1
|
Olivieri PH, Assis IF, Lima AF, Hassan SA, Torquato RJS, Hayashi JY, Tashima AK, Nader HB, Salvati A, Justo GZ, Sousa AA. Glycocalyx Interactions Modulate the Cellular Uptake of Albumin-Coated Nanoparticles. ACS APPLIED BIO MATERIALS 2024. [PMID: 39470630 DOI: 10.1021/acsabm.4c01012] [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: 10/30/2024]
Abstract
Albumin-based nanoparticles (ABNPs) represent promising drug carriers in nanomedicine due to their versatility and biocompatibility, but optimizing their effectiveness in drug delivery requires understanding their interactions with and uptake by cells. Notably, albumin interacts with the cellular glycocalyx, a phenomenon particularly studied in endothelial cells. This observation suggests that the glycocalyx could modulate ABNP uptake and therapeutic efficacy, although this possibility remains unrecognized. In this study, we elucidate the critical role of the glycocalyx in the cellular uptake of a model ABNP system consisting of silica nanoparticles (NPs) coated with native, cationic, and anionic albumin variants (BSA, BSA+, and BSA-). Using various methodologies-including fluorescence anisotropy, dynamic light scattering, microscale thermophoresis, surface plasmon resonance spectroscopy, and computer simulations─we found that both BSA and BSA+, but not BSA-, interact with heparin, a model glycosaminoglycan (GAG). To explore the influence of albumin-GAG interactions on NP uptake, we performed comparative uptake studies in wild-type and GAG-mutated Chinese hamster ovary cells (CHO), along with complementary approaches such as enzymatic GAG cleavage in wild-type cells, chemical inhibition, and competition assays with exogenous heparin. We found that the glycocalyx enhances the cell uptake of NPs coated with BSA and BSA+, while serving as a barrier to the uptake of NPs coated with BSA-. Furthermore, we showed that harnessing albumin-GAG interactions increases cancer cell death induced by paclitaxel-loaded albumin-coated NPs. These findings underscore the importance of albumin-glycocalyx interactions in the rational design and optimization of albumin-based drug delivery systems.
Collapse
Affiliation(s)
- Paulo H Olivieri
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Isabela F Assis
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Andre F Lima
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Sergio A Hassan
- Bioinformatics and Computational Biosciences Branch, OCICB, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ricardo J S Torquato
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Jackelinne Y Hayashi
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Alexandre K Tashima
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Helena B Nader
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Anna Salvati
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Giselle Z Justo
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| | - Alioscka A Sousa
- Department of Biochemistry, Federal University of São Paulo, São Paulo, São Paulo 04044-020, Brazil
| |
Collapse
|
2
|
Zhu S, Wu Q, Ying Y, Mao Y, Lu W, Xu J, Cai X, He H, Wu J. Tissue-Adaptive BSA Hydrogel with Dual Release of PTX and bFGF Promotes Spinal Cord Injury Repair via Glial Scar Inhibition and Axon Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2401407. [PMID: 39385643 DOI: 10.1002/smll.202401407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Spinal cord injury (SCI) is a severe clinical disease usually accompanied by activated glial scar, neuronal axon rupture, and disabled motor function. To mimic the microenvironment of the SCI injury site, a hydrogel system with a comparable mechanical property to the spinal cord is desirable. Therefore, a novel elastic bovine serum albumin (BSA) hydrogel is fabricated with excellent adhesive, injectable, and biocompatible properties. The hydrogel is used to deliver paclitaxel (PTX) together with basic fibroblast growth factor (bFGF) to inhibit glial scar formation as well as promote axon regeneration and motor function for SCI repair. Due to the specific interaction of BSA with both drugs, bFGF, and PTX can be controllably released from the hydrogel system to achieve an effective concentration at the wound site during the SCI regeneration process. Moreover, benefiting from the combination of PTX and bFGF, this bFGF/PTX@BSA system significantly aided axon repair by promoting the elongation of axons across the glial scar with reduced reactive astrocyte secretion. In addition, remarkable anti-apoptosis of nerve cells is evident with the bFGF/PTX@BSA system. Subsequently, this multi-functionalized drug system significantly improved the motor function of the rats after SCI. These results reveal that bFGF/PTX@BSA is an ideal functionalized material for nerve repair in SCI.
Collapse
Affiliation(s)
- Sipin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Qiuji Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yibo Ying
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuqin Mao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenjie Lu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiong Cai
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huacheng He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Jiang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
3
|
Gong Z, Fu Y, Gao Y, Jiao F, Su Q, Sang X, Chen B, Deng X, Liu X. "Abraxane-Like" Radiosensitizer for In Situ Oral Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309569. [PMID: 38973195 PMCID: PMC11425904 DOI: 10.1002/advs.202309569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/06/2024] [Indexed: 07/09/2024]
Abstract
Radiotherapy plays a vital role in cancer therapy. However, the hypoxic microenvironment of tumors greatly limits the effectiveness, thus it is crucial to develop a simple, efficient, and safe radiosensitizer to reverse hypoxia and ameliorate the efficacy of radiotherapy. Inspired by the structure of canonical nanodrug Abraxane, herein, a native HSA-modified CaO2 nanoparticle system (CaO2-HSA) prepared by biomineralization-induced self-assembly is developed. CaO2-HSA will accumulate in tumor tissue and decompose to produce oxygen, altering the hypoxic condition inside the tumor. Simultaneously, ROS and calcium ions will lead to calcium overload and further trigger immunogenic cell death. Notably, its sensitizing enhancement ratio (SER = 3.47) is much higher than that of sodium glycididazole used in the clinic. Furthermore, in animal models of in situ oral cancer, CaO2-HSA can effectively inhibit tumor growth. With its high efficacy, facile preparation, and heavy-metal free biosafety, the CaO2-HSA-based radiosensitizer holds enormous potential for oral cancer therapy.
Collapse
Affiliation(s)
- Zijian Gong
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yixuan Fu
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yuan Gao
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Fei Jiao
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Qinzhi Su
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiao Sang
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Binglin Chen
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Biomedical Engineering DepartmentPeking UniversityBeijing100191P. R. China
| | - Xinyu Liu
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
4
|
Khakpour S, Hosano N, Moosavi-Nejad Z, Farajian AA, Hosano H. Advancing Tumor Therapy: Development and Utilization of Protein-Based Nanoparticles. Pharmaceutics 2024; 16:887. [PMID: 39065584 PMCID: PMC11279530 DOI: 10.3390/pharmaceutics16070887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Protein-based nanoparticles (PNPs) in tumor therapy hold immense potential, combining targeted delivery, minimal toxicity, and customizable properties, thus paving the way for innovative approaches to cancer treatment. Understanding the various methods available for their production is crucial for researchers and scientists aiming to harness these nanoparticles for diverse applications, including tumor therapy, drug delivery, imaging, and tissue engineering. This review delves into the existing techniques for producing PNPs and PNP/drug complexes, while also exploring alternative novel approaches. The methods outlined in this study were divided into three key categories based on their shared procedural steps: solubility change, solvent substitution, and thin flow methods. This classification simplifies the understanding of the underlying mechanisms by offering a clear framework, providing several advantages over other categorizations. The review discusses the principles underlying each method, highlighting the factors influencing the nanoparticle size, morphology, stability, and functionality. It also addresses the challenges and considerations associated with each method, including the scalability, reproducibility, and biocompatibility. Future perspectives and emerging trends in PNPs' production are discussed, emphasizing the potential for innovative strategies to overcome current limitations, which will propel the field forward for biomedical and therapeutic applications.
Collapse
Affiliation(s)
- Shirin Khakpour
- Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan;
| | - Nushin Hosano
- Department of Biomaterials and Bioelectrics, Institute of Industrial Nanomaterials, Kumamoto University, Kumamoto 860-8555, Japan;
| | - Zahra Moosavi-Nejad
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran 1993893973, Iran
| | - Amir A. Farajian
- Department of Mechanical and Materials Engineering, Wright State University, Dayton, OH 45435, USA;
| | - Hamid Hosano
- Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan;
- Department of Biomaterials and Bioelectrics, Institute of Industrial Nanomaterials, Kumamoto University, Kumamoto 860-8555, Japan;
| |
Collapse
|
5
|
Mukai R, Okuyama H, Uchimura M, Sakao K, Matsuhiro M, Ikeda-Imafuku M, Ishima Y, Nishikawa M, Ikushiro S, Tai A. The binding selectivity of quercetin and its structure-related polyphenols to human serum albumin using a fluorescent dye cocktail for multiplex drug-site mapping. Bioorg Chem 2024; 145:107184. [PMID: 38364549 DOI: 10.1016/j.bioorg.2024.107184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 02/18/2024]
Abstract
Human serum albumin (HSA) is a serum protein that carries flavonoids in blood circulation. In this report, the binding selectivity and strength of interactions to HSA-binding sites (sites I or II) by flavonoids were evaluated using competition experiments and the specific fluorescent dyes, dansylamide and BD140. Most tested flavonoids bound site I preferentially, with the binding strength dependent on the mother structure in the order flavonol > flavone > flavanone > flavan 3-ols. Glycosylation or glucuronidation reduced the binding of quercetin to site I of HSA, whereas sulfation increased binding. Quercetin 7-sulfate showed the strongest binding and molecular docking simulations supported this observation. Prenylation at any position or glucuronidation and sulfation at the C-4' or C-7 position of quercetin facilitated stronger binding to site II. The binding affinity of flavonoids toward site I correlated with the partition coefficient value (logP), whereas no corresponding correlation was observed for site II.
Collapse
Affiliation(s)
- Rie Mukai
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan.
| | - Hitomi Okuyama
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan
| | - Miku Uchimura
- Department of Food Science and Technology, Graduate School of Agriculture, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-0065, Japan.
| | - Kozue Sakao
- Department of Food Science and Technology, Graduate School of Agriculture, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-0065, Japan.
| | - Miyu Matsuhiro
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan.
| | - Mayumi Ikeda-Imafuku
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-cho, Wakayama 640-8156, Japan.
| | - Yu Ishima
- Laboratory of Biopharmaceutics, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Miyu Nishikawa
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan.
| | - Shinichi Ikushiro
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan.
| | - Akihiro Tai
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan.
| |
Collapse
|
6
|
Akbari V, Ghobadi S. Evaluation of the effect of phenylpropanoids on the binding of heparin to human serum albumin and glycosylated human serum albumin concerning anticoagulant activity: A comparison study. Int J Biol Macromol 2024; 257:128732. [PMID: 38092116 DOI: 10.1016/j.ijbiomac.2023.128732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
The nonenzymatic advanced glycation end products (AGEs) and the accumulation of AGEs are the two main factors associated with the long-term pathogenesis of diabetes. Human serum albumin (HSA) as the most abundant serum protein has a higher fortuity to be modified by nonenzymatic glycation. In this study, the interaction of three phenylpropanoids (caffeic acid (Caf), p-coumaric acid (Cou), and cinnamic acid (Cin)) toward HSA and glycosylated HSA (gHSA) was analyzed by multiple spectroscopic techniques combined with molecular docking. The formation of fibrils in HSA and gHSA was confirmed by the Thioflavin T (ThT) assay. The phenylpropanoids have shown anti-fibrillation properties in vitro. The obtained thermodynamic parameters indicated that hydrogen bonding and van der Waals forces are the main forces in the binding interaction, and the quenching mechanism of the protein fluorescence is static. Molecular docking results, as well as the in vitro results, showed that Caf, Cou, and Cin exhibit more stable interactions with HSA, respectively. In addition, molecular docking analysis showed that Caf and Cou interact well with K199. Given the critical role of K199 in HSA glycosylation in diabetic patients, this process inhibits the interaction of stabilizer compounds and thus accelerates gHSA aggregation.
Collapse
Affiliation(s)
- Vali Akbari
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran.
| | - Sirous Ghobadi
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran.
| |
Collapse
|
7
|
Digklia A, Kollár A, Dietrich D, Kronig MN, Britschgi C, Rordorf T, Joerger M, Krasniqi F, Metaxas Y, Colombo I, Ribi K, Rothermundt C. SAKK57/16 Nab-paclitaxel and Gemcitabine in Soft Tissue Sarcoma (NAPAGE): a phase I/II trial. Eur J Cancer 2024; 197:113470. [PMID: 38096656 DOI: 10.1016/j.ejca.2023.113470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/02/2024]
Abstract
BACKGROUND To determine whether the combination of nab-paclitaxel with gemcitabine has activity in patients with pretreated soft tissue sarcoma (STS). PATIENTS AND METHODS NAPAGE is a phase Ib/II clinical trial investigating the combination of nab-paclitaxel (nab-pc) with gemcitabine employing two cohorts. One of a dose-de-escalation phase and one of expansion. In phase I, nab-pc was given at 150 mg/m2 in combination with gemcitabine 1000 mg/m2 every two weeks, until disease progression or unacceptable toxicity. This dose was recommended for phase II (RP2D), as there was no dose limiting toxicity (DLT) or discontinuations due to adverse events (AEs). The primary endpoint of the phase II was progression-free rate (PFR) at 3 months (H0: 20%, H1:40%). The secondary endpoints included progression free survival (PFS), overall survival (OS), AEs, objective response and patient-reported outcomes (PRO). Efficacy analysis was by intention to treat. RESULTS The 3-month PFR was 56.4% (95% confidence interval CI: 39.6-72.2%). The 3-month and 6-month PFS were 58.4% (95% CI: 41.3-72.1%) and 44.6% (95% CI: 28.4-59.5%), respectively. Median PFS was 5.3 months (95% CI: 1.4-8.2) and median OS was 12.8 months (95% CI: 10.5-39.2). The most common treatment-related grade ≥ 3 AE were neutropenia (18%), followed by anemia (2.6%), hypertension (2.6%) and alanine aminotransferase increase (2.6%). Grade 1 and grade 2 peripheral sensory neuropathy (PNP) occurred in 15.4% and 20.5%, respectively. No grade 3-4 PNP was reported. CONCLUSIONS Combining nab-pc and gemcitabine is safe. Promising activity is observed in pretreated STS patients with manageable toxicity. This regimen should be considered for further exploration.
Collapse
Affiliation(s)
- A Digklia
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.
| | - A Kollár
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - D Dietrich
- Swiss Group for Clinical Cancer Research (SAKK) Competence Center, Bern, Switzerland
| | - M N Kronig
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - C Britschgi
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - T Rordorf
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - M Joerger
- Department of Medical Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - F Krasniqi
- Department of Medical Oncology, University Hospital of Basel, Basel, Switzerland
| | - Y Metaxas
- Department of Medical Oncology, Cantonal Hospital, Grison Chur, Switzerland, now at Cantonal Hospital Muensterlingen, Muensterlingen, Switzerland
| | - I Colombo
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - K Ribi
- International Breast Cancer Study Group IBCSG (IBCSG), Bern, Switzerland
| | - C Rothermundt
- Department of Medical Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
8
|
Su Y, Gao J, Dong X, Wheeler KA, Wang Z. Neutrophil-Mediated Delivery of Nanocrystal Drugs via Photoinduced Inflammation Enhances Cancer Therapy. ACS NANO 2023; 17:15542-15555. [PMID: 37577982 PMCID: PMC10480050 DOI: 10.1021/acsnano.3c02013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The efficient delivery of anticancer agents into tumor microenvironments is critical for the success of cancer therapies, but it is a prerequisite that drug carriers should overcome tumor vasculature and possess high drug contents. Here, we found that photoinduced inflammation response caused the migration of neutrophils into tumor microenvironments and neutrophils transported neutrophil-targeted nanoparticles (NPs) across the tumor blood barrier. The results showed that tumor delivery efficiencies of NPs were 5% ID/g, and they were independent of particle sizes (30-200 nm) and their doses (108-1011 NPs). To efficiently deliver anticancer agents into tumors via neutrophils, we fabricated carrier-free paclitaxel nanocrystals (PTX NC). The results showed that neutrophil uptake of PTX NC did not impair neutrophil tumor infiltration, and the sustainable release of PTX from PTX NC in tumors was regulated by paclitaxel protein complexes, thus improving the mouse survival in two preclinical models. Our studies demonstrate that delivery of nanocrystal drugs via neutrophils is a promising method to effectively treat a wide range of cancers, and we have also identified a mechanism of drug release from neutrophils in tumors.
Collapse
Affiliation(s)
- Yujie Su
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Jin Gao
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Xinyue Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Kraig A Wheeler
- Department of Chemistry, Whitworth University, Spokane, Washington 99251, United States
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| |
Collapse
|
9
|
Hueppe N, Wurm FR, Landfester K. Nanocarriers with Multiple Cargo Load-A Comprehensive Preparation Guideline Using Orthogonal Strategies. Macromol Rapid Commun 2023; 44:e2200611. [PMID: 36098551 DOI: 10.1002/marc.202200611] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/11/2022] [Indexed: 11/06/2022]
Abstract
Multifunctional nanocarriers enhance the treatment efficacy for modern therapeutics and have gained increasing importance in biomedical research. Codelivery of multiple bioactive molecules enables synergistic therapies. Coencapsulation of cargo molecules into one nanocarrier system is challenging due to different physicochemical properties of the cargo molecules. Additionally, coencapsulation of multiple molecules simultaneously shall proceed with high control and efficiency. Orthogonal approaches for the preparation of nanocarriers are essential to encapsulate sensitive bioactive molecules while preserving their bioactivity. Preparation of nanocarriers by physical processes (i.e., self-assembly or coacervation) and chemical reactions (i.e., click reactions, polymerizations, etc.) are considered as orthogonal methods to most cargo molecules. This review shall act as a guideline to allow the reader to select a suitable preparation protocol for a desired nanocarrier system. This article helps to select for combinations of cargo molecules (hydrophilic-hydrophobic, small-macro, organic-inorganic) with nanocarrier material and synthesis protocols. The focus of this article lies on the coencapsulation of multiple cargo molecules into biocompatible and biodegradable nanocarriers prepared by orthogonal strategies. With this toolbox, the selection of a preparation method for a known set of cargo molecules to prepare the desired biodegradable and loaded nanocarrier shall be provided.
Collapse
Affiliation(s)
- Natkritta Hueppe
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Frederik R Wurm
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Sustainable Polymer Chemistry, Department of Molecules and Materials, Faculty of Science and Technology, MESA+ Institute for Nanotechnology, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| |
Collapse
|
10
|
Mohammad SN, Choi YS, Chung JY, Cedrone E, Neun BW, Dobrovolskaia MA, Yang X, Guo W, Chew YC, Kim J, Baek S, Kim IS, Fruman DA, Kwon YJ. Nanocomplexes of doxorubicin and DNA fragments for efficient and safe cancer chemotherapy. J Control Release 2023; 354:91-108. [PMID: 36572154 DOI: 10.1016/j.jconrel.2022.12.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023]
Abstract
Cancer-targeted therapy by a chemotherapeutic agent formulated in a nanoscale platform has been challenged by complex and inefficient manufacturing, low drug loading, difficult characterization, and marginally improved therapeutic efficacy. This study investigated facile-to-produce nanocomplexes of doxorubicin (DOX), a widely used cancer drug, and clinically approved DNA fragments that are extracted from a natural source. DOX was found to self-assemble DNA fragments into relatively monodispersed nanocomplexes with a diameter of ∼70 nm at 14.3% (w/w) drug loading by simple and scalable mixing. The resulting DOX/DNA nanocomplexes showed sustained DOX release, unlike overly stable Doxil®, cellular uptake via multiple endocytosis pathways, and high hematological and immunological compatibility. DOX/DNA nanocomplexes eradicated EL4 T lymphoma cells in a time-dependent manner, eventually surpassing free DOX. Extended circulation of DOX/DNA nanocomplexes, while avoiding off-target accumulation in the lung and being cleared from the liver, resulted in rapid accumulation in tumor and lowered cardio toxicity. Finally, tumor growth of EL4-challenged C57BL/6 mice (syngeneic model) and OPM2-challenged NSG mice (human xenograft model) were efficiently inhibited by DOX/DNA nanocomplexes with enhanced overall survival, in comparison with free DOX and Doxil®, especially upon repeated administrations. DOX/DNA nanocomplexes are a promising chemotherapeutics delivery platform for their ease of manufacturing, high biocompatibility, desired drug release and accumulation, efficient tumor eradication with improved safety, and further engineering versatility for extended therapeutic applications.
Collapse
Affiliation(s)
- Saad N Mohammad
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States
| | - Yeon Su Choi
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States
| | - Jee Young Chung
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States
| | - Edward Cedrone
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, United States
| | - Barry W Neun
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, United States
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, United States
| | - Xiaojing Yang
- Zymo Research Corporation, Irvine, CA 92604, United States
| | - Wei Guo
- Zymo Research Corporation, Irvine, CA 92604, United States
| | - Yap Ching Chew
- Zymo Research Corporation, Irvine, CA 92604, United States
| | - Juwan Kim
- Pharma Research, Co, Ltd., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seunggul Baek
- Pharma Research, Co, Ltd., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Ik Soo Kim
- Pharma Research, Co, Ltd., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States; Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, United States; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States; Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, United States.
| |
Collapse
|
11
|
Paul M, Itoo AM, Ghosh B, Biswas S. Current trends in the use of human serum albumin for drug delivery in cancer. Expert Opin Drug Deliv 2022; 19:1449-1470. [PMID: 36253957 DOI: 10.1080/17425247.2022.2134341] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Human serum albumin is the most abundant transport protein in plasma, which has recently been extensively utilized to form nanoparticles for drug delivery in cancer. The primary reason for selecting albumin protein as drug delivery cargo is its excellent biocompatibility, biodegradability, and non-immunogenicity. Moreover, the albumin structure containing three homologous domains constituted of a single polypeptide (585 amino acid) incorporates various hydrophobic drugs by non-covalent interactions. Albumin shows active tumor targeting via their interaction with gp60 and SPARC proteins abundant in the tumor-associated endothelial cells and the tumor microenvironment. AREAS COVERED The review discusses the importance of albumin as a drug-carrier system, general procedures to prepare albumin NPs, and the current trends in using albumin-based nanomedicines to deliver various chemotherapeutic agents. The various applications of albumin in the nanomedicines, such as NPs surface modifier and fabrication of hybrid/active-tumor targeted NPs, are delineated based on current trends. EXPERT OPINION Nanomedicines have the potential to revolutionize cancer treatment. However, clinical translation is limited majorly due to the lack of suitable nanomaterials offering systemic stability, optimum drug encapsulation, tumor-targeted delivery, sustained drug release, and biocompatibility. The potential of albumin could be explored in nanomedicines fabrication for superior treatment outcomes in cancer.
Collapse
Affiliation(s)
- Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| | - Asif Mohd Itoo
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| |
Collapse
|
12
|
Wang Y, Fens MH, van Kronenburg NCH, Shi Y, Lammers T, Heger M, van Nostrum CF, Hennink WE. Magnetic beads for the evaluation of drug release from biotinylated polymeric micelles in biological media. J Control Release 2022; 349:954-962. [PMID: 35931210 DOI: 10.1016/j.jconrel.2022.07.044] [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: 06/10/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 11/16/2022]
Abstract
To improve the reliability of in vitro release studies of drug delivery systems, we developed a novel in vitro method for the evaluation of drug release from polymeric micelles in complex biological media. Polymeric micelles based on poly(N-2-hydroxypropyl methacrylamide)-block-poly(N-2-benzoyloxypropyl methacrylamide) (p(HPMAm)-b-p(HPMAm-Bz)) of which 10% of the chains was functionalized with biotin at the p(HPMAm) terminus were prepared using a solvent extraction method. The size of the micelles when loaded with a hydrophobic agent, namely paclitaxel (a clinically used cytostatic drug) or curcumin (a compound with multiple pharmacological activities), was around 65 nm. The biotin decoration allowed the binding of the micelles to streptavidin-coated magnetic beads which occurred within 10 min and reached a binding efficiency of 90 ± 6%. Drug release in different media was studied after the magnetic separation of micelles bound to the streptavidin-coated beads, by determination of the released drug in the media as well as the retained drug in the micellar fraction bound to the beads. The in vitro release of paclitaxel and curcumin at 37 °C in PBS, PBS containing 2% v/v Tween 80, PBS containing 4.5% w/v bovine serum albumin, mouse plasma, and whole mouse blood was highly medium-dependent. In all media studied, paclitaxel showed superior micellar retention compared to curcumin. Importantly, the presence of serum proteins accelerated the release of both paclitaxel and curcumin. The results presented in this study show great potential for predicting drug release from nanomedicines in biological media which in turn is crucial for their further pharmaceutical development.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands
| | - Marcel H Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany
| | - Michal Heger
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands; Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing 314001, Zhejiang, PR China
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| |
Collapse
|
13
|
Synthesis, characterization and investigating the binding mechanism of novel coumarin derivatives with human serum albumin: Spectroscopic and computational approach. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132366] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
14
|
Tian Z, Yao W. Albumin-Bound Paclitaxel: Worthy of Further Study in Sarcomas. Front Oncol 2022; 12:815900. [PMID: 35223497 PMCID: PMC8866444 DOI: 10.3389/fonc.2022.815900] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/20/2022] [Indexed: 12/11/2022] Open
Abstract
Taxanes (paclitaxel and docetaxel) play an important role in the treatment of advanced sarcomas. Albumin-bound paclitaxel (nab-paclitaxel) is a new kind of taxane and has many advantages compared with paclitaxel and docetaxel. Nab-paclitaxel is currently approved for the treatment of advanced breast, non-small cell lung, and pancreatic cancers. However, the efficacy of nab-paclitaxel in sarcomas has not been reviewed. In this review, we first compare the similarities and differences among nab-paclitaxel, paclitaxel, and docetaxel and then summarize the efficacy of nab-paclitaxel against various non-sarcoma malignancies based on clinical trials with reported results. The efficacy and clinical research progress on nab-paclitaxel in sarcomas are also summarized. This review will serve as a good reference for the application of nab-paclitaxel in clinical sarcoma treatment studies and the design of clinical trials.
Collapse
Affiliation(s)
| | - Weitao Yao
- Department of Orthopedics, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
15
|
Paclitaxel Drug Delivery Systems: Focus on Nanocrystals' Surface Modifications. Polymers (Basel) 2022; 14:polym14040658. [PMID: 35215570 PMCID: PMC8875890 DOI: 10.3390/polym14040658] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 12/13/2022] Open
Abstract
Paclitaxel (PTX) is a chemotherapeutic agent that belongs to the taxane family and which was approved to treat various kinds of cancers including breast cancer, ovarian cancer, advanced non-small-cell lung cancer, and acquired immunodeficiency syndrome (AIDS)-related Kaposi’s sarcoma. Several delivery systems for PTX have been developed to enhance its solubility and pharmacological properties involving liposomes, nanoparticles, microparticles, micelles, cosolvent methods, and the complexation with cyclodextrins and other materials that are summarized in this article. Specifically, this review discusses deeply the developed paclitaxel nanocrystal formulations. As PTX is a hydrophobic drug with inferior water solubility properties, which are improved a lot by nanocrystal formulation. Based on that, many studies employed nano-crystallization techniques not only to improve the oral delivery of PTX, but IV, intraperitoneal (IP), and local and intertumoral delivery systems were also developed. Additionally, superior and interesting properties of PTX NCs were achieved by performing additional modifications to the NCs, such as stabilization with surfactants and coating with polymers. This review summarizes these delivery systems by shedding light on their route of administration, the methods used in the preparation and modifications, the in vitro or in vivo models used, and the advantages obtained based on the developed formulations.
Collapse
|
16
|
Huq M, Rosales-Solano H, Pawliszyn J. Investigation of binding of fatty acids to serum albumin to determine free concentrations: Experimental and in-silico approaches. Anal Chim Acta 2022; 1192:339370. [DOI: 10.1016/j.aca.2021.339370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022]
|
17
|
Progress of albumin-polymer conjugates as efficient drug carriers. PURE APPL CHEM 2022. [DOI: 10.1515/pac-2021-2006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Albumin is a protein that has garnered wide attention in nanoparticle-based drug delivery of cancer therapeutics due to its natural abundance and unique cancer-targeting ability. The propensity of albumin to naturally accumulate in tumours, further augmented by the incorporation of targeting ligands, has made the field of albumin-polymer conjugate development a much pursued one. Polymerization techniques such as RAFT and ATRP have paved the path to incorporate various polymers in the design of albumin-polymer hybrids, indicating the advancement of the field since the first instance of PEGylated albumin in 1977. The synergistic combination of albumin and polymer endows manifold features to these macromolecular hybrids to evolve as next generation therapeutics. The current review is successive to our previously published review on drug delivery vehicles based on albumin-polymer conjugates and aims to provide an update on the progress of albumin-polymer conjugates. This review also highlights the alternative of exploring albumin-polymer conjugates formed via supramolecular, non-covalent interactions. Albumin-based supramolecular polymer systems provide a versatile platform for functionalization, thereby, holding great potential in enhancing cytotoxicity and controlled delivery of therapeutic agents.
Collapse
|
18
|
Miao Y, Yang T, Yang S, Yang M, Mao C. Protein nanoparticles directed cancer imaging and therapy. NANO CONVERGENCE 2022; 9:2. [PMID: 34997888 PMCID: PMC8742799 DOI: 10.1186/s40580-021-00293-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/29/2021] [Indexed: 05/10/2023]
Abstract
Cancer has been a serious threat to human health. Among drug delivery carriers, protein nanoparticles are unique because of their mild and environmentally friendly preparation methods. They also inherit desired characteristics from natural proteins, such as biocompatibility and biodegradability. Therefore, they have solved some problems inherent to inorganic nanocarriers such as poor biocompatibility. Also, the surface groups and cavity of protein nanoparticles allow for easy surface modification and drug loading. Besides, protein nanoparticles can be combined with inorganic nanoparticles or contrast agents to form multifunctional theranostic platforms. This review introduces representative protein nanoparticles applicable in cancer theranostics, including virus-like particles, albumin nanoparticles, silk protein nanoparticles, and ferritin nanoparticles. It also describes the common methods for preparing them. It then critically analyzes the use of a variety of protein nanoparticles in improved cancer imaging and therapy.
Collapse
Affiliation(s)
- Yao Miao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
| | - Tao Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, Zhejiang, China.
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK, 73019-5251, USA.
| |
Collapse
|
19
|
Yu L, Hua Z, Luo X, Zhao T, Liu Y. Systematic interaction of plasma albumin with the efficacy of chemotherapeutic drugs. Biochim Biophys Acta Rev Cancer 2021; 1877:188655. [PMID: 34780933 DOI: 10.1016/j.bbcan.2021.188655] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 02/07/2023]
Abstract
Albumin, as the most abundant plasma protein, plays an integral role in the transport of a variety of exogenous and endogenous ligands in the bloodstream and extravascular spaces. For exogenous drugs, especially chemotherapeutic drugs, binding to and being delivered by albumin can significantly affect their efficacy. Meanwhile, albumin can also bind to many endogenous ligands, such as fatty acids, with important physiological significance that can affect tumor proliferation and metabolism. In this review, we summarize how albumin with unique properties affects chemotherapeutic drugs efficacy from the aspects of drug outcome in blood, toxicity, tumor accumulation and direct or indirect interactions with fatty acids, plus application of albumin-based carriers for anti-tumor drug delivery.
Collapse
Affiliation(s)
- Liuchunyang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
20
|
Hwang D, Vinod N, Skoczen SL, Ramsey JD, Snapp KS, Montgomery SA, Wang M, Lim C, Frank JE, Sokolsky-Papkov M, Li Z, Yuan H, Stern ST, Kabanov AV. Bioequivalence assessment of high-capacity polymeric micelle nanoformulation of paclitaxel and Abraxane® in rodent and non-human primate models using a stable isotope tracer assay. Biomaterials 2021; 278:121140. [PMID: 34634661 PMCID: PMC10726948 DOI: 10.1016/j.biomaterials.2021.121140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 02/06/2023]
Abstract
The in vivo fate of nanoformulated drugs is governed by the physicochemical properties of the drug and the functionality of nanocarriers. Nanoformulations such as polymeric micelles, which physically encapsulate poorly soluble drugs, release their payload into the bloodstream during systemic circulation. This results in three distinct fractions of the drug-nanomedicine: encapsulated, protein-bound, and free drug. Having a thorough understanding of the pharmacokinetic (PK) profiles of each fraction is essential to elucidate mechanisms of nanomedicine-driven changes in drug exposure and PK/PD relationships pharmacodynamic activity. Here, we present a comprehensive preclinical assessment of the poly (2-oxazoline)-based polymeric micelle of paclitaxel (PTX) (POXOL hl-PM), including bioequivalence comparison to the clinically approved paclitaxel nanomedicine, Abraxane®. Physicochemical characterization and toxicity analysis of POXOL hl-PM was conducted using standardized protocols by the Nanotechnology Characterization Laboratory (NCL). The bioequivalence of POXOL hl-PM to Abraxane® was evaluated in rats and rhesus macaques using the NCL's established stable isotope tracer ultrafiltration assay (SITUA) to delineate the plasma PK of each PTX fraction. The SITUA study revealed that POXOL hl-PM and Abraxane® had comparable PK profiles not only for total PTX but also for the distinct drug fractions, suggesting bioequivalence in given animal models. The comprehensive preclinical evaluation of POXOL hl-PM in this study showcases a series of widely applicable standardized studies by NCL for assessing nanoformulations prior to clinical investigation.
Collapse
Affiliation(s)
- Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Natasha Vinod
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States; Joint UNC/NC State Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, 27599, United States
| | - Sarah L Skoczen
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, 21702, United States
| | - Jacob D Ramsey
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Kelsie S Snapp
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, 21702, United States
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Mengzhe Wang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Chaemin Lim
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Jonathan E Frank
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Zibo Li
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Hong Yuan
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Stephan T Stern
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, MD, 21702, United States
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, 27599, United States; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M. V. Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|
21
|
Menezes TM, Garcia YS, Dias de Assis CR, Ventura GT, de Queiroz RM, Dias WB, Todeschini AR, Neves JL. Evaluation of europium-based carbon nanocomposites as bioimaging probes: Preparation, NMR relaxivities, binding effects over plasma proteins and cytotoxic aspects. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Menezes TM, Neto AMDS, Gubert P, Neves JL. Effects of human serum albumin glycation on the interaction with the tyrosine kinase inhibitor pazopanib unveiled by multi-spectroscopic and bioinformatic tools. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116843] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
23
|
Zaheer Y, Vorup‐Jensen T, Webster TJ, Ahmed M, Khan WS, Ihsan A. Protein based nanomedicine: Promising therapeutic modalities against inflammatory disorders. NANO SELECT 2021. [DOI: 10.1002/nano.202100214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Yumna Zaheer
- National Institute for Biotechnology and Genetic Engineering College Pakistan Institute of Engineering and Applied Sciences (NIBGE‐C, PIEAS) Faisalabad Punjab 38000 Pakistan
| | - Thomas Vorup‐Jensen
- Department of Biomedicine and Interdisciplinary Nanoscience Center Aarhus University Aarhus Denmark
| | - Thomas J. Webster
- Department of Chemical Engineering Northeastern University Boston Massachusetts USA
| | - Mukhtiar Ahmed
- Chemistry of Interfaces Luleå University of Technology Luleå Sweden
| | - Waheed S. Khan
- National Institute for Biotechnology and Genetic Engineering College Pakistan Institute of Engineering and Applied Sciences (NIBGE‐C, PIEAS) Faisalabad Punjab 38000 Pakistan
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College Pakistan Institute of Engineering and Applied Sciences (NIBGE‐C, PIEAS) Faisalabad Punjab 38000 Pakistan
| |
Collapse
|
24
|
Heide F, McDougall M, Harder-Viddal C, Roshko R, Davidson D, Wu J, Aprosoff C, Moya-Torres A, Lin F, Stetefeld J. Boron rich nanotube drug carrier system is suited for boron neutron capture therapy. Sci Rep 2021; 11:15520. [PMID: 34330984 PMCID: PMC8324832 DOI: 10.1038/s41598-021-95044-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Boron neutron capture therapy (BNCT) is a two-step therapeutic process that utilizes Boron-10 in combination with low energy neutrons to effectively eliminate targeted cells. This therapy is primarily used for difficult to treat head and neck carcinomas; recent advances have expanded this method to cover a broader range of carcinomas. However, it still remains an unconventional therapy where one of the barriers for widespread adoption is the adequate delivery of Boron-10 to target cells. In an effort to address this issue, we examined a unique nanoparticle drug delivery system based on a highly stable and modular proteinaceous nanotube. Initially, we confirmed and structurally analyzed ortho-carborane binding into the cavities of the nanotube. The high ratio of Boron to proteinaceous mass and excellent thermal stability suggest the nanotube system as a suitable candidate for drug delivery into cancer cells. The full physicochemical characterization of the nanotube then allowed for further mechanistic molecular dynamic studies of the ortho-carborane uptake and calculations of corresponding energy profiles. Visualization of the binding event highlighted the protein dynamics and the importance of the interhelical channel formation to allow movement of the boron cluster into the nanotube. Additionally, cell assays showed that the nanotube can penetrate outer membranes of cancer cells followed by localization around the cells' nuclei. This work uses an integrative approach combining experimental data from structural, molecular dynamics simulations and biological experiments to thoroughly present an alternative drug delivery device for BNCT which offers additional benefits over current delivery methods.
Collapse
Affiliation(s)
- Fabian Heide
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Matthew McDougall
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Candice Harder-Viddal
- Department of Chemistry and Physics, Canadian Mennonite University, Winnipeg, MB, R3P 2N2, Canada
| | - Roy Roshko
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - David Davidson
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Jiandong Wu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Camila Aprosoff
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Aniel Moya-Torres
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Jörg Stetefeld
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| |
Collapse
|
25
|
Harder-Viddal C, Heide F, Roshko RM, Stetefeld J. Molecular dynamics simulations of ortho-carborane nano-diamond storage within the nonpolar channel cavities of a right-handed coiled-coil tetrabrachion nanotube. Comput Struct Biotechnol J 2021; 19:3531-3541. [PMID: 34194676 PMCID: PMC8220585 DOI: 10.1016/j.csbj.2021.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 10/26/2022] Open
Abstract
Molecular dynamics simulations have been performed on a complex in which clusters of boron in the form of molecules of the nanodiamond ortho-carborane ( C 2 B 10 H 12 ) have been inserted into the four large nonpolar cavities of a nanotube of the right-handed coiled-coil ( R H C C ) t e t r a b r a c h i o n . The techniques of multi-configurational thermodynamic integration, steered molecular dynamics and umbrella sampling have been combined to investigate the energetics of storage of ortho-carborane in the cavities and to map out the free energy landscape of the RHCC - t e t r a b r a c h i o n - o r t h o - c a r b o r a n e complex along the central channel and along directions transverse to the central channel. The purpose of the study was to explore potential pathways for the diffusion of ortho-carborane between the cavities and the solvent and to assess the stability of the complex as a possible drug delivery system for boron neutron capture therapy (BNCT). The investigation reveals a complex free energy landscape with a multitude of peaks and valleys, all of which can be related to specific architectural elements of the RHCC - n a n o t u b e , and the activation barriers for ortho-carborane capture and release support the requirements for rapid cargo uptake coupled with tight binding to the cavities.
Collapse
Affiliation(s)
- C Harder-Viddal
- Department of Chemistry and Physics, Canadian Mennonite University, 500 Shaftesbury Blvd, Winnipeg, Manitoba, Canada
| | - F Heide
- Department of Chemistry, University of Manitoba, 144 Dysart Rd, Winnipeg, Manitoba, Canada
| | - R M Roshko
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, Winnipeg, Manitoba, Canada
| | - J Stetefeld
- Department of Chemistry, University of Manitoba, 144 Dysart Rd, Winnipeg, Manitoba, Canada.,Center for Oil and Gas Research and Development (COGRAD), Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Canada.,Department of Human Anatomy and Cell Science, University of Manitoba, Canada
| |
Collapse
|
26
|
Ribeiro AG, Alves JEF, Soares JCS, dos Santos KL, Jacob ÍTT, da Silva Ferreira CJ, dos Santos JC, de Azevedo RDS, de Almeida SMV, de Lima MDCA. Albumin roles in developing anticancer compounds. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02748-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Zeeshan F, Madheswaran T, Panneerselvam J, Taliyan R, Kesharwani P. Human Serum Albumin as Multifunctional Nanocarrier for Cancer Therapy. J Pharm Sci 2021; 110:3111-3117. [PMID: 33989679 DOI: 10.1016/j.xphs.2021.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 01/05/2023]
Abstract
Human serum albumin or simply called albumin is a flexible protein employed as a carrier in the fabrication of albumin-based nanocarriers (ANCs) for the administration of cancer therapeutics. Albumin can contribute enhanced tumour specificity, reduced drug induced cytotoxicity and retain concentration of the therapeutically active agent such as drug, peptide, protein, and gene for a prolonged time duration. Nevertheless, apart from cancer management, ANCs are also employed in the diagnosis, imaging, and multimodal cancer therapy. This article figures out salient characteristics, design as well as categories of ANCs in the context of their application in cancer management. In addition, this review article discusses the fabrication methods of ANCs, use of ANCs in gene, cancer, and multimodal therapy along with cancer diagnosis and imaging. Lastly, this review also briefly discusses about (ANCs) formulations, commercial products, and those under clinical testing.
Collapse
Affiliation(s)
- Farrukh Zeeshan
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
| | - Jithendra Panneerselvam
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi - 110062, India.
| |
Collapse
|
28
|
Taguchi K, Okamoto Y, Matsumoto K, Otagiri M, Chuang VTG. When Albumin Meets Liposomes: A Feasible Drug Carrier for Biomedical Applications. Pharmaceuticals (Basel) 2021; 14:ph14040296. [PMID: 33810483 PMCID: PMC8065628 DOI: 10.3390/ph14040296] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
Albumin, the most abundant protein in plasma, possesses some inherent beneficial structural and physiological characteristics that make it suitable for use as a drug delivery agent, such as an extraordinary drug-binding capacity and long blood retention, with a high biocompatibility. The use of these characteristics as a nanoparticle drug delivery system (DDS) offers several advantages, including a longer circulation time, lower toxicity, and more significant drug loading. To date, many innovative liposome preparations have been developed in which albumin is involved as a DDS. These novel albumin-containing liposome preparations show superior deliverability for genes, hydrophilic/hydrophobic substances and proteins/peptides to the targeting area compared to original liposomes by virtue of their high biocompatibility, stability, effective loading content, and the capacity for targeting. This review summarizes the current status of albumin applications in liposome-based DDS, focusing on albumin-coated liposomes and albumin-encapsulated liposomes as a DDS carrier for potential medical applications.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.T.); (K.M.)
| | - Yuko Okamoto
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan; (Y.O.); (M.O.)
| | - Kazuaki Matsumoto
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.T.); (K.M.)
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan; (Y.O.); (M.O.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan
| | - Victor Tuan Giam Chuang
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA 6102, Australia
- Correspondence:
| |
Collapse
|
29
|
Sato T, Okazaki M, Sano J, Kato C, Shimizu K, Kitagawa M. Binding affinities of paclitaxel and docetaxel for generic and nanoparticle albumin-bound paclitaxel-derived albumin from human serum. Biomed Rep 2021; 14:35. [PMID: 33732454 DOI: 10.3892/br.2021.1411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/21/2021] [Indexed: 01/02/2023] Open
Abstract
Nanoparticle albumin-bound (nab)-paclitaxel is a 130-nm formulation containing human serum albumin (HSA). The clinical efficacy of this formulation is considered to depend on its affinity for HSA. The high pressure employed during the manufacture of nab-paclitaxel HSA (nab HSA) may influence its conformation and/or oligomerization, and ultimately its affinity for HSA. Therefore, studies are required to evaluate whether the affinity of paclitaxel for nab HSA is similar to that of generic HSA (control HSA). In the present study, nab HSA was isolated from nab-paclitaxel by gel filtration, and the binding affinities (KDs) were determined by surface plasmon resonance. Furthermore, the affinity of docetaxel for nab HSA and control HSA was measured, as their binding sites are similar. Paclitaxel showed KDs of 8.93±8.60 and 7.39±5.81 µM for nab HSA and control HSA, respectively, whereas the corresponding KDs for docetaxel were 44.3±9.50 and 55.9±2.28 µM, respectively. This suggests that the paclitaxel binding site was not modified during the nab-paclitaxel manufacturing process. Additionally, nab HSA likely does not affect paclitaxel and blood HSA binding, as evidenced by the similar affinities of paclitaxel and docetaxel for nab HSA and control HSA. In conclusion, the binding affinities of paclitaxel and docetaxel for nab HSA and control HSA were found to be comparable. Additionally, the manufacturing process did not influence the paclitaxel binding affinity for nab HSA. These results also suggest that nab HSA may not affect the clinical effectiveness of nab-paclitaxel.
Collapse
Affiliation(s)
- Takamichi Sato
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., Tokyo 115-0042, Japan
| | - Manami Okazaki
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., Tokyo 115-0042, Japan
| | - Junko Sano
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., Tokyo 115-0042, Japan
| | - Chihiro Kato
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., Tokyo 115-0042, Japan
| | - Kazuhisa Shimizu
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., Tokyo 115-0042, Japan
| | - Masayuki Kitagawa
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., Tokyo 115-0042, Japan
| |
Collapse
|
30
|
Li B, Chen X, Ding T, Liu Y, Ma T, Zhang G, Wang X. Nanoparticle albumin-bound paclitaxel versus solvent-based paclitaxel in breast cancer: A protocol for systemic review and meta-analysis. Medicine (Baltimore) 2021; 100:e24514. [PMID: 33607781 PMCID: PMC7899816 DOI: 10.1097/md.0000000000024514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Nanoparticle albumin-bound paclitaxel (nab-PTX) has exhibited clinical efficacy in breast cancer treatment, but toxicities can be yielded more at the same time. We did this meta-analysis aiming to unambiguously compare nab-PTX with conventional solvent-based paclitaxel in breast cancer patients of all stages. METHOD Pubmed, EMBASE, Cochrane Library, Chinese Biomedical database, Chinese National Knowledge Infrastructure, Chinese Science and Technology Periodical database, and WangFang database were searched for head-to-head randomized controlled trials of nab-PTX and solvent-based paclitaxel in breast cancer. Other sources will also be searched like Google Scholar and gray literatures. Two researchers will independently search the database and extract data from the articles. Risk of bias will be assessed using the Cochrane Collaboration's tool. Objective tumor response rate, chemotherapy completion rate after 4 or 6 cycles, and toxicity will be primary outcomes. Disease control rate, overall survival, and progression-free survival/disease-free survival will be included in secondary outcomes. Risk ratio with 95% confidence interval was used for dichotomous variables while hazard ratio was used for time-to-event outcomes. The following 3 data sets will all be considered when synthesizing the data: intention-to-treat population, those who actually received taxanes treatment, and those who were actually assessed. All the analyses were done using Review Manager Software 5.3. Any disagreements in study selection, data collection, and analysis will be resolved by a third investigator. RESULTS AND CONCLUSION This study is aim to evaluate the efficacy and safety of nab-PTX compared with PTX in breast cancer treatment as well as to find the best dose or schedule and identify the benefit population. This meta-analysis could provide evidence for clinicians to make a better choice between nab-PTX and PTX in different specific contexts. PROSPERO REGISTRATION NUMBER CRD42019117912.
Collapse
Affiliation(s)
- Bingxue Li
- Beijing University of Chinese Medicine, Chaoyang District
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Dongcheng District, Beijing, China
| | - Xinjie Chen
- Beijing University of Chinese Medicine, Chaoyang District
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Dongcheng District, Beijing, China
| | - Tongjing Ding
- Beijing University of Chinese Medicine, Chaoyang District
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Dongcheng District, Beijing, China
| | - Yihua Liu
- Beijing University of Chinese Medicine, Chaoyang District
| | - Tingting Ma
- Beijing University of Chinese Medicine, Chaoyang District
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Dongcheng District, Beijing, China
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Dongcheng District, Beijing, China
| | - Xiaomin Wang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Dongcheng District, Beijing, China
| |
Collapse
|
31
|
Infante P, Malfanti A, Quaglio D, Balducci S, De Martin S, Bufalieri F, Mastrotto F, Basili I, Garofalo M, Lospinoso Severini L, Mori M, Manni I, Moretti M, Nicoletti C, Piaggio G, Caliceti P, Botta B, Ghirga F, Salmaso S, Di Marcotullio L. Glabrescione B delivery by self-assembling micelles efficiently inhibits tumor growth in preclinical models of Hedgehog-dependent medulloblastoma. Cancer Lett 2020; 499:220-231. [PMID: 33249196 DOI: 10.1016/j.canlet.2020.11.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022]
Abstract
Aberrant activation of the Hedgehog (Hh) pathway leads to the development of several tumors, including medulloblastoma (MB), the most common pediatric brain malignancy. Hh inhibitors acting on GLI1, the final effector of Hh signaling, offer a valuable opportunity to overcome the pitfalls of the existing therapies to treat Hh-driven cancers. In this study, the toxicity, delivery, biodistribution, and anticancer efficacy of Glabrescione B (GlaB), a selective GLI1 inhibitor, were investigated in preclinical models of Hh-dependent MB. To overcome its poor water solubility, GlaB was formulated with a self-assembling amphiphilic polymer forming micelles, called mPEG5kDa-cholane. mPEG5kDa-cholane/GlaB showed high drug loading and stability, low cytotoxicity, and long permanence in the bloodstream. We found that mPEG5kDa-cholane efficiently enhanced the solubility of GlaB, thus avoiding the use of organic solvents. mPEG5kDa-cholane/GlaB possesses favorable pharmacokinetics and negligible toxicity. Remarkably, GlaB encapsulated in mPEG5kDa-cholane micelles was delivered through the blood-brain barrier and drastically inhibited tumor growth in both allograft and orthotopic models of Hh-dependent MB. Our findings reveal that mPEG5kDa-cholane/GlaB is a good candidate for the treatment of Hh-driven tumors and provide relevant implications for the translation of GlaB into clinical practice.
Collapse
Affiliation(s)
- Paola Infante
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Roma, Italy
| | - Alessio Malfanti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, University La Sapienza, Roma, Italy
| | - Silvia Balducci
- Department of Chemistry and Technology of Drugs, University La Sapienza, Roma, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Irene Basili
- Department of Molecular Medicine, University La Sapienza, Roma, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Isabella Manni
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Marta Moretti
- Department of Experimental Medicine, University La Sapienza, Roma, Italy
| | - Carmine Nicoletti
- DAHFMO-Unit of Histology and Medical Embryology, University La Sapienza, Roma, Italy; Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, University La Sapienza, Roma, Italy
| | - Giulia Piaggio
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, University La Sapienza, Roma, Italy
| | - Francesca Ghirga
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Roma, Italy.
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University La Sapienza, Roma, Italy; Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, University La Sapienza, Roma, Italy.
| |
Collapse
|
32
|
Yu J, Mu Q, Perazzolo S, Griffin JI, Zhu L, McConnachie LA, Shen DD, Ho RJ. Novel Long-Acting Drug Combination Nanoparticles Composed of Gemcitabine and Paclitaxel Enhance Localization of Both Drugs in Metastatic Breast Cancer Nodules. Pharm Res 2020; 37:197. [PMID: 32968837 PMCID: PMC8686529 DOI: 10.1007/s11095-020-02888-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE To develop drug-combination nanoparticles (DcNPs) composed of hydrophilic gemcitabine (G) and hydrophobic paclitaxel (T) and deliver both drugs to metastatic cancer cells. METHODS GT DcNPs were evaluated based on particle size and drug association efficiency (AE%). The effect of DcNP on GT plasma time-course and tissue distribution was characterized in mice and a pharmacokinetic model was developed. A GT distribution study into cancer nodules (derived from 4 T1 cells) was performed. RESULTS An optimized GT DcNP composition (d = 59.2 nm ±9.2 nm) was found to be suitable for IV formulation. Plasma exposure of G and T were enhanced 61-fold and 3.8-fold when given in DcNP form compared to the conventional formulation, respectively. Mechanism based pharmacokinetic modeling and simulation show that both G and T remain highly associated to DcNPs in vivo (G: 98%, T:75%). GT DcNPs have minimal distribution to healthy organs with selective distribution and retention in tumor burdened tissue. Tumor bearing lungs had a 5-fold higher tissue-to-plasma ratio of gemcitabine in GT DcNPs compared to healthy lungs. CONCLUSIONS DcNPs can deliver hydrophilic G and hydrophobic T together to cancer nodules and produce long acting exposure, likely due to stable GT association to DcNPs in vivo.
Collapse
Affiliation(s)
- Jesse Yu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Qingxin Mu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Simone Perazzolo
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - James I Griffin
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Linxi Zhu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Lisa A McConnachie
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Danny D Shen
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Rodney Jy Ho
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA.
- Departments of Bioengineering, University of Washington, Seattle, Washington, 98195, USA.
| |
Collapse
|
33
|
Hassanin IA, Elzoghby AO. Self-assembled non-covalent protein-drug nanoparticles: an emerging delivery platform for anti-cancer drugs. Expert Opin Drug Deliv 2020; 17:1437-1458. [DOI: 10.1080/17425247.2020.1813713] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Islam A. Hassanin
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Ahmed O. Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology (HST), Cambridge, MA, USA
| |
Collapse
|
34
|
Mu Q, Lin G, Stephen ZR, Chung S, Wang H, Patton VK, Gebhart RN, Zhang M. In vivo Serum Enabled Production of Ultrafine Nanotherapeutics for Cancer Treatment. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2020; 38:10-23. [PMID: 33716549 PMCID: PMC7944405 DOI: 10.1016/j.mattod.2020.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Systemic delivery of hydrophobic anti-cancer drugs with nanocarriers, particularly for drug-resistant and metastatic cancer, remain a challenge because of the difficulty to achieve high drug loading, while maintaining a small hydrodynamic size and colloid stability in blood to ensure delivery of an efficacious amount of drug to tumor cells. Here we introduce a new approach to address this challenge. In this approach, nanofibers of larger size with good drug loading capacity are first constructed by a self-assembly process, and upon intravascular injection and interacting with serum proteins in vivo, these nanofibers break down into ultra-fine nanoparticles of smaller size that inherit the drug loading property from their parent nanofibers. We demonstrate the efficacy of this approach with a clinically available anti-cancer drug: paclitaxel (PTX). In vitro, the PTX-loaded nanoparticles enter cancer cells and induce cellular apoptosis. In vivo, they demonstrate prolonged circulation in blood, induce no systemic toxicity, and show high potency in inhibiting tumor growth and metastasis in both mouse models of aggressive, drug-resistant breast cancer and melanoma. This study points to a new strategy toward improved anti-cancer drug delivery and therapy.
Collapse
Affiliation(s)
- Qingxin Mu
- Department of Materials Science and Engineering, University
of Washington, Seattle, Washington, 98195, USA
| | - Guanyou Lin
- Department of Materials Science and Engineering, University
of Washington, Seattle, Washington, 98195, USA
| | - Zachary R. Stephen
- Department of Materials Science and Engineering, University
of Washington, Seattle, Washington, 98195, USA
| | - Steve Chung
- Department of Materials Science and Engineering, University
of Washington, Seattle, Washington, 98195, USA
| | - Hui Wang
- Department of Materials Science and Engineering, University
of Washington, Seattle, Washington, 98195, USA
| | - Victoria K. Patton
- Department of Chemical Engineering, University of
Washington, Seattle, Washington, 98195, USA
| | - Rachel N. Gebhart
- Department of Chemistry, University of Washington, Seattle,
Washington, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University
of Washington, Seattle, Washington, 98195, USA
| |
Collapse
|
35
|
Li X, Vieweger M, Guo P. Self-assembly of four generations of RNA dendrimers for drug shielding with controllable layer-by-layer release. NANOSCALE 2020; 12:16514-16525. [PMID: 32729600 PMCID: PMC7448292 DOI: 10.1039/d0nr02614j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Chemical dendrimers have been shown to be a promising drug delivery platform due to their advantageous properties such as monodispersity, multivalency and branched structure. Taking advantage of self-assembly and its intrinsic negative charge, we used RNA as the building block for dendrimer construction to eliminate complex synthesis procedures and cationic charge-related toxicity. Oligo ribonucleotides produced by solid phase chemical synthesis allow the large-scale manufacture of homologous RNA dendrimers. Employing concepts from RNA nanotechnology enabled the controllable production of dendrimers with generations from G1, G2, G3, to G4 with layer-by-layer release capability. The conjugation of functional groups into individual RNA strands and the incorporation of functionalized RNA strands into the dendrimers at different sites have been reported. Anticancer drugs loaded into RNA dendrimers showed comparable cancer cell inhibition effect to free drugs. Encapsulation of cell binding ligands and hydrophobic drugs within the dendrimer significantly reduced the efficiency of cell binding and protein binding respectively, demonstrating the shielding effect of RNA dendrimers. The results imply a potential application of RNA dendrimer for delivery, shielding and controlled release of hydrophobic drugs in vivo.
Collapse
Affiliation(s)
- Xin Li
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH 43210, USA.
| | | | | |
Collapse
|
36
|
Haider MS, Lübtow MM, Endres S, Forster S, Flegler VJ, Böttcher B, Aseyev V, Pöppler AC, Luxenhofer R. Think Beyond the Core: Impact of the Hydrophilic Corona on Drug Solubilization Using Polymer Micelles. ACS APPLIED MATERIALS & INTERFACES 2020; 12:24531-24543. [PMID: 32378873 DOI: 10.1021/acsami.9b22495] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Polymeric micelles are typically characterized as core-shell structures. The hydrophobic core is considered as a depot for hydrophobic molecules, and the corona-forming block acts as a stabilizing and solubilizing interface between the core and aqueous milieu. Tremendous efforts have been made to tune the hydrophobic block to increase the drug loading and stability of micelles, whereas the role of hydrophilic blocks is rarely investigated in this context, with poly(ethylene glycol) (PEG) being the gold standard of hydrophilic polymers. To better understand the role of the hydrophilic corona, a small library of structurally similar A-B-A-type amphiphiles based on poly(2-oxazoline)s and poly(2-oxazine)s is investigated by varying the hydrophilic block A utilizing poly(2-methyl-2-oxazoline) (pMeOx; A) or poly(2-ethyl-2-oxazoline) (pEtOx; A*). In terms of hydrophilicity, both polymers closely resemble PEG. The more hydrophobic block B bears either a poly(2-oxazoline) and poly(2-oxazine) backbone with C3 (propyl) and C4 (butyl) side chains. Surprisingly, major differences in loading capacities from A-B-A > A*-B-A > A*-B-A* is observed for the formulation with two poorly water-soluble compounds, curcumin and paclitaxel, highlighting the importance of the hydrophilic corona of polymer micelles used for drug formulation. The formulations are also characterized by various nuclear magnetic resonance spectroscopy methods, dynamic light scattering, cryogenic transmission electron microscopy, and (micro) differential scanning calorimetry. Our findings suggest that the interaction between the hydrophilic block and the guest molecule should be considered an important, but previously largely ignored, factor for the rational design of polymeric micelles.
Collapse
Affiliation(s)
- Malik Salman Haider
- Functional Polymer Materials, Chair for Chemical Technology of Material Synthesis and Bavarian Polymer Institute, Faculty of Chemistry and Pharmacy, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Michael M Lübtow
- Functional Polymer Materials, Chair for Chemical Technology of Material Synthesis and Bavarian Polymer Institute, Faculty of Chemistry and Pharmacy, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Sebastian Endres
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Stefan Forster
- Functional Polymer Materials, Chair for Chemical Technology of Material Synthesis and Bavarian Polymer Institute, Faculty of Chemistry and Pharmacy, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Vanessa J Flegler
- Biocenter and Rudolf Virchow Centre, University of Würzburg, Haus D15, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Bettina Böttcher
- Biocenter and Rudolf Virchow Centre, University of Würzburg, Haus D15, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Vladimir Aseyev
- Department of Chemistry, University of Helsinki, PB 55, Helsinki FIN-00014, Finland
| | - Ann-Christin Pöppler
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Robert Luxenhofer
- Functional Polymer Materials, Chair for Chemical Technology of Material Synthesis and Bavarian Polymer Institute, Faculty of Chemistry and Pharmacy, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
- Department of Chemistry, University of Helsinki, PB 55, Helsinki FIN-00014, Finland
| |
Collapse
|
37
|
Suh MS, Patil SM, Kozak D, Pang E, Choi S, Jiang X, Rodriguez JD, Keire DA, Chen K. An NMR Protocol for In Vitro Paclitaxel Release from an Albumin-Bound Nanoparticle Formulation. AAPS PharmSciTech 2020; 21:136. [PMID: 32419122 DOI: 10.1208/s12249-020-01669-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/26/2020] [Indexed: 01/24/2023] Open
Abstract
The paclitaxel protein-bound particles for injectable suspension (marketed under the brand name Abraxane®) contains nanosized complexes of paclitaxel and albumin. The molecular interaction between paclitaxel and albumin within the higher-order nanostructure is analytically challenging to assess, as is any correlation of differences to differences in therapeutic effect. However, because the higher-order nanostructures may affect the paclitaxel release, a suitable in vitro assay to detect potential differences in paclitaxel release between comparator lots and products is desirable. Herein, solution NMR spectroscopy with a T2-filtering technique was developed to detect paclitaxel signal while suppressing albumin signals to follow the released paclitaxel in the NMR tube upon dilution. The non-invasive nature of NMR allows for precise measurement of a full range of dilution-induced drug release percentage from 14 to 92% without any sample extraction. The critical concentration of the drug product (DP) at 50% of release was 0.63 ± 0.04 mg/mL in PBS buffer. In addition, 2D diffusion ordered NMR spectroscopy (DOSY) results revealed that the released paclitaxel experiencing slightly slowed diffusion rates than free paclitaxel, which was attributed to paclitaxel in equilibrium with albumin-bound states. Collectively, the dilution-based NMR method offered an analytical approach to investigate physicochemical attributes of complex injectable products with minimal needed sample preparation and perturbation to nanoparticle formulation.
Collapse
|
38
|
Selva Sharma A, Viswadevarayalu A, Bharathi AC, Anand K, Ali S, Li H, Ibrahim BS, Chen Q. Unravelling the distinctive mode of cooperative and independent interaction mechanism of 1-butyl-2,3-dimethylimidazolium tetrafluoroborate ionic liquid with model transport proteins by comprehensive spectroscopic and computational studies. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
39
|
Carvalho AM, Fernandes E, Gonçalves H, Giner-Casares JJ, Bernstorff S, Nieder JB, Real Oliveira MECD, Lúcio M. Prediction of paclitaxel pharmacokinetic based on in vitro studies: Interaction with membrane models and human serum albumin. Int J Pharm 2020; 580:119222. [PMID: 32194209 DOI: 10.1016/j.ijpharm.2020.119222] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 01/10/2023]
Abstract
Interactions of paclitaxel (PTX) with models mimicking biological interfaces (lipid membranes and serum albumin, HSA) were investigated to test the hypothesis that the set of in vitro assays proposed can be used to predict some aspects of drug pharmacokinetics (PK). PTX membrane partitioning was studied by derivative spectrophotometry; PTX effect on membrane biophysics was evaluated by dynamic light scattering, fluorescence anisotropy, atomic force microscopy and synchrotron small/wide-angle X-ray scattering; PTX distribution/molecular orientation in membranes was assessed by steady-state/time-resolved fluorescence and computer simulations. PTX binding to HSA was studied by fluorescence quenching, derivative spectrophotometry and dynamic/electrophoretic light scattering. PTX high membrane partitioning is consistent with its efficacy crossing cellular membranes and its off-target distribution. PTX is closely located in the membrane phospholipids headgroups, also interacting with the hydrophobic chains, and causes a major distortion of the alignment of the membrane phospholipids, which, together with its fluidizing effect, justifies some of its cellular toxic effects. PTX binds strongly to HSA, which is consistent with its reduced distribution in target tissues and toxicity by bioaccumulation. In conclusion, the described set of biomimetic models and techniques has the potential for early prediction of PK issues, alerting for the required drug optimizations, potentially minimizing the number of animal tests used in the drug development process.
Collapse
Affiliation(s)
- Ana M Carvalho
- CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Departamento de Física da Universidade do Minho, Campus of Gualtar, 4710-057 Braga, Portugal; Nanophotonics Department, Ultrafast Bio- and Nanophotonics Group, INL - International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Eduarda Fernandes
- CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Departamento de Física da Universidade do Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | | | - Juan J Giner-Casares
- Department of Physical Chemistry and Applied Thermodynamics, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, Córdoba E-14014, Spain.
| | - Sigrid Bernstorff
- Elettra-Sincrotrone Trieste S.C.p.A., Strada Statale 14, km 163.5, in Area Science Park, I-34149 Basovizza, Trieste, Italy.
| | - Jana B Nieder
- Nanophotonics Department, Ultrafast Bio- and Nanophotonics Group, INL - International Iberian Nanotechnology Laboratory, Braga, Portugal.
| | - M Elisabete C D Real Oliveira
- CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Departamento de Física da Universidade do Minho, Campus of Gualtar, 4710-057 Braga, Portugal.
| | - Marlene Lúcio
- CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Departamento de Física da Universidade do Minho, Campus of Gualtar, 4710-057 Braga, Portugal; CBMA, Centro de Biologia Molecular e Ambiental, Departamento de Biologia, Universidade do Minho, 4710-057 Braga, Portugal.
| |
Collapse
|
40
|
Albumin nanoparticles as nanocarriers for drug delivery: Focusing on antibody and nanobody delivery and albumin-based drugs. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101471] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
41
|
Gobeaux F, Bizeau J, Samson F, Marichal L, Grillo I, Wien F, Yesylevsky SO, Ramseyer C, Rouquette M, Lepêtre-Mouelhi S, Desmaële D, Couvreur P, Guenoun P, Renault JP, Testard F. Albumin-driven disassembly of lipidic nanoparticles: the specific case of the squalene-adenosine nanodrug. NANOSCALE 2020; 12:2793-2809. [PMID: 31961354 DOI: 10.1039/c9nr06485k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In the field of nanomedicine, nanostructured nanoparticles (NPs) made of self-assembling prodrugs emerged in the recent years with promising properties. In particular, squalene-based drug nanoparticles have already shown their efficiency through in vivo experiments. However, a complete pattern of their stability and interactions in the blood stream is still lacking. In this work we assess the behavior of squalene-adenosine (SQAd) nanoparticles - whose neuroprotective effect has already been demonstrated in murine models - in the presence of fetal bovine serum (FBS) and of bovine serum albumin (BSA), the main protein of blood plasma. Extensive physicochemical characterizations were performed using Small Angle Neutron Scattering (SANS), cryogenic transmission electron microscopy (Cryo-TEM), circular dichroism (CD), steady-state fluorescence spectroscopy (SSFS) and isothermal titration calorimetry (ITC) as well as in silico by means of ensemble docking simulations with human serum albumin (HSA). Significant changes in the colloidal stability of the nanoparticles in the presence of serum albumin were observed. SANS, CD and SSFS analyses demonstrated an interaction between SQAd and BSA, with a partial disassembly of the nanoparticles in the presence of BSA and the formation of a complex between SQAd and BSA. The interaction free energy of SQAd nanoparticles with BSA derived from ITC experiments, is about -8 kcal mol-1 which is further supported in silico by ensemble docking simulations. Overall, our results show that serum albumin partially disassembles SQAd nanoparticles by extracting individual SQAd monomers from them. As a consequence, the SQAd nanoparticles would act as a circulating reservoir in the blood stream. The approach developed in this study could be extended to other soft organic nanoparticles.
Collapse
Affiliation(s)
- Frédéric Gobeaux
- LIONS - NIMBE CEA, CNRS, Université Paris-Saclay, CEA Saclay, 91191 Gif-sur-Yvette Cedex, France.
| | - Joëlle Bizeau
- LIONS - NIMBE CEA, CNRS, Université Paris-Saclay, CEA Saclay, 91191 Gif-sur-Yvette Cedex, France.
| | - Firmin Samson
- LIONS - NIMBE CEA, CNRS, Université Paris-Saclay, CEA Saclay, 91191 Gif-sur-Yvette Cedex, France.
| | - Laurent Marichal
- LIONS - NIMBE CEA, CNRS, Université Paris-Saclay, CEA Saclay, 91191 Gif-sur-Yvette Cedex, France. and I2BC, JOLIOT, DRF, CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Isabelle Grillo
- Institut Laue Langevin, 71 avenue des martyrs, B.P. 156, 38042 Grenoble Cedex 9, France
| | | | - Semen O Yesylevsky
- Department of Physics of Biological Systems, Institute of Physics of the National Academy of Sciences of Ukraine, Prospect Nauky 46, 03028 Kyiv, Ukraine
| | - Christophe Ramseyer
- Laboratoire Chrono Environnement UMR CNRS 6249, Université de Bourgogne Franche-Comté, 16 route de Gray, 25030 Besançon Cedex, France
| | - Marie Rouquette
- Institut Galien Paris-Sud, UMR 8612, CNRS, Université Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France
| | - Sinda Lepêtre-Mouelhi
- Institut Galien Paris-Sud, UMR 8612, CNRS, Université Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France
| | - Didier Desmaële
- Institut Galien Paris-Sud, UMR 8612, CNRS, Université Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR 8612, CNRS, Université Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France
| | - Patrick Guenoun
- LIONS - NIMBE CEA, CNRS, Université Paris-Saclay, CEA Saclay, 91191 Gif-sur-Yvette Cedex, France.
| | - Jean-Philippe Renault
- LIONS - NIMBE CEA, CNRS, Université Paris-Saclay, CEA Saclay, 91191 Gif-sur-Yvette Cedex, France.
| | - Fabienne Testard
- LIONS - NIMBE CEA, CNRS, Université Paris-Saclay, CEA Saclay, 91191 Gif-sur-Yvette Cedex, France.
| |
Collapse
|
42
|
Dunne M, Regenold M, Allen C. Hyperthermia can alter tumor physiology and improve chemo- and radio-therapy efficacy. Adv Drug Deliv Rev 2020; 163-164:98-124. [PMID: 32681862 DOI: 10.1016/j.addr.2020.07.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/20/2022]
Abstract
Hyperthermia has demonstrated clinical success in improving the efficacy of both chemo- and radio-therapy in solid tumors. Pre-clinical and clinical research studies have demonstrated that targeted hyperthermia can increase tumor blood flow and increase the perfused fraction of the tumor in a temperature and time dependent manner. Changes in tumor blood circulation can produce significant physiological changes including enhanced vascular permeability, increased oxygenation, decreased interstitial fluid pressure, and reestablishment of normal physiological pH conditions. These alterations in tumor physiology can positively impact both small molecule and nanomedicine chemotherapy accumulation and distribution within the tumor, as well as the fraction of the tumor susceptible to radiation therapy. Hyperthermia can trigger drug release from thermosensitive formulations and further improve the accumulation, distribution, and efficacy of chemotherapy.
Collapse
|
43
|
Naik R, Jaldappagari S. Spectral and computational attributes: Binding of a potent anticancer agent, dasatinib to a transport protein. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2019.111492] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
44
|
Lu C, Li X, Liang X, Zhang X, Yin T, Gou J, He H, Zhang Y, Tang X. Liver Targeting Albumin-Coated Silybin-Phospholipid Particles Prepared by Nab™ Technology for Improving Treatment Effect of Acute Liver Damage in Intravenous Administration. AAPS PharmSciTech 2019; 20:293. [PMID: 31432294 DOI: 10.1208/s12249-019-1504-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/05/2019] [Indexed: 01/29/2023] Open
Abstract
In this study, a novel human serum albumin nanoparticle loading silybin-phospholipid complex (SLNPs) was developed for liver targeting after intravenous administration. The preparation of the drug delivery system consisted of two steps; initially, a silybin-phospholipid complex (SLC) was produced to improve the lipophilicity of SLB to then achieve enhanced encapsulation of SLB in albumin nanoparticles. FT-IR and XRD analysis confirmed the successful formation of SLC. The complex ratio of SLC in the first step was 99.6%. The encapsulation efficiency and drug loading of SLNPs in the second step were 96.2% and 5.6%, respectively. SLNPs were spherical and well-dispersed, with a zeta potential of approximately - 10 mV, and a mean particle size around 200 nm. An in vivo tissue distribution experiment and a pharmacodynamic experiment showed that, compared with SLB solution, SLNPs had an improved SLB accumulation in the liver. The hepatoprotective effect of SLNPs on CCl4-induced acute liver damage was evaluated. CCl4-damaged mice showed an increased enzymatic activity of ALT and AST; however, enzyme levels returned to near-normal levels in high-dose SLNP-treated mice. As SLNPs combine the enhanced oil solubility of SLC and the passive targeting of albumin nanoparticles, they possess great potential for the treatment of acute liver damage.
Collapse
|
45
|
Okamoto Y, Taguchi K, Sakuragi M, Imoto S, Yamasaki K, Otagiri M. Preparation, Characterization, and in Vitro/in Vivo Evaluation of Paclitaxel-Bound Albumin-Encapsulated Liposomes for the Treatment of Pancreatic Cancer. ACS OMEGA 2019; 4:8693-8700. [PMID: 31459959 PMCID: PMC6649292 DOI: 10.1021/acsomega.9b00537] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/09/2019] [Indexed: 05/28/2023]
Abstract
Paclitaxel (PTX)-loaded liposomes were developed with the goal of enhancing the effects of cancer treatment. Although loading substances into the lipid membrane of liposome cause some destabilization of the lipid membrane, PTX was nearly exclusively embedded in the lipid membrane of liposomes, due to its low water solubility. Hydrophobic drugs can be encapsulated into the inner core of bovine serum albumin (BSA)-encapsulated liposomes (BSA-liposome) via noncovalent binding to albumin. Since PTX is able to noncovalently bind to albumin, we attempted to prepare PTX-loaded BSA-liposome (PTX-BSA-liposome). The amount of PTX loaded in the BSA-liposome could be increased substantially by using ethanol, since ethanol increases PTX solubility in BSA solutions via prompting the binding PTX to BSA. On the basis of the results of transmission electron microscopy and small-angle X-ray scattering, PTX-BSA-liposome formed unilamellar vesicles that were spherical in shape and the PTX was encapsulated into the inner aqueous core of the liposome as a form of PTX-BSA complex. In addition, the PTX-BSA-liposome, as well as nab-PTX, showed cytotoxicity against human pancreatic cancer cells, AsPC-1 cells, in a PTX concentration-dependent manner. The in vivo antitumor effect of PTX-BSA-liposomes was also observed in a mouse model that had been subcutaneously inoculated with pancreatic cancer cells by virtue of its high accumulation at the tumor site via the enhanced permeability retention effect. These results suggest that PTX-BSA-liposomes have the potential for serving as a novel PTX preparation method for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yuko Okamoto
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Kazuaki Taguchi
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
- Faculty
of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Mina Sakuragi
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Shuhei Imoto
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| |
Collapse
|
46
|
Wan X, Beaudoin JJ, Vinod N, Min Y, Makita N, Bludau H, Jordan R, Wang A, Sokolsky M, Kabanov AV. Co-delivery of paclitaxel and cisplatin in poly(2-oxazoline) polymeric micelles: Implications for drug loading, release, pharmacokinetics and outcome of ovarian and breast cancer treatments. Biomaterials 2019; 192:1-14. [PMID: 30415101 PMCID: PMC6331221 DOI: 10.1016/j.biomaterials.2018.10.032] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/16/2022]
Abstract
Concurrent delivery of multiple drugs using nanoformulations can improve outcomes of cancer treatments. Here we demonstrate that this approach can be used to improve the paclitaxel (PTX) and alkylated cisplatin prodrug combination therapy of ovarian and breast cancer. The drugs are co-loaded in the polymeric micelle system based on amphiphilic block copolymer poly(2-methyl-2-oxazoline-block-2-butyl-2-oxazoline-block-2-methyl-2-oxazoline) (P(MeOx-b-BuOx-b-MeOx). A broad range of drug mixing ratios and exceptionally high two-drug loading of over 50 wt.% drug in a stable micellar solution is demonstrated. The drugs co-loading in the micelles result in a slowed-down release to serum, improved pharmacokinetics and increased tumor distribution for both drugs. A superior anti-tumor activity of co-loaded PTX/CP drug micelles compared to single drug micelles or their mixture was demonstrated in cisplatin-resistant human ovarian carcinoma A2780/CisR xenograft tumor and multidrug resistant breast cancer LCC-6-MDR orthotopic tumor models. The improved tumor delivery of co-loaded drugs was related to decreased drug release rates as confirmed by simulation for micelle, serum and tumor compartments in a three-compartmental model. Overall, the results provide support for the use of PTX and cisplatin co-loaded micelles as a strategy for improved chemotherapy of ovarian and breast cancer and potential for the clinical translation.
Collapse
Affiliation(s)
- Xiaomeng Wan
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - James J Beaudoin
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Natasha Vinod
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Naoki Makita
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Herdis Bludau
- Chair of Macromolecular Chemistry, Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstr. 4, 01069 Dresden, Germany
| | - Rainer Jordan
- Chair of Macromolecular Chemistry, Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstr. 4, 01069 Dresden, Germany
| | - Andrew Wang
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Xuzhou Medical College, Xuzhou, China
| | - Marina Sokolsky
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|
47
|
Bastiancich C, Bozzato E, Luyten U, Danhier F, Bastiat G, Préat V. Drug combination using an injectable nanomedicine hydrogel for glioblastoma treatment. Int J Pharm 2019; 559:220-227. [PMID: 30703501 DOI: 10.1016/j.ijpharm.2019.01.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/25/2023]
Abstract
Lauroyl-gemcitabine lipid nanocapsules (GemC12-LNC) hydrogel, administered intratumorally or perisurgically in the tumor resection cavity, increases animal survival in several orthotopic GBM models. We hypothesized that GemC12-LNC can be used as nanodelivery platform for other drugs, to obtain a combined local therapeutic approach for GBM. Paclitaxel (PTX) was selected as a model molecule and PTX-GemC12-LNC formulation was evaluated in terms of physicochemical and mechanical properties. The PTX-GemC12-LNC hydrogel stability and drug release were evaluated over time showing no significant differences compared to GemC12-LNC. The drug combination was evaluated on several GBM cell lines showing increased cytotoxic activity compared to the original formulation and synergy between PTX and GemC12. Our results suggest that GemC12-LNC hydrogel can be used as nanodelivery platform for dual drug delivery to encapsulate active agents with different mechanisms of action to achieve a better antitumor efficacy against GBM or other solid tumors.
Collapse
Affiliation(s)
- Chiara Bastiancich
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Elia Bozzato
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Urszula Luyten
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Fabienne Danhier
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Guillaume Bastiat
- Micro & Nanomedecines Translationnelles - MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, UBL Université Bretagne Loire, Angers, France
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium.
| |
Collapse
|
48
|
Lugert S, Unterweger H, Mühlberger M, Janko C, Draack S, Ludwig F, Eberbeck D, Alexiou C, Friedrich RP. Cellular effects of paclitaxel-loaded iron oxide nanoparticles on breast cancer using different 2D and 3D cell culture models. Int J Nanomedicine 2018; 14:161-180. [PMID: 30613144 PMCID: PMC6306067 DOI: 10.2147/ijn.s187886] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Magnetic drug targeting (MDT) is an effective alternative for common drug applications, which reduces the systemic drug load and maximizes the effect of, eg, chemotherapeutics at the site of interest. After the conjugation of a magnetic carrier to a chemotherapeutic agent, the intra-arterial injection into a tumor-afferent artery in the presence of an external magnetic field ensures the accumulation of the drug within the tumor tissue. MATERIALS AND METHODS In this study, we used superparamagnetic iron oxide nanoparticles (SPIONs) coated with lauric acid and human serum albumin as carriers for paclitaxel (SPIONLA-HSA-Ptx). To investigate whether this particle system is suitable for a potential treatment of cancer, we investigated its physicochemical properties by dynamic light scattering, ζ potential measurements, isoelectric point titration, infrared spectroscopy, drug release quantification, and magnetic susceptibility measurements. The cytotoxic effects were evaluated using extensive toxicological methods using flow cytometry, IncuCyte® live-cell imaging, and growth experiments on different human breast cancer cell lines in two- and three-dimensional cell cultures. CONCLUSION The data showed that next to their high magnetization capability, SPIONLA-HSA-Ptx have similar cytostatic effects on human breast cancer cells as pure paclitaxel, suggesting their usage for future MDT-based cancer therapy.
Collapse
Affiliation(s)
- Stephan Lugert
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany,
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Harald Unterweger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany,
| | - Marina Mühlberger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany,
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany,
| | - Sebastian Draack
- Institut für Elektrische Messtechnik und Grundlagen der Elektrotechnik, TU Braunschweig, Braunschweig, Germany
| | - Frank Ludwig
- Institut für Elektrische Messtechnik und Grundlagen der Elektrotechnik, TU Braunschweig, Braunschweig, Germany
| | - Dietmar Eberbeck
- Physikalisch-Technische Bundesanstalt Braunschweig und Berlin, Berlin, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany,
| | - Ralf P Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Erlangen, Germany,
| |
Collapse
|
49
|
Nontoxic silver nanocluster-induced folding, fibrillation, and aggregation of blood plasma proteins. Int J Biol Macromol 2018; 119:838-848. [DOI: 10.1016/j.ijbiomac.2018.07.177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/27/2018] [Accepted: 07/28/2018] [Indexed: 11/18/2022]
|
50
|
Tang B, Qian Y, Gou Y, Cheng G, Fang G. VE-Albumin Core-Shell Nanoparticles for Paclitaxel Delivery to Treat MDR Breast Cancer. Molecules 2018; 23:E2760. [PMID: 30366367 PMCID: PMC6278303 DOI: 10.3390/molecules23112760] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/18/2018] [Accepted: 10/20/2018] [Indexed: 11/17/2022] Open
Abstract
Multi-drug resistance (MDR) presents a serious problem in cancer chemotherapy. In this study, Vitamin E (VE)-Albumin core-shell nanoparticles were developed for paclitaxel (PTX) delivery to improve the chemotherapy efficacy in an MDR breast cancer model. The PTX-loaded VE-Albumin core-shell nanoparticles (PTX-VE NPs) had small particle sizes (about 100 nm), high drug entrapment efficiency (95.7%) and loading capacity (12.5%), and showed sustained release profiles, in vitro. Docking studies indicated that the hydrophobic interaction and hydrogen bonds play a significant role in the formation of the PTX-VE NPs. The results of confocal laser scanning microscopy analysis demonstrated that the cell uptake of PTX was significantly increased by the PTX-VE NPs, compared with the NPs without VE (PTX NPs). The PTX-VE NPs also exhibited stronger cytotoxicity, compared with PTX NPs with an increased accumulation of PTX in the MCF-7/ADR cells. Importantly, the PTX-VE NPs showed a higher anti-cancer efficacy in MCF-7/ADR tumor xenograft model than the PTX NPs and the PTX solutions. Overall, the VE-Albumin core-shell nanoparticles could be a promising nanocarrier for PTX delivery to improve the chemotherapeutic efficacy of MDR cancer.
Collapse
Affiliation(s)
- Bo Tang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Yu Qian
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Yi Gou
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Gang Cheng
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, Liaoning, China.
| | - Guihua Fang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|