1
|
Sun Y, Wu X, Li J, Radiom M, Mezzenga R, Verma CS, Yu J, Miserez A. Phase-separating peptide coacervates with programmable material properties for universal intracellular delivery of macromolecules. Nat Commun 2024; 15:10094. [PMID: 39572548 PMCID: PMC11582321 DOI: 10.1038/s41467-024-54463-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Phase-separating peptides (PSPs) self-assembling into coacervate microdroplets (CMs) are a promising class of intracellular delivery vehicles that can release macromolecular modalities deployed in a wide range of therapeutic treatments. However, the molecular grammar governing intracellular uptake and release kinetics of CMs remains elusive. Here, we systematically manipulate the sequence of PSPs to unravel the relationships between their molecular structure, the physical properties of the resulting CMs, and their delivery efficacy. We show that a few amino acid alterations are sufficient to modulate the viscoelastic properties of CMs towards either a gel-like or a liquid-like state as well as their binding interaction with cellular membranes, collectively enabling to tune the kinetics of intracellular cargo release. We also demonstrate that the optimized PSPs CMs display excellent transfection efficiency in hard-to-transfect cells such as primary fibroblasts and immune cells. Our findings provide molecular guidelines to precisely program the material properties of PSP CMs and achieve tunable cellular uptake and release kinetics depending on the cargo modality, with broad implications for therapeutic applications such as protein, gene, and immune cell therapies.
Collapse
Affiliation(s)
- Yue Sun
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
| | - Xi Wu
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
| | - Jianguo Li
- Bioinformatics Institute, Agency for Science, Technology and Research, 30 Biopolis Street, Matrix, 138671, Singapore, Singapore
- Singapore Eye Research Institute, 169856, Singapore, Singapore
| | - Milad Radiom
- Department of Health Sciences & Technology, ETH Zurich, 8092, Zürich, Switzerland
| | - Raffaele Mezzenga
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
- Department of Health Sciences & Technology, ETH Zurich, 8092, Zürich, Switzerland
- Department of Materials, ETH Zurich, 8092, Zürich, Switzerland
| | - Chandra Shekhar Verma
- Bioinformatics Institute, Agency for Science, Technology and Research, 30 Biopolis Street, Matrix, 138671, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, 117558, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore, Singapore
| | - Jing Yu
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore, Singapore
| | - Ali Miserez
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore.
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore, Singapore.
| |
Collapse
|
2
|
Liang C, Zhang G, Guo L, Ding X, Yang H, Zhang H, Zhang Z, Hou L. Spatiotemporal transformable nano-assembly for on-demand drug delivery to enhance anti-tumor immunotherapy. Asian J Pharm Sci 2024; 19:100888. [PMID: 38434719 PMCID: PMC10904913 DOI: 10.1016/j.ajps.2024.100888] [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: 11/06/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 03/05/2024] Open
Abstract
Induction of tumor cell senescence has become a promising strategy for anti-tumor immunotherapy, but fibrotic matrix severely blocks senescence inducers penetration and immune cells infiltration. Herein, we designed a cancer-associated fibroblasts (CAFs) triggered structure-transformable nano-assembly (HSD-P@V), which can directionally deliver valsartan (Val, CAFs regulator) and doxorubicin (DOX, senescence inducer) to the specific targets. In detail, DOX is conjugated with hyaluronic acid (HA) via diselenide bonds (Se-Se) to form HSD micelles, while CAFs-sensitive peptide is grafted onto the HSD to form a hydrophilic polymer, which is coated on Val nanocrystals (VNs) surface for improving the stability and achieving responsive release. Once arriving at tumor microenvironment and touching CAFs, HSD-P@V disintegrates into VNs and HSD micelles due to sensitive peptide detachment. VNs can degrade the extracellular matrix, leading to the enhanced penetration of HSD. HSD targets tumor cells, releases DOX to induce senescence, and recruits effector immune cells. Furthermore, senescent cells are cleared by the recruited immune cells to finish the integrated anti-tumor therapy. In vitro and in vivo results show that the nano-assembly remarkably inhibits tumor growth as well as lung metastasis, and extends tumor-bearing mice survival. This work provides a promising paradigm of programmed delivering multi-site nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Chenglin Liang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Ge Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Linlin Guo
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinyi Ding
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Heng Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Hongling Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Lin Hou
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
3
|
Zhang R, Zhao X, Jia A, Wang C, Jiang H. Hyaluronic acid-based prodrug nanomedicines for enhanced tumor targeting and therapy: A review. Int J Biol Macromol 2023; 249:125993. [PMID: 37506794 DOI: 10.1016/j.ijbiomac.2023.125993] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Hyaluronic acid (HA) represents a natural polysaccharide which has attracted significant attention owing to its improved tumor targeting capacity, enzyme degradation capacity, and excellent biocompatibility. Its receptors, such as CD44, are overexpressed in diverse cancer cells and are closely related with tumor progress and metastasis. Accordingly, numerous researchers have designed various kinds of HA-based drug delivery platforms for CD44-mediated tumor targeting. Specifically, the HA-based nanoprodrugs possess distinct advantages such as good bioavailability, long circulation time, and controlled drug release and retention ability and have been extensively studied during the past years. In this review, the potential strategies and applications of HA-modified nanoprodrugs for drug molecule delivery in anti-tumor therapy are summarized.
Collapse
Affiliation(s)
- Renshuai Zhang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China
| | - Xiaohua Zhao
- Department of Thoracic surgery, Affiliated Hospital of Weifang Medical University, No.2428, Yuhe road, Kuiwen district, Weifang 261000, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Chao Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| |
Collapse
|
4
|
Xiao Q, Li X, Liu C, Jiang Y, He Y, Zhang W, Azevedo HS, Wu W, Xia Y, He W. Improving cancer immunotherapy via co-delivering checkpoint blockade and thrombospondin-1 downregulator. Acta Pharm Sin B 2023; 13:3503-3517. [PMID: 37655330 PMCID: PMC10465872 DOI: 10.1016/j.apsb.2022.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/24/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
The use of checkpoint-blockade antibodies is still restricted in several malignancies due to the modest efficacy, despite considerable success in anti-tumor immunotherapy. The poor response of cancer cells to immune destruction is an essential contributor to the failure of checkpoint therapy. We hypothesized that combining checkpoint therapy with natural-product chemosensitizer could enhance immune response. Herein, a targeted diterpenoid derivative was integrated with the checkpoint blockade (anti-CTLA-4) to improve immunotherapy using thermosensitive liposomes as carriers. In vivo, the liposomes enabled the co-delivery of the two drug payloads into the tumor. Consequently, the regulatory T cell proliferation was restrained, the cytotoxic T cell infiltration was enhanced, and the profound immunotherapeutic effect was achieved. In addition, the immunotherapeutic effect of another clinically used checkpoint antibody, anti-PD-1, also benefited from the diterpenoid derivative. Of note, our mechanism study revealed that the targeted diterpenoid derivative increased the sensitivity of cancer cells to immune attack via THBS1 downregulation and the resultant destruction of THBS1-CD47 interaction. Collectively, co-delivering THBS1 inhibitor and checkpoint blockade is promising to boost cancer immunotherapy. We first time discovered that THBS1 suppression could strengthen checkpoint therapy.
Collapse
Affiliation(s)
- Qingqing Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chang Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yuxin Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yonglong He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wanting Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London E1 4NS, UK
| | - Wei Wu
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuanzheng Xia
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
5
|
Curcio M, Vittorio O, Bell JL, Iemma F, Nicoletta FP, Cirillo G. Hyaluronic Acid within Self-Assembling Nanoparticles: Endless Possibilities for Targeted Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12162851. [PMID: 36014715 PMCID: PMC9413373 DOI: 10.3390/nano12162851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 05/27/2023]
Abstract
Self-assembling nanoparticles (SANPs) based on hyaluronic acid (HA) represent unique tools in cancer therapy because they combine the HA targeting activity towards cancer cells with the advantageous features of the self-assembling nanosystems, i.e., chemical versatility and ease of preparation and scalability. This review describes the key outcomes arising from the combination of HA and SANPs, focusing on nanomaterials where HA and/or HA-derivatives are inserted within the self-assembling nanostructure. We elucidate the different HA derivatization strategies proposed for this scope, as well as the preparation methods used for the fabrication of the delivery device. After showing the biological results in the employed in vivo and in vitro models, we discussed the pros and cons of each nanosystem, opening a discussion on which approach represents the most promising strategy for further investigation and effective therapeutic protocol development.
Collapse
Affiliation(s)
- Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Orazio Vittorio
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Jessica Lilian Bell
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
6
|
Ye J, Yi Y, Wang H, Wang G, Sun Y, Liu E, Tao X, He C. A Study of Glutathione-Responsive Dual-Drug-Loaded Nanoparticles in Anti-Osteosarcoma Treatment. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We connected polyglutamic acid and methotrexate (MTX) through disulfide bonds to prepare glutathione-responsive nanoparticles (MTX NPs) and encapsulated doxorubicin (DOX) to obtain dual drug-loaded NPs (DOX/MTX NPs) (Fig. 1). The appearance of the carbonyl stretching vibration peak
at approximately 1640 cm−1 in the results of the infrared spectrum proved the successful synthesis of three kinds of nanoparticles (NPs) with different feeding ratios. The particle sizes of NPs with different feeding ratios were 100–200 nm, and the encapsulation of DOX
slightly increased the size, while the surface charge was always negative. The release of MTX at 10 mM glutathione (GSH) was as high as 91.45%, and that of DOX was 89.44%, suggesting that the breakage of disulfide bonds leads to the disintegration of NPs. The results of the cell experiment
showed that the encapsulation of DOX effectively increased toxicity and side effects in 143B cells and significantly induced cell apoptosis, and the inhibition of the migration rate increased as the feeding ratio increased. In animal experiments, DOX/MTX NPs significantly induced tumor cell
apoptosis and inhibited cell proliferation and tumor growth. The nanoparticles had excellent tumor-targeting properties. Tumor-targeted NPs with the combined action of the two drugs provided a good strategy for the efficient and precise treatment of osteosarcoma.
Collapse
Affiliation(s)
- Jia Ye
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Yangfei Yi
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Hongyi Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Guowei Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Yuting Sun
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Enze Liu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Xiaojun Tao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Chunlian He
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, China
| |
Collapse
|
7
|
Gupta N, Malviya R. Role of Polysaccharides Mimetic Components in Targeted Cancer Treatment. Curr Drug Targets 2022; 23:856-868. [PMID: 35156570 DOI: 10.2174/1389450123666220214121505] [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: 09/14/2021] [Revised: 11/09/2021] [Accepted: 12/02/2021] [Indexed: 11/22/2022]
Abstract
Organic or inorganic compounds are synthesized or formulated in a manner that they completely show their therapeutic actions like as a natural polysaccharide in the body. Polysaccharides, the major type of natural polymers, are efficiently biologically active, non-toxic, hydrophilic, and biodegradable and show various properties. In this manuscript, the main focus is on delivering anticancer drugs with the help of mimetic components of polysaccharides. All data collected for this manuscript was from PubMed, Elsevier, Taylor, and Francis Bentham science journals. Most chemotherapeutics are therapeutically toxin to the human body, have a narrow therapeutic index, sluggish pharmaceutical delivery mechanisms, and are poorly soluble in water. The use of mimetic components of polysaccharides leads to the enhancement of the solubility of drugs in the biological environment. The manuscript summarizes the use of mimetic components of polysaccharides along with anticancer agents which are capable to inhibit the growth of cancerous cells in the body which shows lesser adverse effects in the biological system compared to other therapies.
Collapse
Affiliation(s)
- Nandan Gupta
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| |
Collapse
|
8
|
Sun Y, Lau SY, Lim ZW, Chang SC, Ghadessy F, Partridge A, Miserez A. Phase-separating peptides for direct cytosolic delivery and redox-activated release of macromolecular therapeutics. Nat Chem 2022; 14:274-283. [PMID: 35115657 DOI: 10.1038/s41557-021-00854-4] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022]
Abstract
Biomacromolecules are highly promising therapeutic modalities to treat various diseases. However, they suffer from poor cellular membrane permeability, limiting their access to intracellular targets. Strategies to overcome this challenge often employ nanoscale carriers that can get trapped in endosomal compartments. Here we report conjugated peptides that form pH- and redox-responsive coacervate microdroplets by liquid-liquid phase separation that readily cross the cell membrane. A wide range of macromolecules can be quickly recruited within the microdroplets, including small peptides, enzymes as large as 430 kDa and messenger RNAs (mRNAs). The therapeutic-loaded coacervates bypass classical endocytic pathways to enter the cytosol, where they undergo glutathione-mediated release of payload, the bioactivity of which is retained in the cell, while mRNAs exhibit a high transfection efficiency. These peptide coacervates represent a promising platform for the intracellular delivery of a large palette of macromolecular therapeutics that have potential for treating various pathologies (for example, cancers and metabolic diseases) or as carriers for mRNA-based vaccines.
Collapse
Affiliation(s)
- Yue Sun
- Biological and Biomimetic Material Laboratory (BBML), Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore, Singapore
| | - Sze Yi Lau
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), Neuros/Immunos, Singapore, Singapore
| | - Zhi Wei Lim
- Biological and Biomimetic Material Laboratory (BBML), Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore, Singapore
| | - Shi Chieh Chang
- Translation Medicine Research Centre, MSD International, Singapore, Singapore
| | - Farid Ghadessy
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), Neuros/Immunos, Singapore, Singapore
| | - Anthony Partridge
- Translation Medicine Research Centre, MSD International, Singapore, Singapore
| | - Ali Miserez
- Biological and Biomimetic Material Laboratory (BBML), Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore, Singapore. .,School of Biological Sciences, NTU, Singapore, Singapore.
| |
Collapse
|
9
|
Xiang J, Liu X, Yuan G, Zhang R, Zhou Q, Xie T, Shen Y. Nanomedicine from amphiphilizedprodrugs: Concept and clinical translation. Adv Drug Deliv Rev 2021; 179:114027. [PMID: 34732344 DOI: 10.1016/j.addr.2021.114027] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/30/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
Nanomedicines generally consisting of carrier materials with small fractions of active pharmaceutical ingredients (API) have long been used to improve the pharmacokinetics and biodistributions, augment the therapeutic efficacies and mitigate the side effects. Amphiphilizing hydrophobic/hydrophilic drugs to prodrugs capable of self-assembly into well-defined nanostructures has emerged as a facile approach to fabricating nanomedicines because this amphiphilized prodrug (APD) strategy presents many advantages, including minimized use of inert carrier materials, well-characterized prodrug structures, fixed and high drug loading contents, 100% loading efficiency, and burst-free but controlled drug release. This review comprehensively summarizes recent advances in APDs and their nanomedicines, from the rationale and the stimuli-responsive linker chemistry for on-demand drug release to their progress to the clinics, clinical performance of APDs, as well as the challenges and perspective on future development.
Collapse
|
10
|
Novel drug delivery systems based on silver nanoparticles, hyaluronic acid, lipid nanoparticles and liposomes for cancer treatment. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-02018-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
Bhattacharya DS, Bapat A, Svechkarev D, Mohs AM. Water-Soluble Blue Fluorescent Nonconjugated Polymer Dots from Hyaluronic Acid and Hydrophobic Amino Acids. ACS OMEGA 2021; 6:17890-17901. [PMID: 34308024 PMCID: PMC8296014 DOI: 10.1021/acsomega.1c01343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/24/2021] [Indexed: 05/04/2023]
Abstract
Fluorescent polymers have been increasingly investigated to improve their water solubility and biocompatibility to enhance their performance in drug delivery and theranostic applications. However, the environmentally friendly synthesis and dual functionality of such systems remain a challenge due to the complicated synthesis of conventional fluorescent materials. Herein, we generated a novel blue fluorescent polymer dot through chemical conjugation of hydrophobic amino acids to hyaluronic acid (HA) under one-pot green chemistry conditions. These nonconjugated fluorescent polymer dots (NCPDs) are water soluble, nontoxic to cells, have high fluorescence quantum yield, and can be used for in vitro bioimaging. HA-derived NCPDs exhibit excitation wavelength-dependent fluorescent properties. In addition, the NCPDs also show enhanced doxorubicin loading and delivery in naive and drug-resistant breast cancer cells in 2D and 3D tumor cellular systems. These results demonstrate the potential for successful synthetic scale-up and applications for HA-derived NCPDs.
Collapse
Affiliation(s)
- Deep S. Bhattacharya
- Department
of Pharmaceutical Sciences, University of
Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Aishwarya Bapat
- Department
of Pharmaceutical Sciences, University of
Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Denis Svechkarev
- Department
of Pharmaceutical Sciences, University of
Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Aaron M. Mohs
- Department
of Pharmaceutical Sciences, University of
Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Fred
and Pamela Buffett Cancer Center, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department
of Biochemistry and Molecular Biology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
12
|
pH-dependent reversibly activatable cell-penetrating peptides improve the antitumor effect of artemisinin-loaded liposomes. J Colloid Interface Sci 2020; 586:391-403. [PMID: 33189320 DOI: 10.1016/j.jcis.2020.10.103] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 12/16/2022]
Abstract
Artemisinin (ART) is well known as an antimalarial drug, and it can also be used to treat inflammation as well as cancer. Although many researchers have reported the antitumor activity of ART, most of these studies were investigated in vitro. In addition, ART is sparingly soluble in water, limiting its clinical relevance in drug development. Based on the data from our preliminary study, ART is not cytotoxic at low micromolar concentrations. Thus, we hypothesized that smart nanocarriers are beneficial for not only increasing the solubility of ART but also elevating the concentration of the drug at the target, thereby inducing the ideal antitumor effect. In this article, a reversibly activatable cell-penetrating peptide ((HE)10-G5-R6 or HE-R6) was introduced to modify artemisinin (ART)-loaded liposomes (ART-Lip-HE-R6) against tumors, and in vitro and in vivo performance were investigated. ART-Lip-HE-R6 exhibited sustained release under different pH conditions. The internalization and cytotoxicity of liposomes were enhanced at low pH, i.e., 6.5, after modification with HE-R6 versus nonmodified liposomes. Moreover, a longer retention time in tumors could be observed in the ART-Lip-HE-R6 group, followed by higher efficiency of tumor suppression. In conclusion, Lip-HE-R6 might be a promising delivery system for ART in cancer therapy.
Collapse
|
13
|
Karaosmanoglu S, Zhou M, Shi B, Zhang X, Williams GR, Chen X. Carrier-free nanodrugs for safe and effective cancer treatment. J Control Release 2020; 329:805-832. [PMID: 33045313 DOI: 10.1016/j.jconrel.2020.10.014] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022]
Abstract
Clinical applications of many anti-cancer drugs are restricted due to their hydrophobic nature, requiring use of harmful organic solvents for administration, and poor selectivity and pharmacokinetics resulting in off-target toxicity and inefficient therapies. A wide variety of carrier-based nanoparticles have been developed to tackle these issues, but such strategies often fail to encapsulate drug efficiently and require significant amounts of inorganic and/or organic nanocarriers which may cause toxicity problems in the long term. Preparation of nano-formulations for the delivery of water insoluble drugs without using carriers is thus desired, requiring elegantly designed strategies for products with high quality, stability and performance. These strategies include simple self-assembly or involving chemical modifications via coupling drugs together or conjugating them with various functional molecules such as lipids, carbohydrates and photosensitizers. During nanodrugs synthesis, insertion of redox-responsive linkers and tumor targeting ligands endows them with additional characteristics like on-target delivery, and conjugation with immunotherapeutic reagents enhances immune response alongside therapeutic efficacy. This review aims to summarize the methods of making carrier-free nanodrugs from hydrophobic drug molecules, evaluating their performance, and discussing the advantages, challenges, and future development of these strategies.
Collapse
Affiliation(s)
- Sena Karaosmanoglu
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK
| | - Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Bingyang Shi
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xiujuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, PR China.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Xianfeng Chen
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK.
| |
Collapse
|
14
|
Dong C, Jiang Q, Qian X, Wu W, Wang W, Yu L, Chen Y. A self-assembled carrier-free nanosonosensitizer for photoacoustic imaging-guided synergistic chemo-sonodynamic cancer therapy. NANOSCALE 2020; 12:5587-5600. [PMID: 32100776 DOI: 10.1039/c9nr10735e] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
As one of the most promising noninvasive therapeutic modalities, sonodynamic therapy (SDT) can focus the ultrasound energy on tumor sites located in deep tissue and locally activate the preloaded sonosensitizer to kill tumor cells. However, exploring sonosensitizers with high SDT efficacy and desirable biosafety is still a significant challenge. Herein, we utilized the hydrophilic-hydrophobic self-assembly technology to assemble the hydrophobic organic dye Ce6 and broad spectral anti-cancer agent Paclitaxel with hydrophilic organic dye IR783 to generate a nanoscale sonosensitizer, Ce6-PTX@IR783, without the introduction of extra nanomaterials into the fabrication to guarantee high therapeutic biosafety and further potential clinical translation. The constructed nanodrug was endowed with an external ultrasound-activatable chemo-sonodynamic effect and photoacoustic imaging performance via integrating multiple moieties into one nanosystem. Ce6 could enhance the sonodynamic effect, while PTX exerted a chemotherapeutic effect, and IR783 was applied to increase tumor-specific accumulation and assist in fulfilling photoacoustic imaging. In particular, the small particle size (70 nm) of Ce6-PTX@IR783 contributed to the increased tumor accumulation via the enhanced permeability and retention effect. The high synergistically chemo-sonodynamic therapeutic efficacy has been successfully demonstrated in vitro and in vivo, in addition to the demonstrated high biodegradability, biocompatibility and biosafety. This facile self-assembly procedure provides an intriguing strategy for highly efficient utilization of hydrophobic drugs and is liable to realize large-scale production and further clinical translation.
Collapse
Affiliation(s)
- Caihong Dong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China. and Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Qvzi Jiang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Xiaoqin Qian
- Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Wencheng Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China. and Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Luodan Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| |
Collapse
|
15
|
Liu Y, Khan AR, Du X, Zhai Y, Tan H, Zhai G. Progress in the polymer-paclitaxel conjugate. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
16
|
Gagliardi A, Bonacci S, Paolino D, Celia C, Procopio A, Fresta M, Cosco D. Paclitaxel-loaded sodium deoxycholate-stabilized zein nanoparticles: characterization and in vitro cytotoxicity. Heliyon 2019; 5:e02422. [PMID: 31517130 PMCID: PMC6734341 DOI: 10.1016/j.heliyon.2019.e02422] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/18/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
Paclitaxel (PTX) is one of the most successful antineoplastic drugs and is widely used for the treatment of many forms of advanced and refractory cancer. Unfortunately, various drawbacks including non-selective cytotoxicity, poor water solubility and low bioavailability limit its clinical use. The aim of this study was to characterize a novel colloidal system made up of the natural protein zein, that would be able to efficiently retain the anticancer compound and increase its in vitro pharmacological effects. In fact, zein has promising characteristics that render it a potential material to be used in drug delivery application. The influences of temperature, pH and serum incubation on the stability of these particles, entrapment efficiency of PTX and in vitro toxicity on different cancer cell lines were evaluated. The nanosystems containing PTX demonstrated suitable storage stability, and were not destabilized by temperatures of up to 50 °C, pH alterations, the freeze-drying process or serum proteins. The encapsulation of PTX did not destabilize the structure of the zein nanoparticles and a suitable drug entrapment efficiency resulted. PTX-loaded zein nanoparticles showed an increased toxicity on different cancer cell lines with respect to the free drug, confirming its potential application in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Agnese Gagliardi
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", I-88100, Catanzaro, Italy
| | - Sonia Bonacci
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", I-88100, Catanzaro, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", I-88100, Catanzaro, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", via dei Vestini 31, 66100, Chieti, Italy
| | - Antonio Procopio
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", I-88100, Catanzaro, Italy
| | - Massimo Fresta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", I-88100, Catanzaro, Italy
| | - Donato Cosco
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", I-88100, Catanzaro, Italy
| |
Collapse
|
17
|
Prodrugs in combination with nanocarriers as a strategy for promoting antitumoral efficiency. Future Med Chem 2019; 11:2131-2150. [DOI: 10.4155/fmc-2018-0388] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prodrug entrapment into nanocarriers for tumor delivery is a strategy to achieve a valid therapy with high efficiency. The prodrug contains anticancer agents conjugating with functional moieties or ligands so that the active component is released after metabolism in the body or tumor. The advantages of nanosystems for loading prodrugs include high loading, increased prodrug stability, improved bioavailability and enhanced targeting to tumor cells. In the present article, we introduce the prodrug delivery approaches according to nanomedicine and the recent advances in prodrug-loaded nanocarriers. First, we discuss the conceptional design of combined prodrugs and nanocarriers in response to the obstruction in anticancer therapy. Then we describe the cases of prodrug-loaded nanoparticles for cancer treatment during the past 5 years.
Collapse
|
18
|
Hayes AJ, Melrose J. Glycosaminoglycan and Proteoglycan Biotherapeutics in Articular Cartilage Protection and Repair Strategies: Novel Approaches to Visco‐supplementation in Orthobiologics. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research HubCardiff School of BiosciencesCardiff University Cardiff CF10 3AX Wales UK
| | - James Melrose
- Graduate School of Biomedical EngineeringUNSW Sydney Sydney NSW 2052 Australia
- Raymond Purves Bone and Joint Research LaboratoriesKolling Institute of Medical ResearchRoyal North Shore Hospital and The Faculty of Medicine and HealthUniversity of Sydney St. Leonards NSW 2065 Australia
- Sydney Medical SchoolNorthernRoyal North Shore HospitalSydney University St. Leonards NSW 2065 Australia
| |
Collapse
|
19
|
Zhang W, Cai J, Wu B, Shen Z. pH-responsive hyaluronic acid nanoparticles coloaded with sorafenib and cisplatin for treatment of hepatocellular carcinoma. J Biomater Appl 2019; 34:219-228. [PMID: 31084233 DOI: 10.1177/0885328219849711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Wei Zhang
- Tianjin First Center Hospital, Tianjin, China
| | - Jinzhen Cai
- Tianjin First Center Hospital, Tianjin, China
| | - Bin Wu
- Tianjin First Center Hospital, Tianjin, China
| | | |
Collapse
|
20
|
Aldawsari HM, Dhaliwal HK, Aljaeid BM, Alhakamy NA, Banjar ZM, Amiji MM. Optimization of the Conditions for Plasmid DNA Delivery and Transfection with Self-Assembled Hyaluronic Acid-Based Nanoparticles. Mol Pharm 2018; 16:128-140. [PMID: 30525660 DOI: 10.1021/acs.molpharmaceut.8b00904] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polymeric systems have been extensively studied as polyelectrolyte complexes to enhance the cellular delivery and transfection efficiency of genetic materials, such as plasmid DNA (pDNA). Here, self-assembled nanoparticles were formulated by complexation of hyaluronic acid (HA)-conjugated poly(ethylene glycol) (HA-PEG) and poly(ethylenimine) (HA-PEI), respectively, with pDNA creating relatively small, stable, and multifunctional nanoparticle complex formulations with high transfection efficiency. This formulation strategy offers high gene expression efficiency and negligible cytotoxicity in HeLa and A549 human lung cancer cell lines. To develop the ideal formulation, in vitro transfection efficiency was studied for three different nanoparticle formulations (HA-PEI/HA-PEG, HA-PEI, and HA-PEG) with different concentrations. The combination of the three polymers (HA, PEG, and PEI) was significant for the formulation to achieve the maximum gene expression results. The nanoparticles were found to be stable for up to a week at 4 °C conditions. Overall, these HA-based nanoparticles showed promising aspects that can be utilized in the designing of gene delivery vectors for cancer therapy.
Collapse
Affiliation(s)
- Hibah M Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , KSA
| | - Harkiranpreet Kaur Dhaliwal
- Department of Pharmaceutical Sciences, School of Pharmacy , Northeastern University , Boston , Massachusetts 02115 , United States
| | - Bader Mubarak Aljaeid
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , KSA
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , KSA
| | - Zainy Mohammad Banjar
- Department of Department of Clinical and Biochemistry, Faculty of Medicine , King Abdulaziz University , Jeddah 21589 , KSA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy , Northeastern University , Boston , Massachusetts 02115 , United States
| |
Collapse
|
21
|
A potential carrier for anti-tumor targeted delivery-hyaluronic acid nanoparticles. Carbohydr Polym 2018; 208:356-364. [PMID: 30658811 DOI: 10.1016/j.carbpol.2018.12.074] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 12/03/2018] [Accepted: 12/21/2018] [Indexed: 11/22/2022]
Abstract
In recent years, biomacromolecules have been widely used in anti-tumor delivery systems due to the biocompatibility and biodegradability. However, their applications are limited due to the lack of specific targeting. Hyaluronic acid (HA) is a natural polysaccharide and presents in extracellular matrix and synovial fluid which can specifically recognize receptors over-expressed by tumor cells. In addition, they can self-assemble into nanoparticles. HA nanoparticles provide new hierarchical targeting strategies: passively targeting tumor tissue by enhanced permeability and retention effect, actively targeting tumor cells by cluster determinant 44 (CD44) receptor, and then entering cells through receptor-mediated endocytosis. In this review, the synthesis of HA nanoparticles is described in detail from several aspects and applications are also discussed for improving the delivery of hydrophobic drugs, nucleic acids and photosensitizers into the tumor cells. In addition, the modification of HA for improving the targeting and drug releasing characteristics are also discussed.
Collapse
|
22
|
Jin X, Asghar S, Zhang M, Chen Z, Huang L, Ping Q, Xiao Y. N-acetylcysteine modified hyaluronic acid-paclitaxel conjugate for efficient oral chemotherapy through mucosal bioadhesion ability. Colloids Surf B Biointerfaces 2018; 172:655-664. [DOI: 10.1016/j.colsurfb.2018.09.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022]
|
23
|
Su Y, Liu Y, Xu X, Zhou J, Xu L, Xu X, Wang D, Li M, Chen K, Wang W. On-Demand Versatile Prodrug Nanomicelle for Tumor-Specific Bioimaging and Photothermal-Chemo Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:38700-38714. [PMID: 30360090 DOI: 10.1021/acsami.8b11349] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Photothermal therapy is a promising approach for antitumor application although regrettably restricted by available photothermal agents. Physical entrapment of organic near-infrared dyes into nanosystems was extensively studied to reverse the dilemma. However, problems still remained, such as drug bursting and leakage. We developed here an amphiphilic prodrug conjugate by chemically modifying indocyanine green derivative (ICG-COOH) and paclitaxel (PTX) to hyaluronic acid (HA) backbone for integration of photothermal-chemotherapy and specific tumor imaging. The prepared ICG-HA-PTX conjugates could self-assemble into nanomicelles to improve the stability and reduce systemic toxicity of the therapeutic agents. The high local concentration of ICG-COOH in nanomicelles resulted in fluorescence self-quenching, leading to no fluorescence signal being detected in circulation. When the nanomicelles reached the tumor site via electron paramagnetic resonance effect and HA-mediated active targeting, the overexpressed esterase in tumor cells ruptured the ester linkage between drugs and HA, achieving tumor-targeted therapy and specific imaging. A series of in vitro and in vivo experiments demonstrated that the easily prepared ICG- HA-PTX nanomicelles with high stability, smart release behavio r, and excellent tumor targeting ability showed formidable synergy in tumor inhibition, which provided new thoughts in developing an organic near-infrared-dye-based multifunctional delivery system for tumor theranostics.
Collapse
Affiliation(s)
- Yujie Su
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Yuan Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Xiangting Xu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Lin Xu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Xiaole Xu
- Department of Pharmacology , Nantong University Pharmacy College , Nantong 226000 , China
| | - Dun Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Min Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Kerong Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , China
| |
Collapse
|
24
|
Evaluation of the PEG Density in the PEGylated Chitosan Nanoparticles as a Drug Carrier for Curcumin and Mitoxantrone. NANOMATERIALS 2018; 8:nano8070486. [PMID: 29966380 PMCID: PMC6071138 DOI: 10.3390/nano8070486] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 12/11/2022]
Abstract
Polyethylene glycolated (PEGylated)curcumin-grafted-chitosan (PCC) conjugates were synthesized with three PEG/chitosan feed molar ratios (1/5, 1/7.5, and 1/10), namely PCC1, PCC2 and PCC3. Chemical structures of these conjugates were characterized by Fourier transform infrared (FTIR) and proton nuclear magnetic resonance (¹H NMR). The degrees of substitution (DS) of PEG were 0.75%, 0.45% and 0.33%, respectively, for PCC1, PCC2 and PCC3by ¹H NMR analysis. Self-assembled PCC nanoparticles (NPs) were spherical as observed in transmission electron microscope images. Mitoxantrone (MTO)-loaded PCC NPs were prepared to analyze the particle size, zeta potential, drug loading, drug release and in vitro cytotoxicity. The MTO-loaded PCC3 NP (DS = 0.33%) possessed the smallest size (~183.1 nm), highest zeta potential (~+34.0 mV) and the largest loading capacity of curcumin (CUR, ~16.1%) and MTO (~8.30%). The release results showed that MTO-loaded PCC3 NP demonstrated the lowest percentage of MTO release and increased as pH decreased, but the CUR release could only be detected at pH 4.0. In the cytotoxicity study, MTO-loaded PCC3 NP displayed the highest cytotoxicity in HepG2 cell line and the best synergistic effect among the tested NPs. Our results suggest that the DS of PEG has impacts on the structures and functions of PCC NPs: the smaller DS of PEG was associated with the smaller size, the higher zeta potential, the slower drug release, and the higher cytotoxicity of NPs.
Collapse
|
25
|
Lv Y, Zhao X, Zhu L, Li S, Xiao Q, He W, Yin L. Targeting intracellular MMPs efficiently inhibits tumor metastasis and angiogenesis. Am J Cancer Res 2018; 8:2830-2845. [PMID: 29774078 PMCID: PMC5957012 DOI: 10.7150/thno.23209] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 02/27/2018] [Indexed: 12/21/2022] Open
Abstract
Treatment for metastatic cancer is a great challenge throughout the world. Commonly, directed inhibition of extracellular matrix metalloproteinases (MMPs) secreted by cancer cells can reduce metastasis. Here, a novel nanoplatform (HPMC NPs) assembled from hyaluronic acid (HA)-paclitaxel (PTX) prodrug and marimastat (MATT)/β-casein (CN) complexes was established to cure a 4T1 metastatic cancer model via targeting CD44 and intracellular, rather than extracellular, MMPs. Methods: HPMC NPs were prepared by assembling the complexes and prodrug under ultrasonic treatment, which the interaction between them was evaluated by förster resonance energy transfer, circular dichroism and fluorescence spectra. The developed nanoplatform was characterized via dynamic light scattering and transmission electron microscopy, and was evaluated in terms of MMP-sensitive release and stability. Subsequently, the cellular uptake, trafficking, and in vitro invasion were studied by flow cytometry, confocal laser microscopy and transwell assay. MMP expression and activity was determined by western blotting and gelatin zymography. Finally, the studies of biodistribution and antitumor efficacy in vivo were performed in a mouse 4T1 tumor breast model, followed by in vivo safety study in normal mouse. Results: The interaction between the prodrug and complexes is strong with a high affinity, resulting in the assembly of these two components into hybrid nanoparticles (250 nm). Compared with extracellular incubation with MATT, HPMC NP treatment markedly reduced the expression (100%) and activity (50%) of MMPs in 4T1 cells and in the tumor. HPMC NPs exhibited 1.4-fold tumor accumulation, inhibited tumor-growth by >8-fold in volume with efficient apoptosis and proliferation, and suppressed metastasis (>5-fold) and angiogenesis (>3-fold). Overall, HPMC NPs were efficient in metastatic cancer therapy. Conclusions: According to the assembly of polymer prodrug and protein-drug complexes, this study offers a new strategy for constructing nanoparticles for targeted drug delivery, biomedical imaging, and combinatorial treatment. Importantly, via inhibition of intracellular MMPs, metastasis and angiogenesis can be potently blocked, benefiting the rational design of nanomedicine for cancer treatment.
Collapse
|
26
|
Lv Y, Xu C, Zhao X, Lin C, Yang X, Xin X, Zhang L, Qin C, Han X, Yang L, He W, Yin L. Nanoplatform Assembled from a CD44-Targeted Prodrug and Smart Liposomes for Dual Targeting of Tumor Microenvironment and Cancer Cells. ACS NANO 2018; 12:1519-1536. [PMID: 29350904 DOI: 10.1021/acsnano.7b08051] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The tumor microenvironment (TME) plays a critical role in tumor initiation, progression, invasion, and metastasis. Therefore, a therapy that combines chemotherapeutic drugs with a TME modulator could be a promising route for cancer treatment. This paper reports a nanoplatform self-assembled from a hyaluronic acid (HA)-paclitaxel (PTX) (HA-PTX) prodrug and marimastat (MATT)-loaded thermosensitive liposomes (LTSLs) (MATT-LTSLs) for the dual targeting of the TME and cancer cells. Interestingly, the prodrug HA-PTX can self-assemble on both positively and negatively charged liposomes, forming hybrid nanoparticles (HNPs, 100 nm). Triggered by mild hyperthermia, HA-PTX/MATT-LTSLs HNPs rapidly release their payloads into the extracellular environment, and the released HA-PTX quickly enters 4T1 cells through a CD44-HA affinity. The HNPs possess promoted tumor accumulation (1.6-fold), exhibit deep tumor penetration, and significantly inhibit the tumor growth (10-fold), metastasis (100%), and angiogenesis (10-fold). Importantly, by targeting the TME and maintaining its integrity via inhibiting the expression and activity of matrix metalloproteinases (>5-fold), blocking the fibroblast activation by downregulating the TGF-β1 expression (5-fold) and suppressing the degradation of extracellular matrix, the HNPs allow for significant metastasis inhibition. Overall, these findings indicate that a prodrug of an HA-hydrophobic-active compound and liposomes can be self-assembled into a smart nanoplatform for the dual targeting of the TME and tumor cells and efficient combined treatment; additionally, the co-delivery of MATT and HA-PTX with the HNPs is a promising approach for the treatment of metastatic cancer. This study creates opportunities for fabricating multifunctional nanodevices and offers an efficient strategy for disease therapy.
Collapse
Affiliation(s)
- Yaqi Lv
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chaoran Xu
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xiangmei Zhao
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chenshi Lin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xin Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xiaofei Xin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Li Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chao Qin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Xiaopeng Han
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Lei Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Lifang Yin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University , Nanjing 210009, P.R. China
| |
Collapse
|
27
|
Farah S, Domb AJ. Crystalline paclitaxel coated DES with bioactive protective layer development. J Control Release 2018; 271:107-117. [PMID: 29289571 DOI: 10.1016/j.jconrel.2017.12.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/29/2017] [Accepted: 12/27/2017] [Indexed: 01/01/2023]
Abstract
Drug eluting stents (DES) based on polymeric-carriers currently lead the market, however, reports on clinical complications encourage the development of safer and more effective DES. We recently reported on carrier-free DES based on rapamycin crystalline coating as a potential therapeutic solution. Here, we report for the first time surface crystallization of paclitaxel (PT) onto metallic stents. The physicochemical principles of crystallization and key process parameters were extensively studied for fabrication of controllable and homogeneous crystalline coatings on stent scaffolds. Stents loaded with nearly 100μg PT were chosen as a potential therapeutic device with a multilayer coating of 4-7μm thickness. In vitro PT release from these coated stents shows constant release for at least 28days with 10% cumulatively released. The effect of fast dissolving top coating on the physical stability of the coated stent was determined. The top coating enhances the mechanical stability of the crystalline coating during deployment and expansion simulations. Also, incorporating PT in the protective top coating for developing bioactive top coating for multilayer controlled release purpose was intensively studied. This process has wide applications that can be further implemented for other drugs for effective local drug delivery from implantable medical devices.
Collapse
Affiliation(s)
- Shady Farah
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, Center for Nanoscience and Nanotechnology and The Alex Grass Center for Drug Design and Synthesis, The Hebrew University of Jerusalem, 91120, Israel.
| | - Abraham J Domb
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, Center for Nanoscience and Nanotechnology and The Alex Grass Center for Drug Design and Synthesis, The Hebrew University of Jerusalem, 91120, Israel.
| |
Collapse
|
28
|
Fan Y, Wang Q, Lin G, Shi Y, Gu Z, Ding T. Combination of using prodrug-modified cationic liposome nanocomplexes and a potentiating strategy via targeted co-delivery of gemcitabine and docetaxel for CD44-overexpressed triple negative breast cancer therapy. Acta Biomater 2017; 62:257-272. [PMID: 28859899 DOI: 10.1016/j.actbio.2017.08.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/18/2017] [Accepted: 08/27/2017] [Indexed: 12/31/2022]
Abstract
In this study, novel prodrug-modified cationic liposome nanocomplexes (Combo NCs) were reported for gemcitabine (GEM) and docetaxel (DTX) co-delivery. This nanoplatform exhibited multiple favorable characteristics, such as a 'green' fabrication with a one-step chemical reaction, appropriate size (∼200nm) and distribution (PDI<0.2), low zeta potential (-31.1mv), high drug-loading efficiency (9.3% GEM plus 3.1% DTX, wt%) and pH and enzymatic dual-stimulus-responsive release properties. Immunofluorescence and cellular uptake studies showed that Combo NCs efficiently targeted overexpressed CD44 in MDA-MB-231 carcinoma. In vitro studies revealed that Combo NCs played a critical role in the synergistic induction of cytotoxicity, apoptosis and inhibition of wound healing. Combo NCs were confirmed to exhibit great potency for increasing S phase arrest and remodeling the CDA and dCK balance by decreasing the mRNA expression of CDA down to 0.09-fold and increasing the mRNA expression of dCK by 1.36-fold, remarkably increasing the dCK/CDA ratio to 15.3-fold compared with the blank control. The biodistribution results obtained in vivo revealed an effective accumulation in tumor foci. All of these advantages of Combo NCs contributed to their remarkable anti-tumor efficacy without systemic toxicity as well as their apoptosis-enhancing and anti-proliferative capacities, as determined by TUNEL and Ki67 immunohistochemistry in vivo. Consequently, such a rationally contemplated co-delivery system demonstrated the promising potential of clinical applications for triple-negative breast cancer therapy. STATE OF SIGNIFICANCE The Combo NCs were innovatively applied for co-delivery of hydrophilic GEM and hydrophobic DTX. The ester bond linking and shielding effect of HA-GEM made the carriers achieve synchronous release properties, which was determined in in vitro release study. Due to the HA modification, the vectors own great potency for positive targeting to CD44 overexpressed triple-negative breast cancer cells MDA-MB-231. Cytotoxicity and apoptosis studies confirmed the targeting effect and synergism between two drugs. Interestingly, we found in cell cycle study, drug combinations (free combination or Combo NCs) didn't show a rise in G2M phase, which was significantly higher when treated DTX alone. We further discovered the role of DTX in combinations may involve in modulating GEM associated enzymes thus enhancing the efficacy of GEM. Consequently, this nanoplatform provided a novel solution for achieving targeted co-delivery and potentiating effect in cancer therapy.
Collapse
|
29
|
Liu Y, Zhou C, Wang W, Yang J, Wang H, Hong W, Huang Y. CD44 Receptor Targeting and Endosomal pH-Sensitive Dual Functional Hyaluronic Acid Micelles for Intracellular Paclitaxel Delivery. Mol Pharm 2016; 13:4209-4221. [PMID: 27796093 DOI: 10.1021/acs.molpharmaceut.6b00870] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A novel CD44 receptor targeting and endosome pH-sensitive dual functional hyaluronic acid-deoxycholic acid-histidine (HA-DOCA-His) micellar system was designed for intracellular paclitaxel (PTX) delivery. The HA-DOCA-His micelles exhibited desirable endosome pH (5.0-6.0)-induced aggregation and deformation behavior verified by size distribution, critical micellar concentration, and zeta potential changes. The HA-DOCA-His micelles presented excellent encapsulation efficiency and loading capacity of 90.0% and 18.9% for PTX, respectively. The PTX release from HA-DOCA-His micelles was pH-dependent, with more rapid PTX release at pH 6.0 and 5.0 than those at pH 7.4 and 6.5. The cellular uptake performance of HA-DOCA-His micelles was enhanced comparing with pH-insensitive HA-DOCA micelles by qualitative and quantitative measurements. HA-DOCA-His micelles could be taken up via CD44-receptor mediated endocytosis, transported into endosomes, and triggered drug release to cytoplasm. In vitro cytotoxicity study exhibited PTX-loaded HA-DOCA-His micelles were more active in tumor cell growth inhibition in MCF-7 cells at pH 5.8 than those at pH 6.8 and pH 7.4. A superior antitumor efficacy was demonstrated with HA-DOCA-His micelles in a MCF-7 breast tumor model. These indicated that the dual functional HA-DOCA-His micelles combined targeted intracellular delivery and endosomal release strategies could be developed as a promising nanocarrier for anticancer efficacy improvement of PTX.
Collapse
Affiliation(s)
- Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University , No. 1160, Shengli Street, Yinchuan 750004, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University , Yinchuan 750004, China
| | - Chengming Zhou
- Department of Pharmacy, Tumor Hospital of General Hospital, Ningxia Medical University , Yinchuan 750004, China
| | - Wenping Wang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University , No. 1160, Shengli Street, Yinchuan 750004, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University , Yinchuan 750004, China
| | - Jianhong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University , No. 1160, Shengli Street, Yinchuan 750004, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University , Yinchuan 750004, China
| | - Hao Wang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University , No. 1160, Shengli Street, Yinchuan 750004, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University , Yinchuan 750004, China
| | - Wei Hong
- School of Chemistry and Chemical Engineering, Beifang University of Nationalities , Yinchuan 750021, China
| | - Yu Huang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University , No. 1160, Shengli Street, Yinchuan 750004, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University , Yinchuan 750004, China
| |
Collapse
|
30
|
Yu D, Li W, Zhang Y, Zhang B. Anti-tumor efficiency of paclitaxel and DNA when co-delivered by pH responsive ligand modified nanocarriers for breast cancer treatment. Biomed Pharmacother 2016; 83:1428-1435. [PMID: 27592131 DOI: 10.1016/j.biopha.2016.08.061] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/16/2016] [Accepted: 08/24/2016] [Indexed: 12/13/2022] Open
Abstract
CONTEXT Combination of chemotherapy and nucleic acid therapy generally take advantage of drugs anti-tumor activity together with DNA capacity to transfect cancer cells, showing great promise in cancer treatment. However, effective co-delivery of drugs and DNA in a single carrier for cancer treatment remains a challenge. OBJECTIVE This study aimed to design a tumor targeted, pH sensitive nanocarriers for the co-delivery of gene and drug. MATERIALS AND METHODS Hyaluronic acid - acid sensitive linker - 1,2-distearoyl phosphatideylethanolamine copolymers (HA-as-DSPE) were synthesized. HA-as-DSPE modified, paclitaxel and pDNA loaded solid lipid nanoparticles (HA-PTX/pDNA SLN) was prepared. The physicochemical properties like morphology, size, and zeta potential as well as release properties were evaluated. The ability and therapeutic effects of the novel system for the co-delivery of PTX and pDNA were demonstrated in vitro and in vivo. RESULTS In vitro experiments and in vivo animal studies both confirmed that the HA-PTX/pDNA SLN system could promote the inhibition of tumor, at the same time deliver and transfect gene into the cancer cells. DISCUSSION AND CONCLUSION Highest efficiency achieved by HA-PTX/pDNA SLN might result from the HA ligands that targeted the receptors on the cancer cells, the enhanced cellular uptake by the SLN formulations and also the pH sensitive bound of the carriers let the drug release more in the tumor cells. It could be concluded that HA-PTX/pDNA SLN could be used as a promising delivery system for drug and gene combination therapy.
Collapse
Affiliation(s)
- DongMei Yu
- Department of Public Health, Shandong Jining No. 1 People's Hospital, Jining, 272011, Shandong, PR China
| | - Wei Li
- Department of Science Research, Shandong Jining No. 1 People's Hospital, Jining, 272011, Shandong, PR China
| | - Yueying Zhang
- Department of Experimental Pathology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, Shandong, PR China
| | - Bin Zhang
- Department of Oncology, Shandong Jining No. 1 People's Hospital, Jining, 272011, Shandong, PR China.
| |
Collapse
|
31
|
Cadete A, Alonso MJ. Targeting cancer with hyaluronic acid-based nanocarriers: recent advances and translational perspectives. Nanomedicine (Lond) 2016; 11:2341-57. [PMID: 27526874 DOI: 10.2217/nnm-2016-0117] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Hyaluronic acid is a natural polysaccharide that has been widely explored for the development of anticancer therapies due to its ability to target cancer cells. Moreover, advances made in the last decade have revealed the versatility of this biomaterial in the design of multifunctional carriers, intended for the delivery of a variety of bioactive molecules, including polynucleotides, immunomodulatory drugs and imaging agents. In this review, we aim to provide an overview of the major recent achievements in this field, highlighting the application of the newly developed nanostructures in combination therapies, immunomodulation and theranostics. Finally, we will discuss the main challenges and technological advances that will allow these carriers to be considered as candidates for clinical development.
Collapse
Affiliation(s)
- Ana Cadete
- NanoBioFar Group, Center for Research in Molecular Medicine & Chronic Diseases, Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacy & Pharmaceutical Technology, School of Pharmacy, Campus Vida, University of Santiago de Compostela (USC), Avenida Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - María José Alonso
- NanoBioFar Group, Center for Research in Molecular Medicine & Chronic Diseases, Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacy & Pharmaceutical Technology, School of Pharmacy, Campus Vida, University of Santiago de Compostela (USC), Avenida Barcelona s/n, 15782 Santiago de Compostela, Spain
| |
Collapse
|
32
|
Pro-haloacetate Nanoparticles for Efficient Cancer Therapy via Pyruvate Dehydrogenase Kinase Modulation. Sci Rep 2016; 6:28196. [PMID: 27323896 PMCID: PMC4914936 DOI: 10.1038/srep28196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/31/2016] [Indexed: 12/21/2022] Open
Abstract
Anticancer agents based on haloacetic acids are developed for inhibition of pyruvate dehydrogenase kinase (PDK), an enzyme responsible for reversing the suppression of mitochondria-dependent apoptosis. Through molecular docking studies mono- and dihaloacetates are identified as potent PDK2 binders and matched their efficiency with dichloroacetic acid. In silico screening directed their conversion to phospholipid prodrugs, which were subsequently self-assembled to pro-haloacetate nanoparticles. Following a thorough physico-chemical characterization, the functional activity of these novel agents was established in wide ranges of human cancer cell lines in vitro and in vivo in rodents. Results indicated that the newly explored PDK modulators can act as efficient agent for cancer regression. A Pyruvate dehydrogenase (PDH) assay mechanistically confirmed that these agents trigger their activity through the mitochondria-dependent apoptosis.
Collapse
|
33
|
Dheer D, Arora D, Jaglan S, Rawal RK, Shankar R. Polysaccharides based nanomaterials for targeted anti-cancer drug delivery. J Drug Target 2016; 25:1-16. [DOI: 10.3109/1061186x.2016.1172589] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
34
|
Mogoşanu GD, Grumezescu AM, Bejenaru C, Bejenaru LE. Polymeric protective agents for nanoparticles in drug delivery and targeting. Int J Pharm 2016; 510:419-29. [PMID: 26972379 DOI: 10.1016/j.ijpharm.2016.03.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 03/09/2016] [Indexed: 01/08/2023]
Abstract
Surface modification/functionalization of nanoparticles (NPs) using polymeric protective agents is an issue of great importance and actuality for drug delivery and targeting. Improving the blood circulation half-life of surface-protected nanocarriers is closely related to the elimination of main biological barriers and limiting factors (protein absorption and opsonization), due to the phagocytic activity of reticuloendothelial system. For passive or active targeted delivery, in biomedical area, surface-functionalized NPs with tissue-recognition ligands were designed and optimized as a result of modern research techniques. Also, multi-functionalized nanostructures are characterized by enhanced bioavailability, efficacy, targeted localization, active cellular uptake, and low side effects. Surface-protected NPs are obtained from biocompatible, biodegradable and less toxic natural polymers (dextran, β-cyclodextrin, chitosan, hyaluronic acid, heparin, gelatin) or synthetic polymers, such as poly(lactic acid), poly(lactic-co-glycolic) acid, poly(ε-caprolactone) and poly(alkyl cyanoacrylates). PEGylation is one of the most important functionalization methods providing steric stabilization, long circulating and 'stealth' properties for both polymeric and inorganic-based nanosystems. In addition, for their antimicrobial, antiviral and antitumor effects, cutting-edge researches in the field of pharmaceutical nanobiotechnology highlighted the importance of noble metal (platinum, gold, silver) NPs decorated with biopolymers.
Collapse
Affiliation(s)
- George Dan Mogoşanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxidic Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania.
| | - Cornelia Bejenaru
- Department of Vegetal & Animal Biology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Ludovic Everard Bejenaru
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| |
Collapse
|
35
|
Yi X, Zhao D, Zhang Q, Xu J, Yuan G, Zhuo R, Li F. A co-delivery system based on a reduction-sensitive polymeric prodrug capable of loading hydrophilic and hydrophobic drugs for combination chemotherapy. Polym Chem 2016. [DOI: 10.1039/c6py00900j] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A reduction-sensitive polymeric prodrug PEG-b-PMPMC-g-PTX was designed. The self-assemblies of polymeric prodrug could deliver drugs with different action mechanisms into tumor cells, leading to the apoptosis of tumor cells effectively.
Collapse
Affiliation(s)
- Xiaoqing Yi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Dan Zhao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Quan Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Jiaqi Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Gongdao Yuan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Renxi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Feng Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| |
Collapse
|