1
|
Qiao JX, Guo DY, Tian H, Wang ZP, Fan QQ, Tian Y, Sun J, Zhang XF, Zou JB, Cheng JX, Luan F, Zhai BT. Research progress of paclitaxel nanodrug delivery system in the treatment of triple-negative breast cancer. Mater Today Bio 2024; 29:101358. [PMID: 39677523 PMCID: PMC11638641 DOI: 10.1016/j.mtbio.2024.101358] [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: 07/16/2024] [Revised: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by the loss or low expression of estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2) and progesterone receptor (PR). Due to the lack of clear therapeutic targets, paclitaxel (PTX) is often used as a first-line standard chemotherapy drug for the treatment of high-risk and locally advanced TNBC. PTX is a diterpenoid alkaloid extracted and purified from Taxus plants, functioning as an anticancer agent by inducing and promoting tubulin polymerization, inhibiting spindle formation in cancer cells, and preventing mitosis. However, its clinical application is limited by low solubility and high toxicity. Nanodrug delivery system (NDDS) is one of the feasible methods to improve the water solubility of PTX and reduce side effects. In this review, we summarize the latest advancements in PTX-targeted NDDS, as well as its combination with other codelivery therapies for TNBC treatment. NDDS includes passive targeting, active targeting, stimuli-responsive, codelivery, and multimode strategies. These systems have good prospects in improving the bioavailability of PTX, enhancing tumor targeting, reducing toxicity, controlling drug release, and reverse tumor multidrug resistance (MDR). This review provides valuable insights into the clinical development and application of PTX-targeted NDDS in the treatment of TNBC.
Collapse
Affiliation(s)
- Jia-xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Department of Pharmacy, National Old Pharmacist Inheritance Studio, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Zhan-peng Wang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Qiang-qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yuan Tian
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Fei Luan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| |
Collapse
|
2
|
Onishchenko NR, Moskovtsev AA, Kobanenko MK, Tretiakova DS, Alekseeva AS, Kolesov DV, Mikryukova AA, Boldyrev IA, Kapkaeva MR, Shcheglovitova ON, Bovin NV, Kubatiev AA, Tikhonova OV, Vodovozova EL. Protein Corona Attenuates the Targeting of Antitumor Sialyl Lewis X-Decorated Liposomes to Vascular Endothelial Cells under Flow Conditions. Pharmaceutics 2023; 15:1754. [PMID: 37376203 DOI: 10.3390/pharmaceutics15061754] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/23/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Previously, we showed in the human umbilical vein endothelial cells (HUVECs) model that a liposome formulation of melphalan lipophilic prodrug (MlphDG) decorated with selectin ligand tetrasaccharide Sialyl Lewis X (SiaLeX) undergoes specific uptake by activated cells and in an in vivo tumor model causes a severe antivascular effect. Here, we cultured HUVECs in a microfluidic chip and then applied the liposome formulations to study their interactions with the cells in situ under hydrodynamic conditions close to capillary blood flow using confocal fluorescent microscopy. The incorporation of 5 to 10% SiaLeX conjugate in the bilayer of MlphDG liposomes increased their consumption exclusively by activated endotheliocytes. The increase of serum concentration from 20 to 100% in the flow resulted in lower liposome uptake by the cells. To elucidate the possible roles of plasma proteins in the liposome-cell interactions, liposome protein coronas were isolated and analyzed by shotgun proteomics and immunoblotting of selected proteins. Proteomic analysis showed that a gradual increase in SiaLeX content correlated with the overall enrichment of the liposome-associated proteins with several apolipoproteins, including the most positively charged one, ApoC1, and serum amyloid A4, associated with inflammation, on the one hand, and a decrease in the content of bound immunoglobulins, on the other. The article discusses the potential interference of the proteins in the binding of liposomes to selectins of endothelial cells.
Collapse
Affiliation(s)
- Natalia R Onishchenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Alexey A Moskovtsev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Maria K Kobanenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Daria S Tretiakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Anna S Alekseeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Dmitry V Kolesov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Anna A Mikryukova
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Ivan A Boldyrev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Marina R Kapkaeva
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, ul. Gamaleya 18, 123098 Moscow, Russia
| | - Olga N Shcheglovitova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, ul. Gamaleya 18, 123098 Moscow, Russia
| | - Nicolai V Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Aslan A Kubatiev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Olga V Tikhonova
- Institute of Biomedical Chemistry, ul. Pogodinskaya 10, 119121 Moscow, Russia
| | - Elena L Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
3
|
Lin Z, Chu B, Qu Y, Wei X, Huang J, Wang F, Feng Y, Wang X, Luo H, Zhai X, Xu J, Liu X, Zhang L, Chen F, Wu Y, Zheng Y. Liposome-Encapsulated Melphalan Exhibits Potent Antimyeloma Activity and Reduced Toxicity. ACS OMEGA 2023; 8:1693-1701. [PMID: 36643473 PMCID: PMC9835516 DOI: 10.1021/acsomega.2c07555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
Multiple myeloma (MM), a plasma cell cancer in bone marrow, remains an incurable disease. Melphalan, an alkylating agent, is a conventional anticancer drug that is still widely used for MM treatment in clinics. However, melphalan-induced organ toxicity and side effects are common. In this study, we loaded melphalan into a liposomal capsule and constituted liposomal melphalan (liposomal MEL). Liposomal MEL particles were approximately 120 nm in size and stable in vitro. The liposomal particles could be effectively taken up by MM cells. In vitro cytotoxicity assays using MM cell lines and primary MM cells showed that liposomal MEL exhibited similar anti-MM activity compared to an equivalent amount of free melphalan (free MEL) compound. In animal models, liposomal particles had bone marrow enrichment and prolonged half-life in vivo. Liposomal MEL exposure resulted in less liver and colon organ toxicity than exposure to an equivalent amount of free MEL-treated mice. Importantly, liposomal MEL had potent anti-MM activity in vivo in a human MM xenograft mouse model. Overall, our findings suggested that liposome-encapsulated melphalan was an effective drug modification of the melphalan compound and showed promise in MM treatment.
Collapse
Affiliation(s)
- Zhimei Lin
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
- Department
of Hematology, The Affiliated Hospital of
Chengdu University, Chengdu610081, P. R. China
| | - Bingyang Chu
- State
Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan610041, P. R. China
| | - Ying Qu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
- State
Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan610041, P. R. China
| | - Xue Wei
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Jingcao Huang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Fangfang Wang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Yu Feng
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Xin Wang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Hongmei Luo
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Xinyu Zhai
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Juan Xu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Xiang Liu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Li Zhang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Fengjiao Chen
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Yu Wu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Yuhuan Zheng
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| |
Collapse
|
4
|
Gupta U, Saren BN, Khaparkhuntikar K, Madan J, Singh PK. Applications of lipid-engineered nanoplatforms in the delivery of various cancer therapeutics to surmount breast cancer. J Control Release 2022; 348:1089-1115. [PMID: 35640765 DOI: 10.1016/j.jconrel.2022.05.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/18/2022] [Accepted: 05/21/2022] [Indexed: 11/30/2022]
Abstract
Breast cancer (BC) is the most extensively accounted malignancy among the women across the globe and is treatable in 70-80% of patients with early-stage, non-metastatic cancer. The current available therapies have been found to be less effective to treat distant organ metastases and advanced breast cancers. The clinical efficacy hugely suffers from chemoresistance, non-specific toxicity, relapse and other associated adverse effects. Furthermore, lack of controlled delivery and effective temporospatial presence of chemotherapeutics has resulted in suboptimal therapeutic response. Nanotechnology based approaches have been widely used over the period as they are nanometric, offer controlled and site-specific drug release along with reduced toxicity, improved half-life, and stability. Lipid-based nanoplatforms have grabbed a tremendous attention for delivering cancer therapeutics as they are cost-effective, scalable and provide better entrapment efficiency. In this review, all the promising applications of lipid-engineered nanotechnological tools for breast cancer will be summarized and discussed. Subsequently, BC therapy achieved with the aid of chemotherapeutics, phytomedicine, genes, peptides, photosensitizers, diagnostic and immunogenic agents etc. will be reviewed and discussed. This review gives tabular information on all the results obtained pertaining to the physicochemical properties of the lipidic nanocarrier, in vitro studies conferring to mechanistic drug release profile, cell viability, cellular apoptosis and in vivo studies referring to cellular internalisation, reduction of tumor volume, PK-PD profile, bioavailability achieved and anti-tumor activity in detail. It also gives complete information on the most relevant clinical trials done on lipidic nanoplatforms over two decades in tabular form. The review highlights the current status and future prospects of lipidic nanoplatforms with streamlined focus on cancer nanotherapeutics.
Collapse
Affiliation(s)
- Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Brojendra Nath Saren
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Kedar Khaparkhuntikar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
5
|
Tretiakova D, Svirshchevskaya E, Onishchenko N, Alekseeva A, Boldyrev I, Kamyshinsky R, Natykan A, Lokhmotov A, Arantseva D, Shobolov D, Vodovozova E. Liposomal Formulation of a Melphalan Lipophilic Prodrug: Studies of Acute Toxicity, Tolerability, and Antitumor Efficacy. Curr Drug Deliv 2021; 17:312-323. [PMID: 32056524 DOI: 10.2174/1567201817666200214105357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/13/2019] [Accepted: 02/02/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recently we developed a scalable scheme of synthesis of melphalan ester conjugate with 1,2-dioleoyl-sn-glycerol (MlphDG) and a protocol for the fabrication of its lyophilized liposomal formulation. OBJECTIVE Herein we compared this new convenient in use formulation of MlphDG with parent drug Alkeran® in rats concerning several toxicological parameters and evaluated its antitumor efficacy in the model of breast cancer in mice. METHOD Liposomes of approximately 100 nm in diameter, consisting of egg phosphatidylcholine, soybean phosphatidylinositol, and MlphDG, or placebo liposomes without the drug were produced by extrusion and lyophilized. Alkeran® or liposomes recovered by the addition of water were injected into the tail vein of animals. Clinical examination of rats consisted of detailed inspection of the behavior, general status, and hematological parameters. Mice with transplanted breast cancer WNT-1 were subjected to multiple treatments with the drugs; tumor growth inhibition was assessed, together with cellular immunity parameters. RESULTS Liposomes showed approximately two times lower acute toxicity and better tolerability than Alkeran® in terms of behavioral criteria. The toxic effects of liposomes on hemopoiesis were manifested at higher doses than in the case of Alkeran®, proportionally to the difference in LD50 values. The formulation inhibited tumor growth significantly more effectively than Alkeran®, delaying the start of the exponential growth phase and exhibiting no additional toxic effects toward bone marrow. CONCLUSION Lower toxicity of the liposomal formulation of MlphDG promises improved quality of life for cancer patients in need of treatment with melphalan. Presumably, the list of indications for melphalan therapy could be extended.
Collapse
Affiliation(s)
- Daria Tretiakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Elena Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Natalia Onishchenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Anna Alekseeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Ivan Boldyrev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Roman Kamyshinsky
- National Research Center "Kurchatov Institute", Moscow, Russian Federation
| | - Alexey Natykan
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Anton Lokhmotov
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Diana Arantseva
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Dmitry Shobolov
- Drugs Technology Ltd., Khimki, Мoscow Region, Russian Federation
| | - Elena Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
6
|
Wang Y, van Steenbergen MJ, Beztsinna N, Shi Y, Lammers T, van Nostrum CF, Hennink WE. Biotin-decorated all-HPMA polymeric micelles for paclitaxel delivery. J Control Release 2020; 328:970-984. [PMID: 32926885 DOI: 10.1016/j.jconrel.2020.09.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/21/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022]
Abstract
To avoid poly(ethylene glycol)-related issues of nanomedicines such as accelerated blood clearance, fully N-2-hydroxypropyl methacrylamide (HPMAm)-based polymeric micelles decorated with biotin for drug delivery were designed. To this end, a biotin-functionalized chain transfer agent (CTA), 4-cyano-4-[(dodecylsulfanylthiocarbonyl)-sulfanyl]pentanoic acid (biotin-CDTPA), was synthesized for reversible addition-fragmentation chain-transfer (RAFT) polymerization. Amphiphilic poly(N-2-hydroxypropyl methacrylamide)-block-poly(N-2-benzoyloxypropyl methacrylamide) (p(HPMAm)-b-p(HPMAm-Bz)) with molecular weights ranging from 8 to 24 kDa were synthesized using CDTPA or biotin-CDTPA as CTA and 2,2'-azobis(2-methylpropionitrile) as initiator. The copolymers self-assembled in aqueous media into micelles with sizes of 40-90 nm which positively correlated to the chain length of the hydrophobic block in the polymers, whereas the critical micelle concentrations decreased with increasing hydrophobic block length. The polymer with a molecular weight of 22.1 kDa was used to prepare paclitaxel-loaded micelles which had sizes between 61 and 70 nm, and a maximum loading capacity of around 10 wt%. A549 lung cancer cells overexpressing the biotin receptor, internalized the biotin-decorated micelles more efficiently than non-targeted micelles, while very low internalization of both types of micelles by HEK293 human embryonic kidney cells lacking the biotin receptor was observed. As a consequence, the paclitaxel-loaded micelles with biotin decoration exhibited stronger cytotoxicity in A549 cells than non-targeted micelles. Overall, a synthetic pathway to obtain actively targeted poly(ethylene glycol)-free micelles fully based on a poly(HPMAm) backbone was established. These polymeric micelles are promising systems for the delivery of hydrophobic anticancer drugs.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Mies J van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Nataliia Beztsinna
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbecktrasse 55, 52074 Aachen, Germany.
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, the Netherlands.
| |
Collapse
|
7
|
Kopeček J, Yang J. Polymer nanomedicines. Adv Drug Deliv Rev 2020; 156:40-64. [PMID: 32735811 PMCID: PMC7736172 DOI: 10.1016/j.addr.2020.07.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
Polymer nanomedicines (macromolecular therapeutics, polymer-drug conjugates, drug-free macromolecular therapeutics) are a group of biologically active compounds that are characterized by their large molecular weight. This review focuses on bioconjugates of water-soluble macromolecules with low molecular weight drugs and selected proteins. After analyzing the design principles, different structures of polymer carriers are discussed followed by the examination of the efficacy of the conjugates in animal models and challenges for their translation into the clinic. Two innovative directions in macromolecular therapeutics that depend on receptor crosslinking are highlighted: a) Combination chemotherapy of backbone degradable polymer-drug conjugates with immune checkpoint blockade by multivalent polymer peptide antagonists; and b) Drug-free macromolecular therapeutics, a new paradigm in drug delivery.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
8
|
Norouzi M, Amerian M, Amerian M, Atyabi F. Clinical applications of nanomedicine in cancer therapy. Drug Discov Today 2020; 25:107-125. [DOI: 10.1016/j.drudis.2019.09.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/03/2019] [Accepted: 09/24/2019] [Indexed: 12/23/2022]
|
9
|
Chen L, Mignani S, Caminade AM, Majoral JP. Metal-based phosphorus dendrimers as novel nanotherapeutic strategies to tackle cancers: A concise overview. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1577. [PMID: 31392836 DOI: 10.1002/wnan.1577] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/26/2019] [Accepted: 06/29/2019] [Indexed: 02/03/2023]
Abstract
Several metal-based phosphorus dendrimers were prepared. The first series developed by us was the Cu(II) series. In this series, the most potent is the third generation-Cu(II) showing original mechanism of action with activation of the pro-apoptotic Bax protein. To our knowledge, it is the first example of nanoparticles displaying Bax protein activation and then cell death through apoptosis process. Interestingly, this dendritic-Cu(II) complex showed synergistic effect with doxorubicin. Based on these interesting anti-proliferative activities, we developed Au(III)-conjugated phosphorus dendrimers. The most potent is the third generation-Au(III) dendrimer which represents also a new and promising first-in-class anti-proliferative agent against both solid and liquid tumor cell lines. Then, in order to analyze the influence of the metal moiety distribution of Cu(II) and Au(III) on the surface of dendrimers, mix Cu(II)-Au(III)-conjugated phosphorus dendrimers were also prepared and tested as anti-proliferative agents. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Liang Chen
- Laboratoire de Chimie de Coordination du CNRS, Toulouse, France.,LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France.,State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, PR China
| | - Serge Mignani
- Department of Pharmacy, Zhengzhou Railway Vocational & Technical College, Zhengzhou, PR China.,Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, Université Paris Descartes, Paris, France.,Centro de Química da Madeira (CQM), MMRG, Universidade da Madeira, Funchal, Portugal.,Glycovax Pharma, Montreal, Quebec, Canada
| | - Anne-Marie Caminade
- Laboratoire de Chimie de Coordination du CNRS, Toulouse, France.,LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, Toulouse, France.,LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France.,Department of Pharmacy, Zhengzhou Railway Vocational & Technical College, Zhengzhou, PR China
| |
Collapse
|
10
|
Zhu D, Long Q, Xu Y, Xing J. Evaluating Nanoparticles in Preclinical Research Using Microfluidic Systems. MICROMACHINES 2019; 10:mi10060414. [PMID: 31234335 PMCID: PMC6631852 DOI: 10.3390/mi10060414] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
Nanoparticles (NPs) have found a wide range of applications in clinical therapeutic and diagnostic fields. However, currently most NPs are still in the preclinical evaluation phase with few approved for clinical use. Microfluidic systems can simulate dynamic fluid flows, chemical gradients, partitioning of multi-organs as well as local microenvironment controls, offering an efficient and cost-effective opportunity to fast screen NPs in physiologically relevant conditions. Here, in this review, we are focusing on summarizing key microfluidic platforms promising to mimic in vivo situations and test the performance of fabricated nanoparticles. Firstly, we summarize the key evaluation parameters of NPs which can affect their delivery efficacy, followed by highlighting the importance of microfluidic-based NP evaluation. Next, we will summarize main microfluidic systems effective in evaluating NP haemocompatibility, transport, uptake and toxicity, targeted accumulation and general efficacy respectively, and discuss the future directions for NP evaluation in microfluidic systems. The combination of nanoparticles and microfluidic technologies could greatly facilitate the development of drug delivery strategies and provide novel treatments and diagnostic techniques for clinically challenging diseases.
Collapse
Affiliation(s)
- Derui Zhu
- Research Center of Basic Medical Sciences, Medical College, Qinghai University, Xining 810016, China.
| | - Qifu Long
- Research Center of Basic Medical Sciences, Medical College, Qinghai University, Xining 810016, China.
| | - Yuzhen Xu
- Department of Basic Medical Sciences, Medical College, Qinghai University, Xining 810016, China.
| | - Jiangwa Xing
- Research Center of Basic Medical Sciences, Medical College, Qinghai University, Xining 810016, China.
| |
Collapse
|
11
|
Farjadian F, Ghasemi A, Gohari O, Roointan A, Karimi M, Hamblin MR. Nanopharmaceuticals and nanomedicines currently on the market: challenges and opportunities. Nanomedicine (Lond) 2019; 14:93-126. [PMID: 30451076 PMCID: PMC6391637 DOI: 10.2217/nnm-2018-0120] [Citation(s) in RCA: 333] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022] Open
Abstract
There has been a revolution in nanotechnology and nanomedicine. Since 1980, there has been a remarkable increase in approved nano-based pharmaceutical products. These novel nano-based systems can either be therapeutic agents themselves, or else act as vehicles to carry different active pharmaceutical agents into specific parts of the body. Currently marketed nanostructures include nanocrystals, liposomes and lipid nanoparticles, PEGylated polymeric nanodrugs, other polymers, protein-based nanoparticles and metal-based nanoparticles. A range of issues must be addressed in the development of these nanostructures. Ethics, market size, possibility of market failure, costs and commercial development, are some topics which are on the table to be discussed. After passing all the ethical and biological assessments, and satisfying the investors as to future profitability, only a handful of these nanoformulations, successfully obtained marketing approval. We survey the range of nanomedicines that have received regulatory approval and are marketed. We discuss ethics, costs, commercial development and possible market failure. We estimate the global nanomedicine market size and future growth. Our goal is to summarize the different approved nanoformulations on the market, and briefly cover the challenges and future outlook.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Amir Ghasemi
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran 11365-9466, Iran
- Advances Nanobiotechnology & Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14496-4535, Iran
| | - Omid Gohari
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran 11365-9466, Iran
| | - Amir Roointan
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Science, Shiraz 71348-14336, Iran
| | - Mahdi Karimi
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Harvard – MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA
| |
Collapse
|
12
|
Yang J, Li L, Kopeček J. Biorecognition: A key to drug-free macromolecular therapeutics. Biomaterials 2018; 190-191:11-23. [PMID: 30391799 DOI: 10.1016/j.biomaterials.2018.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/02/2018] [Accepted: 10/07/2018] [Indexed: 12/13/2022]
Abstract
This review highlights a new paradigm in macromolecular nanomedicine - drug-free macromolecular therapeutics (DFMT). The effectiveness of the new system is based on biorecognition events without the participation of low molecular weight drugs. Apoptosis of cells can be initiated by the biorecognition of complementary peptide/oligonucleotide motifs at the cell surface resulting in the crosslinking of slowly internalizing receptors. B-cell CD20 receptors and Non-Hodgkin lymphoma (NHL) were chosen as the first target. Exposing cells to a conjugate of one motif with a targeting ligand decorates the cells with this motif. Further exposure of decorated cells to a macromolecule (synthetic polymer or human serum albumin) containing multiple copies of the complementary motif as grafts results in receptor crosslinking and apoptosis induction in vitro and in vivo. The review focuses on recent developments and explores the mechanism of action of DFMT. The altered molecular signaling pathways demonstrated the great potential of DFMT to overcome rituximab resistance resulting from either down-regulation of CD20 or endocytosis and trogocytosis of rituximab/CD20 complexes. The suitability of this approach for the treatment of blood borne cancers is confirmed. In addition, the widespread applicability of DFMT as a new concept in macromolecular therapeutics for numerous diseases is exposed.
Collapse
Affiliation(s)
- Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA.
| | - Lian Li
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA
| | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
13
|
Dong YD, Tchung E, Nowell C, Kaga S, Leong N, Mehta D, Kaminskas LM, Boyd BJ. Microfluidic preparation of drug-loaded PEGylated liposomes, and the impact of liposome size on tumour retention and penetration. J Liposome Res 2017; 29:1-9. [DOI: 10.1080/08982104.2017.1391285] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Yao-Da Dong
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Estefania Tchung
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Cameron Nowell
- Drug Delivery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Sadik Kaga
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
- Department of Chemistry, Bogazici University, Istanbul, Turkey
| | - Nathania Leong
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Dharmini Mehta
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Lisa M. Kaminskas
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
- School of Biomedical Sciences, University of Queensland, St Lucia, Australia
| | - Ben J. Boyd
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
- ARC Centre of Excellence in Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
14
|
Duro-Castano A, Gallon E, Decker C, Vicent MJ. Modulating angiogenesis with integrin-targeted nanomedicines. Adv Drug Deliv Rev 2017; 119:101-119. [PMID: 28502767 DOI: 10.1016/j.addr.2017.05.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/12/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022]
Abstract
Targeting angiogenesis-related pathologies, which include tumorigenesis and metastatic processes, has become an attractive strategy for the development of efficient guided nanomedicines. In this respect, integrins are cell-adhesion molecules involved in angiogenesis signaling pathways and are overexpressed in many angiogenic processes. Therefore, they represent specific biomarkers not only to monitor disease progression but also to rationally design targeted nanomedicines. Arginine-glycine-aspartic (RGD) containing peptides that bind to specific integrins have been widely utilized to provide ligand-mediated targeting capabilities to small molecules, peptides, proteins, and antibodies, as well as to drug/imaging agent-containing nanomedicines, with the final aim of maximizing their therapeutic index. Within this review, we aim to cover recent and relevant examples of different integrin-assisted nanosystems including polymeric nanoconstructs, liposomes, and inorganic nanoparticles applied in drug/gene therapy as well as imaging and theranostics. We will also critically address the overall benefits of integrin-targeting.
Collapse
Affiliation(s)
- Aroa Duro-Castano
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Elena Gallon
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Caitlin Decker
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
15
|
Ozcelikkale A, Moon HR, Linnes M, Han B. In vitro microfluidic models of tumor microenvironment to screen transport of drugs and nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1460. [PMID: 28198106 PMCID: PMC5555839 DOI: 10.1002/wnan.1460] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/14/2016] [Accepted: 12/17/2016] [Indexed: 12/16/2022]
Abstract
Advances in nanotechnology have enabled numerous types of nanoparticles (NPs) to improve drug delivery to tumors. While many NP systems have been proposed, their clinical translation has been less than anticipated primarily due to failure of current preclinical evaluation techniques to adequately model the complex interactions between the NP and physiological barriers of tumor microenvironment. This review focuses on microfluidic tumor models for characterization of delivery efficacy and toxicity of cancer nanomedicine. Microfluidics offer significant advantages over traditional macroscale cell cultures by enabling recapitulation of tumor microenvironment through precise control of physiological cues such as hydrostatic pressure, shear stress, oxygen, and nutrient gradients. Microfluidic systems have recently started to be adapted for screening of drugs and NPs under physiologically relevant settings. So far the two primary application areas of microfluidics in this area have been high-throughput screening using traditional culture settings such as single cells or multicellular tumor spheroids, and mimicry of tumor microenvironment for study of cancer-related cell-cell and cell-matrix interactions. These microfluidic technologies are also useful in modeling specific steps in NP delivery to tumor and characterize NP transport properties and outcomes by systematic variation of physiological conditions. Ultimately, it will be possible to design drug-screening platforms uniquely tailored for individual patient physiology using microfluidics. These in vitro models can contribute to development of precision medicine by enabling rapid and patient-specific evaluation of cancer nanomedicine. WIREs Nanomed Nanobiotechnol 2017, 9:e1460. doi: 10.1002/wnan.1460 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Altug Ozcelikkale
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Hye-ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Michael Linnes
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA,
| |
Collapse
|
16
|
Santos MA, Goertz DE, Hynynen K. Focused Ultrasound Hyperthermia Mediated Drug Delivery Using Thermosensitive Liposomes and Visualized With in vivo Two-Photon Microscopy. Am J Cancer Res 2017; 7:2718-2731. [PMID: 28819458 PMCID: PMC5558564 DOI: 10.7150/thno.19662] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/28/2017] [Indexed: 12/25/2022] Open
Abstract
The future of nanomedicines in oncology requires leveraging more than just the passive drug accumulation in tumors through the enhanced permeability and retention effect. Promising results combining mild hyperthermia (HT) with lyso-thermosensitive liposomal doxorubicin (LTSL-DOX) has led to improved drug delivery and potent antitumor effects in pre-clinical studies. The ultimate patient benefit from these treatments can only be realized when robust methods of HT can be achieved clinically. One of the most promising methods of non-invasive HT is the use of focused ultrasound (FUS) with MRI thermometry for anatomical targeting and feedback. MRI-guided focused ultrasound (MRgFUS) is limited by respiratory motion and large blood vessel cooling. In order to translate exciting pre-clinical results to the clinic, novel heating approaches capable of overcoming the limitations on clinical MRgFUS+HT must be tested and evaluated on their ability to locally release drug from LTSL-DOX. Methods: In this work, a new system is described to integrate focused ultrasound (FUS) into a two-photon microscopy (2PM) setting to image the release of drug from LTSL-DOX in real-time during FUS+HT in vivo. A candidate scheme for overcoming the limitations of respiratory motion and large blood vessel cooling during MRgFUS+HT involves applying FUS+HT to 42°C in short ~30s bursts. The spatiotemporal drug release pattern from LTSL-DOX as a result is quantified using 2PM and compared against continuous (3.5min and 20min at 42°C) FUS+HT schemes and unheated controls. Results: It was observed for the first time in vivo that these short duration temperature elevations could produce substantial drug release from LTSL-DOX. Ten 30s bursts of FUS+HT was able to achieve almost half of the interstitial drug concentration as 20min of continuous FUS+HT. There was no significant difference between the intravascular area under the concentration-time curve for ten 30s bursts of FUS+HT and 3.5min of continuous FUS+HT. Conclusion: We have successfully combined 2PM with FUS+HT for imaging the release of DOX from LTSL-DOX in vivo in real-time, which will permit the investigation of FUS+HT heating schemes to improve drug delivery from LTSL-DOX. We have evaluated the ability to release DOX in short 30s FUS+HT bursts to 42°C as a method to overcome limitations on clinical MRgFUS+HT and have found that such exposures are capable of releasing measurable amounts of drug. Such an exposure has the potential to overcome limitations that hamper conventional MRgFUS+HT treatments in targets that are associated with substantial tissue motion.
Collapse
|
17
|
Houdaihed L, Evans JC, Allen C. Overcoming the Road Blocks: Advancement of Block Copolymer Micelles for Cancer Therapy in the Clinic. Mol Pharm 2017; 14:2503-2517. [DOI: 10.1021/acs.molpharmaceut.7b00188] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Loujin Houdaihed
- Department of Pharmaceutical
Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | - James C. Evans
- Department of Pharmaceutical
Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | - Christine Allen
- Department of Pharmaceutical
Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| |
Collapse
|
18
|
Dubbelboer IR, Lilienberg E, Sjögren E, Lennernäs H. A Model-Based Approach To Assessing the Importance of Intracellular Binding Sites in Doxorubicin Disposition. Mol Pharm 2017; 14:686-698. [PMID: 28182434 DOI: 10.1021/acs.molpharmaceut.6b00974] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Doxorubicin is an anticancer agent, which binds reversibly to topoisomerase I and II, intercalates to DNA base pairs, and generates free radicals. Doxorubicin has a high tissue:plasma partition coefficient and high intracellular binding to the nucleus and other subcellular compartments. The metabolite doxorubicinol has an extensive tissue distribution. This porcine study investigated whether the traditional implementation of tissue binding, described by the tissue:plasma partition coefficient (Kp,t), could be used to appropriately analyze and/or simulate tissue doxorubicin and doxorubicinol concentrations in healthy pigs, when applying a physiologically based pharmacokinetic (PBPK) model approach, or whether intracellular binding is required in the semi-PBPK model. Two semi-PBPK models were developed and evaluated using doxorubicin and doxorubicinol concentrations in healthy pig blood, bile, and urine and kidney and liver tissues. In the generic semi-PBPK model, tissue binding was described using the conventional Kp,t approach. In the binding-specific semi-PBPK model, tissue binding was described using intracellular binding sites. The best semi-PBPK model was validated against a second data set of healthy pig blood and bile concentrations. Both models could be used for analysis and simulations of biliary and urinary excretion of doxorubicin and doxorubicinol and plasma doxorubicinol concentrations in pigs, but the binding-specific model was better at describing plasma doxorubicin concentrations. Porcine tissue concentrations were 400- to 1250-fold better captured by the binding-specific model. This model adequately predicted plasma doxorubicin concentration-time and biliary doxorubicin excretion profiles against the validation data set. The semi-PBPK models applied were similarly effective for analysis of plasma concentrations and biliary and urinary excretion of doxorubicin and doxorubicinol in healthy pigs. Inclusion of intracellular binding in the doxorubicin semi-PBPK models was important to accurately describe tissue concentrations during in vivo conditions.
Collapse
Affiliation(s)
- Ilse R Dubbelboer
- Department of Pharmacy, Uppsala University , Box 580, 751 23 Uppsala, Sweden
| | - Elsa Lilienberg
- Department of Pharmacy, Uppsala University , Box 580, 751 23 Uppsala, Sweden
| | - Erik Sjögren
- Department of Pharmacy, Uppsala University , Box 580, 751 23 Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmacy, Uppsala University , Box 580, 751 23 Uppsala, Sweden
| |
Collapse
|