (HA) facilitated the uptake of HCP@GNS@HA into CD44-rich cancer cells, maintaining the superior photothermal conversion capacity of HCP@GNS nanoparticles for hyperthermia and photothermal eradication of tumor tissues under near-infrared exposure. As a nanocarrier, HCP@GNS@HA exhibited high biocompatibility and hemocompatibility, showing stronger cytotoxicity than either the free drug or photothermal therapy alone when combined with NIR irradiation, especially at high cis-diamminedichloroplatinum (II) (CDDP) concentrations. The chemo-PTT mediated by HCP@GNS@HA effectively curtailed tumor growth without adverse effects, significantly mobilizing B cells, DC cells, macrophages, natural killer (NK) cells, and NKT cells in the distal tumor against tumor growth. In conclusion, the developed hyaluronic acid (HA)-coated gold nanoshells could potentially serve as a promising candidate for nanomedicine, tackling both primary and distal LC growth.
Collapse
Affiliation(s)
- Ke-Cheng Chen
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan; Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
| | - Shu-Jyuan Yang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan.
| | - Shih-Hung Yang
- Department of Oncology, National Taiwan University Hospital and College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
| | - Jui-An Pai
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan; Department of Oncology, National Taiwan University Hospital and College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan.
| |
Collapse
|
2
|
Dang Z, Zheng X, Gao Y, Du Y, Zhang Y, Zhu S. In situ albumin tagging for targeted imaging of endothelial barrier disruption. SCIENCE ADVANCES 2025; 11:eads4412. [PMID: 39951533 PMCID: PMC11827639 DOI: 10.1126/sciadv.ads4412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/14/2025] [Indexed: 02/16/2025]
Abstract
The endothelial barrier (EB) is a critical component of the body's homeostatic mechanisms, thus developing effective imaging techniques to visualize its integrity is essential. The EB disruption is accompanied by the alternations in permeability and even the breakdown of tight junctions (TJs), leading to the leakage of albumin; thus, albumin can serve as a biomarker for EB disruption. Herein, we develop an albumin-specific, covalently tagged near-infrared II (NIR-II) dye, with its high selectivity for endogenous albumin, for targeted imaging EB disruption. Our albumin-tagging dye serves as a chromophore to construct NIR-II fluorescent proteins in situ, with substantially improved brightness. Thus, through in situ dye tagging of endogenous albumin as the efficient "targeting agent," we can precisely image disruptions in various endothelial barriers. Unlike the traditional exogenous targeting agents (e.g., dye-labeled antibodies) with enzymatic degradation or immune system capture issues, in situ albumin tagging demonstrates superhigh performance for targeted imaging.
Collapse
Affiliation(s)
- Zetao Dang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Xue Zheng
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Yanli Gao
- Department of Pediatric Ultrasound, Ultrasound Diagnostic Center, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Yijing Du
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Yuewei Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P.R. China
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun 130021, P.R. China
| |
Collapse
|
3
|
Bordes A, Opalinski I, Thoreau F, Provot O, Hamze A, Alami M, Papot S. β-Glucuronidase-Responsive Albumin-Binding Prodrug of Colchicine-Site Binders for Selective Cancer Therapy. ChemMedChem 2025:e202400969. [PMID: 39836075 DOI: 10.1002/cmdc.202400969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 01/22/2025]
Abstract
The development of novel therapeutic strategies enabling the selective destruction of tumors while sparing healthy tissues is of great interest to improve the efficacy of cancer chemotherapy. In this context, we designed a β-glucuronidase-responsive albumin-binding prodrug programmed to release a potent Isocombretastatin A-4 analog within the tumor microenvironment. When injected at a non-toxic dose in mice bearing orthotopic triple-negative mammary tumors, this prodrug produced a significant anticancer activity, therefore offering a valuable alternative to the systemic administration of the parent drug.
Collapse
Affiliation(s)
- Alexandra Bordes
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)., Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073, Poitiers cedex 9, France
| | - Isabelle Opalinski
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)., Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073, Poitiers cedex 9, France
| | - Fabien Thoreau
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)., Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073, Poitiers cedex 9, France
| | - Olivier Provot
- CNRS, BioCIS, Université Paris-Saclay, 17 avenue des Sciences, 86073 91400, Orsay, France
| | - Abdallah Hamze
- CNRS, BioCIS, Université Paris-Saclay, 17 avenue des Sciences, 86073 91400, Orsay, France
| | - Mouad Alami
- CNRS, BioCIS, Université Paris-Saclay, 17 avenue des Sciences, 86073 91400, Orsay, France
| | - Sébastien Papot
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)., Equipe Labellisée Ligue Contre le Cancer, 4 rue Michel-Brunet, TSA 51106, 86073, Poitiers cedex 9, France
| |
Collapse
|
4
|
Pivarcsik T, Tóth S, Pósa SP, May NV, Kováts É, Spengler G, Kántor I, Rolya A, Feczkó T, Szatmári I, Szakács G, Enyedy ÉA. Organometallic Half-Sandwich Complexes of 8-Hydroxyquinoline-Derived Mannich Bases with Enhanced Solubility: Targeting Multidrug Resistant Cancer. Inorg Chem 2024; 63:23983-23998. [PMID: 39638428 DOI: 10.1021/acs.inorgchem.4c04398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Drug resistance is a major obstacle in cancer treatment. Herein, four novel organometallic complexes, with the general formula [Ru(η6-p-cymene)(HL)Cl]Cl and [Rh(η5-C5Me5)(HL)Cl]Cl, were developed to target multidrug-resistant (MDR) cancer cells, where HL denotes 8-hydroxyquinoline-derived Mannich bases (HQCl-pyr and HQCl-pip). The aim of the complexation was to obtain compounds with improved drug-like properties. The complexes were comprehensively characterized by various spectroscopic methods in terms of their structure, solution speciation and interaction with human serum albumin. The structure of [Rh(η5-C5Me5)(HQCl-pip)Cl]Cl was analyzed by X-ray crystallography. The complexes were found to be highly stable in solution and in various biological matrices, showing enhanced solubility compared with the ligands and significant binding ability to albumin via coordination. The Rh(η5-C5Me5) complexes exhibited strong cytotoxicity against MDR MES-SA/Dx5 cell lines (IC50 = 0.19 and 0.22 μM), demonstrating high MDR-selectivity. Ganglioside-functionalized nanoparticles with the most promising ligand HQCl-pip and its Rh(η5-C5Me5) complex were prepared to enhance the bioavailability. The nanocarriers showed faster drug release at acidic pH than at pH 7.4, and could retain the cytotoxicity and selectivity of the loaded compounds. The encapsulated Rh(η5-C5Me5) complex of HQCl-pip has been identified as an optimal candidate for the pharmacological development of MDR-selective compounds.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7-8, Szeged H-6720 , Hungary
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
| | - Szilárd Tóth
- Drug Resistance Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok krt. 2, Budapest H-1117, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, Budapest H-1117 , Hungary
| | - Szonja P Pósa
- Drug Resistance Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok krt. 2, Budapest H-1117, Hungary
| | - Nóra V May
- Centre for Structural Science, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok krt. 2, Budapest H-1117, Hungary
| | - Éva Kováts
- Institute for Solid State Physics and Optics, HUN-REN Wigner Research Centre for Physics, P.O. Box 49, Budapest H-1525, Hungary
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7-8, Szeged H-6720 , Hungary
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, Szeged H-6725, Hungary
| | - Izolda Kántor
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok krt. 2, Budapest H-1117, Hungary
| | - Alexandra Rolya
- Faculty of Engineering, University of Pannonia, Egyetem u. 10, Veszprém H-8200, Hungary
| | - Tivadar Feczkó
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok krt. 2, Budapest H-1117, Hungary
- Faculty of Engineering, University of Pannonia, Egyetem u. 10, Veszprém H-8200, Hungary
| | - István Szatmári
- Institute of Pharmaceutical Chemistry, HUN-REN-SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, Szeged H-6720, Hungary
| | - Gergely Szakács
- Drug Resistance Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok krt. 2, Budapest H-1117, Hungary
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, Wien, Vienna A-1090 Austria
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7-8, Szeged H-6720 , Hungary
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
| |
Collapse
|
5
|
Kiarashi M, Yasamineh S. Albumin nanoparticles are a promising drug delivery system in dentistry. Biomed Eng Online 2024; 23:122. [PMID: 39605007 PMCID: PMC11600845 DOI: 10.1186/s12938-024-01318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Periodontal infection is a long-lasting inflammatory condition caused by the growth and development of an abnormal and harmful community of microorganisms. This destructive illness leads to the loss of the tissues that support the teeth, degradation of the bone surrounding the teeth, and eventually tooth loss. To treat oral infections, it is necessary to use nonsurgical methods such as antibiotics. However, the indiscriminate and incorrect use of antibiotics results in drug resistance. Among these alternate therapeutic options, using nanoparticles to treat infectious dental disease was particularly significant. Consequently, researchers have worked to develop an effective and satisfactory drug delivery method for treating periodontal and dental illnesses. Albumin nanoparticles serve a considerable function as carriers in the drug delivery of chemical and biomolecular medications, such as anticancer treatments; they have several advantages, including biocompatibility and biodegradability, and they are well-tolerated with no adverse effects. Albumin nanoparticles have several benefits over other nanomaterials. Protein nanocarriers provide advantages such as biocompatibility, biodegradability, reduced immunogenicity, and lower cytotoxicity. Furthermore, this nanoparticle demonstrated significant intrinsic antibacterial properties without being loaded with antibiotic medicines. As a medication and antibacterial nanoparticle delivery method, albumin nanoparticles have substantial applications in periodontal and dental infectious disorders such as periodontal infection, apical periodontitis, and peri-implantitis. As a result, in this article, we studied the usage of albumin nanoparticles in dental disorders.
Collapse
Affiliation(s)
- Mohammad Kiarashi
- College of Dentistry, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| |
Collapse
|
6
|
Lhaglham P, Jiramonai L, Jia Y, Huang B, Huang Y, Gao X, Zhang J, Liang XJ, Zhu M. Drug nanocrystals: Surface engineering and its applications in targeted delivery. iScience 2024; 27:111185. [PMID: 39555405 PMCID: PMC11564948 DOI: 10.1016/j.isci.2024.111185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Drug nanocrystals have received significant attention in drug development due to their enhanced dissolution rate and improved water solubility, making them effective in overcoming issues related to drug hydrophobicity, thereby improving drug bioavailability and treatment effectiveness. Recent advances in preparation techniques have facilitated research on drug surface properties, leading to valuable surface engineering strategies. Surface modification can stabilize drug nanocrystals, making them suitable for versatile drug delivery platforms. Functionalized ligands further enhance the potential for targeted delivery, enabling precision medicine. This review focuses on the surface engineering of drug nanocrystals, discussing various preparation methods, surface ligand design strategies, and their applications in targeted drug delivery, especially for cancer treatments. Finally, challenges and future directions are also discussed to promote the development of drug nanocrystals. The surface engineering of drug nanocrystals promises new opportunities for treating complex and chronic diseases while broadening the application of drug delivery systems.
Collapse
Affiliation(s)
- Phattalapol Lhaglham
- 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
- University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Manufacturing Pharmacy, Faculty of Pharmacy, Mahidol University, Sri-ayudhya Road, Bangkok 10400, Thailand
| | - Luksika Jiramonai
- 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
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaru Jia
- 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, College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Baoying Huang
- MHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, China CDC, Beijing 102206, China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xueyun Gao
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Xing-Jie Liang
- 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
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengliang Zhu
- 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
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
7
|
Zhang SH, Zeng N, Xu JZ, Liu CQ, Xu MY, Sun JX, An Y, Zhong XY, Miao LT, Wang SG, Xia QD. Recent breakthroughs in innovative elements, multidimensional enhancements, derived technologies, and novel applications of PROTACs. Biomed Pharmacother 2024; 180:117584. [PMID: 39427546 DOI: 10.1016/j.biopha.2024.117584] [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: 08/06/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Proteolysis Targeting Chimera (PROTAC) is an emerging and evolving technology based on targeted protein degradation (TPD). Small molecule PROTACs have shown great efficacy in degrading disease-specific proteins in preclinical and clinical studies, but also showed various limitations. In recent years, new technologies and advances in TPD have provided additional optimized strategies based on conventional PROTACs that can overcome the shortcomings of conventional PROTACs in terms of undruggable targets, bioavailability, tissue-specificity, spatiotemporal control, and degradation scope. In addition, some designs of special targeting chimeras and applications based on multidisciplinary science have shed light on novel therapeutic modalities and drug design. However, each improvement has its own advantages, disadvantages and application conditions. In this review, we summarize the exploration of PROTAC elements, depict a landscape of improvements and derived concepts of PROTACs, and expect to provide perspectives for technological innovations, combinations and applications in future targeting chimera design.
Collapse
Affiliation(s)
- Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Na Zeng
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Xing-Yu Zhong
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Lin-Tao Miao
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
8
|
Vander Elst Z, Laenen A, Deberdt J, Delemarre L, Vermeersch P, Frans G, Naulaers G, Gijsen M, Dreesen E, Spriet I, Allegaert K, Smits A. Human serum albumin: prediction model and reference values for preterm and term neonates. Pediatr Res 2024:10.1038/s41390-024-03634-1. [PMID: 39394426 DOI: 10.1038/s41390-024-03634-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Human serum albumin (HSA) concentrations may alter HSA-bound drug distribution. This study aims to describe longitudinal real-world HSA trends, and to develop a prediction model for HSA concentrations using a large neonatal cohort. METHODS Patients admitted to the neonatal intensive care unit of the University Hospitals Leuven (postnatal age (PNA) ≤28days) were retrospectively included. Using linear mixed models, covariate effects on HSA were explored. A multivariable prediction model was developed (backward model selection procedure, 1% significance level). RESULTS In total, 848 neonates were included [median(interquartile range) gestational age (GA) 35(32-38)weeks, birth weight (BW) 2400(1640-3130)grams]. Median HSA concentration was 32.3(28.7-35.6)g/L. Longitudinal analyses demonstrated increasing HSA concentrations with PNA and GA for most GA groups. Univariable analyses revealed significant associations of HSA with PNA, GA, BW, current weight, total and direct bilirubin, total plasma proteins, respiratory support, mechanical ventilation, sepsis, ibuprofen use, and C-reactive protein (p-values < 0.05). A high-performance (R2 = 76.3%) multivariable HSA prediction model was developed, and PNA- and GA-dependent HSA centiles were provided. CONCLUSION Population-specific HSA centiles and an accurate neonatal HSA prediction model were developed, incorporating both maturational and non-maturational covariates. These results can enhance future clinical care and pharmacokinetic analyses to improve pharmacotherapy of HSA-bound drugs in neonates, respectively. IMPACT To improve future pharmacokinetic modeling initiatives, a high-performance human serum albumin (HSA) prediction model was developed for (pre)term neonates, using a large, single-center cohort of real-world data. This prediction model integrates both maturational and non-maturational covariates, resulting in accurate HSA predictions in neonates. Additionally, HSA centiles based on postnatal and gestational age were developed, which can be easily applied in clinical practice when interpreting HSA concentrations of neonates. In general, unbound drug fractions are higher in neonates compared to older populations. To improve pharmacotherapy of HSA-bound drugs in neonates, the obtained results can be integrated in future pharmacokinetic-pharmacodynamic analyses.
Collapse
Affiliation(s)
- Zoë Vander Elst
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Annouschka Laenen
- Leuven Biostatistics and Statistical Bioinformatics Centre (L-Biostat), KU Leuven, Leuven, Belgium
| | | | | | - Pieter Vermeersch
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Glynis Frans
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Gunnar Naulaers
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Matthias Gijsen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Pharmacy Department, UZ Leuven, Leuven, Belgium
| | - Erwin Dreesen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Isabel Spriet
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Pharmacy Department, UZ Leuven, Leuven, Belgium
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Chen Y, Liu F, Pal S, Hu Q. Proteolysis-targeting drug delivery system (ProDDS): integrating targeted protein degradation concepts into formulation design. Chem Soc Rev 2024; 53:9582-9608. [PMID: 39171633 DOI: 10.1039/d4cs00411f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a revolutionary paradigm in drug discovery and development, offering a promising avenue to tackle challenging therapeutic targets. Unlike traditional drug discovery approaches that focus on inhibiting protein function, TPD aims to eliminate proteins of interest (POIs) using modular chimeric structures. This is achieved through the utilization of proteolysis-targeting chimeras (PROTACs), which redirect POIs to E3 ubiquitin ligases, rendering them for degradation by the cellular ubiquitin-proteasome system (UPS). Additionally, other TPD technologies such as lysosome-targeting chimeras (LYTACs) and autophagy-based protein degraders facilitate the transportation of proteins to endo-lysosomal or autophagy-lysosomal pathways for degradation, respectively. Despite significant growth in preclinical TPD research, many chimeras fail to progress beyond this stage in the drug development. Various factors contribute to the limited success of TPD agents, including a significant hurdle of inadequate delivery to the target site. Integrating TPD into delivery platforms could surmount the challenges of in vivo applications of TPD strategies by reshaping their pharmacokinetics and pharmacodynamic profiles. These proteolysis-targeting drug delivery systems (ProDDSs) exhibit superior delivery performance, enhanced targetability, and reduced off-tissue side effects. In this review, we will survey the latest progress in TPD-inspired drug delivery systems, highlight the importance of introducing delivery ideas or technologies to the development of protein degraders, outline design principles of protein degrader-inspired delivery systems, discuss the current challenges, and provide an outlook on future opportunities in this field.
Collapse
Affiliation(s)
- Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fengyuan Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Samira Pal
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
10
|
Shastri D, Raj V, Lee S. Revolutionizing Alzheimer's treatment: Harnessing human serum albumin for targeted drug delivery and therapy advancements. Ageing Res Rev 2024; 99:102379. [PMID: 38901740 DOI: 10.1016/j.arr.2024.102379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder initiated by amyloid-beta (Aβ) accumulation, leading to impaired cognitive function. Several delivery approaches have been improved for AD management. Among them, human serum albumin (HSA) is broadly employed for drug delivery and targeting the Aβ in AD owing to its biocompatibility, Aβ inhibitory effect, and nanoform, which showed blood-brain barrier (BBB) crossing ability via glycoprotein 60 (gp60) receptor and secreted protein acidic and rich in cysteine (SPARC) protein to transfer the drug molecules in the brain. Thus far, there is no previous review focusing on HSA and its drug delivery system in AD. Hence, the reviewed article aimed to critically compile the HSA therapeutic as well as drug delivery role in AD management. It also delivers information on how HSA-incorporated nanoparticles with surfaced embedded ligands such as TAT, GM1, and so on, not only improve BBB permeability but also increase neuron cell targetability in AD brain. Additionally, Aβ and tau pathology, including various metabolic markers likely BACE1 and BACE2, etc., are discussed. Besides, the molecular interaction of HSA with Aβ and its distinctive forms are critically reviewed that HSA can segregate Zn(II) and Cu(II) metal ions from Aβ owing to high affinity. Furthermore, the BBB drug delivery challenges in AD are addressed. Finally, the clinical formulation of HSA for the management of AD is critically discussed on how the HSA inhibits Aβ oligomer and fibril, while glycated HSA participates in amyloid plaque formation, i.e., β-structure sheet formation. This review report provides theoretical background on HSA-based AD drug delivery and makes suggestions for future prospect-related work.
Collapse
Affiliation(s)
- Divya Shastri
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, the Republic of Korea; College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, the Republic of Korea
| | - Vinit Raj
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, the Republic of Korea.
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, the Republic of Korea.
| |
Collapse
|
11
|
Liu Y, Li Y, Shen W, Li M, Wang W, Jin X. Trend of albumin nanoparticles in oncology: a bibliometric analysis of research progress and prospects. Front Pharmacol 2024; 15:1409163. [PMID: 39070787 PMCID: PMC11272567 DOI: 10.3389/fphar.2024.1409163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Background Delivery systems based on albumin nanoparticles (NPs) have recently garnered substantial interest in anti-tumor drug development. However, systematic bibliometric analyses in this field remain lacking. This study aimed to analyze the current research status, hotspots, and frontiers in the application of albumin NPs in the field of oncology from a bibliometric perspective. Methods Using the Web of Science Core Collection (WOSCC) as the data source, retrieved articles were analyzed using software, such as VOSviewer 1.6.18 and CiteSpace 6.1.6, and the relevant visualization maps were plotted. Results From 1 January 2000, to 15 April 2024, 2,262 institutions from 67 countries/regions published 1,624 articles related to the application of albumin NPs in the field of oncology. The USA was a leader in this field and held a formidable academic reputation. The most productive institution was the Chinese Academy of Sciences. The most productive author was Youn YS, whereas Kratz F was the most frequently co-cited author. The most productive journal was the International Journal of Nanomedicine, whereas the Journal of Controlled Release was the most co-cited journal. Future research hotspots and frontiers included "rapid and convenient synthesis methods predominated by self-assembly," "surface modification," "construction of multifunctional NPs for theranostics," "research on natural active ingredients mainly based on phenolic compounds," "combination therapy," and "clinical applications." Conclusion Based on our bibliometric analysis and summary, we obtained an overview of the research on albumin NPs in the field of oncology, identified the most influential countries, institutions, authors, journals, and citations, and discussed the current research hotspots and frontiers in this field. Our study may serve as an important reference for future research in this field.
Collapse
Affiliation(s)
- Ye Liu
- Department of Pharmacy, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yi Li
- Department of Pharmacy, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Wei Shen
- Department of Pharmacy, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Min Li
- Department of Pharmacy, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Wen Wang
- Department of Rheumatology and Immunology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Xin Jin
- Department of Pharmacy, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Ray D, Chamlagai D, Kumar S, Mukhopadhyay S, Chakrabarty S, Aswal VK, Mitra S. Molecular Insights into the Conformational and Binding Behaviors of Human Serum Albumin Induced by Surface-Active Ionic Liquids. J Phys Chem B 2024; 128:6622-6637. [PMID: 38937939 DOI: 10.1021/acs.jpcb.4c01915] [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: 06/29/2024]
Abstract
Extensive research has been carried out to investigate the stability and function of human serum albumin (HSA) when exposed to surface-active ionic liquids (SAILs) with different head groups (imidazolium, morpholinium, and pyridinium) and alkyl chain lengths (ranging from decyl to tetradecyl). Analysis of the protein fluorescence spectra indicates noticeable changes in the secondary structure of HSA with varying concentrations of all SAILs tested. Helicity calculations based on the Fourier transform infrared (FTIR) data show that HSA becomes more organized at the micellar concentration of SAILs, leading to an increased protein activity at this level. Small-angle neutron scattering (SANS) data confirm the formation of a bead-necklace structure between the SAILs and HSA. Atomistic molecular dynamics (MD) simulation results identify several hotspots on the protein surface for interaction with SAIL, which results in the modulation of protein conformational fluctuation and stability. Furthermore, fluorescence resonance energy transfer (FRET) experiments with the intramolecular charge transfer (ICT) probe trans-ethyl p-(dimethylamino) cinnamate (EDAC) demonstrate that higher alkyl chain lengths and SAIL concentrations result in a significantly increased energy transfer efficiency. The findings of this study provide a detailed molecular-level understanding of how the protein structure and function are affected by the presence of SAILs, with potential implications for a wide range of applications involving protein-SAIL composite systems.
Collapse
Affiliation(s)
- Dhiman Ray
- Department of Chemistry, North-Eastern Hill University, Shillong 793 022, India
| | - Dipak Chamlagai
- Department of Chemistry, North-Eastern Hill University, Shillong 793 022, India
| | - Sugam Kumar
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Sutanu Mukhopadhyay
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata 700106, India
| | - Suman Chakrabarty
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata 700106, India
| | - Vinod K Aswal
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Sivaprasad Mitra
- Department of Chemistry, North-Eastern Hill University, Shillong 793 022, India
| |
Collapse
|
13
|
Pun MD, Gallazzi F, Ho KV, Watkinson L, Carmack TL, Iweha E, Li L, Anderson CJ. Albumin-Binding Lutetium-177-Labeled LLP2A Derivatives as Theranostics for Melanoma. Mol Pharm 2024; 21:2960-2969. [PMID: 38680059 DOI: 10.1021/acs.molpharmaceut.4c00095] [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: 05/01/2024]
Abstract
Very late antigen-4 (VLA-4) is a transmembrane integrin protein that is highly expressed in aggressive forms of metastatic melanoma. A small-molecule peptidomimetic, LLP2A, was found to have a low pM affinity binding to VLA-4. Because LLP2A itself does not inhibit cancer cell proliferation and survival, it is an ideal candidate for the imaging and delivery of therapeutic payloads. An analog of [177Lu]Lu-labeled-LLP2A was previously investigated as a therapeutic agent in melanoma tumor-bearing mice, resulting in only a modest improvement in tumor growth inhibition, likely due to rapid clearance of the agent from the tumor. To improve the pharmacokinetic profile, DOTAGA-PEG4-LLP2A with a 4-(p-iodophenyl)butyric acid (pIBA) albumin binding moiety was synthesized. We demonstrate the feasibility of this albumin binding strategy by comparing in vitro cell binding assays and in vivo biodistribution performance of [177Lu]Lu-DOTAGA-PEG4-LLP2A ([177Lu]Lu-1) to the albumin binding [177Lu]Lu-DOTAGA-pIBA-PEG4-LLP2A ([177Lu]Lu-2). In vitro cell binding assay results for [177Lu]Lu-1 and [177Lu]Lu-2 showed Kd values of 0.40 ± 0.07 and 1.75 ± 0.40 nM, with similar Bmax values of 200 ± 6 and 315 ± 15 fmol/mg, respectively. In vivo biodistribution data for both tracers exhibited specific uptake in the tumor, spleen, thymus, and bone due to endogenous expression of VLA-4. Compound [177Lu]Lu-2 exhibited a much longer blood circulation time compared to [177Lu]Lu-1. The tumor uptake for [177Lu]Lu-1 was highest at 1 h (∼15%ID/g) and that for [177Lu]Lu-2 was highest at 4 h (∼23%ID/g). Significant clearance of [177Lu]Lu-1 from the tumor occurs at 24 h (<5%ID/g) while[177Lu]Lu-2 is retained for greater than 96 h (∼10%ID/g). An efficacy study showed that melanoma tumor-bearing mice receiving compound [177Lu]Lu-2 given in two fractions (2 × 14.8 MBq, 14 days apart) had a greater median survival time than mice administered a single 29.6 MBq dose of compound [177Lu]Lu-1, while a single 29.6 MBq dose of [177Lu]Lu-2 imparted hematopoietic toxicity. The in vitro and in vivo data show addition of pIBA to [177Lu]Lu-DOTAGA-PEG4-LLP2A slows blood clearance for a higher tumor uptake, and there is potential of [177Lu]Lu-2 as a theranostic in fractionated administered doses.
Collapse
Affiliation(s)
- Michael D Pun
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Fabio Gallazzi
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Interactions Core, University of Missouri, Columbia, Missouri 65211, United States
| | - Khanh-Van Ho
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Lisa Watkinson
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65211, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Terry L Carmack
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65211, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Ejike Iweha
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Longbo Li
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Carolyn J Anderson
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Department of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri 65212, United States
| |
Collapse
|
14
|
Go EB, Lee JH, Cho JH, Kwon NH, Choi JI, Kwon I. Enhanced therapeutic potential of antibody fragment via IEDDA-mediated site-specific albumin conjugation. J Biol Eng 2024; 18:23. [PMID: 38576037 PMCID: PMC10996255 DOI: 10.1186/s13036-024-00418-3] [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: 11/11/2023] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND The use of single-chain variable fragments (scFvs) for treating human diseases, such as cancer and immune system disorders, has attracted significant attention. However, a critical drawback of scFv is its extremely short serum half-life, which limits its therapeutic potential. Thus, there is a critical need to prolong the serum half-life of the scFv for clinical applications. One promising serum half-life extender for therapeutic proteins is human serum albumin (HSA), which is the most abundant protein in human serum, known to have an exceptionally long serum half-life. However, conjugating a macromolecular half-life extender to a small protein, such as scFv, often results in a significant loss of its critical properties. RESULTS In this study, we conjugated the HSA to a permissive site of scFv to improve pharmacokinetic profiles. To ensure minimal damage to the antigen-binding capacity of scFv upon HSA conjugation, we employed a site-specific conjugation approach using a heterobifunctional crosslinker that facilitates thiol-maleimide reaction and inverse electron-demand Diels-Alder reaction (IEDDA). As a model protein, we selected 4D5scFv, derived from trastuzumab, a therapeutic antibody used in human epithermal growth factor 2 (HER2)-positive breast cancer treatment. We introduced a phenylalanine analog containing a very reactive tetrazine group (frTet) at conjugation site candidates predicted by computational methods. Using the linker TCO-PEG4-MAL, a single HSA molecule was site-specifically conjugated to the 4D5scFv (4D5scFv-HSA). The 4D5scFv-HSA conjugate exhibited HER2 binding affinity comparable to that of unmodified 4D5scFv. Furthermore, in pharmacokinetic profile in mice, the serum half-life of 4D5scFv-HSA was approximately 12 h, which is 85 times longer than that of 4D5scFv. CONCLUSIONS The antigen binding results and pharmacokinetic profile of 4D5scFv-HSA demonstrate that the site-specifically albumin-conjugated scFv retained its binding affinity with a prolonged serum half-life. In conclusion, we developed an effective strategy to prepare site-specifically albumin-conjugated 4D5scFv, which can have versatile clinical applications with improved efficacy.
Collapse
Affiliation(s)
- Eun Byeol Go
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jae Hun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jeong Haeng Cho
- ProAbTech, Gwangju, 61005, Republic of Korea
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Na Hyun Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jong-Il Choi
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Inchan Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| |
Collapse
|
15
|
Pivarcsik T, Kiss MA, Rapuš U, Kljun J, Spengler G, Frank É, Turel I, Enyedy ÉA. Organometallic Ru(II), Rh(III) and Re(I) complexes of sterane-based bidentate ligands: synthesis, solution speciation, interaction with biomolecules and anticancer activity. Dalton Trans 2024; 53:4984-5000. [PMID: 38406993 DOI: 10.1039/d3dt04138g] [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: 02/27/2024]
Abstract
In this study, we present the synthesis, characterization and in vitro cytotoxicity of six organometallic [Ru(II)(η6-p-cymene)(N,N)Cl]Cl, [Rh(III)(η5-C5Me5)(N,N)Cl]Cl and [Re(I)(CO)3(N,N)Cl] complexes, in which the (N,N) ligands are sterane-based 2,2'-bipyridine derivatives (4-Me-bpy-St-OH, 4-Ph-bpy-St-OH). The solution chemical behavior of the ligands and the complexes was explored by UV-visible spectrophotometry and 1H NMR spectroscopy. The ligands and their Re(I) complexes are neutral at pH = 7.40; this contributes to their highly lipophilic character (log D7.40 > +3). The Ru(II) and Rh(III) half-sandwich complexes are much more hydrophilic, and this property is greatly affected by the actual chloride ion content of the medium. The half-sandwich Ru and Rh complexes are highly stable in 30% (v/v) DMSO/water (<5% dissociation at pH = 7.40); this is further increased in water. The Rh(III)(η5-C5Me5) complexes were characterized by higher water/chloride exchange and pKa constants compared to their Ru(II)(η6-p-cymene) counterparts. The Re(I)(CO)3 complexes are also stable in solution over a wide pH range (2-12) without the release of the bidentate ligand; only the chlorido co-ligand can be replaced with OH- at higher pH values. A comprehensive discussion of the binding affinity of the half-sandwich Ru(II) and Rh(III) complexes toward human serum albumin and calf-thymus DNA is also provided. The Ru(II)(η6-p-cymene) complexes interact with human serum albumin via intermolecular forces, while for the Rh(III)(η5-C5Me5) complexes the coordinative binding mode is suggested as well. They are also able to interact with calf-thymus DNA, most likely via the coordination of the guanine nitrogen. The Ru(II)(η6-p-cymene) complexes were found to be the most promising among the tested compounds as they exhibited moderate-to-strong cytotoxic activity (IC50 = 3-11 μM) in LNCaP as well as in PC3 prostate cells in an androgen receptor-independent manner. They were also significantly cytotoxic in breast and colon adenocarcinoma cancer cell lines and showed good selectivity for cancer cells.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary.
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| | - Márton A Kiss
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| | - Uroš Rapuš
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary.
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Éva Frank
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary.
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| |
Collapse
|
16
|
Zhou TJ, Zhang MM, Liu DM, Huang LL, Yu HQ, Wang Y, Xing L, Jiang HL. Glutathione depletion and dihydroorotate dehydrogenase inhibition actuated ferroptosis-augment to surmount triple-negative breast cancer. Biomaterials 2024; 305:122447. [PMID: 38154441 DOI: 10.1016/j.biomaterials.2023.122447] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/03/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023]
Abstract
Ferroptosis is a promising therapeutic approach for combating malignant cancers, but its effectiveness is limited in clinical due to the adaptability and self-repair abilities of cancer cells. Mitochondria, as the pivotal player in ferroptosis, exhibit tremendous therapeutic potential by targeting the intramitochondrial anti-ferroptotic pathway mediated by dihydroorotate dehydrogenase (DHODH). In this study, an albumin-based nanomedicine was developed to induce augmented ferroptosis in triple-negative breast cancer (TNBC) by depleting glutathione (GSH) and inhibiting DHODH activity. The nanomedicine (ATO/SRF@BSA) was developed by loading sorafenib (SRF) and atovaquone (ATO) into bovine serum albumin (BSA). SRF is an FDA-approved ferroptosis inducer and ATO is the only drug used in clinical that targets mitochondria. By combining the effects of SRF and ATO, ATO/SRF@BSA promoted the accumulation of lipid peroxides within mitochondria by inhibiting the glutathione peroxidase 4 (GPX4)-GSH pathway and downregulating the DHODH-coenzyme Q (CoQH2) defense mechanism, triggers a burst of lipid peroxides. Simultaneously, ATO/SRF@BSA suppressed cancer cell self-repair and enhanced cell death by inhibiting the synthesis of adenosine triphosphate (ATP) and pyrimidine nucleotides. Furthermore, the anti-cancer results showed that ATO/SRF@BSA exhibited tumor-specific killing efficacy, significantly improved the tumor hypoxic microenvironment, and lessened the toxic side effects of SRF. This work presents an efficient and easily achievable strategy for TNBC treatment, which may hold promise for clinical applications.
Collapse
Affiliation(s)
- Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Meng-Meng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Dan-Meng Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Li-Ling Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Hai-Qing Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; College of Pharmacy, Yanbian University, Yanji, 133002, PR China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 210009, PR China.
| |
Collapse
|
17
|
Bernardim B, Conde J, Hakala T, Becher JB, Canzano M, Vasco AV, Knowles TPJ, Cameron J, Bernardes GJL. Cathepsin B Processing Is Required for the In Vivo Efficacy of Albumin-Drug Conjugates. Bioconjug Chem 2024; 35:132-139. [PMID: 38345213 PMCID: PMC10885003 DOI: 10.1021/acs.bioconjchem.3c00478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Targeted drug delivery approaches that selectively and preferentially deliver therapeutic agents to specific tissues are of great interest for safer and more effective pharmaceutical treatments. We investigated whether cathepsin B cleavage of a valine-citrulline [VC(S)]-containing linker is required for the release of monomethyl auristatin E (MMAE) from albumin-drug conjugates. In this study, we used an engineered version of human serum albumin, Veltis High Binder II (HBII), which has enhanced binding to the neonatal Fc (fragment crystallizable) receptor (FcRn) to improve drug release upon binding and FcRn-mediated recycling. The linker-payload was conjugated to cysteine 34 of albumin using a carbonylacrylic (caa) reagent which produced homogeneous and plasma stable conjugates that retained FcRn binding. Two caa-linker-MMAE reagents were synthesized─one with a cleavable [VC(S)] linker and one with a noncleavable [VC(R)] linker─to question whether protease-mediated cleavage is needed for MMAE release. Our findings demonstrate that cathepsin B is required to achieve efficient and selective antitumor activity. The conjugates equipped with the cleavable [VC(S)] linker had potent antitumor activity in vivo facilitated by the release of free MMAE upon FcRn binding and internalization. In addition to the pronounced antitumor activity of the albumin conjugates in vivo, we also demonstrated their preferable tumor biodistribution and biocompatibility with no associated toxicity or side effects. These results suggest that the use of engineered albumins with high FcRn binding combined with protease cleavable linkers is an efficient strategy to target delivery of drugs to solid tumors.
Collapse
Affiliation(s)
- Barbara Bernardim
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - João Conde
- Instituto
de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Tuuli Hakala
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Julie B. Becher
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Mary Canzano
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Aldrin V. Vasco
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Tuomas P. J. Knowles
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Jason Cameron
- Albumedix
Ltd, Mabel Street, Nottingham NG2 3ED, United Kingdom
| | - Gonçalo J. L. Bernardes
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
- Instituto
de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
18
|
de Roode KE, Joosten L, Behe M. Towards the Magic Radioactive Bullet: Improving Targeted Radionuclide Therapy by Reducing the Renal Retention of Radioligands. Pharmaceuticals (Basel) 2024; 17:256. [PMID: 38399470 PMCID: PMC10892921 DOI: 10.3390/ph17020256] [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: 06/23/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Targeted radionuclide therapy (TRT) is an emerging field and has the potential to become a major pillar in effective cancer treatment. Several pharmaceuticals are already in routine use for treating cancer, and there is still a high potential for new compounds for this application. But, a major issue for many radiolabeled low-to-moderate-molecular-weight molecules is their clearance via the kidneys and their subsequent reuptake. High renal accumulation of radioactive compounds may lead to nephrotoxicity, and therefore, the kidneys are often the dose-limiting organs in TRT with these radioligands. Over the years, different strategies have been developed aiming for reduced kidney retention and enhanced therapeutic efficacy of radioligands. In this review, we will give an overview of the efforts and achievements of the used strategies, with focus on the therapeutic potential of low-to-moderate-molecular-weight molecules. Among the strategies discussed here is coadministration of compounds that compete for binding to the endocytic receptors in the proximal tubuli. In addition, the influence of altering the molecular design of radiolabeled ligands on pharmacokinetics is discussed, which includes changes in their physicochemical properties and implementation of cleavable linkers or albumin-binding moieties. Furthermore, we discuss the influence of chelator and radionuclide choice on reabsorption of radioligands by the kidneys.
Collapse
Affiliation(s)
- Kim E. de Roode
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
| | - Martin Behe
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen, Switzerland
| |
Collapse
|
19
|
Ji G, Li Y, Zhang Z, Li H, Sun P. Recent advances of novel targeted drug delivery systems based on natural medicine monomers against hepatocellular carcinoma. Heliyon 2024; 10:e24667. [PMID: 38312669 PMCID: PMC10834828 DOI: 10.1016/j.heliyon.2024.e24667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/22/2023] [Accepted: 01/11/2024] [Indexed: 02/06/2024] Open
Abstract
Hepatocellular carcinoma (HCC), the most prevalent type of liver cancer, is often diagnosed at an advanced stage. Surgical interventions are often ineffective, leading HCC patients to rely on systemic chemotherapy. Unfortunately, commonly used chemotherapeutic drugs have limited efficacy and can adversely affect vital organs, causing significant physical and psychological distress for patients. Natural medicine monomers (NMMs) have shown promising efficacy and safety profiles in HCC treatment, garnering attention from researchers. In recent years, the development of novel targeted drug delivery systems (TDDS) combining NMMs with nanocarriers has emerged. These TDDS aim to concentrate drugs effectively in HCC cells by manipulating the characteristics of nanomedicines, leveraging receptor and ligand interactions, and utilizing endogenous stimulatory responses to promote specific nanomedicines distribution. This comprehensive review presents recent research on TDDS for HCC treatment using NMMs from three perspectives: passive TDDS, active TDDS, and stimuli-responsive drug delivery systems (SDDS). It consolidates the current state of research on TDDS for HCC treatment with NMMs and highlights the potential of these innovative approaches in improving treatment outcomes. Moreover, the review also identifies research gaps in the related fields to provide references for future targeted therapy research in HCC.
Collapse
Affiliation(s)
- Guanjie Ji
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yue Li
- Department of Clinical Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhiyue Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Hui Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Ping Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Grade Three Laboratory of Traditional Chinese Medicine Preparation of the National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| |
Collapse
|
20
|
Volovat SR, Scripcariu DV, Vasilache IA, Stolniceanu CR, Volovat C, Augustin IG, Volovat CC, Ostafe MR, Andreea-Voichița SG, Bejusca-Vieriu T, Lungulescu CV, Sur D, Boboc D. Oncolytic Virotherapy: A New Paradigm in Cancer Immunotherapy. Int J Mol Sci 2024; 25:1180. [PMID: 38256250 PMCID: PMC10816814 DOI: 10.3390/ijms25021180] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Oncolytic viruses (OVs) are emerging as potential treatment options for cancer. Natural and genetically engineered viruses exhibit various antitumor mechanisms. OVs act by direct cytolysis, the potentiation of the immune system through antigen release, and the activation of inflammatory responses or indirectly by interference with different types of elements in the tumor microenvironment, modification of energy metabolism in tumor cells, and antiangiogenic action. The action of OVs is pleiotropic, and they show varied interactions with the host and tumor cells. An important impediment in oncolytic virotherapy is the journey of the virus into the tumor cells and the possibility of its binding to different biological and nonbiological vectors. OVs have been demonstrated to eliminate cancer cells that are resistant to standard treatments in many clinical trials for various cancers (melanoma, lung, and hepatic); however, there are several elements of resistance to the action of viruses per se. Therefore, it is necessary to evaluate the combination of OVs with other standard treatment modalities, such as chemotherapy, immunotherapy, targeted therapies, and cellular therapies, to increase the response rate. This review provides a comprehensive update on OVs, their use in oncolytic virotherapy, and the future prospects of this therapy alongside the standard therapies currently used in cancer treatment.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Dragos Viorel Scripcariu
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania;
| | - Ingrid Andrada Vasilache
- Department of Obstetrics and Gynecology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics—Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania;
| | - Constantin Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | | | | | - Madalina-Raluca Ostafe
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Slevoacă-Grigore Andreea-Voichița
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Toni Bejusca-Vieriu
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | | | - Daniel Sur
- 11th Department of Medical Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania;
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| |
Collapse
|
21
|
Liu Z, Zhou X, Chen Y, Ni Y, Zhu Z, Cao W, Chen K, Yan Y, Ji J, Zhang P. Fibrous capsule-resistant, controllably degradable and functionalizable zwitterion-albumin hybrid hydrogels. Biomater Sci 2024; 12:468-478. [PMID: 38086632 DOI: 10.1039/d3bm01783d] [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: 01/17/2024]
Abstract
Foreign body response (FBR) represents an immune-mediated cascade reaction capable of inducing the rejection of foreign implants, thereby compromising their in vivo performance. Pure zwitterionic hydrogels have demonstrated the ability to resist long-term FBR, owing to their outstanding antifouling capabilities. However, achieving such a robust anti-FBR effect necessitates stringent requirements concerning the purity of zwitterionic materials, which constrains their broader functional applications. Herein, we present a biocompatible, controllably degradable, and functionalizable zwitterion-albumin hybrid hydrogel. The zwitterionic hydrogel crosslinked with serum albumin exhibits controllable degradation and excels in preventing the adsorption of various proteins and adhesion of cells and bacteria. Moreover, the hydrogel significantly alleviates the host's FBR compared with PEG hydrogels and particularly outperforms PEG-based cross-linker crosslinked zwitterionic hydrogels in reducing collagen encapsulation when subcutaneously implanted into mice. The zwitterion-albumin hybrid hydrogel shows potential as a functionalizable anti-FBR material in the context of implantable materials and biomedical devices.
Collapse
Affiliation(s)
- Zuolong Liu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Xianchi Zhou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yongcheng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yanwen Ni
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Zihao Zhu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Wenzhong Cao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Kexin Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yu Yan
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, P. R. China
| | - Peng Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, P. R. China
| |
Collapse
|
22
|
Karami E, Mesbahi Moghaddam M, Kazemi-Lomedasht F. Use of Albumin for Drug Delivery as a Diagnostic and Therapeutic Tool. Curr Pharm Biotechnol 2024; 25:676-693. [PMID: 37550918 DOI: 10.2174/1389201024666230807161200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 08/09/2023]
Abstract
Drug delivery is an important topic that has attracted the attention of researchers in recent years. Albumin nanoparticles play a significant role in drug delivery as a carrier due to their unique characteristics. Albumin is non-toxic, biocompatible, and biodegradable. Its structure is such that it can interact with different drugs, which makes the treatment of the disease faster and also reduces the side effects of the drug. Albumin nanoparticles can be used in the diagnosis and treatment of many diseases, including cancer, diabetes, Alzheimer's, etc. These nanoparticles can connect to some compounds, such as metal nanoparticles, antibodies, folate, etc. and create a powerful nanostructure for drug delivery. In this paper, we aim to investigate albumin nanoparticles in carrier format for drug delivery application. In the beginning, different types of albumin and their preparation methods were discussed, and then albumin nanoparticles were discussed in detail in diagnosing and treating various diseases.
Collapse
Affiliation(s)
- Elmira Karami
- Venom and Biotherapeutics Molecules Laboratory, Department of Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Department of Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Liu LH, Liu YF, Zhang HB, Liu XL, Zhang HW, Huang B, Lin F, Li WH. A Novel ANG-BSA/BCNU/ICG MNPs Integrated for Targeting Therapy of Glioblastoma. Technol Cancer Res Treat 2024; 23:15330338241281321. [PMID: 39444362 PMCID: PMC11526396 DOI: 10.1177/15330338241281321] [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: 10/25/2024] Open
Abstract
PURPOSE Develop an albumin nanoparticle-based nanoprobe for targeted glioblastoma (GBM) diagnosis and treatment, utilizing Angopep-2 for low-density lipoprotein receptor-related protein (LRP) targeting. METHODS Combined albumin-coated superparamagnetic iron oxide (SPIO), Carmustine (BCNU), and indocyanine green (ICG). Assessed morphology, size, Zeta potential, fluorescence, and drug encapsulation. Conducted in vitro fluorescence/MRI imaging and cell viability assays, and in vivo nanoprobe accumulation evaluation in brain tumors. RESULTS ANG-BSA/BCNU/ICG MNPs exhibited superior targeting and cytotoxicity against GBM cells in vitro. In vivo, enhanced brain tumor accumulation during imaging was observed. CONCLUSION This targeted imaging and drug delivery system holds promise for efficient GBM therapy and intraoperative localization, addressing Blood-brain barrier (BBB) limitations with precise drug delivery and imaging capabilities.
Collapse
Affiliation(s)
- Li-Hong Liu
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Yu-Feng Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Hong-Bo Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Xiao-Lei Liu
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Han-Wen Zhang
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Biao Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Fan Lin
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Wei-Hua Li
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| |
Collapse
|
24
|
Cho J, Yang B, Lee JH, Kim H, Kim H, Go EB, Bak DH, Park SJ, Kwon I, Choi JI, Lee K. In vivo study of newly developed albumin-conjugated urate oxidase for gout treatment. Arthritis Res Ther 2023; 25:247. [PMID: 38111075 PMCID: PMC10726570 DOI: 10.1186/s13075-023-03231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Exogenously providing engineered Uox with enhanced half-life is one of the important urate-lowering treatments for gout. The potential of PAT101, a recombinant human albumin (rHA)-conjugated variant, was evaluated and compared as a novel gout treatment through various in vivo studies with PAT101 and competing drugs. METHODS PAT101 was produced by site-specific conjugation of rHA and Aspergillus flavus Uox (AfUox-rHA) through clickable non-natural amino acid (frTet) and Inverse electron demand Diels-Alder (IEDDA) reaction. In vivo pharmacokinetics, efficacy tests and in vitro immunogenetic assay were performed after single or multiple doses of PAT101 and its competitors in BALB/c mice, transgenic (TG) mice, Sprague-Dawley (SD) rats, and non-human primate (NHP). RESULTS The half-life of PAT101 in single-dose treated TG mice was more than doubled compared to pegloticase. In SD rats with 4 weeks of repeated administration of rasburicase, only 24% of Uox activity remained, whereas in PAT101, it was maintained by 86%. In the Uox KO model, the survival rate of PAT101 was comparable to that of pegloticase. In addition, human PBMC-based CD4+/CD8+ T-cell activation analysis demonstrated that PAT101 has a lower immune response compared to the original drug, rasburicase. CONCLUSION All results suggest that this rHA-conjugated AfUox, PAT101, can be provided as a reliable source of Uox for gout treatment.
Collapse
Affiliation(s)
- Jeonghaeng Cho
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Byungseop Yang
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Jae Hun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyunwoo Kim
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Hyeongseok Kim
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Eun Byeol Go
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Dong-Ho Bak
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Su Jin Park
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Inchan Kwon
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jong-Il Choi
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kyunghee Lee
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea.
| |
Collapse
|
25
|
Yang SJ, Pai JA, Yao CJ, Huang CH, Chen JL, Wang CH, Chen KC, Shieh MJ. SN38-loaded nanomedicine mediates chemo-radiotherapy against CD44-expressing cancer growth. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-022-00151-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Abstract
Background
Chemo-radiotherapy is the combined chemotherapy and radiotherapy on tumor treatment to obtain the local radiosensitization and local cytotoxicity of the tumor and to control the microscopic metastatic disease.
Methods
In this study, 7-ethyl-10-hydroxycamptothecin (SN38) molecules could be successfully loaded into human serum albumin (HSA)–hyaluronic acid (HA) nanoparticles (SH/HA NPs) by the hydrophobic side groups of amino acid in HSA.
Results
HSA could be used to increase the biocompatibility and residence time of the nanoparticles in the blood, whereas HA could improve the benefits and overall treatment effect on CD44-expressing colorectal cancer (CRC), and reduce drug side effects. In addition to its role as a chemotherapeutic agent, SN38 could be used as a radiosensitizer, able to arrest the cell cycle, and allowing cells to stay in the G2/M stage, to improve the sensitivity of tumor cells to radiation. In vivo results demonstrated that SH/HA NPs could accumulate in the tumor and produce significant tumor suppression, with no adverse effects observed when combined with γ-ray irradiation. This SH/HA NPs-medicated chemo-radiotherapy could induce an anti-tumor immune response to inhibit the growth of distal tumors, and produce an abscopal effect.
Conclusions
Therefore, this SN38-loaded and HA-incorporated nanoparticle combined with radiotherapy may be a promising therapeutic artifice for CRC in the future.
Collapse
|
26
|
Deng C, Zheng M, Xin J, An F. A nanoparticle composed of totally hospital-available drugs and isotope for fluorescence/SPECT dual-modal imaging-guided photothermal therapy to inhibit tumor metastasis. J Colloid Interface Sci 2023; 651:384-393. [PMID: 37544227 DOI: 10.1016/j.jcis.2023.07.163] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
As primary sites of tumor metastasis, sentinel lymph nodes (SLNs) require a highly biocompatible theranostic platform for precise localization and treatment to inhibit tumor metastasis. Herein, indocyanine green-human serum albumin (ICG-HSA) nanoparticles (NPs) were fabricated by ICG-induced self-assembly and radiolabeled with technetuim-99 m (99mTc). The fabricated NPs were composed of hospital-available drugs and isotopes, making them highly biocompatible for in vivo applications. In a mouse model of SLN metastasis, the prepared NPs exhibited excellent capacity for preoperative planning by single-photon emission computed tomography (SPECT) imaging-enabled SLN localization, near-infrared fluorescence (NIRF) imaging-enabled intraoperative real-time monitoring, and SLN photothermal treatment. Photothermal treatment with SLN enhanced the inhibition of lung metastasis and significantly increased the survival time of mice. The prepared NPs were highly biocompatible and exhibited efficient theranostic properties for inhibiting cancer metastasis, making them promising candidates for clinical translation.
Collapse
Affiliation(s)
- Caiting Deng
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an 710061, Shaanxi, China; School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an 710061, Shaanxi, China
| | - Meichen Zheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an 710061, Shaanxi, China
| | - Jingqi Xin
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an 710061, Shaanxi, China; School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an 710061, Shaanxi, China
| | - Feifei An
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
27
|
Moriyama K, Inomoto N, Moriuchi H, Nihei M, Sato M, Miyagi Y, Tajiri A, Sato T, Tanaka Y, Johno Y, Goto M, Kamiya N. Characterization of enzyme-crosslinked albumin hydrogel for cell encapsulation. J Biosci Bioeng 2023; 136:471-476. [PMID: 37798227 DOI: 10.1016/j.jbiosc.2023.09.007] [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: 06/22/2023] [Revised: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 10/07/2023]
Abstract
Albumin is an attractive component for the development of biomaterials applied as biomedical implants, including drug carriers and tissue engineering scaffolds, because of its high biocompatibility and low immunogenicity. Additionally, albumin-based gelators facilitate cross-linking reactions under mild conditions, which maintains the high viability of encapsulated living cells. In this study, we synthesized albumin derivatives to undergo gelation under physiological conditions via the peroxidase-catalyzed formation of cross-links. Albumin was modified with phenolic hydroxyl groups (Alb-Ph-OH) using carbodiimide chemistry, and the effect of degree of substitution on gelation was investigated. Various properties of the Alb-Ph-OH hydrogels, namely the gelation time, swelling ratio, pore size, storage modulus, and enzymatic degradability, were easily controlled by adjusting the degree of substitution and the polymer concentration. Moreover, the viability of cells encapsulated within the Alb-Ph-OH hydrogel was high. These results demonstrate the potential applicability of Alb-Ph-OH hydrogels as cell-encapsulating materials for biomedical applications, including tissue engineering.
Collapse
Affiliation(s)
- Kousuke Moriyama
- Department of Chemical and Biological Engineering, National Institute of Technology, Sasebo Collage, 1-1 Okishin-cho, Sasebo, Nagasaki 857-1193, Japan.
| | - Noe Inomoto
- Department of Chemical and Biological Engineering, National Institute of Technology, Sasebo Collage, 1-1 Okishin-cho, Sasebo, Nagasaki 857-1193, Japan
| | - Hidetoshi Moriuchi
- Department of Chemical and Biological Engineering, National Institute of Technology, Sasebo Collage, 1-1 Okishin-cho, Sasebo, Nagasaki 857-1193, Japan
| | - Masanobu Nihei
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Miku Sato
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Yoshiki Miyagi
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Ayaka Tajiri
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Takeshi Sato
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Yasuhiko Tanaka
- Department of Chemical and Biological Engineering, National Institute of Technology, Sasebo Collage, 1-1 Okishin-cho, Sasebo, Nagasaki 857-1193, Japan
| | - Yuuki Johno
- Department of Chemical and Biological Engineering, National Institute of Technology, Sasebo Collage, 1-1 Okishin-cho, Sasebo, Nagasaki 857-1193, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
28
|
Wang Z, Wang X, Xu W, Li Y, Lai R, Qiu X, Chen X, Chen Z, Mi B, Wu M, Wang J. Translational Challenges and Prospective Solutions in the Implementation of Biomimetic Delivery Systems. Pharmaceutics 2023; 15:2623. [PMID: 38004601 PMCID: PMC10674763 DOI: 10.3390/pharmaceutics15112623] [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: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Biomimetic delivery systems (BDSs), inspired by the intricate designs of biological systems, have emerged as a groundbreaking paradigm in nanomedicine, offering unparalleled advantages in therapeutic delivery. These systems, encompassing platforms such as liposomes, protein-based nanoparticles, extracellular vesicles, and polysaccharides, are lauded for their targeted delivery, minimized side effects, and enhanced therapeutic outcomes. However, the translation of BDSs from research settings to clinical applications is fraught with challenges, including reproducibility concerns, physiological stability, and rigorous efficacy and safety evaluations. Furthermore, the innovative nature of BDSs demands the reevaluation and evolution of existing regulatory and ethical frameworks. This review provides an overview of BDSs and delves into the multifaceted translational challenges and present emerging solutions, underscored by real-world case studies. Emphasizing the potential of BDSs to redefine healthcare, we advocate for sustained interdisciplinary collaboration and research. As our understanding of biological systems deepens, the future of BDSs in clinical translation appears promising, with a focus on personalized medicine and refined patient-specific delivery systems.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; (Z.W.); (R.L.)
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Wanting Xu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Yongxiao Li
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Ruizhi Lai
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; (Z.W.); (R.L.)
| | - Xiaohui Qiu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Xu Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Zhidong Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Meiying Wu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| |
Collapse
|
29
|
Su Q, Zhang Y, Zhu S. Site-specific albumin tagging with chloride-containing near-infrared cyanine dyes: molecular engineering, mechanism, and imaging applications. Chem Commun (Camb) 2023; 59:13125-13138. [PMID: 37850230 DOI: 10.1039/d3cc04200f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Near-infrared dyes, particularly cyanine dyes, have shown great potential in biomedical imaging due to their deep tissue penetration, high resolution, and minimal tissue autofluorescence/scattering. These dyes can be adjusted in terms of absorption and emission wavelengths by modifying their chemical structures. The current issues with cyanine dyes include aggregation-induced quenching, poor photostability, and short in vivo circulation time. Encapsulating cyanine dyes with albumin, whether exogenous or endogenous, has been proven to be an effective strategy for improving their brightness and pharmacokinetics. In detail, the chloride-containing (Cl-containing) cyanine dyes have been found to selectively bind to albumin to achieve site-specific albumin tagging, resulting in enhanced optical properties and improved biosafety. This feature article provides an overview of the progress in the covalent binding of Cl-containing cyanine dyes with albumin, including molecular engineering methods, binding sites, and the selective binding mechanism. The improved optical properties of cyanine dyes and albumin complexes have led to cutting-edge applications in biological imaging, such as tumor imaging (diagnostics) and imaging-guided surgery.
Collapse
Affiliation(s)
- Qi Su
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China.
| | - Yuewei Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P. R. China.
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China.
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P. R. China.
| |
Collapse
|
30
|
Jiang T, Wang J, Xie L, Zhou S, Huang Y, Chen Y, Gao X, Xiao W, Chen J. Biguanide-anchored albumin-based nanoplatform inhibits epithelial-mesenchymal transition and reduces the stemness phenotype for metastatic cancer therapy. Acta Biomater 2023; 171:565-579. [PMID: 37716479 DOI: 10.1016/j.actbio.2023.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
In clinical chemotherapy, albumin-bound paclitaxel (Abraxane) can improve the tumor targeting property and therapeutic efficacy of paclitaxel (PTX) against orthotopic malignancies. However, patients with metastatic cancer have a poor prognosis, probably due to the instability, chemoresistance, and inability of albumin-bound paclitaxel to alter the tumor microenvironment. Here we propose a new biguanide-modified albumin-based nanoplatform that encapsulates paclitaxel for the effective treatment of metastatic cancer. The PTX is encapsulated in poly (lactic-co-glycolic acid) cores coated with biguanide-modified albumin (HSA-NH). The functionalized nanoparticles (HSA-NH NPs) exhibit a remarkable stable profile with low drug release (P < 0.05 versus Abraxane), target tumor tissues, suppress epithelial-mesenchymal transition (EMT) events for anti-metastatic effects, and reduce the phenotype of cancer stem cells. As a result, HSA-NH NPs effectively prolong animal survival (55 days) by inhibiting not only primary tumor growth but also metastasis. This study provides proof of concept that the biguanide-anchored albumin-based nanoplatform encapsulating PTX is a powerful, safe, and clinically translational strategy for the treatment of metastatic cancer. STATEMENT OF SIGNIFICANCE: Albumin-bound paclitaxel (Abraxane) can increase paclitaxel's tumor targeting and therapeutic efficacy in clinical cancer treatments such as breast cancer. However, the instability, chemoresistance, and lack of tumor microenvironment modulation of albumin-bound paclitaxel may lead to poor therapeutic efficacy in metastatic cancer patients. Here we develop biguanide-anchored albumin-based nanoplatforms that encapsulate paclitaxel (HSA-NH NPs) for metastatic cancer treatment. Poly(lactic-co-glycolic acid) (PLGA) cores encapsulating paclitaxel improve the stability of HSA-NH NPs. Based on the activities of metformin, biguanide-anchored albumin adsorbed on PLGA cores improves paclitaxel efficacy, inhibits various aberrant changes during epithelial-mesenchymal transition, and reduces tumor cell stemness. The biguanide-anchored albumin-based nanoplatform encapsulating PTX can serve as a potent, safe, and clinically translational approach for metastatic cancer therapies.
Collapse
Affiliation(s)
- Tianze Jiang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Marine Drugs, Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jiahao Wang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Laozhi Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Yukun Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yu Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Wenze Xiao
- Department of Rheumatology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai 201399, China.
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|
31
|
Cheng Y, Pang X, Wu J, Zhou L, Cao J, Wang L, Qian K, Yang P, Xu M, Sheng D, Meng R, Wang P, Guo Q, Xu S, Wei Y, Zhang Q. Medium-chain triglyceride-stabilized docetaxel-loaded HSA nanoparticles effectively inhibited metastatic non-small cell lung cancer. Drug Deliv Transl Res 2023; 13:2869-2884. [PMID: 37204680 DOI: 10.1007/s13346-023-01355-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 05/20/2023]
Abstract
Metastatic non-small cell lung cancer (NSCLC) is refractory with a very poor prognosis. Docetaxel (DTX) injection (Taxotere®) has been approved for the treatment of locally advanced or metastatic NSCLC. However, its clinical application is restricted by severe adverse effects and non-selective tissue distribution. In this study, we successfully developed DTX-loaded human serum albumin (HSA) nanoparticles (DNPs) with modified Nab technology, by introducing medium-chain triglyceride (MCT) as a stabilizer. The optimized formulation had a particle size of approximately 130 nm and a favorable stabilization time of more than 24 h. DNPs dissociated in circulation in a concentration-dependent manner and slowly released DTX. Compared with DTX injection, DNPs were more effectively taken up by NSCLC cells, thus exerting stronger inhibitory effects on their proliferation, adhesion, migration, and invasion. In addition, DNPs showed prolonged blood retention and increased tumor accumulation relative to DTX injection. Ultimately, DNPs produced more potent inhibitory effects on primary or metastatic tumor foci than DTX injections but caused markedly lower organ toxicity and hematotoxicity. Overall, these results support that DNPs hold great potential for the treatment of metastatic NSCLC in clinical.
Collapse
Affiliation(s)
- Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xiaoying Pang
- Department of Pharmacy, the Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jinxu Cao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Liuchang Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qian Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shuting Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
32
|
Tincu (Iurciuc) CE, Andrițoiu CV, Popa M, Ochiuz L. Recent Advancements and Strategies for Overcoming the Blood-Brain Barrier Using Albumin-Based Drug Delivery Systems to Treat Brain Cancer, with a Focus on Glioblastoma. Polymers (Basel) 2023; 15:3969. [PMID: 37836018 PMCID: PMC10575401 DOI: 10.3390/polym15193969] [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: 08/14/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive malignant tumor, and the most prevalent primary malignant tumor affecting the brain and central nervous system. Recent research indicates that the genetic profile of GBM makes it resistant to drugs and radiation. However, the main obstacle in treating GBM is transporting drugs through the blood-brain barrier (BBB). Albumin is a versatile biomaterial for the synthesis of nanoparticles. The efficiency of albumin-based delivery systems is determined by their ability to improve tumor targeting and accumulation. In this review, we will discuss the prevalence of human glioblastoma and the currently adopted treatment, as well as the structure and some essential functions of the BBB, to transport drugs through this barrier. We will also mention some aspects related to the blood-tumor brain barrier (BTBB) that lead to poor treatment efficacy. The properties and structure of serum albumin were highlighted, such as its role in targeting brain tumors, as well as the progress made until now regarding the techniques for obtaining albumin nanoparticles and their functionalization, in order to overcome the BBB and treat cancer, especially human glioblastoma. The albumin drug delivery nanosystems mentioned in this paper have improved properties and can overcome the BBB to target brain tumors.
Collapse
Affiliation(s)
- Camelia-Elena Tincu (Iurciuc)
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| | - Călin Vasile Andrițoiu
- Apitherapy Medical Center, Balanesti, Nr. 336-337, 217036 Gorj, Romania;
- Specialization of Nutrition and Dietetics, Faculty of Pharmacy, Vasile Goldis Western University of Arad, Liviu Rebreanu Street, 86, 310045 Arad, Romania
| | - Marcel Popa
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Faculty of Dental Medicine, “Apollonia” University of Iasi, 11, Pacurari Street, 700511 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Lăcrămioara Ochiuz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| |
Collapse
|
33
|
Liu L, Yang B, Yuan H, Yu N, Feng Y, Zhang Y, Yin T, He H, Gou J, Tang X. Human Serum Albumin Nanoparticles as a Carrier of 20( S)-Protopanaxadiol via Intramuscular Injection to Alleviate Cyclophosphamide-Induced Myelosuppression. Mol Pharm 2023; 20:5125-5134. [PMID: 37647098 DOI: 10.1021/acs.molpharmaceut.3c00409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Myelosuppression is a prevalent and potentially life-threatening side effect during chemotherapy. As the main active component of ginseng, 20(S)-protopanaxadiol (PPD) is capable of relieving myelosuppression by restoring hematopoiesis and immunity. In this study, PPD was encapsulated in human albumin nanoparticles (PPD-HSA NPs) by nanoparticle albumin-bound (Nab) technology for intramuscular injection to optimize its pharmacokinetic properties and promote recovery of myelosuppression. The prepared PPD-HSA NPs had a particle size of about 280 nm with a narrow size distribution. PPD dispersed as an amorphous state within the PPD-HSA NPs, and the NPs exhibited in vitro sustained release behavior. PPD-HSA NPs showed a favorable pharmacokinetic profile with high absolute bioavailability, probably due to the fact that NPs entered into the blood circulation via lymphatic circulation and were eliminated slowly. In vivo distribution experiments demonstrated that PPD-HSA NPs were mainly distributed in the liver and spleen, but a strong fluorescence signal was also found in the inguinal lymph node, indicating drug absorption via a lymph route. The myelosuppressive model was established using cyclophosphamide as the inducer. Pharmacodynamic studies confirmed that PPD-HSA NPs were effective in promoting the level of white blood cells. Moreover, the neutrophil and lymphocyte counts were significantly higher in the PPD-HSA NPs group compared with the control group. This preliminary investigation revealed that PPD-HSA NPs via intramuscular administration may be an effective intervention strategy to alleviate myelosuppression.
Collapse
Affiliation(s)
- Lei Liu
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Bing Yang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haoyang Yuan
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Nini Yu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yupeng Feng
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Yu Zhang
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Tian Yin
- Department of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Haibing He
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Jingxin Gou
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Xing Tang
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| |
Collapse
|
34
|
Vinck R, Dömötör O, Karges J, Jakubaszek M, Seguin J, Tharaud M, Guérineau V, Cariou K, Mignet N, Enyedy ÉA, Gasser G. In Situ Bioconjugation of a Maleimide-Functionalized Ruthenium-Based Photosensitizer to Albumin for Photodynamic Therapy. Inorg Chem 2023; 62:15510-15526. [PMID: 37708255 DOI: 10.1021/acs.inorgchem.3c01984] [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: 09/16/2023]
Abstract
Maleimide-containing prodrugs can quickly and selectively react with circulating serum albumin following their injection in the bloodstream. The drug-albumin complex then benefits from longer blood circulation times and better tumor accumulation. Herein, we have applied this strategy to a previously reported highly phototoxic Ru polypyridyl complex-based photosensitizer to increase its accumulation at the tumor, reduce off-target cytotoxicity, and therefore improve its pharmacological profile. Specifically, two complexes were synthesized bearing a maleimide group: one complex with the maleimide directly incorporated into the bipyridyl ligand, and the other has a hydrophilic linker between the ligand and the maleimide group. Their interaction with albumin was studied in-depth, revealing their ability to efficiently bind both covalently and noncovalently to the plasma protein. A crucial finding is that the maleimide-functionalized complexes exhibited significantly lower cytotoxicity in noncancerous cells under dark conditions compared to the nonfunctionalized complex, which is a highly desirable property for a photosensitizer. The binding to albumin also led to a decrease in the phototoxicity of the Ru bioconjugates in comparison to the nonfunctionalized complex, probably due to a decreased cellular uptake. Unfortunately, this decrease in phototoxicity was not compensated by a dramatic increase in tumor accumulation, as was demonstrated in a tumor-bearing mouse model using inductively coupled plasma mass spectrometry (ICP-MS) studies. Consequently, this study provides valuable insight into the future design of in situ albumin-binding complexes for photodynamic therapy in order to maximize their effectiveness and realize their full potential.
Collapse
Affiliation(s)
- Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Orsolya Dömötör
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7. H-6720 Szeged, Hungary
| | - Johannes Karges
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Marta Jakubaszek
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Johanne Seguin
- Université Paris Cité, UTCBS, INSERM, CNRS, 75006 Paris, France
| | - Mickaël Tharaud
- Biogéochimie à l'Anthropocène des Eléments et Contaminants Emergents, Institut de Physique du Globe de Paris, 75005 Paris, France
| | - Vincent Guérineau
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Sud, Université Paris-Saclay, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Nathalie Mignet
- Université Paris Cité, UTCBS, INSERM, CNRS, 75006 Paris, France
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7. H-6720 Szeged, Hungary
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| |
Collapse
|
35
|
Liu H, Zhao J, Xue Y, Zhang J, Bai H, Pan S, Peng B, Li L, Voelcker NH. X-Ray-Induced Drug Release for Cancer Therapy. Angew Chem Int Ed Engl 2023; 62:e202306100. [PMID: 37278399 DOI: 10.1002/anie.202306100] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/07/2023]
Abstract
Drug delivery systems (DDSs) are designed to deliver therapeutic agents to specific target sites while minimizing systemic toxicity. Recent developments in drug-loaded DDSs have demonstrated promising characteristics and paved new pathways for cancer treatment. Light, a prevalent external stimulus, is widely utilized to trigger drug release. However, conventional light sources primarily concentrate on the ultraviolet (UV) and visible light regions, which suffer from limited biological tissue penetration. This limitation hinders applications for deep-tissue tumor drug release. Given their deep tissue penetration and well-established application technology, X-rays have recently received attention for the pursuit of controlled drug release. With precise spatiotemporal and dosage controllability, X-rays stand as an ideal stimulus for achieving controlled drug release in deep-tissue cancer therapy. This article explores the recent advancements in using X-rays for stimulus-triggered drug release in DDSs and delves into their action mechanisms.
Collapse
Affiliation(s)
- Hui Liu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jun Zhao
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yufei Xue
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jiaxin Zhang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Sijun Pan
- The Institute of Flexible Electronics, IFE, Future Technologies), Xiamen University, Xiamen, 361005, Fujian, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
- Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, 399 Royal Parade, Parkville, Victoria, 3052, Australia
- Wuhan National Laboratory for Optoelectronics, Advanced Biomedical Imaging Facility, 13 Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Lin Li
- The Institute of Flexible Electronics, IFE, Future Technologies), Xiamen University, Xiamen, 361005, Fujian, China
| | - Nicolas H Voelcker
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
- Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, 399 Royal Parade, Parkville, Victoria, 3052, Australia
| |
Collapse
|
36
|
Arojojoye AS, Walker B, Dewahare JC, Afrifa MAO, Parkin S, Awuah SG. Circumventing Physicochemical Barriers of Cyclometalated Gold(III) Dithiocarbamate Complexes with Protein-Based Nanoparticle Delivery to Enhance Anticancer Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43607-43620. [PMID: 37698293 PMCID: PMC11264193 DOI: 10.1021/acsami.3c10025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Optimizing the bioavailability of drug candidates is crucial to successful drug development campaigns, especially for metal-derived chemotherapeutic agents. Nanoparticle delivery strategies can be deployed to overcome physicochemical limitations associated with drugs to improve bioavailability, pharmacokinetics, efficacy, and minimize toxicity. Biodegradable albumin nanoconstructs offer pragmatic solutions for drug delivery of metallodrugs with translational benefits in the clinic. In this work, we explored a logical approach to investigate and resolve the physicochemical drawbacks of gold(III) complexes with albumin nanoparticle delivery to improve solubility, enhance intracellular accumulation, circumvent premature deactivation, and enhance anticancer activity. We synthesized and characterized stable gold(III) dithiocarbamate complexes with a variable degree of cyclometalation such as phenylpyridine (C^N) or biphenyl (C^C) Au(III) framework and different alkyl chain lengths. We noted that extended alkyl chain lengths impaired the solubility of these complexes in biological media, thus adversely impacting potency. Encapsulation of these complexes in bovine serum albumin (BSA) reversed solubility limitations and improved cancer cytotoxicity by ∼25-fold. Further speciation and mechanism of action studies demonstrate the stability of the compounds and alteration of mitochondria bioenergetics, respectively. We postulate that this nanodelivery strategy is a relevant approach for translational small-molecule gold drug delivery.
Collapse
Affiliation(s)
| | - Breyanna Walker
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | - James C. Dewahare
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | | | - Sean Parkin
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
- Center for Pharmaceutical Research and Innovation and Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
| |
Collapse
|
37
|
Fujikawa Y, Fukuo Y, Nishimura K, Tsujino K, Kashiwagi H, Hiramatsu R, Nonoguchi N, Furuse M, Takami T, Hu N, Miyatake SI, Takata T, Tanaka H, Watanabe T, Suzuki M, Kawabata S, Nakamura H, Wanibuchi M. Evaluation of the Effectiveness of Boron Neutron Capture Therapy with Iodophenyl-Conjugated closo-Dodecaborate on a Rat Brain Tumor Model. BIOLOGY 2023; 12:1240. [PMID: 37759639 PMCID: PMC10525593 DOI: 10.3390/biology12091240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
High-grade gliomas present a significant challenge in neuro-oncology because of their aggressive nature and resistance to current therapies. Boron neutron capture therapy (BNCT) is a potential treatment method; however, the boron used by the carrier compounds-such as 4-borono-L-phenylalanine (L-BPA)-have limitations. This study evaluated the use of boron-conjugated 4-iodophenylbutanamide (BC-IP), a novel boron compound in BNCT, for the treatment of glioma. Using in vitro drug exposure experiments and in vivo studies, we compared BC-IP and BPA, with a focus on boron uptake and retention characteristics. The results showed that although BC-IP had a lower boron uptake than BPA, it exhibited superior retention. Furthermore, despite lower boron accumulation in tumors, BNCT mediated by BC-IP showed significant survival improvement in glioma-bearing rats compared to controls (not treated animals and neutrons only). These results suggest that BC-IP, with its unique properties, may be an alternative boron carrier for BNCT. Further research is required to optimize this potential treatment modality, which could significantly contribute to advancing the treatment of high-grade gliomas.
Collapse
Affiliation(s)
- Yoshiki Fujikawa
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Yusuke Fukuo
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Kai Nishimura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan; (K.N.); (H.N.)
| | - Kohei Tsujino
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Hideki Kashiwagi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Ryo Hiramatsu
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Naosuke Nonoguchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Motomasa Furuse
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Toshihiro Takami
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Naonori Hu
- Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (N.H.); (S.-I.M.)
| | - Shin-Ichi Miyatake
- Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (N.H.); (S.-I.M.)
| | - Takushi Takata
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka 590-0494, Japan; (T.T.); (H.T.); (T.W.); (M.S.)
| | - Hiroki Tanaka
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka 590-0494, Japan; (T.T.); (H.T.); (T.W.); (M.S.)
| | - Tsubasa Watanabe
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka 590-0494, Japan; (T.T.); (H.T.); (T.W.); (M.S.)
| | - Minoru Suzuki
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka 590-0494, Japan; (T.T.); (H.T.); (T.W.); (M.S.)
| | - Shinji Kawabata
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| | - Hiroyuki Nakamura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan; (K.N.); (H.N.)
| | - Masahiko Wanibuchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (Y.F.); (Y.F.); (K.T.); (H.K.); (R.H.); (N.N.); (M.F.); (T.T.); (M.W.)
| |
Collapse
|
38
|
Péraudeau E, Renoux B, Emambux S, Poinot P, Châtre R, Thoreau F, Riss Yaw B, Tougeron D, Clarhaut J, Papot S. Combination of Targeted Therapies for Colorectal Cancer Treatment. Mol Pharm 2023; 20:4537-4545. [PMID: 37579031 DOI: 10.1021/acs.molpharmaceut.3c00224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
The design of innovative therapeutic strategies enabling the selective destruction of tumor cells while sparing healthy tissues remains highly challenging in cancer therapy. Here, we show that the combination of two targeted therapies, including bevacizumab (Bev), and a β-glucuronidase-responsive albumin-binding prodrug of monomethyl auristatin E (MMAE), is efficient for the treatment of colorectal cancer implanted in mice. This combined therapy produces a therapeutic activity superior to that of the association of FOLFOX and Bev currently used to treat patients with this pathology. The increased anticancer efficacy is due to either a synergistic or an additive effect between Bev and MMAE selectively released from the glucuronide prodrug in the tumor microenvironment. Since numerous drug delivery systems such as antibody-drug conjugates employ MMAE as a cytotoxic payload, this finding may be of great interest for improving their therapeutic index by combining them with Bev, particularly for the therapy of colorectal cancer.
Collapse
Affiliation(s)
- Elodie Péraudeau
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
- CHU de Poitiers, 86021 Poitiers, France
| | - Brigitte Renoux
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
| | - Sheik Emambux
- CHU de Poitiers, 86021 Poitiers, France
- Department of Medical Oncology, Poitiers University Hospital, 86021 Poitiers, France
| | - Pauline Poinot
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
| | - Rémi Châtre
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
| | - Fabien Thoreau
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
| | - Benjamin Riss Yaw
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
| | - David Tougeron
- CHU de Poitiers, 86021 Poitiers, France
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, 86021 Poitiers, France
| | - Jonathan Clarhaut
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
- CHU de Poitiers, 86021 Poitiers, France
| | - Sébastien Papot
- Equipe Labellisée Ligue Contre le Cancer, Université de Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), 4 rue Michel-Brunet, TSA 51106, 86073 Poitiers, Cedex 9, France
| |
Collapse
|
39
|
Shukla VN, Vikas, Mehata AK, Setia A, Kumari P, Mahto SK, Muthu MS, Mishra SK. EGFR targeted albumin nanoparticles of oleanolic acid: In silico screening of nanocarrier, cytotoxicity and pharmacokinetics for lung cancer therapy. Int J Biol Macromol 2023; 246:125719. [PMID: 37419266 DOI: 10.1016/j.ijbiomac.2023.125719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/12/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
This study aimed to develop cetuximab (CTX) functionalized albumin nanoparticles (ALB-NPs) of oleanolic acid for EGFR targeted lung cancer therapy. The molecular docking methodology has been applied for a selection of suitable nanocarrier. Various physicochemical parameters like particle size, polydispersity, zeta potential, morphology, entrapment efficiency, and in-vitro drug release of all the ALB-NPs were analyzed. Furthermore, the in-vitro qualitative and quantitative cellular uptake study revealed that higher uptake of CTX conjugated ALB-NPs than nontargeted ALB-NPs in A549 cells. The in-vitro MTT assay revealed that the IC50 value of CTX-OLA-ALB-NPs (4.34 ± 1.90 μg/mL) was significantly reduced (p < 0.001) than OLA-ALB-NPs (13.87 ± 1.28 μg/mL) in A-549 cells. CTX-OLA-ALB-NPs caused apoptosis in A-549 cells at concentrations equivalent to its IC50 value and blocked the cell cycle in the G0/G1 phases. The hemocompatibility, histopathology and lung safety study confirmed the biocompatibility of the developed NPs. In vivo ultrasound and photoacoustic imaging confirmed the targeted delivery of the NPs to lung cancer. The findings demonstrated that CTX-OLA-ALB-NPs have potential for site-specific delivery of OLA for effective and targeted therapy of lung carcinoma.
Collapse
Affiliation(s)
- Vishwa Nath Shukla
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Vikas
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Aseem Setia
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Pooja Kumari
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Sanjeev Kumar Mahto
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India.
| | - Sunil Kumar Mishra
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India.
| |
Collapse
|
40
|
Li C, Zhang D, Pan Y, Chen B. Human Serum Albumin Based Nanodrug Delivery Systems: Recent Advances and Future Perspective. Polymers (Basel) 2023; 15:3354. [PMID: 37631411 PMCID: PMC10459149 DOI: 10.3390/polym15163354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
With the success of several clinical trials of products based on human serum albumin (HSA) and the rapid development of nanotechnology, HSA-based nanodrug delivery systems (HBNDSs) have received extensive attention in the field of nanomedicine. However, there is still a lack of comprehensive reviews exploring the broader scope of HBNDSs in biomedical applications beyond cancer therapy. To address this gap, this review takes a systematic approach. Firstly, it focuses on the crystal structure and the potential binding sites of HSA. Additionally, it provides a comprehensive summary of recent progresses in the field of HBNDSs for various biomedical applications over the past five years, categorized according to the type of therapeutic drugs loaded onto HSA. These categories include small-molecule drugs, inorganic materials and bioactive ingredients. Finally, the review summarizes the characteristics and current application status of HBNDSs in drug delivery, and also discusses the challenges that need to be addressed for the clinical transformation of HSA formulations and offers future perspectives in this field.
Collapse
Affiliation(s)
- Changyong Li
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China;
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Dagui Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Yujing Pan
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Biaoqi Chen
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China;
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| |
Collapse
|
41
|
Dou WT, Qiu P, Shi Y, Zhu L, Guo C, Li N, Zang Y, Liu T, Zhao S, Pan Y, Dong L, Sessler JL, Tan Y, Li J, Wang H, Tian H, He XP. Orthogonally Engineered Albumin with Attenuated Macrophage Phagocytosis for the Targeted Visualization and Phototherapy of Liver Cancer. J Am Chem Soc 2023; 145:17377-17388. [PMID: 37497917 DOI: 10.1021/jacs.3c05052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
The five-year survival rate of hepatocellular carcinoma (HCC) remains unsatisfactory. This reflects, in part, the paucity of effective methods that allow the target-specific diagnosis and therapy of HCC. Here, we report a strategy based on engineered human serum albumin (HSA) that permits the HCC-targeted delivery of diagnostic and therapeutic agents. Covalent cysteine conjugation combined with the exploitation of host-guest chemistry was used to effect the orthogonal functionalization of HSA with two functionally independent peptides. One of these peptides targets glypican-3 (GPC-3), an HCC-specific biomarker, while the second reduces macrophage phagocytosis through immune-checkpoint stimulation. This orthogonally engineered HSA proved effective for the GPC-3-targeted delivery of near-infrared fluorescent and phototherapeutic agents, thus permitting target-specific optical visualization and photodynamic ablation of HCC in vivo. This study thus offers new insights into specificity-enhanced fluorescence-guided surgery and phototherapy of HCC through the orthogonal engineering of biocompatible proteins.
Collapse
Affiliation(s)
- Wei-Tao Dou
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, P. R. China
| | - Peng Qiu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, P. R. China
| | - Yuanyuan Shi
- National Center for Liver Cancer, The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai 200433, P. R. China
| | - Ling Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, P. R. China
| | - Chen Guo
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, P. R. China
| | - Na Li
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, 333 Haike Rd, Pudong New District, Shanghai 201210, P. R. China
| | - Yi Zang
- National Centre for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd, Shanghai 201203, P. R. China
- Lingang laboratory, Shanghai 201203, P. R. China
| | - Tingting Liu
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Rd, Pudong New District, Shanghai 201210, P. R. China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Rd, Pudong New District, Shanghai 201210, P. R. China
| | - Yufei Pan
- National Center for Liver Cancer, The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai 200433, P. R. China
| | - Liwei Dong
- National Center for Liver Cancer, The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai 200433, P. R. China
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street-A5300, Austin, Texas 78712-1224, United States of America
| | - Yexiong Tan
- National Center for Liver Cancer, The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai 200433, P. R. China
| | - Jia Li
- National Centre for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd, Shanghai 201203, P. R. China
| | - Hongyang Wang
- National Center for Liver Cancer, The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai 200433, P. R. China
| | - He Tian
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, P. R. China
- National Center for Liver Cancer, The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, the Second Military Medical University, Shanghai 200433, P. R. China
| |
Collapse
|
42
|
Song X, Huang H, Xia L, Jia W, Yang S, Wang C, Chen Y. Engineering 2D Multienzyme-Mimicking Pyroptosis Inducers for Ultrasound-Augmented Catalytic Tumor Nanotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301279. [PMID: 37350357 PMCID: PMC10460896 DOI: 10.1002/advs.202301279] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/17/2023] [Indexed: 06/24/2023]
Abstract
Overcoming apoptosis resistance is necessary to ensure an effective cancer treatment; however, it is currently very difficult to achieve. A desirable alternative for cancer treatment is the targeted activation of pyroptosis, a unique type of programmed cell death. However, the pyroptosis inducers that are efficient for cancer therapy are limited. This work reports the engineering of 2D NiCoOx nanosheets as inducers of the production of harmful reactive oxygen species (ROS), which promote intense cell pyroptosis, and that can be applied to ultrasound (US)-augmented catalytic tumor nanotherapy. The main therapeutic task is carried out by the 2D NiCoOx nanosheets, which have four multienzyme-mimicking activities: peroxidase- (POD), oxidase- (OXD), glutathione peroxidase- (GPx), and catalase- (CAT) mimicking activities. These activities induce the reversal of the hypoxic microenvironment, endogenous glutathione depletion, and a continuous ROS output. The ROS-induced pyroptosis process is carried out via the ROS-NLRP3-GSDMD pathway, and the exogenous US activation boosts the multienzyme-mimicking activities and favors the incremental ROS generation, thus inducing mitochondrial dysfunction. The anti-cancer experimental results support the dominance of NiCoOx nanosheet-induced pyroptosis. This work expands on the biomedical applications of engineering 2D materials for US-augmented catalytic breast cancer nanotherapy and deepens the understanding of the multienzyme activities of nanomaterials.
Collapse
Affiliation(s)
- Xinran Song
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Hui Huang
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Lili Xia
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Wencong Jia
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| | - Shaoling Yang
- Department of Ultrasound MedicineShanghai Eighth People's HospitalShanghai200235P. R. China
| | - Chenglong Wang
- Department of Orthopedic SurgeryXinHua Hospital Affiliated with Shanghai Jiaotong University School of MedicineShanghai200082P. R. China
| | - Yu Chen
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
| |
Collapse
|
43
|
Wang L, Wang X, Wu Y, Wang J, Zhou W, Wang J, Guo H, Zhang N, Zhang L, Hu X, Zhao Y, Miao J, Zhang Z, Chard Dunmall LS, Zhang D, Lemoine NR, Cheng Z, Wang Y. A novel microenvironment regulated system CAR-T (MRS.CAR-T) for immunotherapeutic treatment of esophageal squamous carcinoma. Cancer Lett 2023; 568:216303. [PMID: 37422126 DOI: 10.1016/j.canlet.2023.216303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/23/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023]
Abstract
Chimeric antigen receptor T cell immunotherapy has achieved promising therapeutic effects in the treatment of hematological malignancies. However, there are still many obstacles, including on-target off-tumor antigen expression, that prevent successful application to solid tumors. We designed a tumor microenvironment (TME) regulated system chimeric antigen receptor T (MRS.CAR-T) which can only be auto-activated in the solid TME. B7-H3 was selected as the target antigen for esophageal carcinoma. An element comprising a human serum albumin (HSA) binding peptide and a matrix metalloproteases (MMPs) cleavage site was inserted between the 5' terminal signal peptide and single chain fragment variable (scFv) of the CAR skeleton. Upon administration, HSA bound the binding peptide in MRS.B7-H3.CAR-T effectively and promoted proliferation and differentiation into memory cells. MRS.B7-H3.CAR-T was not cytotoxic in normal tissues expressing B7-H3 as the antigen recognition site in the scFv was cloaked by HSA. The anti-tumor function of MRS.B7-H3.CAR-T was recovered once the cleavage site was cleaved by MMPs in the TME. The anti-tumor efficacy associated with MRS.B7-H3.CAR-T cells was improved compared to classic B7-H3.CAR-T cells in vitro and less IFN-γ was released, suggesting a treatment that may induce less extent of cytokine release syndrome-mediated toxicity. In vivo, MRS.B7-H3.CAR-T cells had strong anti-tumor activity and were safe. MRS.CAR-T represents a novel strategy to improve the efficacy and safety of CAR-T therapy in solid tumors.
Collapse
Affiliation(s)
- Lihong Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaosa Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yangyang Wu
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingjing Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wenping Zhou
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianyao Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Haoran Guo
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Zhang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lufang Zhang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuanyu Hu
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Zhao
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Zifang Zhang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Danhua Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Nicholas R Lemoine
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China; Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China; Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
44
|
Paladino O, Moranda A, Falugi C. Spatiotemporal role of muscarinic signaling in early chick development: exposure to cholinomimetic agents by a mathematical model. Cell Biol Toxicol 2023; 39:1453-1469. [PMID: 36098822 PMCID: PMC10425487 DOI: 10.1007/s10565-022-09770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/26/2022] [Indexed: 11/02/2022]
Abstract
Awareness is growing that, besides several neurotoxic effects, cholinomimetic drugs able to interfere the cholinergic neurotransmitter system may exert a teratogen effect in developing embryos of vertebrate and invertebrate organisms. Cholinomimetic substances exert their toxic activity on organisms as they inhibit the functionality of the cholinergic system by completely or partially replacing the ACh molecule both at the level of the AChE active site and at the level of acetylcholine receptors. In this work, we focused the attention on the effects of muscarinic antagonist (atropine) and agonist (carbachol) drugs during the early development and ontogenesis of chick embryos. An unsteady-state mathematical model of the drug release and fate was developed, to synchronize exposure to a gradient of drug concentrations with the different developmental events. Since concentration measures in time and space cannot be taken without damaging the embryo itself, the diffusion model was the only way to establish at each time-step the exact concentration of drug at the different points of the embryo body (considered two-dimensional up to the 50 h stage). This concentration depends on the distance and position of the embryo with respect to the releasing source. The exposure to carbachol generally enhanced dimensions and stages of the embryos, while atropine mainly caused delay in development and small size of the embryos. Both the drugs were able to cause developmental anomalies, depending on the moment of development, in a time- and dose-dependent way, regardless the expression of genes driving each event. 1. Early chick embryos were exposed to muscarinic drugs in a spatial-temporal context. 2. Effects were stage-(time) dependent, according to distance and position of the source. 3. Atropine inhibited growth, mainly interfering with the cephalic process formation and heart differentiation; carbachol increased growth reducing differentiation. 4. Interferences may be exerted by alteration of calcium responses to naturally occurring morphogen-driven mechanisms.
Collapse
Affiliation(s)
- Ombretta Paladino
- Department of Civil, Chemical and Environmental Engineering (DICCA), University of Genoa, Via Opera Pia 15, 16145 Genoa, Italy
| | - Arianna Moranda
- Department of Civil, Chemical and Environmental Engineering (DICCA), University of Genoa, Via Opera Pia 15, 16145 Genoa, Italy
| | - Carla Falugi
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Corso Europa 26, 16132 Genoa, Italy
| |
Collapse
|
45
|
Fazal T, Murtaza BN, Shah M, Iqbal S, Rehman MU, Jaber F, Dera AA, Awwad NS, Ibrahium HA. Recent developments in natural biopolymer based drug delivery systems. RSC Adv 2023; 13:23087-23121. [PMID: 37529365 PMCID: PMC10388836 DOI: 10.1039/d3ra03369d] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023] Open
Abstract
Targeted delivery of drug molecules to diseased sites is a great challenge in pharmaceutical and biomedical sciences. Fabrication of drug delivery systems (DDS) to target and/or diagnose sick cells is an effective means to achieve good therapeutic results along with a minimal toxicological impact on healthy cells. Biopolymers are becoming an important class of materials owing to their biodegradability, good compatibility, non-toxicity, non-immunogenicity, and long blood circulation time and high drug loading ratio for both macros as well as micro-sized drug molecules. This review summarizes the recent trends in biopolymer-based DDS, forecasting their broad future clinical applications. Cellulose chitosan, starch, silk fibroins, collagen, albumin, gelatin, alginate, agar, proteins and peptides have shown potential applications in DDS. A range of synthetic techniques have been reported to design the DDS and are discussed in the current study which is being successfully employed in ocular, dental, transdermal and intranasal delivery systems. Different formulations of DDS are also overviewed in this review article along with synthesis techniques employed for designing the DDS. The possibility of these biopolymer applications points to a new route for creating unique DDS with enhanced therapeutic qualities for scaling up creative formulations up to the clinical level.
Collapse
Affiliation(s)
- Tanzeela Fazal
- Department of Chemistry, Abbottabad University of Science and Technology Pakistan
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology Pakistan
| | - Mazloom Shah
- Department of Chemistry, Faculty of Science, Grand Asian University Sialkot Pakistan
| | - Shahid Iqbal
- Department of Chemistry, School of Natural Sciences (SNS), National University of Science and Technology (NUST) H-12 Islamabad 46000 Pakistan
| | - Mujaddad-Ur Rehman
- Department of Microbiology, Abbottabad University of Science & Technology Pakistan
| | - Fadi Jaber
- Department of Biomedical Engineering, Ajman University Ajman UAE
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University Ajman UAE
| | - Ayed A Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University Abha Saudi Arabia
| | - Nasser S Awwad
- Chemistry Department, Faculty of Science, King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| | - Hala A Ibrahium
- Biology Department, Faculty of Science, King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| |
Collapse
|
46
|
Yurt F, Özel D, Tunçel A, Gokbayrak O, Aktas S. Synthesis and Optimization of the Docetaxel-Loaded and Durvalumab-Targeted Human Serum Albumin Nanoparticles, In Vitro Characterization on Triple-Negative Breast Cancer Cells. ACS OMEGA 2023; 8:26287-26300. [PMID: 37521641 PMCID: PMC10372957 DOI: 10.1021/acsomega.3c02682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Triple-negative breast cancer (TNBC) tends to behave more aggressively compared to other breast cancer subtypes due to the lack of receptors and its limited targeting therapy. In recent years, nanotechnology advancement has led to the development of various nanoparticle platforms for the targeted treatment of cancers. Especially, HSA-NPs have specific advantages such as biocompatibility, adjustable size during production, and relatively easy synthesis. In this study, HSA-NPs were encapsulated with docetaxel (DTX) and functionalized with polyethylene glycol (PEG), also becoming a targeting nanoplatform modified with durvalumab (DVL), and the whole nanostructure was well characterized. Subsequently, drug release studies and various in vitro cell culture studies such as determining the cytotoxicity and apoptotic levels of the nanoplatforms and PD-L1 using ELISA test were conducted on MDA-MB-468, MDA-MB-231, and MCF-7 cells. According to the results, HSA-DTX@PEG-DVL NPs showed better cytotoxicity compared to DTX in all the three cell lines. In addition, it was observed that the HSA-DTX@PEG-DVL NPs did not lead the cells to late apoptosis but were effective in the early apoptotic stage. Moreover, the ELISA data showed a significantly induced PD-L1 expression due to the presence of DVL in the nanostructure, which indicates that DVL antibodies successfully bind to the HSA-DTX@PEG-DVL nanostructure.
Collapse
Affiliation(s)
- Fatma Yurt
- Department
of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Derya Özel
- Department
of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Ayça Tunçel
- Department
of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Ozde Gokbayrak
- Department
of Basic Oncology, Institute of Oncology, DokuzEylül University, 35340 Izmir, Turkey
| | - Safiye Aktas
- Department
of Basic Oncology, Institute of Oncology, DokuzEylül University, 35340 Izmir, Turkey
| |
Collapse
|
47
|
Traverso AN, Fragale DJ, Viale DL, Garate O, Torres P, Valverde G, Berra A, Torbidoni AV, Yakisich JS, Grasselli M, Radrizzani M. Two-Step Preparation of Protein-Decorated Biohybrid Quantum Dot Nanoparticles for Cellular Uptake. Pharmaceutics 2023; 15:1651. [PMID: 37376099 DOI: 10.3390/pharmaceutics15061651] [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: 04/27/2023] [Revised: 05/26/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Decoration of nanoparticles with specific molecules such as antibodies, peptides, and proteins that preserve their biological properties is essential for the recognition and internalization of their specific target cells. Inefficient preparation of such decorated nanoparticles leads to nonspecific interactions diverting them from their desired target. We report a simple two-step procedure for the preparation of biohybrid nanoparticles containing a core of hydrophobic quantum dots coated with a multilayer of human serum albumin. These nanoparticles were prepared by ultra-sonication, crosslinked using glutaraldehyde, and decorated with proteins such as human serum albumin or human transferrin in their native conformations. These nanoparticles were homogeneous in size (20-30 nm), retained the fluorescent properties of quantum dots, and did not show a "corona effect" in the presence of serum. The uptake of transferrin-decorated quantum dot nanoparticles was observed in A549 lung cancer and SH-SY5Y neuroblastoma cells but not in non-cancerous 16HB14o- or retinoic acid dopaminergic neurons differentiated SH-SY5Y cells. Furthermore, digitoxin-loaded transferrin-decorated nanoparticles decreased the number of A549 cells without effect on 16HB14o-. Finally, we analyzed the in vivo uptake of these biohybrids by murine retinal cells, demonstrating their capacity to selectively target and deliver into specific cell types with excellent traceability.
Collapse
Affiliation(s)
- Agata Noelia Traverso
- Neuro and Molecular Cytogenetics Laboratory, Institute of Emerging Technologies and Applied Sciences (ITECA), National Council for Scientific and Technical Research (CONICET), School of Science and Technology, National University of San Martín, Av. Gral. Paz 5445, San Martín B1650, Argentina
| | - David José Fragale
- Neuro and Molecular Cytogenetics Laboratory, Institute of Emerging Technologies and Applied Sciences (ITECA), National Council for Scientific and Technical Research (CONICET), School of Science and Technology, National University of San Martín, Av. Gral. Paz 5445, San Martín B1650, Argentina
| | - Diego Luis Viale
- Neuro and Molecular Cytogenetics Laboratory, Institute of Emerging Technologies and Applied Sciences (ITECA), National Council for Scientific and Technical Research (CONICET), School of Science and Technology, National University of San Martín, Av. Gral. Paz 5445, San Martín B1650, Argentina
| | - Octavio Garate
- Nanomateriales Funcionales, INTI-Micro y Nanotecnología, Instituto Nacional de Tecnología Industrial, San Martín B1650, Argentina
| | - Pablo Torres
- Science and Technology Institute Cesar Milstein, Fundación Pablo Cassará-National Council for Scientific and Technical Research (CONICET) Saladillo 2452, Ciudad Autónoma de Buenos Aires C1440, Argentina
| | - Gastón Valverde
- Translational Laboratory of Immunopathology and Ophthalmology, Department of Pathology, Faculty of Medicine, Universidad de Buenos Aires, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1121, Argentina
| | - Alejandro Berra
- Translational Laboratory of Immunopathology and Ophthalmology, Department of Pathology, Faculty of Medicine, Universidad de Buenos Aires, Paraguay 2155, Ciudad Autónoma de Buenos Aires C1121, Argentina
| | - Ana Vanesa Torbidoni
- Laboratorio de Biología Celular y Molecular, Instituto Argentino de Veterinaria, Ambiente y Salud (IAVAS) Universidad Juan Agustín Maza (UMaza), Mendoza M5519, Argentina
| | - Juan Sebastián Yakisich
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23693, USA
| | - Mariano Grasselli
- Biotechnological Materials Laboratory (LaMaBio), Department of Science and Technology, National University of Quilmes, GBEyB, Grupo Vinculado IMBICE-CONICET, Roque Sáenz Peña 352, Buenos Aires B1876, Argentina
| | - Martín Radrizzani
- Neuro and Molecular Cytogenetics Laboratory, Institute of Emerging Technologies and Applied Sciences (ITECA), National Council for Scientific and Technical Research (CONICET), School of Science and Technology, National University of San Martín, Av. Gral. Paz 5445, San Martín B1650, Argentina
| |
Collapse
|
48
|
Sun Q, Yang Z, Qi X. Design and Application of Hybrid Polymer-Protein Systems in Cancer Therapy. Polymers (Basel) 2023; 15:polym15092219. [PMID: 37177365 PMCID: PMC10181109 DOI: 10.3390/polym15092219] [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: 02/28/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Polymer-protein systems have excellent characteristics, such as non-toxic, non-irritating, good water solubility and biocompatibility, which makes them very appealing as cancer therapeutics agents. Inspiringly, they can achieve sustained release and targeted delivery of drugs, greatly improving the effect of cancer therapy and reducing side effects. However, many challenges, such as reducing the toxicity of materials, protecting the activities of proteins and controlling the release of proteins, still need to be overcome. In this review, the design of hybrid polymer-protein systems, including the selection of polymers and the bonding forms of polymer-protein systems, is presented. Meanwhile, vital considerations, including reaction conditions and the release of proteins in the design process, are addressed. Then, hybrid polymer-protein systems developed in the past decades for cancer therapy, including targeted therapy, gene therapy, phototherapy, immunotherapy and vaccine therapy, are summarized. Furthermore, challenges for the hybrid polymer-protein systems in cancer therapy are exemplified, and the perspectives of the field are covered.
Collapse
Affiliation(s)
- Qi Sun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
| | - Zhenzhen Yang
- Drug Clinical Trial Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Xianrong Qi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
49
|
Khan I, Gril B, Paranjape AN, Robinson CM, Difilippantonio S, Biernat W, Bieńkowski M, Pęksa R, Duchnowska R, Jassem J, Brastianos PK, Metellus P, Bialecki E, Woodroofe CC, Wu H, Swenson RE, Steeg PS. Comparison of Three Transcytotic Pathways for Distribution to Brain Metastases of Breast Cancer. Mol Cancer Ther 2023; 22:646-658. [PMID: 36912773 PMCID: PMC10164055 DOI: 10.1158/1535-7163.mct-22-0815] [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: 12/20/2022] [Revised: 02/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Advances in drug treatments for brain metastases of breast cancer have improved progression-free survival but new, more efficacious strategies are needed. Most chemotherapeutic drugs infiltrate brain metastases by moving between brain capillary endothelial cells, paracellular distribution, resulting in heterogeneous distribution, lower than that of systemic metastases. Herein, we tested three well-known transcytotic pathways through brain capillary endothelial cells as potential avenues for drug access: transferrin receptor (TfR) peptide, low-density lipoprotein receptor 1 (LRP1) peptide, albumin. Each was far-red labeled, injected into two hematogenous models of brain metastases, circulated for two different times, and their uptake quantified in metastases and uninvolved (nonmetastatic) brain. Surprisingly, all three pathways demonstrated distinct distribution patterns in vivo. Two were suboptimal: TfR distributed to uninvolved brain but poorly in metastases, while LRP1 was poorly distributed. Albumin distributed to virtually all metastases in both model systems, significantly greater than in uninvolved brain (P < 0.0001). Further experiments revealed that albumin entered both macrometastases and micrometastases, the targets of treatment and prevention translational strategies. Albumin uptake into brain metastases was not correlated with the uptake of a paracellular probe (biocytin). We identified a novel mechanism of albumin endocytosis through the endothelia of brain metastases consistent with clathrin-independent endocytosis (CIE), involving the neonatal Fc receptor, galectin-3, and glycosphingolipids. Components of the CIE process were found on metastatic endothelial cells in human craniotomies. The data suggest a reconsideration of albumin as a translational mechanism for improved drug delivery to brain metastases and possibly other central nervous system (CNS) cancers.In conclusion, drug therapy for brain metastasis needs improvement. We surveyed three transcytotic pathways as potential delivery systems in brain-tropic models and found that albumin has optimal properties. Albumin used a novel endocytic mechanism.
Collapse
Affiliation(s)
- Imran Khan
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Brunilde Gril
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Anurag N. Paranjape
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Christina M. Robinson
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD
| | | | | | - Rafał Pęksa
- Department of Pathology, Medical University of Gdańsk, Poland
| | - Renata Duchnowska
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Jacek Jassem
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Poland
| | - Priscilla K. Brastianos
- Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Philippe Metellus
- Ramsay Général de Santé, Hôpital Privé Clairval, Département de Neurochirurgie and Aix-Marseille Université, Institut de Neurophysiopathologie – UMR 7051, Marseille, France
| | - Emilie Bialecki
- Ramsay Général de Santé, Hôpital Privé Clairval, Département de Neurochirurgie and Aix-Marseille Université, Institut de Neurophysiopathologie – UMR 7051, Marseille, France
| | - Carolyn C. Woodroofe
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD
| | - Haitao Wu
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD
| | - Patricia S. Steeg
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
50
|
He Y, Yang M, Yang L, Hao M, Wang F, Li X, Taylor EW, Zhang X, Zhang J. Preparation and anticancer actions of CuET-nanoparticles dispersed by bovine serum albumin. Colloids Surf B Biointerfaces 2023; 226:113329. [PMID: 37156027 DOI: 10.1016/j.colsurfb.2023.113329] [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: 02/02/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Diethyldithiocarbamate-copper complex (CuET) shows promising anticancer effect; nonetheless, preclinical evaluations of CuET are hindered due to poor solubility. We prepared bovine serum albumin (BSA)-dispersed CuET nanoparticles (CuET-NPs) to overcome the shortcoming. Results from a cell-free redox system demonstrated that CuET-NPs reacted with glutathione, leading to form hydroxyl radical. Glutathione-mediated production of hydroxyl radicals may help explain why CuET selectively kills drug-resistant cancer cells with higher levels of glutathione. CuET-NPs dispersed by autoxidation products of green tea epigallocatechin gallate (EGCG) also reacted with glutathione; however, the autoxidation products eradicated hydroxyl radicals; consequently, such CuET-NPs exhibited largely compromised cytotoxicity, suggesting that hydroxyl radical is a crucial mediator of CuET anticancer activity. In cancer cells, BSA-dispersed CuET-NPs exhibited cytotoxic activities equivalent to CuET and induced protein poly-ubiquitination. Moreover, the reported powerful inhibition of CuET on colony formation and migration of cancer cells could be replicated by CuET-NPs. These similarities demonstrate BSA-dispersed CuET-NPs is identical to CuET. Thus, we advanced to pilot toxicological and pharmacological evaluations. CuET-NPs caused hematologic toxicities in mice and induced protein poly-ubiquitination and apoptosis of cancer cells inoculated in mice at a defined pharmacological dose. Given high interest in CuET and its poor solubility, BSA-dispersed CuET-NPs pave the way for preclinical evaluations.
Collapse
Affiliation(s)
- Yufeng He
- Laboratory of Redox Biology, State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei 230036, China; Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 310008, China
| | - Mingchuan Yang
- Laboratory of Redox Biology, State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei 230036, China
| | - Lumin Yang
- Laboratory of Redox Biology, State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei 230036, China
| | - Meng Hao
- Laboratory of Redox Biology, State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei 230036, China
| | - Fuming Wang
- Laboratory of Redox Biology, State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei 230036, China
| | - Xiuli Li
- Laboratory of Redox Biology, State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei 230036, China
| | - Ethan Will Taylor
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | - Xiangchun Zhang
- Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 310008, China.
| | - Jinsong Zhang
- Laboratory of Redox Biology, State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|