101
|
Fan N, Zhao J, Zhao W, Shen Y, Song Q, Shum HC, Wang Y, Rong J. Biodegradable celastrol-loaded albumin nanoparticles ameliorate inflammation and lipid accumulation in diet-induced obese mice. Biomater Sci 2022; 10:984-996. [PMID: 35019905 DOI: 10.1039/d1bm01637g] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Obesity is hallmarked by endoplasmic reticulum (ER) stress, chronic inflammation and metabolic dysfunctions. The control of obesity is the key to preventing the onset of non-alcoholic fatty liver disease, diabetes, cerebro-cardiovascular diseases and cancers. As a promising anti-obesity drug, plant-derived celastrol is challenged by poor water solubility and low oral bioavailability in clinical applications. The present study was designed to develop a biocompatible albumin-based nanoparticle carrier system for the controlled release of celastrol in diet-induced obese mice. Celastrol was loaded into bovine serum albumin (BSA) nanoparticles to yield celastrol-BSA-NPs by high pressure homogenization. Celastrol-BSA-NPs exhibited spherical morphology, narrow size distribution with a diameter of 125.6 ± 2.2 nm, satisfactory drug-loading efficiency at 13.88 ± 0.12% and a sustained-release profile over a period of 168 h. Compared with free celastrol, celastrol-BSA-NPs effectively improved cellular uptake, intestinal absorption and hepatic deposition. In animal experiments, celastrol-BSA-NPs outperformed free celastrol in lowering lipid accumulation, improving insulin sensitivity, and reducing inflammation in diet-induced obesity. Collectively, celastrol-BSA-NPs exhibited better bioavailability and in vivo efficacy in the treatment of diet-induced obesity. Importantly, such albumin-based nanoparticles may be a general biocompatible drug carrier system for the controlled release of hydrophobic compounds (e.g., celastrol) for the treatment of obesity and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Ni Fan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China.
| | - Jia Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China.
| | - Wei Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China.
| | - Yanting Shen
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong 999077, China
| | - Qingchun Song
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong 999077, China
| | - Ho Cheung Shum
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong 999077, China
| | - Yu Wang
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam Road, Hong Kong 999077, China
| | - Jianhui Rong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China. .,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen 518000, China
| |
Collapse
|
102
|
Lee WT, Yoon J, Kim SS, Kim H, Nguyen NT, Le XT, Lee ES, Oh KT, Choi HG, Youn YS. Combined Antitumor Therapy Using In Situ Injectable Hydrogels Formulated with Albumin Nanoparticles Containing Indocyanine Green, Chlorin e6, and Perfluorocarbon in Hypoxic Tumors. Pharmaceutics 2022; 14:pharmaceutics14010148. [PMID: 35057044 PMCID: PMC8781012 DOI: 10.3390/pharmaceutics14010148] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/28/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Combined therapy using photothermal and photodynamic treatments together with chemotherapeutic agents is considered one of the most synergistic treatment protocols to ablate hypoxic tumors. Herein, we sought to fabricate an in situ-injectable PEG hydrogel system having such multifunctional effects. This PEG hydrogel was prepared with (i) nabTM-technique-based paclitaxel (PTX)-bound albumin nanoparticles with chlorin-e6 (Ce6)-conjugated bovine serum albumin (BSA-Ce6) and indocyanine green (ICG), named ICG/PTX/BSA-Ce6-NPs (~175 nm), and (ii) an albumin-stabilized perfluorocarbon (PFC) nano-emulsion (BSA-PFC-NEs; ~320 nm). This multifunctional PEG hydrogel induced moderate and severe hyperthermia (41−42 °C and >48 °C, respectively) at the target site under two different 808 nm laser irradiation protocols, and also induced efficient singlet oxygen (1O2) generation under 660 nm laser irradiation supplemented by oxygen produced by ultrasound-triggered PFC. Due to such multifunctionality, our PEG hydrogel formula displayed significantly enhanced killing of three-dimensional 4T1 cell spheroids and also suppressed the growth of xenografted 4T1 cell tumors in mice (tumor volume: 47.7 ± 11.6 and 63.4 ± 13.0 mm3 for photothermal and photodynamic treatment, respectively, vs. PBS group (805.9 ± 138.5 mm3), presumably based on sufficient generation of moderate heat as well as 1O2/O2 even under hypoxic conditions. Our PEG hydrogel formula also showed excellent hyperthermal efficacy (>50 °C), ablating the 4T1 tumors when the irradiation duration was extended and output intensity was increased. We expect that our multifunctional PEG hydrogel formula will become a prototype for ablation of otherwise poorly responsive hypoxic tumors.
Collapse
Affiliation(s)
- Woo Tak Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Johyun Yoon
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Sung Soo Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Hanju Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Nguyen Thi Nguyen
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Xuan Thien Le
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Eun Seong Lee
- Department of Biotechnology and Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si 14662, Gyeonggi-do, Korea;
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Gyeonggi-do, Korea;
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
- Correspondence: ; Tel.: +82-31-290-7785
| |
Collapse
|
103
|
Tkachenko V, Kunemann P, Malval JP, Petithory T, Pieuchot L, Vidal L, Chemtob A. Kinetically stable sub-50 nm fluorescent block copolymer nanoparticles via photomediated RAFT dispersion polymerization for cellular imaging. NANOSCALE 2022; 14:534-545. [PMID: 34935832 DOI: 10.1039/d1nr04934h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Self-assembled block copolymer nanoparticles (NPs) have emerged as major potential nanoscale vehicles for fluorescence bioimaging. The preparation of NPs with high yields possessing high kinetic stability to prevent the leakage of fluorophore molecules is crucial to their practical implementation. Here, we report a photomediated RAFT polymerization-induced self-assembly (PISA) yielding uniform and nanosized poly((oligo(ethylene glycol) acrylate)-block-poly(benzyl acrylate) particles (POEGA-b-PBzA) with a concentration of 22 wt%, over 20 times more than with micellization and nanoprecipitation. The spherical diblock copolymer nanoparticles have an average size of 10-50 nm controllable through the degree of polymerization of the stabilizing POEGA block. Subsequent dialysis against water and swelling with Nile red solution led to highly stable fluorescent NPs able to withstand the changes in concentration, ionic strength, pH or temperature. A PBzA/water interfacial tension of 48.6 mN m-1 hinders the exchange between copolymer chains, resulting in the trapping of NPs in a "kinetically frozen" state responsible for high stability. A spectroscopic study combining fluorescence and UV-vis absorption agrees with a preferential distribution of fluorophores in the outer POEGEA shell despite its hydrophobic nature. Nile red-doped POEGA-b-PBzA micelles without initiator residues and unimers but with high structural stability turn out to be noncytotoxic, and can be used for the optical imaging of cells. Real-time confocal fluorescence microscopy shows a fast cellular uptake using C2C12 cell lines in minutes, and a preferential localization in the perinuclear region, in particular in the vesicles.
Collapse
Affiliation(s)
- Vitalii Tkachenko
- Université de Haute-Alsace, CNRS, IS2M UMR7361, F-68100 Mulhouse, France.
- Université de Strasbourg, France
| | - Philippe Kunemann
- Université de Haute-Alsace, CNRS, IS2M UMR7361, F-68100 Mulhouse, France.
- Université de Strasbourg, France
| | - Jean Pierre Malval
- Université de Haute-Alsace, CNRS, IS2M UMR7361, F-68100 Mulhouse, France.
- Université de Strasbourg, France
| | - Tatiana Petithory
- Université de Haute-Alsace, CNRS, IS2M UMR7361, F-68100 Mulhouse, France.
- Université de Strasbourg, France
| | - Laurent Pieuchot
- Université de Haute-Alsace, CNRS, IS2M UMR7361, F-68100 Mulhouse, France.
- Université de Strasbourg, France
| | - Loïc Vidal
- Université de Haute-Alsace, CNRS, IS2M UMR7361, F-68100 Mulhouse, France.
- Université de Strasbourg, France
| | - Abraham Chemtob
- Université de Haute-Alsace, CNRS, IS2M UMR7361, F-68100 Mulhouse, France.
- Université de Strasbourg, France
| |
Collapse
|
104
|
Mao W, Wang S, Mao D, Liu Y, Li L, Ma D. Supramolecular complexation with kinetic stabilization: cucurbit[6]uril encapsulated doxorubicin-based prodrugs for pH-responsive controlled release. NEW J CHEM 2022. [DOI: 10.1039/d1nj06237a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Kinetically-stabilized host–guest complexation for the construction of a pH-responsive drug delivery system.
Collapse
Affiliation(s)
- Weipeng Mao
- School of Pharmaceutical and Materials Engineering & Institute for Advanced Studies, Taizhou University, 1139 Shifu Avenue, Jiaojiang, Zhejiang 318000, China
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Shuyi Wang
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Dake Mao
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Yamin Liu
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Libai Li
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Da Ma
- School of Pharmaceutical and Materials Engineering & Institute for Advanced Studies, Taizhou University, 1139 Shifu Avenue, Jiaojiang, Zhejiang 318000, China
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai, 200433, China
| |
Collapse
|
105
|
Moreno-Lanceta A, Medrano-Bosch M, Edelman ER, Melgar-Lesmes P. Polymeric Nanoparticles for Targeted Drug and Gene Delivery Systems. NANOTECHNOLOGY IN THE LIFE SCIENCES 2022:561-608. [DOI: 10.1007/978-3-031-12658-1_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
106
|
Yan XJ, Li Z, Liu HB, Wang ZG, Fan J, Xie CZ, Li QZ, Xu JY. A chromone hydrazide Schiff base fluorescence probe with high selectivity and sensitivity for the detection and discrimination of human serum albumin (HSA) and bovine serum albumin (BSA). J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2021.113576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
107
|
Cho H, Shim MK, Yang S, Song S, Moon Y, Kim J, Byun Y, Ahn CH, Kim K. Cathepsin B-Overexpressed Tumor Cell Activatable Albumin-Binding Doxorubicin Prodrug for Cancer-Targeted Therapy. Pharmaceutics 2021; 14:83. [PMID: 35056979 PMCID: PMC8780658 DOI: 10.3390/pharmaceutics14010083] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 11/18/2022] Open
Abstract
Prodrugs are bioreversible medications that should undergo an enzymatic or chemical transformation in the tumor microenvironment to release active drugs, which improve cancer selectivity to reduce toxicities of anticancer drugs. However, such approaches have been challenged by poor therapeutic efficacy attributed to a short half-life and low tumor targeting. Herein, we propose cathepsin B-overexpressed tumor cell activatable albumin-binding doxorubicin prodrug, Al-ProD, that consists of a albumin-binding maleimide group, cathepsin B-cleavable peptide (FRRG), and doxorubicin. The Al-ProD binds to in situ albumin, and albumin-bound Al-ProD indicates high tumor accumulation with prolonged half-life, and selctively releases doxorubicin in cathepsin B-overexpressed tumor cells, inducing a potent antitumor efficacy. Concurrently, toxicity of Al-ProD toward normal tissues with innately low cathepsin B expression is significantly reduced by maintaining an inactive state, thereby increasing the safety of chemotherapy. This study offers a promising approach for effective and safe chemotherapy, which may open new avenues for drug design and translational medicine.
Collapse
Affiliation(s)
- Hanhee Cho
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.C.); (M.K.S.); (S.Y.); (S.S.); (Y.M.); (J.K.)
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Man Kyu Shim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.C.); (M.K.S.); (S.Y.); (S.S.); (Y.M.); (J.K.)
| | - Suah Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.C.); (M.K.S.); (S.Y.); (S.S.); (Y.M.); (J.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Sukyung Song
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.C.); (M.K.S.); (S.Y.); (S.S.); (Y.M.); (J.K.)
- Department of Biosystems & Biotechnology, Korea University, Seoul 02841, Korea
| | - Yujeong Moon
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.C.); (M.K.S.); (S.Y.); (S.S.); (Y.M.); (J.K.)
- Department of Bioengineering, Korea University, Seoul 02841, Korea
| | - Jinseong Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.C.); (M.K.S.); (S.Y.); (S.S.); (Y.M.); (J.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea;
| | - Cheol-Hee Ahn
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Kwangmeyung Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (H.C.); (M.K.S.); (S.Y.); (S.S.); (Y.M.); (J.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
108
|
Yang SJ, Huang HT, Huang CH, Pai JA, Wang CH, Shieh MJ. The synergistic effect of chemo-photothermal therapies in SN-38-loaded gold-nanoshell-based colorectal cancer treatment. Nanomedicine (Lond) 2021; 17:23-40. [PMID: 34918941 DOI: 10.2217/nnm-2021-0187] [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: 11/21/2022] Open
Abstract
Aim: 7-Ethyl-10-hydroxycamptothecin (SN-38)-loaded gold nanoshells nanoparticles (HSP@Au NPs) were developed for combined chemo-photothermal therapy to treat colorectal cancer. Materials & methods: SN-38-loaded nanoparticles (HSP NPs) were prepared by the lyophilization-hydration method, and then developed into gold nanoshells. The nanoparticles were characterized and assessed for photothermal properties, cytotoxicity and hemocompatibility in vitro. In vivo anticancer activity was tested in a tumor mouse model. Results: The HSP@Au NPs (diameter 186.9 nm, zeta potential 33.4 mV) led to significant cytotoxicity in cancer cells exposed to a near-infrared laser. Moreover, the HSP@Au NP-mediated chemo-photothermal therapy displayed significant tumor growth suppression and disappearance (25% of tumor clearance rate) without adverse side effects in vivo. Conclusion: HSP@Au NPs may be promising in the treatment of colorectal cancer in the future.
Collapse
Affiliation(s)
- Shu-Jyuan Yang
- Institute of Biomedical Engineering, College of Medicine & College of Engineering, National Taiwan University, Taipei, 100, Taiwan
| | - Hsiao-Ting Huang
- Institute of Biomedical Engineering, College of Medicine & College of Engineering, National Taiwan University, Taipei, 100, Taiwan
| | - Chung-Huan Huang
- Institute of Biomedical Engineering, College of Medicine & College of Engineering, National Taiwan University, Taipei, 100, Taiwan
| | - Jui-An Pai
- Institute of Biomedical Engineering, College of Medicine & College of Engineering, National Taiwan University, Taipei, 100, Taiwan
| | - Chung-Hao Wang
- Gene'e Tech Co. Ltd. 2F., No. 661, Bannan Rd., Zhonghe District, New Taipei City, 235, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, College of Medicine & College of Engineering, National Taiwan University, Taipei, 100, Taiwan.,Department of Oncology, National Taiwan University Hospital & College of Medicine, Taipei, 100, Taiwan
| |
Collapse
|
109
|
Solanki R, Rostamabadi H, Patel S, Jafari SM. Anticancer nano-delivery systems based on bovine serum albumin nanoparticles: A critical review. Int J Biol Macromol 2021; 193:528-540. [PMID: 34655592 DOI: 10.1016/j.ijbiomac.2021.10.040] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 01/23/2023]
Abstract
Among the health-promotional protein-based vehicles, bovine serum albumin nanoparticles (BSA NPs) are particularly interesting. Meeting requirements e. g., non-toxicity, non-immunogenicity, biodegradability, biocompatibility, and high drug-binding capacity, has introduced BSA NPs as a promising candidate for efficient anti-cancer drug delivery and its application is now a rapidly-growing strategy to promote cancer therapy. Nevertheless, the leverage of such carriers requires an in-depth understanding of structural/physicochemical features of the BSA molecule and its derived nanovehicles, together with the utilized nano-formulation approaches, effective variables in delivery mechanism, specific shortfalls, and recent nanoencapsulation progresses. The current review highlights the novel advances in the application of BSA NPs to engineer drug vehicles for delivering anti-cancer agents. The factors influencing the efficiency of the therapeutics in such nano-delivery systems, alongside their advantaged and limitations are also discussed.
Collapse
Affiliation(s)
- Raghu Solanki
- School of Life Sciences, Central University of Gujarat, Sector-30, Gandhinagar 382030, India
| | - Hadis Rostamabadi
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sunita Patel
- School of Life Sciences, Central University of Gujarat, Sector-30, Gandhinagar 382030, India.
| | - Seid Mahdi Jafari
- Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran.
| |
Collapse
|
110
|
Adeel M, Saorin G, Boccalon G, Sfriso AA, Parisi S, Moro I, Palazzolo S, Caligiuri I, Granchi C, Corona G, Cemazar M, Canzonieri V, Tuccinardi T, Rizzolio F. A carrier free delivery system of a monoacylglycerol lipase hydrophobic inhibitor. Int J Pharm 2021; 613:121374. [PMID: 34906647 DOI: 10.1016/j.ijpharm.2021.121374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 11/19/2022]
Abstract
Monoacylglycerol lipase (MAGL) is an emerging therapeutic target for cancer. It is involved in lipid metabolism and its inhibition impairs many hallmarks of cancer including cell proliferation, migration/invasion and tumor growth. For these reasons, our group has recently developed a potent reversible MAGL inhibitor (MAGL23), which showed promising anticancer activities. Here in, to improve its pharmacological properties, a nanoformulation based on nanocrystals coated with albumin was prepared for therapeutic applications. MAGL23 was solubilized by a nanocrystallization method with Pluronic F-127 as surfactant into an organic solvent and was recovered as nanocrystals in water after solvent evaporation. Finally, the solubilized nanocrystals were stabilized by human serum albumin to create a smart delivery carrier. An in-silico prediction (lipophilicity, structure at different pH and solubility in water), as well as experimental studies (solubility), have been performed to check the chemical properties of the inhibitor and nanocrystals. The solubility in water increases from less than 0.01 mg/mL (0.0008 mg/mL, predicted) up to 0.82 mg/mL in water. The formulated inhibitor maintained its potency in ovarian and colon cancer cell lines as the free drug. Furthermore, the system was thoroughly observed at each step of the solubilization process till the final formulation stage by different spectroscopic techniques and a comparative study was performed to check the effects of Pluronic F-127 and CTAB as surfactants. The formulated system is favorable to release the drug at physiological pH conditions (at pH 7.4, after 24 h, less than 20% of compound is released). In vivo studies have shown that albumin-complexed nanocrystals increase the therapeutic window of MAGL23 along with a favorable biodistribution. As per our knowledge, we are reporting the first ever nanoformulation of a MAGL inhibitor, which is promising as a therapeutic system where the MAGL enzyme is involved, especially for cancer therapeutic applications.
Collapse
Affiliation(s)
- Muhammad Adeel
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy; Dotoctoral School in Science and Technology of Bio and Nanomaterials, Ca'Foscari University of Venice, Venezia-Mestre, Italy; Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Gloria Saorin
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy; Dotoctoral School in Science and Technology of Bio and Nanomaterials, Ca'Foscari University of Venice, Venezia-Mestre, Italy
| | - Giacomo Boccalon
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy
| | | | - Salvatore Parisi
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy; Doctoral School in Molecular Biomedicine, University of Trieste, Trieste, Italy
| | - Isabella Moro
- Department of Biology, University of Padua, Padua, Italy
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | | | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Pisa, Italy; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca'Foscari University of Venice, Venezia-Mestre, Italy; Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy.
| |
Collapse
|
111
|
Iikuni S, Kitano A, Watanabe H, Ono M. Feasibility of using a 99mTc-hydroxamamide complex containing an albumin binder moiety for in vivo albumin labeling-based tumor imaging. Bioorg Med Chem Lett 2021; 53:128417. [PMID: 34710623 DOI: 10.1016/j.bmcl.2021.128417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 11/29/2022]
Abstract
Human serum albumin (HSA), which is distributed throughout the blood, is used as a carrier for transporting drugs to tumors based on the enhanced permeability and retention (EPR) effect. To develop an agent for the in vivo radiolabeling of endogenous albumin, we designed and synthesized novel hydroxamamide (Ham)-based technetium-99m (99mTc) complexes, which contained a monovalent or bivalent 4-(4-iodophenyl)butyric acid (IA) derivative as an albumin binder (ALB) moiety ([99mTc]AB2 and [99mTc]ALB2, respectively), and evaluated their utility for in vivo tumor imaging. In an in vitro HSA-binding assay, [99mTc]AB2 and [99mTc]ALB2 showed greater binding to HSA than [99mTc]BHam, a 99mTc-Ham complex without an ALB moiety. In an in vivo biodistribution assay, [99mTc]ALB2 showed marked blood and tumor retention (25.13 and 4.61% injected dose (ID)/g, respectively, at 1 h postinjection), suggesting that the EPR effect had been induced. However, [99mTc]AB2 showed no marked blood or tumor retention (4.16 and 0.75% ID/g, respectively, at 1 h postinjection), probably because the affinity of the monovalent IA derivative for albumin was insufficient to induce the EPR effect. These findings indicated that the multivalent interactions of [99mTc]ALB2 had enhanced its affinity for albumin. 99mTc-complexes containing multivalent ALB moieties may be useful for tumor imaging.
Collapse
Affiliation(s)
- Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Anna Kitano
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
112
|
Promoted antitumor therapy on pancreatic cancer by a novel recombinant human albumin-bound miriplatin nanoparticle. Eur J Pharm Sci 2021; 167:106000. [PMID: 34517105 DOI: 10.1016/j.ejps.2021.106000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/27/2021] [Accepted: 09/07/2021] [Indexed: 11/20/2022]
Abstract
Pancreatic cancer is an aggressive and highly lethal disease with a very poor prognosis. Our previous study found miriplatin can inhibit proliferation of various tumor cells, including pancreatic cancer cells. For the chemotherapy of pancreatic cancer, a novel recombinant human serum albumin (rHSA)-bound miriplatin nanoparticles (rHSA-miPt) were constructed by emulsion-diffusion evaporation method. The optimal formulation was composed of 150 mg of rHSA and 30 mg of miriplatin. The key parameters in rHSA-miPt production were 10 min of high-pressure homogenization in a solution with volume ratio of 10:2 of 5% glucose and chloroform. The rHSA-miPt was characterized with a particle size of 61 ± 10 nm, a zeta potential value of -18 ± 5 mV, encapsulation efficiency of 98.4%, drug loading of 16.4%, T1/2 of 13.3 h and Vd of 0.5 L in Sprague Dawley rats. The concentrations of platinum (Pt) in the tumors were 15 and 22-fold higher than those in the blood at 24 and 72 h in tumor-bearing mice, respectively. The internalization of rHSA-miPt through caveolae-dependent pathway. In vitro, the half-maximal inhibitory concentration (IC50) of rHSA-miPt was 12.7 μM vs more than 100 μM of gemcitabine (Gem). The inhibition rate of tumor growth was 76% of rHSA-miPt and 51% of Gem, respectively. Compared with Gem, rHSA-miPt was identified to be safer and less toxic based on body weight loss in mice (0% vs 20%), the survival rate of mice (100% vs 80%) and hematological and biochemical parameters of the mice including leukocytes, lymphocytes, neutrophils, monocytes, serum alanine aminotransferase and aspartate aminotransferase. The present study revealed that rHSA-miPt might be a promising candidate for pancreatic cancer therapy.
Collapse
|
113
|
de Hoyos-Vega JM, Hong HJ, Stybayeva G, Revzin A. Hepatocyte cultures: From collagen gel sandwiches to microfluidic devices with integrated biosensors. APL Bioeng 2021; 5:041504. [PMID: 34703968 PMCID: PMC8519630 DOI: 10.1063/5.0058798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes are parenchymal cells of the liver responsible for drug detoxification, urea and bile production, serum protein synthesis, and glucose homeostasis. Hepatocytes are widely used for drug toxicity studies in bioartificial liver devices and for cell-based liver therapies. Because hepatocytes are highly differentiated cells residing in a complex microenvironment in vivo, they tend to lose hepatic phenotype and function in vitro. This paper first reviews traditional culture approaches used to rescue hepatic function in vitro and then discusses the benefits of emerging microfluidic-based culture approaches. We conclude by reviewing integration of hepatocyte cultures with bioanalytical or sensing approaches.
Collapse
Affiliation(s)
- Jose M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
114
|
Interaction of Supramolecular Congo Red and Congo Red-Doxorubicin Complexes with Proteins for Drug Carrier Design. Pharmaceutics 2021; 13:pharmaceutics13122027. [PMID: 34959309 PMCID: PMC8707210 DOI: 10.3390/pharmaceutics13122027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Targeted immunotherapy has expanded to simultaneous delivery of drugs, including chemotherapeutics. The aim of the presented research is to design a new drug carrier system. Systems based on the use of proteins as natural components of the body offer the chance to boost safety and efficacy of targeted drug delivery and excess drug removal. Congo red (CR) type supramolecular, self-assembled ribbon-like structures (SRLS) were previously shown to interact with some proteins, including albumin and antibodies complexed with antigen. CR can intercalate some chemotherapeutics including doxorubicin (Dox). The goal of this work was to describe the CR-Dox complexes, to analyze their interaction with some proteins, and to explain the mechanism of this interaction. In the present experiments, a model system composed of heated immunoglobulin light chain Lλ capable of CR binding was used. Heat aggregated immunoglobulins (HAI) and albumin were chosen as another model system. The results of experiments employing methods such as gel filtration chromatography and dynamic light scattering confirmed the formation of the CR-Dox complex of large size and properties different from the free CR structures. Electrophoresis and chromatography experiments have shown the binding of free CR to heated Lλ while CR-Dox mixed structures were not capable of forming such complexes. HAI was able to bind both free CR and CR-Dox complexes. Albumin also bound both CR and its complex with Dox. Additionally, we observed that albumin-bound CR-Dox complexes were transferred from albumin to HAI upon addition of HAI. DLS analyses showed that interaction of CR with Dox distinctly increased the hydrodynamic diameter of CR-Dox compared with a free CR supramolecular structure. To our knowledge, individual small proteins such as Lλ may bind upon heating a few molecules of Congo red tape penetrating protein body due to the relatively low cohesion of the dye micelle. If, however, the compactness is high (in the case of, e.g., CR-Dox) large ribbon-like, micellar structures appear. They do not divide easily into smaller portions and cannot attach to proteins where there is no room for binding large ligands. Such binding is, however, possible by albumin which is biologically adapted to form complexes with different large ligands and by tightly packed immune complexes and heat aggregated immunoglobulin-specific protein complex structures of even higher affinity for Congo red than albumin. The CR clouds formed around them also bind the CR-Dox complexes. The presented research is essential in the search for optimum solutions for SRLS application in immuno-targeting therapeutic strategies, especially with the use of chemotherapeutics.
Collapse
|
115
|
Gevenois PJLY, De Pauw P, Schoonooghe S, Delporte C, Sebti T, Amighi K, Muyldermans S, Wauthoz N. Development of Neutralizing Multimeric Nanobody Constructs Directed against IL-13: From Immunization to Lead Optimization. THE JOURNAL OF IMMUNOLOGY 2021; 207:2608-2620. [PMID: 34645688 DOI: 10.4049/jimmunol.2100250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/16/2021] [Indexed: 11/19/2022]
Abstract
IL-13 is a pleiotropic cytokine mainly secreted by Th2 cells. It reacts with many different types of cells involved in allergy, inflammation, and fibrosis, e.g., mastocytes, B cells, and fibroblasts. The role of IL-13 in conditions involving one or several of these phenotypes has therefore been extensively investigated. The inhibition of this cytokine in animal models for various pathologies yielded highly promising results. However, most human trials relying on anti-IL-13 conventional mAbs have failed to achieve a significant improvement of the envisaged disorders. Where some studies might have suffered from several weaknesses, the strategies themselves, such as targeting only IL-13 using conventional mAbs or employing a systemic administration, could be questioned. Nanobodies are recombinant Ag-binding fragments derived from the variable part of H chain-only Abs occurring in Camelidae. Thanks to their single-domain structure, small size (≈15 kDa), good stability, and solubility, they can be engineered into multispecific constructs for combined therapies or for use in new strategies such as formulations for local administration, e.g., pulmonary administration. In this study, we describe the generation of 38 nanobodies that can be subdivided into five CDR3 families. Nine nanobodies were found to have a good affinity profile (KD = 1-200 nM), but none were able to strongly inhibit IL-13 biological activity in vitro (IC50 > 50 µM: HEK-Blue IL-13/IL-4 cells). Multimeric constructs were therefore designed from these inhibitors and resulted in an up to 36-fold improvement in affinity and up to 300-fold enhancement of the biological activity while conserving a high specificity toward IL-13.
Collapse
Affiliation(s)
- Philippe J-L Y Gevenois
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium;
| | - Pieter De Pauw
- Laboratory of Cellular and Molecular Immunology, Free University of Brussels, Ixelles, Belgium
| | - Steve Schoonooghe
- Flemish Institute for Biotechnology Nanobody Core, Free University of Brussels, Brussels, Belgium
| | - Cédric Delporte
- Unit of Pharmacognosy, Bioanalysis and Drug Discovery, RD3 and Analytical Platform of the Faculty of Pharmacy, Free University of Brussels, Brussels, Belgium; and
| | | | - Karim Amighi
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Free University of Brussels, Ixelles, Belgium
| | - Nathalie Wauthoz
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium
| |
Collapse
|
116
|
Recent advances in supramolecular activatable phthalocyanine-based photosensitizers for anti-cancer therapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214155] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
117
|
Hornok V. Serum Albumin Nanoparticles: Problems and Prospects. Polymers (Basel) 2021; 13:3759. [PMID: 34771316 PMCID: PMC8586933 DOI: 10.3390/polym13213759] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
The present paper aims to summarize the results regarding serum albumin-based nanoparticles (NPs) for drug delivery purposes. In particular, it focuses on the relationship between their preparation techniques and synthesis parameters, as well as their successful clinical application. In spite of the huge amount of consumed material and immaterial sources and promising possibilities, products made from different types of albumin NPs, with the exception of a few, still have not been invented. In the present paper, promising applications of serum albumin nanoparticles (SANPs) for different biomedical purposes, such as carriers, delivery systems and contrast agents, are also discussed. The most frequent utilization of the NPs for certain diseases, i.e., cancer therapy, and future prospects are also detailed in this study.
Collapse
Affiliation(s)
- Viktória Hornok
- Department of Physical Chemistry and Materials Science, University of Szeged, Rerrich B. Square 1, H-6720 Szeged, Hungary; ; Tel.: +36-62-544211
- MTA Premium Post Doctoral Research Program, Rerrich B. Square 1, H-6720 Szeged, Hungary
| |
Collapse
|
118
|
Popova T, Dymova MA, Koroleva LS, Zakharova OD, Lisitskiy VA, Raskolupova VI, Sycheva T, Taskaev S, Silnikov VN, Godovikova TS. Homocystamide Conjugates of Human Serum Albumin as a Platform to Prepare Bimodal Multidrug Delivery Systems for Boron Neutron Capture Therapy. Molecules 2021; 26:molecules26216537. [PMID: 34770947 PMCID: PMC8586956 DOI: 10.3390/molecules26216537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
Boron neutron capture therapy is a unique form of adjuvant cancer therapy for various malignancies including malignant gliomas. The conjugation of boron compounds and human serum albumin (HSA)-a carrier protein with a long plasma half-life-is expected to extend systemic circulation of the boron compounds and increase their accumulation in human glioma cells. We report on the synthesis of fluorophore-labeled homocystamide conjugates of human serum albumin and their use in thiol-'click' chemistry to prepare novel multimodal boronated albumin-based theranostic agents, which could be accumulated in tumor cells. The novelty of this work involves the development of the synthesis methodology of albumin conjugates for the imaging-guided boron neutron capture therapy combination. Herein, we suggest using thenoyltrifluoroacetone as a part of an anticancer theranostic construct: approximately 5.4 molecules of thenoyltrifluoroacetone were bound to each albumin. Along with its beneficial properties as a chemotherapeutic agent, thenoyltrifluoroacetone is a promising magnetic resonance imaging agent. The conjugation of bimodal HSA with undecahydro-closo-dodecaborate only slightly reduced human glioma cell line viability in the absence of irradiation (~30 μM of boronated albumin) but allowed for neutron capture and decreased tumor cell survival under epithermal neutron flux. The simultaneous presence of undecahydro-closo-dodecaborate and labeled amino acid residues (fluorophore dye and fluorine atoms) in the obtained HSA conjugate makes it a promising candidate for the combination imaging-guided boron neutron capture therapy.
Collapse
Affiliation(s)
- Tatyana Popova
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Maya A Dymova
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
| | - Ludmila S Koroleva
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
| | - Olga D Zakharova
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
| | - Vladimir A Lisitskiy
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
| | - Valeria I Raskolupova
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Tatiana Sycheva
- Budker Institute of Nuclear Physics, SB RAS, 630090 Novosibirsk, Russia
| | - Sergei Taskaev
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
- Budker Institute of Nuclear Physics, SB RAS, 630090 Novosibirsk, Russia
| | - Vladimir N Silnikov
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
| | - Tatyana S Godovikova
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
119
|
Enantioresolution and Binding Affinity Studies on Human Serum Albumin: Recent Applications and Trends. CHEMOSENSORS 2021. [DOI: 10.3390/chemosensors9110304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interaction between proteins and drugs or other bioactive compounds has been widely explored over the past years. Several methods for analysis of this phenomenon have been developed and improved. Nowadays, increasing attention is paid to innovative methods, such as high performance affinity liquid chromatography (HPALC) and affinity capillary electrophoresis (ACE), taking into account various advantages. Moreover, the development of separation methods for the analysis and resolution of chiral drugs has been an area of ongoing interest in analytical and medicinal chemistry research. In addition to bioaffinity binding studies, both HPALC and ACE al-low one to perform other type of analyses, namely, displacement studies and enantioseparation of racemic or enantiomeric mixtures. Actually, proteins used as chiral selectors in chromatographic and electrophoretic methods have unique enantioselective properties demonstrating suitability for the enantioseparation of a large variety of chiral drugs or other bioactive compounds. This review is mainly focused in chromatographic and electrophoretic methods using human serum albumin (HSA), the most abundant plasma protein, as chiral selector for binding affinity analysis and enantioresolution of drugs. For both analytical purposes, updated examples are presented to highlight recent applications and current trends.
Collapse
|
120
|
Pivarcsik T, Dömötör O, Mészáros JP, May NV, Spengler G, Csuvik O, Szatmári I, Enyedy ÉA. 8-Hydroxyquinoline-Amino Acid Hybrids and Their Half-Sandwich Rh and Ru Complexes: Synthesis, Anticancer Activities, Solution Chemistry and Interaction with Biomolecules. Int J Mol Sci 2021; 22:ijms222011281. [PMID: 34681939 PMCID: PMC8570331 DOI: 10.3390/ijms222011281] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022] Open
Abstract
Solution chemical properties of two novel 8-hydroxyquinoline-D-proline and homo-proline hybrids were investigated along with their complex formation with [Rh(η5-C5Me5)(H2O)3]2+ and [Ru(η6-p-cymene)(H2O)3]2+ ions by pH-potentiometry, UV-visible spectrophotometry and 1H NMR spectroscopy. Due to the zwitterionic structure of the ligands, they possess excellent water solubility as well as their complexes. The complexes exhibit high solution stability in a wide pH range; no significant dissociation occurs at physiological pH. The hybrids and their Rh(η5-C5Me5) complexes displayed enhanced cytotoxicity in human colon adenocarcinoma cell lines and exhibited multidrug resistance selectivity. In addition, the Rh(η5-C5Me5) complexes showed increased selectivity to the chemosensitive cancer cells over the normal cells; meanwhile, the Ru(η6-p-cymene) complexes were inactive, most likely due to arene loss. Interaction of the complexes with human serum albumin (HSA) and calf-thymus DNA (ct-DNA) was investigated by capillary electrophoresis, fluorometry and circular dichroism. The complexes are able to bind strongly to HSA and ct-DNA, but DNA cleavage was not observed. Changing the five-membered proline ring to the six-membered homoproline resulted in increased lipophilicity and cytotoxicity of the Rh(η5-C5Me5) complexes while changing the configuration (L vs. D) rather has an impact on HSA or ct-DNA binding.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (O.D.); (J.P.M.); (G.S.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
| | - Orsolya Dömötör
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (O.D.); (J.P.M.); (G.S.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
| | - János P. Mészáros
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (O.D.); (J.P.M.); (G.S.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
| | - Nóra V. May
- Centre for Structural Science, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary;
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (O.D.); (J.P.M.); (G.S.)
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis U. 6, H-6725 Szeged, Hungary
| | - Oszkár Csuvik
- Institute of Pharmaceutical Chemistry and Stereochemistry Research Group of Hungarian Academy of Sciences, University of Szeged, Eötvös U. 6, H-6720 Szeged, Hungary; (O.C.); (I.S.)
| | - István Szatmári
- Institute of Pharmaceutical Chemistry and Stereochemistry Research Group of Hungarian Academy of Sciences, University of Szeged, Eötvös U. 6, H-6720 Szeged, Hungary; (O.C.); (I.S.)
| | - Éva A. Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (O.D.); (J.P.M.); (G.S.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
121
|
Singh AK, Italiya KS, Narisepalli S, Chitkara D, Mittal A. Role of Chain Length and Degree of Unsaturation of Fatty Acids in the Physicochemical and Pharmacological Behavior of Drug-Fatty Acid Conjugates in Diabetes. J Med Chem 2021; 64:14217-14229. [PMID: 34581574 DOI: 10.1021/acs.jmedchem.1c00391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Several drug-fatty acid (FA) prodrugs have been reported to exhibit desirable physicochemical and pharmacological profile; however, comparative beneficial effects rendered by different FAs have not been explored. In the present study, four different FAs (linoleic acid, oleic acid, palmitic acid, and α-lipoic acid) were selected based on their chain length and degree of unsaturation and conjugated to Lisofylline (LSF), an antidiabetic molecule to obtain different drug-FA prodrugs and characterized for molecular weight, hydrophobicity, purity, self-assembly, and efficacy in vitro and in vivo in type 1 diabetes model. Prodrugs demonstrated a 2- to 6-fold increase in the plasma half-life of LSF. Diabetic animals treated with prodrugs, once daily for 5 weeks, maintained a steady fasting blood glucose level with a significant increase in insulin level, considerable restoration of biochemical parameters, and preserved β-cells integrity. Among the different LSF-FA prodrugs, LSF-OA and LSF-PA demonstrated the most favorable physicochemical, systemic pharmacokinetic, and pharmacodynamic profiles.
Collapse
Affiliation(s)
- Arihant Kumar Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Kishan S Italiya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Saibhargav Narisepalli
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| |
Collapse
|
122
|
Experimental and Computational Validation of Structural Features and BSA Binding Tendency of 5‐Hydroxy‐5‐trifluoromethyl‐3‐arylpyrazolines**. ChemistrySelect 2021. [DOI: 10.1002/slct.202102669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
123
|
Molecular-Level Release of Coumarin-3-Carboxylic Acid and Warfarin-Derivatives from BSA-Based Hydrogels. Pharmaceutics 2021; 13:pharmaceutics13101661. [PMID: 34683955 PMCID: PMC8539358 DOI: 10.3390/pharmaceutics13101661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 11/17/2022] Open
Abstract
This investigation aimed at developing BSA hydrogels as a controlled release system to study the release behavior of spin-labeled coumarin-3-carboxylic acid (SL-CCS) and warfarin (SL-WFR). The release profiles of these spin-labeled (SL-) pharmaceuticals from BSA hydrogels prepared with different procedures are compared in detail. The mechanical properties of the gels during formation and release were studied via rheology, while a nanoscopic view on the release behavior was achieved by analyzing SL-drugs–BSA interaction using continuous wave electron paramagnetic resonance (CW EPR) spectroscopy. The influence of type of drug, drug concentration, duration of gel formation, and gelation methods on release behavior were characterized by CW EPR spectroscopy, EPR imaging (EPRI), and dynamic light scattering (DLS), which provide information on the interaction of BSA with SL-drugs, the percentage of drug inside the hydrogel and the nature and size of the released structures, respectively. We found that the release rate of SL-CCS and SL-WFR from BSA hydrogels is tunable through drug ratios, hydrogel incubation time and gelation procedures. All of the results indicate that BSA hydrogels can be potentially exploited in controlled drug delivery applications.
Collapse
|
124
|
Wang S, Liu X, Wang S, Ouyang L, Li H, Ding J, Deng G, Zhou W. Imatinib co-loaded targeted realgar nanocrystal for synergistic therapy of chronic myeloid leukemia. J Control Release 2021; 338:190-200. [PMID: 34428479 DOI: 10.1016/j.jconrel.2021.08.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/26/2022]
Abstract
Discovery of BCR-ABL1 tyrosine kinase inhibitors (TKIs) has revolutionized the therapy of chronic myeloid leukemia (CML), a malignant myeloproliferative disease characterized by abnormal activation of BCR-ABL fusion oncoprotein with protein tyrosine kinase (PTK) activity. However, the long-term treatment outcomes with TKIs are strongly limited by multiple drug resistances, resulting in relapse albeit with initial high response rate. Here, we reported a realgar (As4S4) nanocrystal-based delivery system to reverse drug resistance for synergistic CML therapy. While As4S4 is extremely insoluble in water, bovine serum albumin (BSA) was rationally screened to effectively stabilize As4S4 nanocrystal with uniformed size of ~40 nm. Imatinib (IMA), a representative TKIs, can be readily loaded into the hydrophobic domain of BSA to develop As4S4/IMA co-delivery system. Mechanistically, IMA inhibits PTK activity, while As4S4 degrades BCR-ABL1, which co-contribute to tumor suppression via complementary pathways for synergistic effect. Moreover, the nanosystem was modified with folic acid (FA) to enable tumor targetability, which has been demonstrated both in vitro and in vivo, resulting in robust tumor growth inhibition and significantly prolonged mice survival without any noticeable adverse effects. This work designed a synergistic nanoplatform for targeted CML therapy, provided a strategy to address the key limitation of As4S4 for biomedical applications, and highlighted the advantages of the combination between traditional Chinese and western medicine for diseases treatment.
Collapse
Affiliation(s)
- Shengmei Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Xuanjun Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Shengfeng Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China; Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Linqi Ouyang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Hui Li
- Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412008, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Guiming Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Wenhu Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
125
|
Gao Y, Yue Y, Xiong S. An Albumin-Binding Domain Peptide Confers Enhanced Immunoprotection Against Viral Myocarditis by CVB3 VP1 Vaccine. Front Immunol 2021; 12:666594. [PMID: 34630378 PMCID: PMC8492941 DOI: 10.3389/fimmu.2021.666594] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Coxsackievirus B3 (CVB3)-induced viral myocarditis is a common clinical cardiovascular disease without effective available vaccine. In this study, we tried to potentiate the immunoprotection efficacy of our previous CVB3-specific VP1 protein vaccine by introducing a streptococcal protein G-derived, draining lymph nodes (dLNs)-targeting albumin-binding domain (ABD) peptide. We found that compared with the original VP1 vaccine, ABD-fused VP1 (ABD-VP1) vaccine gained the new ability to efficiently bind murine albumin both in vitro and in vivo, possessed a much longer serum half-life in serum and exhibited more abundance in the dLNs after immunization. Accordingly, ABD-VP1 immunization not only significantly facilitated the enrichment and maturation of dendritic cells (DCs), induced higher percentages of IFN-γ+ CD8 + cells in the dLNs, but also robustly promoted VP1-induced T cell proliferation and cytotoxic T lymphocyte (CTL) responses in the spleens. More importantly, ABD-VP1 also elicited higher percentages of protective CD44hi CD62Lhi memory T cells in dLNs and spleens. Consequently, obvious protective effect against viral myocarditis was conferred by ABD-VP1 vaccine compared to the VP1 vaccine, reflected by the less body weight loss, improved cardiac function, alleviated cardiac histomorphological changes and an increased 28-day survival rate. Our results indicated that the ABD might be a promising immune-enhancing regime for vaccine design and development.
Collapse
Affiliation(s)
| | - Yan Yue
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| | - Sidong Xiong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| |
Collapse
|
126
|
Han S, Mei L, Quach T, Porter C, Trevaskis N. Lipophilic Conjugates of Drugs: A Tool to Improve Drug Pharmacokinetic and Therapeutic Profiles. Pharm Res 2021; 38:1497-1518. [PMID: 34463935 DOI: 10.1007/s11095-021-03093-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/05/2021] [Indexed: 01/19/2023]
Abstract
Lipophilic conjugates (LCs) of small molecule drugs have been used widely in clinical and pre-clinical studies to achieve a number of pharmacokinetic and therapeutic benefits. For example, lipophilic derivatives of drugs are employed in several long acting injectable products to provide sustained drug exposure for hormone replacement therapy and to treat conditions such as neuropsychiatric diseases. LCs can also be used to modulate drug metabolism, and to enhance drug permeation across membranes, either by increasing lipophilicity to enhance passive diffusion or by increasing protein-mediated active transport. Furthermore, such conjugation strategies have been employed to promote drug association with endogenous macromolecular carriers (e.g. albumin and lipoproteins), and this in turn results in altered drug distribution and pharmacokinetic profiles, where the changes can be 'general' (e.g. prolonged plasma half-life) or 'specific' (e.g. enhanced delivery to specific tissues in parallel with the macromolecular carriers). Another utility of LCs is to enhance the encapsulation of drugs within engineered nanoscale drug delivery systems, in order to best take advantage of the targeting and pharmacokinetic benefits of nanomedicines. The current review provides a summary of the mechanisms by which lipophilic conjugates, including in combination with delivery vehicles, can be used to control drug delivery, distribution and therapeutic profiles. The article is structured into sections which highlight a specific benefit of LCs and then demonstrate this benefit with case studies. The review attempts to provide a toolbox to assist researchers to design and optimise drug candidates, including consideration of drug-formulation compatibility.
Collapse
Affiliation(s)
- Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
- Suzhou Institute of Drug Innovation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China.
| | - Lianghe Mei
- Suzhou Institute of Drug Innovation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Tim Quach
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- PureTech Health, 6 Tide Street, Boston, MA, 02210, USA
| | - Chris Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Natalie Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
| |
Collapse
|
127
|
Robinson-Duggon J, McTiernan CD, Muñoz M, Guerra D, Escobar Álvarez E, Andrade-Villalobos F, Fierro A, Edwards AM, Alarcon EI, Fuentealba D. Biosupramolecular complexes of amphiphilic photosensitizers with human serum albumin and cucurbit[7]uril as carriers for photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 223:112284. [PMID: 34450362 DOI: 10.1016/j.jphotobiol.2021.112284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/21/2021] [Accepted: 08/09/2021] [Indexed: 01/17/2023]
Abstract
In the present work, we evaluated the supramolecular interactions between three photosensitizers, namely toluidine blue O (TBO, positively charged) and two fatty acid conjugates of 6 and 14 carbon atoms chain lengths (TBOC6 and TBOC14), with human serum albumin (HSA) and the macrocycle cucurbit[7]uril (CB[7]), alone or in combination within a biosupramolecular system as potential carriers of photosensitizers for Photodynamic therapy (PDT). Binding studies were carried out using photophysical and calorimetric techniques and accompanied with molecular docking simulations. Amphiphilic photosensitizers, particularly TBOC14, showed stronger binding to HSA and (CB[7]). Comparing the different delivery systems, (CB[7]) had a marginal effect on cell uptake and phototoxicity in HeLa cells, while HSA showed enhanced cell uptake with phototoxicities that depended on the photosensitizer. Despite low cell uptake, the combination of both (CB[7]) and HSA was the most phototoxic, which illustrates the potential of combining these systems for PDT applications.
Collapse
Affiliation(s)
- José Robinson-Duggon
- Laboratorio de Química Biosupramolecular, Departamento de Química Física, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile; Departamento de Bioquímica, Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Panamá 0824, Panamá.
| | - Christopher D McTiernan
- BEaTS Research Laboratory, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada
| | - Marcelo Muñoz
- BEaTS Research Laboratory, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada
| | - Daniel Guerra
- Laboratorio de Química Biosupramolecular, Departamento de Química Física, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile
| | - Elizabeth Escobar Álvarez
- Laboratorio de Química Biosupramolecular, Departamento de Química Física, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile
| | - Felipe Andrade-Villalobos
- Laboratorio de Química Biosupramolecular, Departamento de Química Física, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile; Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile
| | - Angélica Fierro
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile
| | - Ana María Edwards
- Laboratorio de Química Biosupramolecular, Departamento de Química Física, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile
| | - Emilio I Alarcon
- BEaTS Research Laboratory, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Denis Fuentealba
- Laboratorio de Química Biosupramolecular, Departamento de Química Física, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile.
| |
Collapse
|
128
|
TiO 2@BSA nano-composites investigated through orthogonal multi-techniques characterization platform. Colloids Surf B Biointerfaces 2021; 207:112037. [PMID: 34416445 DOI: 10.1016/j.colsurfb.2021.112037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/30/2021] [Accepted: 08/12/2021] [Indexed: 11/22/2022]
Abstract
Biocompatible coating based on bovine serum albumin (BSA) was applied on two different TiO2 nanoparticles (aeroxide P25 and food grade E171) to investigate properties and stability of resulting TiO2@BSA composites, under the final perspective to create a "Safe-by-Design" coating, able to uniform, level off and mitigate surface chemistry related phenomena, as naturally occurring when nano-phases come in touch with proteins enriched biological fluids. The first step towards validating the proposed approach is a detailed characterization of surface chemistry with the quantification of amount and stability of BSA coating deposited on nanoparticles' surfaces. At this purpose, we implemented an orthogonal multi-techniques characterization platform, providing important information on colloidal behavior, particle size distribution and BSA-coating structure of investigated TiO2 systems. Specifically, the proposed orthogonal approach enabled the quantitative determination of bound and free (not adsorbed) BSA, a key aspect for the design of intentionally BSA coated nano-structures, in nanomedicine and, overall, for the control of nano-surface reactivity. In fact, the BSA-coating strategy developed and the orthogonal characterisation performed can be extended to different designed nanomaterials in order to further investigate the protein-corona formation and promote the implementation of BSA engineered coating as a strategy to harmonize the surface reactivity and minimize the biological impact.
Collapse
|
129
|
Thermostable and Long-Circulating Albumin-Conjugated Arthrobacter globiformis Urate Oxidase. Pharmaceutics 2021; 13:pharmaceutics13081298. [PMID: 34452259 PMCID: PMC8400835 DOI: 10.3390/pharmaceutics13081298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Urate oxidase derived from Aspergillus flavus has been investigated as a treatment for tumor lysis syndrome, hyperuricemia, and gout. However, its long-term use is limited owing to potential immunogenicity, low thermostability, and short circulation time in vivo. Recently, urate oxidase isolated from Arthrobacter globiformis (AgUox) has been reported to be thermostable and less immunogenic than the Aspergillus-derived urate oxidase. Conjugation of human serum albumin (HSA) to therapeutic proteins has become a promising strategy to prolong circulation time in vivo. To develop a thermostable and long-circulating urate oxidase, we investigated the site-specific conjugation of HSA to AgUox based on site-specific incorporation of a clickable non-natural amino acid (frTet) and an inverse electron demand Diels-Alder reaction. We selected 14 sites for frTet incorporation using the ROSETTA design, a computational stability prediction program, among which AgUox containing frTet at position 196 (Ag12) exhibited enzymatic activity and thermostability comparable to those of wild-type AgUox. Furthermore, Ag12 exhibited a high HSA conjugation yield without compromising the enzymatic activity, generating well-defined HSA-conjugated AgUox (Ag12-HSA). In mice, the serum half-life of Ag12-HSA was approximately 29 h, which was roughly 17-fold longer than that of wild-type AgUox. Altogether, this novel formulated AgUox may hold enhanced therapeutic efficacy for several diseases.
Collapse
|
130
|
Obara S, Nakane K, Fujimura C, Tomoshige S, Ishikawa M, Sato S. Functionalization of Human Serum Albumin by Tyrosine Click. Int J Mol Sci 2021; 22:ijms22168676. [PMID: 34445381 PMCID: PMC8395410 DOI: 10.3390/ijms22168676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 12/02/2022] Open
Abstract
Human serum albumin (HSA) is a promising drug delivery carrier. Although covalent modification of Cys34 is a well-established method, it is desirable to develop a novel covalent modification method that targets residues other than cysteine to introduce multiple functions into a single HSA molecule. We developed a tyrosine-selective modification of HSA. Three tyrosine selective modification methods, hemin-catalyzed, horseradish peroxidase (HRP)-catalyzed, and laccase-catalyzed reactions were performed, and the modification efficiencies and modification sites of the modified HSAs obtained by these methods were evaluated and compared. We found that the laccase-catalyzed method could efficiently modify the tyrosine residue of HSA under mild reaction conditions without inducing oxidative side reactions. An average of 2.2 molecules of functional groups could be introduced to a single molecule of HSA by the laccase method. Binding site analysis using mass spectrometry suggested Y84, Y138, and Y401 as the main modification sites. Furthermore, we evaluated binding to ibuprofen and found that, unlike the conventional lysine residue modification, the inhibition of drug binding was minimal. These results suggest that tyrosine-residue selective chemical modification is a promising method for covalent drug attachment to HSA.
Collapse
Affiliation(s)
- Satsuki Obara
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan; (S.O.); (K.N.); (S.T.); (M.I.)
| | - Keita Nakane
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan; (S.O.); (K.N.); (S.T.); (M.I.)
| | - Chizu Fujimura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan;
| | - Shusuke Tomoshige
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan; (S.O.); (K.N.); (S.T.); (M.I.)
| | - Minoru Ishikawa
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan; (S.O.); (K.N.); (S.T.); (M.I.)
| | - Shinichi Sato
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan; (S.O.); (K.N.); (S.T.); (M.I.)
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan;
- Correspondence:
| |
Collapse
|
131
|
Dömötör O, Pivarcsik T, Mészáros JP, Szatmári I, Fülöp F, Enyedy ÉA. Critical factors affecting the albumin binding of half-sandwich Ru(ii) and Rh(iii) complexes of 8-hydroxyquinolines and oligopyridines. Dalton Trans 2021; 50:11918-11930. [PMID: 34374386 DOI: 10.1039/d1dt01700d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
There is significant interest today in the interaction of half-sandwich anticancer organometallic complexes with proteins. It is considered as a crucial factor in the transport and mode of action of these compounds; thus it can affect their overall pharmacological and toxicological profiles. Albumin binding of high stability Ru(ii)(η6-p-cymene) and Rh(iii)(η5-C5Me5) complexes formed with 8-hydroxyquinoline, its 5-chloro-7-((proline-1-yl)methyl) substituted derivative, 2,2'-bipyridine and 1,10-phenanthroline is discussed herein. The interaction with human serum albumin in terms of kinetic aspects, binding strength and possible binding sites was studied in detail by means of various methods such as 1H NMR spectroscopy, UV-visible spectrophotometry, steady-state and time-resolved fluorometry, ultrafiltration and capillary zone electrophoresis. Ru(ii)(η6-p-cymene)(2,2'-bipyridine) and Ru(ii)(η6-p-cymene)(1,10-phenanthroline) complexes do not bind to the protein measurably, most probably due to kinetic reasons. However, other complexes bind significantly to albumin with fairly different kinetics to albumin. The binding affinity towards hydrophobic binding pockets shows correlation with lipophilicity along with the actual charge of the respective complexes. The studied complexes preserve their original structure upon interaction with albumin. Formation constants computed for the binding of these metal complexes to histidine-containing model oligopeptides demonstrated significant ternary complex formation, pointing out the importance of histidine coordination in the binding of these types of complexes.
Collapse
Affiliation(s)
- Orsolya Dömötör
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary.
| | | | | | | | | | | |
Collapse
|
132
|
Schoch S, Hadiji M, Pereira SAP, Saraiva MLMFS, Braccini S, Chiellini F, Biver T, Zacchini S, Pampaloni G, Dyson PJ, Marchetti F. A Strategy to Conjugate Bioactive Fragments to Cytotoxic Diiron Bis(cyclopentadienyl) Complexes. Organometallics 2021; 40:2516-2528. [PMID: 34475610 PMCID: PMC8397425 DOI: 10.1021/acs.organomet.1c00270] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Indexed: 12/19/2022]
Abstract
![]()
A series of bioactive
molecules were synthesized from the condensation
of aspirin or chlorambucil with terminal alkynes bearing alcohol or
amine substituents. Insertion of the resulting alkynes into the iron–carbyne
bond of readily accessible diiron bis(cyclopentadienyl) μ-aminocarbyne
complexes, [1a,b]CF3SO3, afforded novel diiron complexes with a bridging vinyliminium ligand,
[2–10]CF3SO3, functionalized with a bioactive moiety. All compounds were characterized
by elemental analysis and IR and multinuclear NMR spectroscopy and
in three cases by single-crystal X-ray diffraction. Moreover, the
D2O solubility, stability in D2O and cell culture
media, and octanol–water partition coefficients of diiron complexes
were determined spectroscopically. The cytotoxicity of the complexes
was assessed in the tumorigenic A2780 and A2780cisR and the nontumorigenic
HEK 293T cell lines. Some complexes exhibit high potency and the ability
to overcome resistance in A2780cisR cells (aspirin complexes) or high
selectivity relative to HEK 293T cells (chlorambucil complexes). Further
studies indicate that the complexes significantly trigger intracellular
ROS production, irrespective of the nature of the bioactive fragment.
DNA alkylation and protein binding studies were also undertaken.
Collapse
Affiliation(s)
- Silvia Schoch
- University of Pisa, Dipartimento di Chimica e Chimica Industriale, 56124 Pisa, Italy
| | - Mouna Hadiji
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sarah A P Pereira
- LAQV, REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia, da Universidade do Porto, Porto, Portugal
| | - M Lúcia M F S Saraiva
- LAQV, REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia, da Universidade do Porto, Porto, Portugal
| | - Simona Braccini
- University of Pisa, Dipartimento di Chimica e Chimica Industriale, 56124 Pisa, Italy
| | - Federica Chiellini
- University of Pisa, Dipartimento di Chimica e Chimica Industriale, 56124 Pisa, Italy
| | - Tarita Biver
- University of Pisa, Dipartimento di Farmacia, 56126 Pisa, Italy.,University of Pisa, Dipartimento di Chimica e Chimica Industriale, 56124 Pisa, Italy
| | - Stefano Zacchini
- University of Bologna, Dipartimento di Chimica Industriale "Toso Montanari", 40136 Bologna, Italy
| | - Guido Pampaloni
- University of Pisa, Dipartimento di Chimica e Chimica Industriale, 56124 Pisa, Italy
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fabio Marchetti
- University of Pisa, Dipartimento di Chimica e Chimica Industriale, 56124 Pisa, Italy
| |
Collapse
|
133
|
Ailuno G, Iacobazzi RM, Lopalco A, Baldassari S, Arduino I, Azzariti A, Pastorino S, Caviglioli G, Denora N. The Pharmaceutical Technology Approach on Imaging Innovations from Italian Research. Pharmaceutics 2021; 13:1214. [PMID: 34452175 PMCID: PMC8402236 DOI: 10.3390/pharmaceutics13081214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022] Open
Abstract
Many modern therapeutic approaches are based on precise diagnostic evidence, where imaging procedures play an essential role. To date, in the diagnostic field, a plethora of agents have been investigated to increase the selectivity and sensitivity of diagnosis. However, the most common drawbacks of conventional imaging agents reside in their non-specificity, short imaging time, instability, and toxicity. Moreover, routinely used diagnostic agents have low molecular weights and consequently a rapid clearance and renal excretion, and this represents a limitation if long-lasting imaging analyses are to be conducted. Thus, the development of new agents for in vivo diagnostics requires not only a deep knowledge of the physical principles of the imaging techniques and of the physiopathological aspects of the disease but also of the relative pharmaceutical and biopharmaceutical requirements. In this scenario, skills in pharmaceutical technology have become highly indispensable in order to respond to these needs. This review specifically aims to collect examples of newly developed diagnostic agents connoting the importance of an appropriate formulation study for the realization of effective products. Within the context of pharmaceutical technology research in Italy, several groups have developed and patented promising agents for fluorescence and radioactive imaging, the most relevant of which are described hereafter.
Collapse
Affiliation(s)
- Giorgia Ailuno
- Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy; (G.A.); (S.B.)
| | - Rosa Maria Iacobazzi
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori “Giovanni Paolo II”, O. Flacco St., 70124 Bari, Italy; (R.M.I.); (A.A.)
| | - Antonio Lopalco
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, Orabona St. 4, 70125 Bari, Italy; (A.L.); (I.A.)
| | - Sara Baldassari
- Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy; (G.A.); (S.B.)
| | - Ilaria Arduino
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, Orabona St. 4, 70125 Bari, Italy; (A.L.); (I.A.)
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori “Giovanni Paolo II”, O. Flacco St., 70124 Bari, Italy; (R.M.I.); (A.A.)
| | - Sara Pastorino
- Nuclear Medicine Unit, S. Andrea Hospital, via Vittorio Veneto 197, 19124 La Spezia, Italy;
| | - Gabriele Caviglioli
- Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy; (G.A.); (S.B.)
| | - Nunzio Denora
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, Orabona St. 4, 70125 Bari, Italy; (A.L.); (I.A.)
| |
Collapse
|
134
|
Kudłacik-Kramarczyk S, Drabczyk A, Głąb M, Gajda P, Czopek A, Zagórska A, Jaromin A, Gubernator J, Makara A, Tyliszczak B. The Development of the Innovative Synthesis Methodology of Albumin Nanoparticles Supported by Their Physicochemical, Cytotoxic and Hemolytic Evaluation. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4386. [PMID: 34442909 PMCID: PMC8400698 DOI: 10.3390/ma14164386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 01/28/2023]
Abstract
Many studies are being performed to develop effective carriers for controlled cytostatic delivery wherein albumin is a promising material due to its tendency to accumulate near cancer cells. The novelty of this work involves the development of the synthesis methodology of albumin nanoparticles and their biological and physicochemical evaluation. Albumin particles were obtained via the salt-induced precipitation and K3PO4 was used as a salting-out agent. Various concentrations of protein and salting-out agent solutions were mixed using a burette or a syringe system. It was proved that the size of the particles depended on the concentrations of the reagents and the methodology applied. As a result of a process performed using a burette and 2 M K3PO4, albumin spheres having a size 5-25 nm were obtained. The size of nanospheres and their spherical shape was confirmed via TEM analysis. The use of a syringe system led to preparation of particles of large polydispersity. The highest albumin concentration allowing for synthesis of homogeneous particles was 2 g/L. The presence of albumin in spheres was confirmed via the FT-IR technique and UV-Vis spectroscopy. All samples showed no cytotoxicity towards normal human dermal fibroblasts and no hemolytic properties against human erythrocytes (the hemolysis did not exceed 2.5%).
Collapse
Affiliation(s)
- Sonia Kudłacik-Kramarczyk
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Anna Drabczyk
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Magdalena Głąb
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Paweł Gajda
- Department of Sustainable Energy Development, Faculty of Energy and Fuels, AGH University of Science and Technology, 30 Mickiewicza Av., 30-059 Krakow, Poland;
| | - Anna Czopek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (A.C.); (A.Z.)
| | - Agnieszka Zagórska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (A.C.); (A.Z.)
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 14a Joliot-Curie St., 50-383 Wroclaw, Poland; (A.J.); (J.G.)
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 14a Joliot-Curie St., 50-383 Wroclaw, Poland; (A.J.); (J.G.)
| | - Agnieszka Makara
- Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska St., 31-155 Krakow, Poland;
| | - Bożena Tyliszczak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| |
Collapse
|
135
|
Shi Y, Lu A, Wang X, Belhadj Z, Wang J, Zhang Q. A review of existing strategies for designing long-acting parenteral formulations: Focus on underlying mechanisms, and future perspectives. Acta Pharm Sin B 2021; 11:2396-2415. [PMID: 34522592 PMCID: PMC8424287 DOI: 10.1016/j.apsb.2021.05.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/03/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
The need for long-term treatments of chronic diseases has motivated the widespread development of long-acting parenteral formulations (LAPFs) with the aim of improving drug pharmacokinetics and therapeutic efficacy. LAPFs have been proven to extend the half-life of therapeutics, as well as to improve patient adherence; consequently, this enhances the outcome of therapy positively. Over past decades, considerable progress has been made in designing effective LAPFs in both preclinical and clinical settings. Here we review the latest advances of LAPFs in preclinical and clinical stages, focusing on the strategies and underlying mechanisms for achieving long acting. Existing strategies are classified into manipulation of in vivo clearance and manipulation of drug release from delivery systems, respectively. And the current challenges and prospects of each strategy are discussed. In addition, we also briefly discuss the design principles of LAPFs and provide future perspectives of the rational design of more effective LAPFs for their further clinical translation.
Collapse
Key Words
- 2′-F, 2′-fluoro
- 2′-O-MOE, 2′-O-(2-methoxyethyl)
- 2′-OMe, 2′-O-methyl
- 3D, three-dimensional
- ART, antiretroviral therapy
- ASO, antisense oligonucleotide
- Biomimetic strategies
- Chemical modification
- DDS, drug delivery systems
- ECM, extracellular matrix
- ENA, ethylene-bridged nucleic acid
- ESC, enhanced stabilization chemistry
- EVA, ethylene vinyl acetate
- Fc/HSA fusion
- FcRn, Fc receptor
- GLP-1, glucagon like peptide-1
- GS, glycine–serine
- HA, hyaluronic acid
- HES, hydroxy-ethyl-starch
- HP, hypoparathyroidism
- HSA, human serum albumin
- Hydrogels
- ISFI, in situ forming implants
- IgG, immunoglobulin G
- Implantable systems
- LAFs, long-acting formulations
- LAPFs, long-acting parenteral formulations
- LNA, locked nucleic acid
- Long-acting
- MNs, microneedles
- Microneedles
- NDS, nanochannel delivery system
- NPs, nanoparticles
- Nanocrystal suspensions
- OA, osteoarthritis
- PCPP-SA, poly(1,3-bis(carboxyphenoxy)propane-co-sebacic-acid)
- PEG, polyethylene glycol
- PM, platelet membrane
- PMPC, poly(2-methyacryloyloxyethyl phosphorylcholine)
- PNAs, peptide nucleic acids
- PS, phase separation
- PSA, polysialic acid
- PTH, parathyroid hormone
- PVA, polyvinyl alcohol
- RBCs, red blood cells
- RES, reticuloendothelial system
- RNAi, RNA interference
- SAR, structure‒activity relationship
- SCID, severe combined immunodeficiency
- SE, solvent extraction
- STC, standard template chemistry
- TNFR2, tumor necrosis factor receptor 2
- hGH, human growth hormone
- im, intramuscular
- iv, intravenous
- mPEG, methoxypolyethylene glycol
- sc, subcutaneous
Collapse
Affiliation(s)
- Yujie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiangyu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
136
|
Azevedo C, Pinto S, Benjakul S, Nilsen J, Santos HA, Traverso G, Andersen JT, Sarmento B. Prevention of diabetes-associated fibrosis: Strategies in FcRn-targeted nanosystems for oral drug delivery. Adv Drug Deliv Rev 2021; 175:113778. [PMID: 33887405 DOI: 10.1016/j.addr.2021.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/29/2021] [Accepted: 04/16/2021] [Indexed: 01/02/2023]
Abstract
Diabetes mellitus is a chronic disease with an elevated risk of micro- and macrovascular complications, such as fibrosis. To prevent diabetes-associated fibrosis, the symptomatology of diabetes must be controlled, which is commonly done by subcutaneous injection of antidiabetic peptides. To minimize the pain and distress associated with such injections, there is an urgent need for non-invasive oral transmucosal drug delivery strategies. However, orally administered peptide-based drugs are exposed to harsh conditions in the gastrointestinal tract and poorly cross the selective intestinal epithelium. Thus, targeting of drugs to receptors expressed in epithelial cells, such as the neonatal Fc receptor (FcRn), may therefore enhance uptake and transport through mucosal barriers. This review compiles how in-depth studies of FcRn biology and engineering of receptor-binding molecules may pave the way for design of new classes of FcRn-targeted nanosystems. Tailored strategies may open new avenues for oral drug delivery and provide better treatment options for diabetes and, consequently, fibrosis prevention.
Collapse
|
137
|
Kwak G, Kim H, Park J, Kim EH, Jang H, Han G, Wang SY, Yang Y, Chan Kwon I, Kim SH. A Trojan-Horse Strategy by In Situ Piggybacking onto Endogenous Albumin for Tumor-Specific Neutralization of Oncogenic MicroRNA. ACS NANO 2021; 15:11369-11384. [PMID: 34191497 DOI: 10.1021/acsnano.1c00799] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
MicroRNAs (miRNAs), a recently discovered class of noncoding RNAs, play pivotal roles in regulating fundamental biological processes by suppressing the expression of target genes. Aberrant miRNA expression is commonly correlated with human diseases, including cancers. Anti-miRNA oligonucleotides provide an innovative therapeutic strategy for silencing disease-associated miRNAs. However, the clinical application of anti-miRNA therapy has been limited by formulation challenges and physiological delivery barriers. Here, to provide the safe and effective tumor-targeted delivery of anti-miRNAs, we designed carrier-free maleimide-functionalized anti-miRNAs (MI-Anti-miRNAs) that enable "piggybacking" onto albumin in vivo. These functionalized MI-Anti-miRNAs covalently bind to cysteine-34 of endogenous albumin within minutes. In addition to resulting in a markedly extended blood circulation lifetime, this strategy allows MI-Anti-miRNAs to "hitchhike" to the tumor site. Importantly, in situ-generated albumin-Anti-miRNAs are capable of intracellularly internalizing highly negatively charged anti-miRNA molecules and knocking down target miRNAs. In particular, MI-Anti-miRNAs that targeted miRNA-21, which is involved in tumor initiation, progression, invasion, and metastasis in several types of cancer, successfully repressed miRNA-21 activity, resulting in a superior antitumor activity in both solid and metastatic tumor models without causing systemic toxicity. This endogenous albumin-piggybacking approach using MI-Anti-miRNAs provides a simple and broadly applicable platform strategy for the systemic delivery of anti-miRNA therapeutics.
Collapse
Affiliation(s)
- Gijung Kwak
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hyosuk Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jooho Park
- Department of Biomedical & Health Science, Konkuk University, 268 Chungwon-daero, Chungju 27478, Republic of Korea
| | - Eun Hye Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Life Sciences, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hochung Jang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Geonhee Han
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Sun Young Wang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Yoosoo Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- KIST-DFCI On-Site-Lab, Department of Cancer Biology, Dana Farber Cancer Institute, 450 Brookline Ave, Boston, Massachusetts 02215, United States
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
138
|
Braccini S, Rizzi G, Biancalana L, Pratesi A, Zacchini S, Pampaloni G, Chiellini F, Marchetti F. Anticancer Diiron Vinyliminium Complexes: A Structure-Activity Relationship Study. Pharmaceutics 2021; 13:1158. [PMID: 34452119 PMCID: PMC8398472 DOI: 10.3390/pharmaceutics13081158] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/31/2022] Open
Abstract
A series of 16 novel diiron complexes of general formula [Fe2Cp2(CO)(μ-CO){μ-η1:η3-C(R')C(R″)CN(R)(Y)}]CF3SO3 (2-7), bearing different substituents on the bridging vinyliminium ligand, was synthesized in 69-95% yields from the reactions of diiron μ-aminocarbyne precursors with various alkynes. The products were characterized by elemental analysis, IR, 1H and 13C NMR spectroscopy; moreover the X-ray structures of 2c (R = Y = CH2Ph, R' = R″ = Me) and 3a (R = CH2CH=CH2, Y = R' = Me, R″ = H) were ascertained by single-crystal X-ray diffraction studies. NMR and UV-Vis methods were used to assess the D2O solubility, the stability in aqueous solution at 37 °C and the octanol-water partition coefficients of the complexes. A screening study evidenced a potent cytotoxicity of 2-7 against the A2780 cancer cell line, with a remarkable selectivity compared to the nontumoral Balb/3T3 cell line; complex 4c (R = Cy, Y = R' = R″ = Me) revealed as the most performant of the series. The antiproliferative activity of a selection of complexes was also assessed on the cisplatin-resistant A2780cisR cancer cell line, and these complexes were capable of inducing a significant ROS production. Moreover, ESI-MS experiments indicated the absence of interaction of selected complexes with cytochrome c and the potentiality to inhibit the thioredoxin reductase enzyme (TrxR).
Collapse
Affiliation(s)
- Simona Braccini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Giorgia Rizzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Lorenzo Biancalana
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Stefano Zacchini
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Viale Risorgimento 4, I-40136 Bologna, Italy;
| | - Guido Pampaloni
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Federica Chiellini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| |
Collapse
|
139
|
Wu CR, Huang YD, Hong YH, Liu YH, Narwane M, Chang YH, Dinh TK, Hsieh HT, Hseuh YJ, Wu PC, Pao CW, Chan TS, Hsu IJ, Chen Y, Chen HC, Chin TY, Lu TT. Endogenous Conjugation of Biomimetic Dinitrosyl Iron Complex with Protein Vehicles for Oral Delivery of Nitric Oxide to Brain and Activation of Hippocampal Neurogenesis. JACS AU 2021; 1:998-1013. [PMID: 34467346 PMCID: PMC8395708 DOI: 10.1021/jacsau.1c00160] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 06/13/2023]
Abstract
Nitric oxide (NO), a pro-neurogenic and antineuroinflammatory gasotransmitter, features the potential to develop a translational medicine against neuropathological conditions. Despite the extensive efforts made on the controlled delivery of therapeutic NO, however, an orally active NO prodrug for a treatment of chronic neuropathy was not reported yet. Inspired by the natural dinitrosyl iron unit (DNIU) [Fe(NO)2], in this study, a reversible and dynamic interaction between the biomimetic [(NO)2Fe(μ-SCH2CH2OH)2Fe(NO)2] (DNIC-1) and serum albumin (or gastrointestinal mucin) was explored to discover endogenous proteins as a vehicle for an oral delivery of NO to the brain after an oral administration of DNIC-1. On the basis of the in vitro and in vivo study, a rapid binding of DNIC-1 toward gastrointestinal mucin yielding the mucin-bound dinitrosyl iron complex (DNIC) discovers the mucoadhesive nature of DNIC-1. A reversible interconversion between mucin-bound DNIC and DNIC-1 facilitates the mucus-penetrating migration of DNIC-1 shielded in the gastrointestinal tract of the stomach and small intestine. Moreover, the NO-release reactivity of DNIC-1 induces the transient opening of the cellular tight junction and enhances its paracellular permeability across the intestinal epithelial barrier. During circulation in the bloodstream, a stoichiometric binding of DNIC-1 to the serum albumin, as another endogenous protein vehicle, stabilizes the DNIU [Fe(NO)2] for a subsequent transfer into the brain. With aging mice under a Western diet as a disease model for metabolic syndrome and cognitive impairment, an oral administration of DNIC-1 in a daily manner for 16 weeks activates the hippocampal neurogenesis and ameliorates the impaired cognitive ability. Taken together, these findings disclose the synergy between biomimetic DNIC-1 and endogenous protein vehicles for an oral delivery of therapeutic NO to the brain against chronic neuropathy.
Collapse
Affiliation(s)
- Cheng-Ru Wu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Yi-Da Huang
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yong-Huei Hong
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Ya-Hsin Liu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Manmath Narwane
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Yu-Hsiang Chang
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Trinh Kieu Dinh
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Hsin-Tzu Hsieh
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Yi-Jen Hseuh
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ping-Ching Wu
- Department
of Biomedical Engineering, National Cheng
Kung University, Tainan, Taiwan
| | - Chih-Wen Pao
- National
Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - Ting-Shan Chan
- National
Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - I-Jui Hsu
- Department
of Molecular Science and Engineering, Research and Development Center
of Smart Textile Technology, National Taipei
University of Technology, Taipei, Taiwan
| | - Yunching Chen
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| | - Hung-Chi Chen
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department
of Medicine, College of Medicine, Chang
Gung University, Taoyuan, Taiwan
| | - Ting-Yu Chin
- Department
of Bioscience Technology, Chung Yuan Christian
University, Taoyuan, Taiwan
| | - Tsai-Te Lu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu, Taiwan
| |
Collapse
|
140
|
Li C, Xiang W, Wu M, Zhang H, Cheng J, Yang T, Mai J, Chi X, Gao X, Ding Y, Niu J. A randomized dose-escalation study on the safety, tolerability, immunogenicity, pharmacokinetics and pharmacodynamics of a novel recombinant human albumin in healthy subjects. Eur J Pharm Sci 2021; 165:105923. [PMID: 34229078 DOI: 10.1016/j.ejps.2021.105923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/19/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Recombinant human albumin (rHA) is an alternative to human serum albumin (HSA) for treating ascites in cirrhosis patients. This study was to evaluate the safety, tolerability, immunogenicity, and pharmacokinetics/pharmacodynamics (PK/PD) of rHA in healthy subjects to guide the design for further clinical trials. METHODS Healthy subjects aged 18-55 years were enrolled in this double-blinded, first-in-human, placebo-controlled single ascending dose (SAD) (1.25, 5, 10, 20, or 30g) and positive-controlled multiple-dose study (3-day treatment of 10g/day for three cycles every three weeks). The safety was assessed by adverse events (AEs). Antibodies (IgE and IgD) and cytokines were analyzed for immunogenicity. Serum albumin levels and changes in plasma colloid osmotic pressure (PCOP) and hematocrit (HCT) were measured for PK/PD analysis. RESULTS rHA was well tolerated as all AEs were assessed as mild or moderate. No severe allergy or difference in the incidence of AEs was observed among the different cohorts in the SAD study or in the different cycles in the multiple-dose study. The incidence of AEs was similar for the rHA and HSA cohort. Antibodies or cytokines showed no changes after drug administration. As expected, serum albumin levels and PCOP increases, and HCT ratio decreases were dose-related with significant differences (p < 0.01). No differences were observed between rHA and HSA. CONCLUSION rHA is safe and well-tolerated in healthy Chinese subjects. rHA and HSA exhibited similar safety, tolerability, and PK/PD profiles. The results support further evaluation of rHA treatment in cirrhotic patients with ascites. The clinical trial registration numbers are CTR20191221 (http://www.chinadrugtrials.org.cn/clinicaltrials.searchlistdetail.dhtml).
Collapse
Affiliation(s)
- Cuiyun Li
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China.
| | - Wei Xiang
- Tonghua Anrate Biopharmaceuticals Co., Ltd., Tonghua, Jilin, 134100, China.
| | - Min Wu
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China.
| | - Hong Zhang
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China.
| | - Jianqiu Cheng
- Tonghua Anrate Biopharmaceuticals Co., Ltd., Tonghua, Jilin, 134100, China.
| | - Tao Yang
- Tonghua Anrate Biopharmaceuticals Co., Ltd., Tonghua, Jilin, 134100, China.
| | - Jiajia Mai
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China.
| | - Xiumei Chi
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China.
| | - Xiuzhu Gao
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China.
| | - Yanhua Ding
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China.
| | - Junqi Niu
- Department of Hepatology, First Hospital, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
141
|
Ruseska I, Fresacher K, Petschacher C, Zimmer A. Use of Protamine in Nanopharmaceuticals-A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1508. [PMID: 34200384 PMCID: PMC8230241 DOI: 10.3390/nano11061508] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/18/2022]
Abstract
Macromolecular biomolecules are currently dethroning classical small molecule therapeutics because of their improved targeting and delivery properties. Protamine-a small polycationic peptide-represents a promising candidate. In nature, it binds and protects DNA against degradation during spermatogenesis due to electrostatic interactions between the negatively charged DNA-phosphate backbone and the positively charged protamine. Researchers are mimicking this technique to develop innovative nanopharmaceutical drug delivery systems, incorporating protamine as a carrier for biologically active components such as DNA or RNA. The first part of this review highlights ongoing investigations in the field of protamine-associated nanotechnology, discussing the self-assembling manufacturing process and nanoparticle engineering. Immune-modulating properties of protamine are those that lead to the second key part, which is protamine in novel vaccine technologies. Protamine-based RNA delivery systems in vaccines (some belong to the new class of mRNA-vaccines) against infectious disease and their use in cancer treatment are reviewed, and we provide an update on the current state of latest developments with protamine as pharmaceutical excipient for vaccines.
Collapse
Affiliation(s)
| | | | | | - Andreas Zimmer
- Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Universitätsplatz 1, 8010 Graz, Austria; (I.R.); (K.F.); (C.P.)
| |
Collapse
|
142
|
Pivarcsik T, Tóth G, Szemerédi N, Bogdanov A, Spengler G, Kljun J, Kladnik J, Turel I, Enyedy ÉA. Comparison of Solution Chemical Properties and Biological Activity of Ruthenium Complexes of Selected β-Diketone, 8-Hydroxyquinoline and Pyrithione Ligands. Pharmaceuticals (Basel) 2021; 14:518. [PMID: 34072270 PMCID: PMC8226722 DOI: 10.3390/ph14060518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
In this work, the various biological activities of eight organoruthenium(II) complexes were evaluated to reveal correlations with their stability and reactivity in aqueous media. Complexes with general formula [Ru(η6-p-cymene)(X,Y)(Z)] were prepared, where (X,Y) represents either an O,O-ligand (β-diketone), N,O-ligand (8-hydroxyquinoline) or O,S-pyrithione-type ligands (pyrithione = 1-hydroxypyridine-2(1H)-thione) with Cl- or 1,3,5-triaza-7-phosphaadamantane (PTA) as a co-ligand (Z). The tested complexes inhibit the chlamydial growth on HeLa cells, and one of the complexes inhibits the growth of the human herpes simplex virus-2. The chlorido complexes with N,O- and O,S-ligands displayed strong antibacterial activity on Gram-positive strains including the resistant S. aureus (MRSA) and were cytotoxic in adenocarcinoma cell lines. Effect of the structural variation on the biological properties and solution stability was clearly revealed. The decreased bioactivity of the β-diketone complexes can be related to their lower stability in solution. In contrast, the O,S-pyrithione-type complexes are highly stable in solution and the complexation prevents the oxidation of the O,S-ligands. Comparing the binding of PTA and the chlorido co-ligands, it can be concluded that PTA is generally more strongly coordinated to ruthenium, which at the same time decreased the reactivity of complexes with human serum albumin or 1-methylimidazole as well as diminished their bioactivity.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
| | - Gábor Tóth
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
| | - Nikoletta Szemerédi
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.S.); (A.B.)
| | - Anita Bogdanov
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.S.); (A.B.)
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.S.); (A.B.)
| | - Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - Jerneja Kladnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - Éva A. Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
| |
Collapse
|
143
|
Yang B, Kwon I. Multivalent Albumin-Neonatal Fc Receptor Interactions Mediate a Prominent Extension of the Serum Half-Life of a Therapeutic Protein. Mol Pharm 2021; 18:2397-2405. [PMID: 33983743 DOI: 10.1021/acs.molpharmaceut.1c00231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human serum albumin (HSA) has been used to extend the serum half-life of therapeutic proteins owing to its exceptionally long serum half-life via the neonatal Fc receptor (FcRn)-mediated recycling mechanism. In most cases, only one HSA molecule was conjugated to a therapeutic protein, leading to a limited extension of the serum half-life. In this study, we hypothesized that conjugation of multiple HSA molecules to a therapeutic protein significantly further extends the serum half-life via multivalent HSA-FcRn interactions. We chose urate oxidase (Uox), a tetrameric therapeutic protein used for the treatment of gout, as a model. In previous studies, only one HSA molecule was site-specifically conjugated to one Uox because of poor conjugation yield of the relatively slow bio-orthogonal chemistry, strain-promoted azide-alkyne cycloaddition (SPAAC). To increase the number of HSA molecules conjugated to one Uox, we employed the faster bio-orthogonal chemistry, inverse electron demand Diels-Alder reaction (IEDDA). We site-specifically introduced the phenylalanine analog with a fast-reacting tetrazine group (frTet) into position 174 of each subunit of Uox. We then achieved site-specific HSA conjugation to each subunit of Uox via IEDDA, generating Uox conjugated to four HSA molecules (Uox-HSA4), with a small portion of Uox conjugated to three HSA molecules (Uox-HSA3). We characterized Uox-HSA4 as well as Uox variants conjugated to one or two HSA molecules prepared via SPAAC (Uox-HSA1 or Uox-HSA2). The enzyme activity of all three Uox-HSA conjugates was comparable to that of unmodified Uox. We found out that an increase in HSA molecules conjugated to Uox (multiple albumin-conjugated therapeutic protein) enhanced FcRn binding and consequently prolonged the serum half-life in vivo. In particular, the conjugation of four HSA molecules to Uox led to a prominent extension of the serum half-life (over 21 h), which is about 16-fold longer than that of Uox-WT.
Collapse
Affiliation(s)
- Byungseop Yang
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Inchan Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
144
|
Hama M, Ishima Y, Chuang VTG, Ando H, Shimizu T, Ishida T. Evidence for Delivery of Abraxane via a Denatured-Albumin Transport System. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19736-19744. [PMID: 33881292 DOI: 10.1021/acsami.1c03065] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Abraxane, an albumin-bound paclitaxel nanoparticle formulation, is superior to conventional paclitaxel preparations because it has better efficacy against unresectable pancreatic cancer. Previous reports suggest that this better efficacy of Abraxane than conventional paclitaxel preparation is probably due to its transport through Gp60, an albumin receptor on the surface of vascular endothelial cells. The increased tumor accumulation of Abraxane is also caused by the secreted protein acid and rich in cysteine in the tumor stroma. However, the uptake mechanism of Abraxane remains poorly understood. In this study, we demonstrated that the delivery of Abraxane occurred via different receptor pathways from that of endogenous albumin. Our results showed that the uptake of endogenous albumin was inhibited by a Gp60 pathway inhibitor in the process of endocytosis through endothelial cells or tumor cells. In contrast, the uptake of Abraxane-derived HSA was less affected by the Gp60 pathway inhibitor but significantly reduced by denatured albumin receptor inhibitors. In conclusion, these data indicate that Abraxane-derived HSA was taken up into endothelial cells or tumor cells by a mechanism different from normal endogenous albumin. These new data on distinct cellular transport pathways of denatured albumin via gp family proteins different from those of innate albumin shed light on the mechanisms of tumor delivery and antitumor activity of Abraxane and provide new scientific rationale for the development of a novel albumin drug delivery strategy via a denatured albumin receptor.
Collapse
Affiliation(s)
- Maichi Hama
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Victor Tuan Giam Chuang
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| |
Collapse
|
145
|
Binding Models of Copper(II) Thiosemicarbazone Complexes with Human Serum Albumin: A Speciation Study. Molecules 2021; 26:molecules26092711. [PMID: 34063080 PMCID: PMC8125041 DOI: 10.3390/molecules26092711] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 11/24/2022] Open
Abstract
Copper(II) complexes of thiosemicarbazones (TSCs) often exhibit anticancer properties, and their pharmacokinetic behavior can be affected by their interaction with blood transport proteins. Interaction of copper(II) complexes of an {N,N,S} donor α-N-pyridyl TSC (Triapine) and an {O,N,S} donor 2-hydroxybenzaldehyde TSC (STSC) with human serum albumin (HSA) was investigated by UV–visible and electron paramagnetic resonance spectroscopy at physiological pH. Asp-Ala-His-Lys and the monodentate N-methylimidazole were also applied as binding models. Conditional formation constants were determined for the ternary copper(II)-TSC complexes formed with HSA, DAHK, and N-methylimidazole based on the spectral changes of both charge transfer and d-d bands. The neutral N-methylimidazole displays a similar binding affinity to both TSC complexes. The partially negatively charged tetrapeptide binds stronger to the positively charged Triapine complex in comparison to the neutral STSC complex, while the opposite trend was observed for HSA, which demonstrates the limitations of the use of simple ligands to model the protein binding. The studied TSC complexes are able to bind to HSA in a fast process, and the conditional constants suggest that their binding strength is only weak-to-moderate.
Collapse
|
146
|
Sen S, Perrin MW, Sedgwick AC, Lynch VM, Sessler JL, Arambula JF. Covalent and non-covalent albumin binding of Au(i) bis-NHCs via post-synthetic amide modification. Chem Sci 2021; 12:7547-7553. [PMID: 34163845 PMCID: PMC8171490 DOI: 10.1039/d1sc01055g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/24/2021] [Indexed: 12/21/2022] Open
Abstract
Recent decades have witnessed the emergence of Au(i) bis-N-heterocyclic carbenes (NHCs) as potential anticancer agents. However, these systems exhibit little interaction with serum proteins (e.g., human serum albumin), which presumably impacts their pharmacokinetic profile and tumor exposure. Anticancer drugs bound to human serum albumin (HSA) often benefit from significant advantages, including longer circulatory half-lives, tumor targeted delivery, and easier administration relative to the drug alone. In this work, we present Au(i) bis-NHCs complexes, 7 and 9, capable of binding to HSA. Complex 7 contains a reactive maleimide moiety for covalent protein conjugation, whereas its congener 9 contains a naphthalimide fluorophore for non-covalent binding. A similar drug motif was used in both cases. Complexes 7 and 9 were prepared from a carboxylic acid functionalized Au(i) bis-NHC (complex 2) using a newly developed post-synthetic amide functionalization protocol that allows coupling to both aliphatic and aromatic amines. Analytical, and in vitro techniques were used to confirm protein binding, as well as cellular uptake and antiproliferative activity in A549 human lung cancer cells. The present findings highlight a hitherto unexplored approach to modifying Au(i) bis-NHC drug candidates for protein ligation and serve to showcase the relative benefits of covalent and non-covalent HSA binding.
Collapse
Affiliation(s)
- Sajal Sen
- Department of Chemistry, The University of Texas at Austin 105 E 24th Street A5300 Austin TX 78712-1224 USA
| | - Mark W Perrin
- Department of Chemistry, The University of Texas at Austin 105 E 24th Street A5300 Austin TX 78712-1224 USA
| | - Adam C Sedgwick
- Department of Chemistry, The University of Texas at Austin 105 E 24th Street A5300 Austin TX 78712-1224 USA
| | - Vincent M Lynch
- Department of Chemistry, The University of Texas at Austin 105 E 24th Street A5300 Austin TX 78712-1224 USA
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin 105 E 24th Street A5300 Austin TX 78712-1224 USA
| | - Jonathan F Arambula
- Department of Chemistry, The University of Texas at Austin 105 E 24th Street A5300 Austin TX 78712-1224 USA
| |
Collapse
|
147
|
Takagi K, Somiya M, Jung J, Iijima M, Kuroda S. Polymerized Albumin Receptor of Hepatitis B Virus for Evading the Reticuloendothelial System. Pharmaceuticals (Basel) 2021; 14:ph14050408. [PMID: 33923102 PMCID: PMC8145202 DOI: 10.3390/ph14050408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 01/05/2023] Open
Abstract
Various strategies, such as optimization of surface chemistry, size, shape, and charge, have been undertaken to develop nanoparticles (NPs) as DDS (drug delivery system) nanocarriers for evading the reticuloendothelial system (RES) in vivo. We previously developed a hollow NP composed of hepatitis B virus (HBV) surface antigen L proteins and lipid bilayers, hereinafter referred to as bio-nanocapsule (BNC), as a nonviral DDS nanocarrier. Such a BNC harbors the HBV-derived human hepatic cell-specific infection mechanism, and intravenously injected BNCs by themselves were shown to avoid clearance by RES-rich organs and accumulate in target tissues. In this study, since the surface modification with albumins is known to prolong the circulation time of nanomedicines, we examined whether the polymerized albumin receptor (PAR) of BNCs contributes to RES evasion in mouse liver. Our results show that NPs conjugated with peptides possessing sufficient PAR activity were captured by Kupffer cells less efficiently in vitro and were able to circulate for a longer period of time in vivo. Comparing with polyethylene glycol, PAR peptides were shown to reduce the recognition by RES to equal content. Taken together, our results strongly suggest that the PAR domain of BNCs, as well as HBV, harbors an innate RES evasion mechanism. Therefore, the surface modification with PAR peptides could be an alternative strategy for improving the pharmacodynamics and pharmacokinetics of forthcoming nanomedicines.
Collapse
Affiliation(s)
- Kurumi Takagi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (K.T.); (M.I.)
| | - Masaharu Somiya
- Department of Biomolecular Science and Reaction, The Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan;
| | - Joohee Jung
- College of Pharmacy, Duksung Women’s University, Seoul 132-714, Korea;
| | - Masumi Iijima
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (K.T.); (M.I.)
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo 156-8502, Japan
| | - Shun’ichi Kuroda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (K.T.); (M.I.)
- Department of Biomolecular Science and Reaction, The Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan;
- Correspondence:
| |
Collapse
|
148
|
Jiang L, Jiang Y, Li L, Zheng K, Yu S, Li J, Yuan C, Huang M. A supramolecular nanocarrier for efficient cancer imaging and therapy by targeting at matriptase. J Control Release 2021; 334:153-163. [PMID: 33894302 DOI: 10.1016/j.jconrel.2021.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/30/2021] [Accepted: 04/18/2021] [Indexed: 11/26/2022]
Abstract
Human serum albumin (HSA), a versatile protein carrier for endogenous and exogenous compounds, is a proven macromolecule to form nanoparticles for drug delivery. To render HSA carrier specificity toward tumors, we designed a recombinant HSA protein fused with Kunitz domain 1 (KD1) of hepatocyte growth factor activator inhibitor type 1, which targets to matriptase, a type II transmembrane serine protease overexpressed on tumor cell surface. The carrier was thus named matriptase targeting carrier (MTC). In this study, we showed that MTC displayed the same inhibitory potency as the KD1 againast matriptase, demonstrating the HSA fusion did not affect the KD1 targeting potency. For tumor optical imaging and ablation, MTC was prepared as nanoparticle drug carrier by a novel method via denaturation and refolding to incorporate photosensitizer, CPZ. This matriptase targeting nanoparticles, CPZ:MTC@NPs, showed high specificity and cytotoxicity for matriptase-overexpressing cancer cells in vitro. In tumor-bearing mice, CPZ:MTC@NPs demonstrated selective accumulation and high retention in matriptase-overexpressing tumor. Under illumination, the nanoparticles significantly reduced tumor volumes (79.6%) as compared to saline control. These findings showed that this supramolecular nanocarrier, a new type of tumor targeting self-assembly nanoparticle, had potential as a highly efficient tumor targeting drug carrier for imaging and therapy.
Collapse
Affiliation(s)
- Libin Jiang
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Yunbin Jiang
- State Key Laboratory of Structure Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Linlin Li
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Ke Zheng
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, Shandong 266061, China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fujian 350116, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fujian 350116, China.
| |
Collapse
|
149
|
Zhang H, Zhang Y, Huang Y, Wu L, Guo Q, Wang Q, Liang L, Nishinari K, Zhao M. Interaction between bovine serum albumin and chitooligosaccharides: I. Molecular mechanism. Food Chem 2021; 358:129853. [PMID: 33933970 DOI: 10.1016/j.foodchem.2021.129853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/06/2021] [Accepted: 04/11/2021] [Indexed: 01/27/2023]
Abstract
The interaction between chitooligosaccharides (COS2-6) and bovine serum albumin (BSA) is worthy of investigation, which provides support for improving the physical properties (gelling, foaming, and emulsifying) of food proteins via COS addition and in vivo research on COS bioactivity. Component analysis indicated that COS2 and COS3 were enriched in the COS2-6-BSA precipitate. The fluorescence binding constant (1.73 × 103 M-1), ΔG of isothermal titration calorimetry (-6.7 kJ/mol), and the predicted ΔG of molecular docking (-10 to -5 kJ/mol) confirmed the weak interaction of COS2-6-BSA. Quartz crystal microbalance dissipation and molecular docking indicated that electrostatic and hydrophobic interactions were the main stabilization forces. Molecular docking showed that the predicted ΔG of COS2-6 to BSA decreased with the increasing degree of polymerization. This work clarified the weak and selective interaction between COS2-6 and BSA via various methods, which is useful for the food application of COS.
Collapse
Affiliation(s)
- Hui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China
| | - Yanzhen Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China
| | - Yongqi Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China
| | - Ling Wu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China
| | - Qianwan Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China
| | - Qi Wang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China
| | - Li Liang
- State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Katsuyoshi Nishinari
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China
| | - Meng Zhao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei International Scientific and Technological Cooperation Base of Food Hydrocolloids, Wuhan 430068, China; State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China.
| |
Collapse
|
150
|
Wang Y, Chen S, Yang X, Zhang S, Cui C. Preparation Optimization of Bovine Serum Albumin Nanoparticles and Its Application for siRNA Delivery. Drug Des Devel Ther 2021; 15:1531-1547. [PMID: 33883877 PMCID: PMC8053787 DOI: 10.2147/dddt.s299479] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND siRNA brings hope for cancer therapy. However, there are many obstacles for application of siRNA in clinical. Because of the excellent biocompatibility, non-toxicity and non-immunogenicity of bovine serum albumin (BSA), BSA-based nanoparticles have been widely designed as a drug carrier system. METHODS The optimal formula for BSA NPs preparation was investigated by central composite design response surface methodology (CCD-RSM), BSA-based survivin-siRNA delivery system (BSA NPs/siRNA) was characterized by dynamic light scattering, atomic force microscope, transmission electron microscope and Bradford method. The in vitro anti-tumor effect and mechanism of BSA NPs were investigated by confocal microscopic imaging, MTT assay, RT-qPCR and ELISA analysis. Moreover, the anti-tumor effect, distribution and biosafety of BSA NPs were studied in vivo. RESULTS The optimal formula for BSA NPs was settled to be 20 mg/mL for BSA concentration, 9 for pH value, 136% for crosslinking degree and 1.6 mL/min for speed of ethanol addition. BSA NPs/siRNA could remain stable at 4°C for 4 weeks and protect siRNA from degradation by RNase A. Besides, BSA NPs/siRNA could maintain a sustained release of siRNA and promote the uptake of siRNA significantly. The survivin-mRNA level and the survivin-protein level were decreased by 55% ± 1.6% and 54% ± 1.6% separately. The in vivo tumor inhibition results suggested that the tumor inhibition rate of BSA NPs/siRNA-treated group was 54% ± 12% and was similar with that of DOX-treated group (57% ± 9.2%, P > 0.05). The biosafety results confirmed that BSA NPs/siRNA could not induce significant damages to the main organs and blood in vivo. CONCLUSION These results demonstrated that CCD-RSM was an effective tool for preparation analysis, and the BSA NPs/siRNA was a promising system for siRNA-based gene therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cattle
- Cell Proliferation/drug effects
- Drug Carriers/chemistry
- Drug Delivery Systems
- Drug Screening Assays, Antitumor
- Humans
- MCF-7 Cells
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice, Inbred BALB C
- Mice, Nude
- Nanoparticles/chemistry
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/pharmacology
- Serum Albumin, Bovine/chemistry
- Tumor Cells, Cultured
- Mice
Collapse
Affiliation(s)
- Yifan Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, People’s Republic of China
| | - Si Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, People’s Republic of China
| | - Xin Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, People’s Republic of China
| | - Shuang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, People’s Republic of China
| | - Chunying Cui
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, People’s Republic of China
| |
Collapse
|