1
|
Li C, Zhou L, Yin X. Pathophysiological aspects of transferrin-A potential nano-based drug delivery signaling molecule in therapeutic target for varied diseases. Front Pharmacol 2024; 15:1342181. [PMID: 38500764 PMCID: PMC10944884 DOI: 10.3389/fphar.2024.1342181] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/23/2024] [Indexed: 03/20/2024] Open
Abstract
Transferrin (Tf), widely known for its role as an iron-binding protein, exemplifies multitasking in biological processes. The role of Tf in iron metabolism involves both the uptake of iron from Tf by various cells, as well as the endocytosis mediated by the complex of Tf and the transferrin receptor (TfR). The direct conjugation of the therapeutic compound and immunotoxin studies using Tf peptide or anti-Tf receptor antibodies as targeting moieties aims to prolong drug circulation time and augment efficient cellular drug uptake, diminish systemic toxicity, traverse the blood-brain barrier, restrict systemic exposure, overcome multidrug resistance, and enhance therapeutic efficacy with disease specificity. This review primarily discusses the various biological actions of Tf, as well as the development of Tf-targeted nano-based drug delivery systems. The goal is to establish the use of Tf as a disease-targeting component, accentuating the potential therapeutic applications of this protein.
Collapse
Affiliation(s)
- Chang Li
- Basic Medical College, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Liya Zhou
- Basic Medical College, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xunzhe Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
2
|
Mahdinloo S, Hemmati S, Valizadeh H, Mahmoudian M, Mahmoudi J, Roshangar L, Sarfraz M, Zakeri-Milani P. Synthesis and preparation of vitamin A coupled butein-loaded solid lipid nanoparticles for liver fibrosis therapy in rats. Int J Pharm 2022; 625:122063. [PMID: 35964827 DOI: 10.1016/j.ijpharm.2022.122063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022]
Abstract
The development of a therapeutic system for hepatic fibrosis has become a research hotspot to date. Butein, a simple chalcone derivative, displays anti-fibrotic effects through different pathways. However, impurities, low solubility, and low concentration in the target tissue hinder therapy with herbal ingredients. Hepatic stellate cells (HSCs), the vitamin A (VA) storage cells, as the main contributors to liver fibrogenesis, are not readily accessible to drugs owing to their anatomical location. Targeted delivery of therapeutics to the activated HSCs is therefore critical for successful treatment. For these reasons, the current study aimed at increasing butein delivery to the liver. Hence, high purity butein was synthesized in three steps. A novel VA-Myrj52 ester conjugate was also synthesized using all-trans retinoic acid and a hydrophilic emulsifier (Myrj52) as a targeting agent. Next, butein was encapsulated inside the novel VA-modified solid lipid nanoparticles (VA-SLNs) and studied in vitro and in vivo. According to our evaluations, negatively charged SLNs with a mean diameter of 150 nm and entrapment efficacy of 75 % were successful in liver fibrosis amelioration. Intraperitoneal (i.p.) injection of VA-SLNs in fibrotic rats, for four weeks long, reduced serum AST and ALT by 58% (P, 0.001) and 72% (P, 0.05), respectively, concerning the CCl4 group. Additionally, histologic damage score decline and normalization of tissue oxidative stress markers collectively confirmed the efficacy of formulations in hepatic fibrosis and kidney damage amelioration.
Collapse
Affiliation(s)
- Somayeh Mahdinloo
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran.
| | - Mohammad Mahmoudian
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical sciences, Tabriz 5166614756, Iran
| | - Leyla Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain 64141, United Arab Emirates.
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran.
| |
Collapse
|
3
|
Peng Z, Yuan L, XuHong J, Tian H, Zhang Y, Deng J, Qi X. Chiral nanomaterials for tumor therapy: autophagy, apoptosis, and photothermal ablation. J Nanobiotechnology 2021; 19:220. [PMID: 34294083 PMCID: PMC8299636 DOI: 10.1186/s12951-021-00965-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/16/2021] [Indexed: 01/08/2023] Open
Abstract
Chirality is a fundamental characteristic of natural molecules and a crucial factor in the biochemical reactions of living cells and organisms. Recently, researchers have successfully introduced chiral molecules to the surfaces of nanomaterials, creating chiral nanomaterials that exhibit an upscaling of chiral behavior from the molecular scale to the nanoscale. These chiral nanomaterials can selectively induce autophagy, apoptosis, and photothermal ablation in tumor cells based on their chirality, making them promising for application in anti-tumor therapy. However, these interesting and important phenomena have hitherto received little attention. Accordingly, we herein present a review of recent research progress in the field of chiral nanomaterials for tumor therapy along with brief looks at the mechanistic details of their actions. Finally, the current challenges and future perspectives of chiral nanomaterials in terms of maximizing their potential in tumor therapy are discussed. Thus, this review provides a helpful introduction to the design of chiral nanomaterials and will hopefully highlight the importance of chirality in tumor therapy. ![]()
Collapse
Affiliation(s)
- Zaihui Peng
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Long Yuan
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Juncheng XuHong
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Hao Tian
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Yi Zhang
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400038, China.
| | - Xiaowei Qi
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
4
|
Cano-Garrido O, Álamo P, Sánchez-García L, Falgàs A, Sánchez-Chardi A, Serna N, Parladé E, Unzueta U, Roldán M, Voltà-Durán E, Casanova I, Villaverde A, Mangues R, Vázquez E. Biparatopic Protein Nanoparticles for the Precision Therapy of CXCR4 + Cancers. Cancers (Basel) 2021; 13:2929. [PMID: 34208189 PMCID: PMC8230831 DOI: 10.3390/cancers13122929] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 01/05/2023] Open
Abstract
The accumulated molecular knowledge about human cancer enables the identification of multiple cell surface markers as highly specific therapeutic targets. A proper tumor targeting could significantly avoid drug exposure of healthy cells, minimizing side effects, but it is also expected to increase the therapeutic index. Specifically, colorectal cancer has a particularly poor prognosis in late stages, being drug targeting an appropriate strategy to substantially improve the therapeutic efficacy. In this study, we have explored the potential of the human albumin-derived peptide, EPI-X4, as a suitable ligand to target colorectal cancer via the cell surface protein CXCR4, a chemokine receptor overexpressed in cancer stem cells. To explore the potential use of this ligand, self-assembling protein nanoparticles have been generated displaying an engineered EPI-X4 version, which conferred a modest CXCR4 targeting and fast and high level of cell apoptosis in tumor CXCR4+ cells, in vitro and in vivo. In addition, when EPI-X4-based building blocks are combined with biologically inert polypeptides containing the CXCR4 ligand T22, the resulting biparatopic nanoparticles show a dramatically improved biodistribution in mouse models of CXCR4+ human cancer, faster cell internalization and enhanced target cell death when compared to the version based on a single ligand. The generation of biparatopic materials opens exciting possibilities in oncotherapies based on high precision drug delivery based on the receptor CXCR4.
Collapse
Affiliation(s)
- Olivia Cano-Garrido
- Nanoligent SL, Edifici EUREKA, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Patricia Álamo
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Laura Sánchez-García
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Aïda Falgàs
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Alejandro Sánchez-Chardi
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain
- Servei de Microscòpia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Naroa Serna
- Nanoligent SL, Edifici EUREKA, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Mònica Roldán
- Unitat de Microscòpia Confocal i Imatge Cel·lular, Servei de Medicina Genètica i Molecular, Institut Pediàtric de Malalties Rares (IPER), Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Isolda Casanova
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| |
Collapse
|
5
|
Current Status and Challenges Associated with CNS-Targeted Gene Delivery across the BBB. Pharmaceutics 2020; 12:pharmaceutics12121216. [PMID: 33334049 PMCID: PMC7765480 DOI: 10.3390/pharmaceutics12121216] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/19/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
The era of the aging society has arrived, and this is accompanied by an increase in the absolute numbers of patients with neurological disorders, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Such neurological disorders are serious costly diseases that have a significant impact on society, both globally and socially. Gene therapy has great promise for the treatment of neurological disorders, but only a few gene therapy drugs are currently available. Delivery to the brain is the biggest hurdle in developing new drugs for the central nervous system (CNS) diseases and this is especially true in the case of gene delivery. Nanotechnologies such as viral and non-viral vectors allow efficient brain-targeted gene delivery systems to be created. The purpose of this review is to provide a comprehensive review of the current status of the development of successful drug delivery to the CNS for the treatment of CNS-related disorders especially by gene therapy. We mainly address three aspects of this situation: (1) blood-brain barrier (BBB) functions; (2) adeno-associated viral (AAV) vectors, currently the most advanced gene delivery vector; (3) non-viral brain targeting by non-invasive methods.
Collapse
|
6
|
Paliwal SR, Kenwat R, Maiti S, Paliwal R. Nanotheranostics for Cancer Therapy and Detection: State of the Art. Curr Pharm Des 2020; 26:5503-5517. [PMID: 33200696 DOI: 10.2174/1381612826666201116120422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 08/09/2020] [Indexed: 11/22/2022]
Abstract
Nanotheranostics, an approach of combining both diagnosis and therapy, is one of the latest advances in cancer therapy particularly. Nanocarriers designed and derived from inorganic materials such as like gold nanoparticles, silica nanoparticles, magnetic nanoparticles and carbon nanotubes have been explored for tremendous applications in this area. Similarly, nanoparticles composed of some organic material alone or in combination with inorganic nano-cargos have been developed pre-clinically and possess excellent features desired. Photothermal therapy, MRI, simultaneous imaging and delivery, and combination chemotherapy with a diagnosis are a few of the known methods exploring cancer therapy and detection at organ/tissue/molecular/sub-cellular level. This review comprises an overview of the recent reports meant for nano theranostics purposes. Targeted cancer nanotheranostics have been included for understating tumor micro-environment or cell-specific targeting approach employed. A brief account of various strategies is also included for the readers highlighting the mechanism of cancer therapy.
Collapse
Affiliation(s)
- Shivani Rai Paliwal
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilapsur, CG, India
| | - Rameshroo Kenwat
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, MP, India
| | - Sabyasachi Maiti
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, MP, India
| | - Rishi Paliwal
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, MP, India
| |
Collapse
|
7
|
Lactoferrin coated or conjugated nanomaterials as an active targeting approach in nanomedicine. Int J Biol Macromol 2020; 167:1527-1543. [PMID: 33212102 DOI: 10.1016/j.ijbiomac.2020.11.107] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/08/2020] [Accepted: 11/15/2020] [Indexed: 12/28/2022]
Abstract
A successful drug delivery to a specific site relies on two essential factors including; efficient entrapment of the drug within the carrier and successful delivery of drug- loaded nanocarrier to the target site without opsonisation or drug release in the circulation before reaching the organ of interest. Lactoferrin (LF) is a glycoprotein belonging to the transferrin (TF) family which can bind to TF receptors (TFRs) and LF membrane internalization receptors (LFRs) highly expressed on the cell surface of both highly proliferating cancer cells and blood brain barrier (BBB), which in turn can facilitate its accessibility to the cell nucleus. This merit could be exploited to develop actively targeted drug delivery systems that can easily cross the BBB or internalize into tumor cells. In this review, the most recent advances of utilizing LF as an active targeting ligand for different types of nanocarriers including: inorganic nanoparticles, dendrimers, synthetic biodegradable polymers, lipid nanocarriers, natural polymers, and nanoemulstions will be highlighted. Collectively, LF seems to be a promising targeting ligand in the field of nanomedicine.
Collapse
|
8
|
Chen X, Hu X, Hu J, Qiu Z, Yuan M, Zheng G. Celastrol-Loaded Galactosylated Liposomes Effectively Inhibit AKT/c-Met-Triggered Rapid Hepatocarcinogenesis in Mice. Mol Pharm 2020; 17:738-747. [PMID: 31904241 DOI: 10.1021/acs.molpharmaceut.9b00428] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our previous study proved that celastrol was a potential candidate for hepatocellular carcinoma (HCC) therapy. However, poor water solubility and toxic side effects may restrict its clinical application. To overcome these shortcomings and optimize its antitumor efficacy, we developed galactosylated liposomes using galactose-modified 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) to deliver celastrol (C-GPL). C-GPL improved the water solubility of celastrol and exhibited high encapsulation efficiency, good stability in serum, and slow drug release profile. In vitro studies showed that C-GPL increased the cellular uptake of celastrol through receptor-mediated endocytosis, thereby enhancing celastrol cytotoxicity and cancer cell apoptosis. Particularly, in vivo antitumor activity of C-GPL was assessed in rapid HCC mouse models established via hydrodynamic transfection of the activated forms of AKT and c-Met. Compared to free celastrol, C-GPL significantly prevented liver weight gain, decreased liver damage biomarkers (glutamic-oxalacetic transaminase and alanine aminotransferase) and HCC marker (alpha-fetoprotein), and led to tumor disappearance on the liver surface. The improved therapeutic effect of C-GPL may be attributed to suppression of AKT activation, induction of apoptosis, and retardation of cell proliferation. Importantly, C-GPL exerted low toxicity to normal tissues without causing severe weight loss in mice. Taken together, C-GPL may become a promising drug delivery system for HCC treatment.
Collapse
Affiliation(s)
- Xinyan Chen
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xianxian Hu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Junjie Hu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Zhenpeng Qiu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Yuan
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Guohua Zheng
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
9
|
Targeting Small Molecule Delivery to the Brain and Spinal Cord via Intranasal Administration of Rabies Virus Glycoprotein (RVG29)-Modified PLGA Nanoparticles. Pharmaceutics 2020; 12:pharmaceutics12020093. [PMID: 31991664 PMCID: PMC7076461 DOI: 10.3390/pharmaceutics12020093] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 02/07/2023] Open
Abstract
Alternative routes of administration are one approach that could be used to bypass the blood–brain barrier (BBB) for effective drug delivery to the central nervous system (CNS). Here, we focused on intranasal delivery of polymer nanoparticles. We hypothesized that surface modification of poly(lactic-co-glycolic acid) (PLGA) nanoparticles with rabies virus glycoprotein (RVG29) would increase residence time and exposure of encapsulated payload to the CNS compared to non-targeted nanoparticles. Delivery kinetics and biodistribution were analyzed by administering nanoparticles loaded with the carbocyanine dye 1,1′-Dioctadecyl-3,3,3′,3′-Tetramethylindotricarbocyanine Iodide (DiR) to healthy mice. Intranasal administration yielded minimal exposure of nanoparticle payload to most peripheral organs and rapid, effective delivery to whole brain. Regional analysis of payload delivery within the CNS revealed higher delivery to tissues closest to the trigeminal nerve, including the olfactory bulb, striatum, midbrain, brainstem, and cervical spinal cord. RVG29 surface modifications presented modest targeting benefits to the striatum, midbrain, and brainstem 2 h after administration, although targeting was not observed 30 min or 6 h after administration. Payload delivery to the trigeminal nerve was 3.5× higher for targeted nanoparticles compared to control nanoparticles 2 h after nanoparticle administration. These data support a nose-to-brain mechanism of drug delivery that closely implicates the trigeminal nerve for payload delivery from nanoparticles via transport of intact nanoparticles and eventual diffusion of payload. Olfactory and CSF routes are also observed to play a role. These data advance the utility of targeted nanoparticles for nose-to-brain drug delivery of lipophilic payloads and provide mechanistic insight to engineer effective delivery vectors to treat disease in the CNS.
Collapse
|
10
|
Shrivastava P, Vyas S, Sharma R, Mody N, Gautam L, Jain A, Vishwakarma N, Vyas SP. Nanotechnology for oral drug delivery and targeting. NANOENGINEERED BIOMATERIALS FOR ADVANCED DRUG DELIVERY 2020:473-498. [DOI: 10.1016/b978-0-08-102985-5.00020-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Brain-targeted drug delivery by manipulating protein corona functions. Nat Commun 2019; 10:3561. [PMID: 31395892 PMCID: PMC6687821 DOI: 10.1038/s41467-019-11593-z] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/23/2019] [Indexed: 12/21/2022] Open
Abstract
Protein corona presents a major obstacle to bench-to-bedside translation of targeted drug delivery systems, severely affecting targeting yields and directing unfavorable biodistribution. Corona-mediated targeting provides a new impetus for specific drug delivery by precisely manipulating interaction modes of functional plasma proteins on nano-surface. Here bio-inspired liposomes (SP-sLip) were developed by modifying liposomal surface with a short nontoxic peptide derived from Aβ1-42 that specifically interacts with the lipid-binding domain of exchangeable apolipoproteins. SP-sLip absorb plasma apolipoproteins A1, E and J, consequently exposing receptor-binding domain of apolipoproteins to achieve brain-targeted delivery. Doxorubicin loaded SP-sLip (SP-sLip/DOX) show significant enhancement of brain distribution and anti-brain cancer effect in comparison to doxorubicin loaded plain liposomes. SP-sLip preserve functions of the absorbed human plasma ApoE, and the corona-mediated targeting strategy works in SP modified PLGA nanoparticles. The present study may pave a new avenue to facilitate clinical translation of targeted drug delivery systems. Plasma proteins may severely affect the in vivo performance of liposomes. Here, the authors develop bio-inspired liposomes that specifically absorb brain-targeted apolipoproteins and preserve their bioactivities, thereby achieving efficient brain targeting with minor influence on immunocompatibility of liposomes.
Collapse
|
12
|
Zhang YD, Huang QW, Ma C, Liu XY, Zhang HX. Magnetic fluorescent molecularly imprinted nanoparticles for detection and separation of transferrin in human serum. Talanta 2018; 188:540-545. [DOI: 10.1016/j.talanta.2018.06.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 11/28/2022]
|
13
|
Yuan L, Zhang F, Qi X, Yang Y, Yan C, Jiang J, Deng J. Chiral polymer modified nanoparticles selectively induce autophagy of cancer cells for tumor ablation. J Nanobiotechnology 2018; 16:55. [PMID: 29996877 PMCID: PMC6040058 DOI: 10.1186/s12951-018-0383-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/02/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Autophagy regulation through exogenous materials has aroused intensive attention to develop treatment protocols according to diverse human diseases. However, to the best of our knowledge, few examples have been reported to selectively control autophagy process and ultimately achieve efficient therapeutic potential. RESULTS In this study, monolayers of poly (acryloyl-L, D and racemic valine) (L-PAV-AuNPs, D-PAV-AuNPs and L/D-PAV-AuNPs) chiral molecules were anchored on the surfaces of gold nanoparticles (PAV-AuNPs), and the subsequent chirality-selective effects on autophagy activation were thoroughly studied. The cytotoxicity induced by PAV-AuNPs towards MDA-MB-231 cells (Breast cancer cells) was achieved mainly through autophagy and showed chirality-dependent, with D-PAV-AuNPs exhibiting high autophagy-inducing activity in vitro and in vivo. In contrast, the PAV-AuNPs exhibited autophagy inactivation for normal cells, e.g., 3T3 fibroblasts and HBL-100 cells. The chirality-selective autophagy activation effect in MDA-MB-231 cells was likely attributed to the chirality-variant ROS generation, cellular uptake and their continuous autophagy stimulus. Furthermore, the intratumoral injection of D-PAV-AuNPs could largely suppress the tumor growth but exhibit negligible toxicity in vivo. CONCLUSIONS As the first exploration on stereospecific NPs for autophagy induction, this work not only substantiates that chiral polymer coated NPs can selective induce autophagy-specific in cancer cells and achieve a high tumor eradication efficiency in vivo, but also opens up a new direction in discovering unprecedented stereospecific nanoagents for autophagy-associated tumor treatment.
Collapse
Affiliation(s)
- Long Yuan
- Department of Breast Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Fan Zhang
- Department of Breast Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Xiaowei Qi
- Department of Breast Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Yongjun Yang
- Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Chang Yan
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Jun Jiang
- Department of Breast Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038 China
| |
Collapse
|
14
|
Qi H, Yang L, Li X, Zhan Q, Han D, Zhao J, Hou X, Yuan X. Exosomes separated based on the “STOP” criteria for tumor-targeted drug delivery. J Mater Chem B 2018; 6:2758-2768. [DOI: 10.1039/c8tb00355f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A new exosome-related drug delivery vehicle was explored based on the “STOP” criteria, dramatically promoting the clinical translation of exosomes.
Collapse
Affiliation(s)
- Hongzhao Qi
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Lijun Yang
- School of Pharmaceutical Science and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Xueping Li
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Qi Zhan
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Donglin Han
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Jin Zhao
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Xin Hou
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Xubo Yuan
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| |
Collapse
|
15
|
Hamman J, Demana P, Olivier E. Targeting Receptors, Transporters and Site of Absorption to Improve Oral Drug Delivery. Drug Target Insights 2017. [DOI: 10.1177/117739280700200003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- J.H. Hamman
- School of Pharmacy, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - P.H. Demana
- School of Pharmacy, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - E.I. Olivier
- School of Pharmacy, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| |
Collapse
|
16
|
Parayath NN, Amiji MM. Therapeutic targeting strategies using endogenous cells and proteins. J Control Release 2017; 258:81-94. [DOI: 10.1016/j.jconrel.2017.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/02/2017] [Accepted: 05/04/2017] [Indexed: 01/14/2023]
|
17
|
Zhu WJ, Yang SD, Qu CX, Zhu QL, Chen WL, Li F, Yuan ZQ, Liu Y, You BG, Zhang XN. Low-density lipoprotein-coupled micelles with reduction and pH dual sensitivity for intelligent co-delivery of paclitaxel and siRNA to breast tumor. Int J Nanomedicine 2017; 12:3375-3393. [PMID: 28490877 PMCID: PMC5413542 DOI: 10.2147/ijn.s126310] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multidrug resistance (MDR) is a major obstacle for the clinical therapy of malignant human cancers. The discovery of RNA interference provides efficient gene silencing within tumor cells for reversing MDR. In this study, a new “binary polymer” low-density lipoprotein–N-succinyl chitosan–cystamine–urocanic acid (LDL–NSC–SS–UA) with dual pH/redox sensitivity and targeting effect was synthesized for the co-delivery of breast cancer resistance protein small interfering RNA (siRNA) and paclitaxel (PTX). In vivo, the co-delivering micelles can accumulate in tumor tissue via the enhanced permeability and retention effect and the specific recognition and combination of LDL and LDL receptor, which is overexpressed on the surface of tumor cell membranes. The siRNA–PTX-loaded micelles inhibited gene and drug release under physiological conditions while promoting fast release in an acid microenvironment or in the presence of glutathione. The micelles escaped from the lysosome through the proton sponge effect. Additionally, the micelles exhibited superior antitumor activity and downregulated the protein and mRNA expression levels of breast cancer resistance protein in MCF-7/Taxol cells. The biodistribution and antitumor studies proved that the siRNA–PTX-loaded micelles possessed prolonged circulation time with a remarkable tumor-targeting effect and effectively inhibited tumor growth. Therefore, the novel dual pH/redox-sensitive polymers co-delivering siRNA and PTX with excellent biocompatibility and effective reversal of MDR demonstrate a considerable potential in cancer therapy.
Collapse
Affiliation(s)
- Wen-Jing Zhu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Shu-di Yang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Chen-Xi Qu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Qiao-Ling Zhu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou.,Department of Clinical Medicine, Nanjing Gulou Hospital, Nanjing, People's Republic of China
| | - Wei-Liang Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Fang Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Zhi-Qiang Yuan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Yang Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Ben-Gang You
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| | - Xue-Nong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou
| |
Collapse
|
18
|
Qi H, Jia H, Sang J, Ren Y, Zhao J, Hou X, Yuan X. Using endogenous ligands for direct superparamagnetic nanoparticle cluster-based body fluid exosome separation. RSC Adv 2017. [DOI: 10.1039/c6ra24937j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Using endogenous ligands to directly separate exosomes from body fluids facilitates the in vivo clinical translation of exosomes.
Collapse
Affiliation(s)
- Hongzhao Qi
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Huanhuan Jia
- Tianjin Research Center of Basic Medical Science
- Tianjin Medical University
- Tianjin 300070
- China
| | - Jimeng Sang
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Yu Ren
- Tianjin Research Center of Basic Medical Science
- Tianjin Medical University
- Tianjin 300070
- China
| | - Jin Zhao
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Xin Hou
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| | - Xubo Yuan
- Tianjin Key Laboratory of Composite and Functional Materials
- School of Materials Science and Engineering
- Tianjin University
- Tianjin 300072
- China
| |
Collapse
|
19
|
Afzal SM, Shareef MZ, Kishan V. Transferrin tagged lipid nanoemulsion of docetaxel for enhanced tumor targeting. J Drug Deliv Sci Technol 2016. [DOI: 10.1016/j.jddst.2016.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Noh JK, Naeem M, Cao J, Lee EH, Kim MS, Jung Y, Yoo JW. Herceptin-functionalized pure paclitaxel nanocrystals for enhanced delivery to HER2-postive breast cancer cells. Int J Pharm 2016; 513:543-553. [DOI: 10.1016/j.ijpharm.2016.09.067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/05/2016] [Accepted: 09/24/2016] [Indexed: 01/16/2023]
|
21
|
Garg T, Bhandari S, Rath G, Goyal AK. Current strategies for targeted delivery of bio-active drug molecules in the treatment of brain tumor. J Drug Target 2015; 23:865-87. [PMID: 25835469 DOI: 10.3109/1061186x.2015.1029930] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Brain tumor is one of the most challenging diseases to treat. The major obstacle in the specific drug delivery to brain is blood-brain barrier (BBB). Mostly available anti-cancer drugs are large hydrophobic molecules which have limited permeability via BBB. Therefore, it is clear that the protective barriers confining the passage of the foreign particles into the brain are the main impediment for the brain drug delivery. Hence, the major challenge in drug development and delivery for the neurological diseases is to design non-invasive nanocarrier systems that can assist controlled and targeted drug delivery to the specific regions of the brain. In this review article, our major focus to treat brain tumor by study numerous strategies includes intracerebral implants, BBB disruption, intraventricular infusion, convection-enhanced delivery, intra-arterial drug delivery, intrathecal drug delivery, injection, catheters, pumps, microdialysis, RNA interference, antisense therapy, gene therapy, monoclonal/cationic antibodies conjugate, endogenous transporters, lipophilic analogues, prodrugs, efflux transporters, direct conjugation of antitumor drugs, direct targeting of liposomes, nanoparticles, solid-lipid nanoparticles, polymeric micelles, dendrimers and albumin-based drug carriers.
Collapse
Affiliation(s)
| | - Saurav Bhandari
- b Department of Quality Assurance , ISF College of Pharmacy , Moga , Punjab , India
| | | | | |
Collapse
|
22
|
Ucisik MH, Küpcü S, Breitwieser A, Gelbmann N, Schuster B, Sleytr UB. S-layer fusion protein as a tool functionalizing emulsomes and CurcuEmulsomes for antibody binding and targeting. Colloids Surf B Biointerfaces 2015; 128:132-139. [PMID: 25734967 PMCID: PMC4406452 DOI: 10.1016/j.colsurfb.2015.01.055] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/28/2015] [Accepted: 01/29/2015] [Indexed: 11/13/2022]
Abstract
Selective targeting of tumor cells by nanoparticle-based drug delivery systems is highly desirable because it maximizes the drug concentration at the desired target while simultaneously protecting the surrounding healthy tissues. Here, we show a design for smart nanocarriers based on a biomimetic approach that utilizes the building principle of virus envelope structures. Emulsomes and CurcuEmulsomes comprising a tripalmitin solid core surrounded by phospholipid layers are modified by S-layer proteins that self-assemble into a two-dimensional array to form a surface layer. One significant advantage of this nanoformulation is that it increases the solubility of the lipophilic anti-cancer agent curcumin in the CurcuEmulsomes by a factor of 2700. In order to make the emulsomes specific for IgG, the S-layer protein is fused with two protein G domains. This S-layer fusion protein preserves its recrystallization characteristics, forming an ordered surface layer (square lattice with 13 nm unit-by-unit distance). The GG domains are presented in a predicted orientation and exhibit a selective binding affinity for IgG.
Collapse
Affiliation(s)
- Mehmet H Ucisik
- Institute for Synthetic Bioarchitectures, Department of Nanobiotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 11, 1190 Vienna, Austria; Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Ekinciler Cad. No. 19 Kavacık Kavşağı, Beykoz 34810, Istanbul, Turkey.
| | - Seta Küpcü
- Institute for Synthetic Bioarchitectures, Department of Nanobiotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 11, 1190 Vienna, Austria
| | - Andreas Breitwieser
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 11, 1190 Vienna, Austria
| | | | - Bernhard Schuster
- Institute for Synthetic Bioarchitectures, Department of Nanobiotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 11, 1190 Vienna, Austria
| | - Uwe B Sleytr
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 11, 1190 Vienna, Austria
| |
Collapse
|
23
|
Zhou H, Mohamedali KA, Gonzalez-Angulo AM, Cao Y, Migliorini M, Cheung LH, LoBello J, Lei X, Qi Y, Hittelman WN, Winkles JA, Tran NL, Rosenblum MG. Development of human serine protease-based therapeutics targeting Fn14 and identification of Fn14 as a new target overexpressed in TNBC. Mol Cancer Ther 2014; 13:2688-705. [PMID: 25239934 DOI: 10.1158/1535-7163.mct-14-0346] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cytokine TWEAK and its receptor, Fn14, have emerged as potentially valuable targets for cancer therapy. Granzyme B (GrB)-containing Fn14-targeted constructs were generated containing either the Fn14 ligand TWEAK (GrB-TWEAK) or an anti-Fn14 humanized single-chain antibody (GrB-Fc-IT4) as the targeting moieties. Both constructs showed high affinity and selective cytotoxicity against a panel of Fn14-expressing human tumor cells including triple-negative breast cancer (TNBC) lines. Cellular expression of the GrB inhibitor PI-9 in target cells had no impact on the cytotoxic effect of either construct. Cellular expression of MDR1 showed no cross-resistance to the fusion constructs. GrB-TWEAK and GrB-Fc-IT4 activated intracellular caspase cascades and cytochrome c-related proapoptotic pathways consistent with the known intracellular functions of GrB in target cells. Treatment of mice bearing established HT-29 xenografts with GrB-TWEAK showed significant tumor growth inhibition compared with vehicle alone (P < 0.05). Both GrB-TWEAK and GrB-Fc-IT4 displayed significant tumor growth inhibition when administered to mice bearing orthotopic MDA-MB-231 (TNBC) tumor xenografts. The Cancer Genome Atlas analysis revealed that Fn14 mRNA expression was significantly higher in TNBC and in HER2-positive disease (P < 0.0001) compared with hormone receptor-positive breast cancer, and in basal-like 2 tumors (P = 0.01) compared with other TNBC molecular subtypes. IHC analysis of a 101 patient TNBC tumor microarray showed that 55 of 101 (54%) of tumors stained positive for Fn14, suggesting that this may be an excellent potential target for precision therapeutic approaches. Targeting Fn14 using fully human, GrB-containing fusion constructs may form the basis for a new class of novel, potent, and highly effective constructs for targeted therapeutic applications.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Khalid A Mohamedali
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Ana Maria Gonzalez-Angulo
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, Texas. Department of Systems Biology, MD Anderson Cancer Center, Houston, Texas
| | - Yu Cao
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Mary Migliorini
- Department of Surgery, Center for Vascular and Inflammatory Diseases, and the Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lawrence H Cheung
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Janine LoBello
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Xiudong Lei
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas
| | - Yuan Qi
- Department of Bioinformatics, MD Anderson Cancer Center, Houston, Texas
| | - Walter N Hittelman
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Jeffrey A Winkles
- Department of Surgery, Center for Vascular and Inflammatory Diseases, and the Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Nhan L Tran
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Michael G Rosenblum
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
24
|
Yousefpour P, Chilkoti A. Co-opting biology to deliver drugs. Biotechnol Bioeng 2014; 111:1699-716. [PMID: 24916780 PMCID: PMC4251460 DOI: 10.1002/bit.25307] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/30/2014] [Accepted: 06/05/2014] [Indexed: 01/17/2023]
Abstract
The goal of drug delivery is to improve the safety and therapeutic efficacy of drugs. This review focuses on delivery platforms that are either derived from endogenous pathways, long-circulating biomolecules and cells or that piggyback onto long-circulating biomolecules and cells. The first class of such platforms is protein-based delivery systems--albumin, transferrin, and fusion to the Fc domain of antibodies--that have a long-circulation half-life and are designed to transport different molecules. The second class is lipid-based delivery systems-lipoproteins and exosomes-that are naturally occurring circulating lipid particles. The third class is cell-based delivery systems--erythrocytes, macrophages, and platelets--that have evolved, for reasons central to their function, to exhibit a long life-time in the body. The last class is small molecule-based delivery systems that include folic acid. This article reviews the biology of these systems, their application in drug delivery, and the promises and limitations of these endogenous systems for drug delivery.
Collapse
Affiliation(s)
- Parisa Yousefpour
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| | | |
Collapse
|
25
|
Akhtar MJ, Ahamed M, Alhadlaq HA, Alrokayan SA, Kumar S. Targeted anticancer therapy: Overexpressed receptors and nanotechnology. Clin Chim Acta 2014; 436:78-92. [DOI: 10.1016/j.cca.2014.05.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/07/2014] [Accepted: 05/10/2014] [Indexed: 01/05/2023]
|
26
|
Venkatasubbu GD, Ramasamy S, Reddy GP, Kumar J. In vitro and in vivo anticancer activity of surface modified paclitaxel attached hydroxyapatite and titanium dioxide nanoparticles. Biomed Microdevices 2014; 15:711-726. [PMID: 23615724 DOI: 10.1007/s10544-013-9767-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Targeted drug delivery using nanocrystalline materials delivers the drug at the diseased site. This increases the efficacy of the drug in killing the cancer cells. Surface modifications were done to target the drug to a particular receptor on the cell surface. This paper reports synthesis of hydroxyapatite and titanium dioxide nanoparticles and modification of their surface with polyethylene glycol (PEG) followed by folic acid (FA). Paclitaxel, an anticancer drug, is attached to functionalized hydroxyapatite and titanium dioxide nanoparticles. The pure and functionalised nanoparticles are characterised with XRD, TEM and UV spectroscopy. Anticancer analysis was carried out in DEN induced hepatocarcinoma animals. Biochemical, hematological and histopathological analysis show that the surface modified paclitaxel attached nanoparticles have an higher anticancer activity than the pure paclitaxel and surface modified nanoparticles without paclitaxel. This is due to the targeting of the drug to the folate receptor in the cancer cells.
Collapse
Affiliation(s)
| | - S Ramasamy
- Crystal Growth Centre, Anna University, Chennai, 600025, Tamil Nadu, India.
| | - G Pramod Reddy
- Department of Pharmacology, Siddha Central Research Institute, Chennai, Tamil Nadu, India
| | - J Kumar
- Crystal Growth Centre, Anna University, Chennai, 600025, Tamil Nadu, India
| |
Collapse
|
27
|
Ansar M, Serrano D, Papademetriou I, Bhowmick TK, Muro S. Biological functionalization of drug delivery carriers to bypass size restrictions of receptor-mediated endocytosis independently from receptor targeting. ACS NANO 2013; 7:10597-10611. [PMID: 24237309 PMCID: PMC3901850 DOI: 10.1021/nn404719c] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Targeting of drug carriers to cell-surface receptors involved in endocytosis is commonly used for intracellular drug delivery. However, most endocytic receptors mediate uptake via clathrin or caveolar pathways associated with ≤200-nm vesicles, restricting carrier design. We recently showed that endocytosis mediated by intercellular adhesion molecule 1 (ICAM-1), which differs from clathrin- and caveolae-mediated pathways, allows uptake of nano- and microcarriers in cell culture and in vivo due to recruitment of cellular sphingomyelinases to the plasmalemma. This leads to ceramide generation at carrier binding sites and formation of actin stress-fibers, enabling engulfment and uptake of a wide size-range of carriers. Here we adapted this paradigm to enhance uptake of drug carriers targeted to receptors associated with size-restricted pathways. We coated sphingomyelinase onto model (polystyrene) submicro- and microcarriers targeted to clathrin-associated mannose-6-phosphate receptor. In endothelial cells, this provided ceramide enrichment at the cell surface and actin stress-fiber formation, modifying the uptake pathway and enhancing carrier endocytosis without affecting targeting, endosomal transport, cell-associated degradation, or cell viability. This improvement depended on the carrier size and enzyme dose, and similar results were observed for other receptors (transferrin receptor) and cell types (epithelial cells). This phenomenon also enhanced tissue accumulation of carriers after intravenous injection in mice. Hence, it is possible to maintain targeting toward a selected receptor while bypassing natural size restrictions of its associated endocytic route by functionalization of drug carriers with biological elements mimicking the ICAM-1 pathway. This strategy holds considerable promise to enhance flexibility of design of targeted drug delivery systems.
Collapse
Affiliation(s)
- Maria Ansar
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD
| | - Daniel Serrano
- Department of Cell Biology & Molecular Genetics and Biological Sciences Graduate Program, University of Maryland, College Park, MD
| | - Iason Papademetriou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
| | - Tridib Kumar Bhowmick
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
| |
Collapse
|
28
|
|
29
|
Muro S. Challenges in design and characterization of ligand-targeted drug delivery systems. J Control Release 2012; 164:125-37. [PMID: 22709588 PMCID: PMC3481020 DOI: 10.1016/j.jconrel.2012.05.052] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Revised: 05/19/2012] [Accepted: 05/26/2012] [Indexed: 01/11/2023]
Abstract
Targeting of therapeutic agents to molecular markers expressed on the surface of cells requiring clinical intervention holds promise to improve specificity of delivery, enhancing therapeutic effects while decreasing potential damage to healthy tissues. Drug targeting to cellular receptors involved in endocytic transport facilitates intracellular delivery, a requirement for a number of therapeutic goals. However, after several decades of experimental design, there is still considerable controversy on the practical outcome of drug targeting strategies. The plethora of factors contributing to the relative efficacy of targeting makes the success of these approaches hardly predictable. Lack of fully specific targets, along with selection of targets with spatial and temporal expression well aligned to interventional requirements, pose difficulties to this process. Selection of adequate sub-molecular target epitopes determines accessibility for anchoring of drug conjugates and bulkier drug carriers, as well as proper signaling for uptake within the cell. Targeting design must adapt to physiological variables of blood flow, disease status, and tissue architecture by accommodating physicochemical parameters such as carrier composition, functionalization, geometry, and avidity. In many cases, opposite features need to meet a balance, e.g., sustained circulation versus efficient targeting, penetration through tissues versus uptake within cells, internalization within endocytic compartment to avoid efflux pumps versus accessibility to molecular targets within the cytosol, etc. Detailed characterization of these complex physiological factors and design parameters, along with a deep understanding of the mechanisms governing the interaction of targeted drugs and carriers with the biological environment, are necessary steps toward achieving efficient drug targeting systems.
Collapse
Affiliation(s)
- Silvia Muro
- Fischell Department of Bioengineering, School of Engineering, University of Maryland College Park, College Park, MD 20742, USA.
| |
Collapse
|
30
|
Jain R, Dandekar P, Loretz B, Melero A, Stauner T, Wenz G, Koch M, Lehr CM. Enhanced cellular delivery of idarubicin by surface modification of propyl starch nanoparticles employing pteroic acid conjugated polyvinyl alcohol. Int J Pharm 2011; 420:147-55. [DOI: 10.1016/j.ijpharm.2011.08.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/12/2011] [Accepted: 08/15/2011] [Indexed: 10/17/2022]
|
31
|
Han L, Huang R, Li J, Liu S, Huang S, Jiang C. Plasmid pORF-hTRAIL and doxorubicin co-delivery targeting to tumor using peptide-conjugated polyamidoamine dendrimer. Biomaterials 2011; 32:1242-52. [DOI: 10.1016/j.biomaterials.2010.09.070] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 09/30/2010] [Indexed: 01/20/2023]
|
32
|
Han L, Huang R, Liu S, Huang S, Jiang C. Peptide-Conjugated PAMAM for Targeted Doxorubicin Delivery to Transferrin Receptor Overexpressed Tumors. Mol Pharm 2010; 7:2156-65. [DOI: 10.1021/mp100185f] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Liang Han
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Rongqin Huang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Shuhuan Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Shixian Huang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Li Y, Liu B, Zhao C, Yang B. Common Pathway for K562 Cells Endocytosis and Release of GaC-Tf and Ga2-Tf via a Transferrin Receptor. CHINESE J CHEM 2010. [DOI: 10.1002/cjoc.201090144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
34
|
Myachina GF, Kon’kova TV, Korzhova SA, Ermakova TG, Pozdnyakov AS, Sukhov BG, Arsent’ev KY, Likhoshvai EV, Trofimov BA. Gold nanoparticles stabilized with water-soluble biocompatible poly(1-vinyl-1,2,4-triazole). DOKLADY CHEMISTRY 2010. [DOI: 10.1134/s0012500810030018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
35
|
Abstract
Mannosylated liposomes appear to be a promising and potential carrier system for delivery of proteins, peptides, or nucleic acids. The present chapter describes novel mannosylated liposomes, which increase the intracellular targeting of immunogen to dendritic cells and macrophages possessing the specific receptors. The liposomes used in the present investigation were prepared by hand-shaken method and characterized for size, shape, surface charge, encapsulation efficiency, ligand binding, and specificity and uptake studies. The immune-stimulating activity of the liposomes was studied by measuring antigen-specific antibody titer following subcutaneous administration of different liposomal formulations in BALB/c mice. It was found that O-palmitoyl mannan (OPM)-coated liposomes showed better uptake efficiency. In vivo studies revealed that the OPM-coated liposomes exhibited significant higher serum antibody response and stronger TH1/TH2-based cellular responses. In conclusion, novel vesicular constructs are useful nanosized carriers having superior surface characteristics--for active interaction with the antigen-presenting cells and subsequent processing and presentation of antigen.
Collapse
Affiliation(s)
- Suresh Prasad Vyas
- Department of Pharmaceutical Sciences, Drug Delivery Research Laboratory, Sagar, India
| | | | | |
Collapse
|
36
|
Godin B, Driessen WHP, Proneth B, Lee SY, Srinivasan S, Rumbaut R, Arap W, Pasqualini R, Ferrari M, Decuzzi P. An integrated approach for the rational design of nanovectors for biomedical imaging and therapy. ADVANCES IN GENETICS 2010; 69:31-64. [PMID: 20807601 DOI: 10.1016/s0065-2660(10)69009-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The use of nanoparticles for the early detection, cure, and imaging of diseases has been proved already to have a colossal potential in different biomedical fields, such as oncology and cardiology. A broad spectrum of nanoparticles are currently under development, exhibiting differences in (i) size, ranging from few tens of nanometers to few microns; (ii) shape, from the classical spherical beads to discoidal, hemispherical, cylindrical, and conical; (iii) surface functionalization, with a wide range of electrostatic charges and biomolecule conjugations. Clearly, the library of nanoparticles generated by combining all possible sizes, shapes, and surface physicochemical properties is enormous. With such a complex scenario, an integrated approach is here proposed and described for the rational design of nanoparticle systems (nanovectors) for the intravascular delivery of therapeutic and imaging contrast agents. The proposed integrated approach combines multiscale/multiphysics mathematical models with in vitro assays and in vivo intravital microscopy (IVM) experiments and aims at identifying the optimal combination of size, shape, and surface properties that maximize the nanovectors localization within the diseased microvasculature.
Collapse
Affiliation(s)
- Biana Godin
- Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wu F, Wuensch SA, Azadniv M, Ebrahimkhani MR, Crispe IN. Galactosylated LDL nanoparticles: a novel targeting delivery system to deliver antigen to macrophages and enhance antigen specific T cell responses. Mol Pharm 2009; 6:1506-17. [PMID: 19637876 PMCID: PMC2782853 DOI: 10.1021/mp900081y] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We aim to define the role of Kupffer cells in intrahepatic antigen presentation, using the selective delivery of antigen to Kupffer cells rather than other populations of liver antigen-presenting cells. To achieve this we developed a novel antigen delivery system that can target antigens to macrophages, based on a galactosylated low-density lipoprotein nanoscale platform. Antigen was delivered via the galactose particle receptor (GPr), internalized, degraded and presented to T cells. The conjugation of fluoresceinated ovalbumin (FLUO-OVA) and lactobionic acid with LDL resulted in a substantially increased uptake of FLUO-OVA by murine macrophage-like ANA1 cells in preference to NIH3T3 cells, and by primary peritoneal macrophages in preference to primary hepatic stellate cells. Such preferential uptake led to enhanced proliferation of OVA specific T cells, showing that the galactosylated LDL nanoscale platform is a successful antigen carrier, targeting antigen to macrophages but not to all categories of antigen presenting cells. This system will allow targeted delivery of antigen to macrophages in the liver and elsewhere, addressing the question of the role of Kupffer cells in liver immunology. It may also be an effective way of delivering drugs or vaccines directly at macrophages.
Collapse
Affiliation(s)
- Fang Wu
- David H. Smith Center for Vaccine Biology and Immunology, The Aab Institute for Biomedical Research, Department of Microbiology, University of Rochester Medical Center, Rochester, New York 14642, USA.
| | | | | | | | | |
Collapse
|
38
|
Khatri K, Rawat A, Mahor S, Gupta PN, Vyas SP. Hepatitis B surface protein docked vesicular carrier for site specific delivery to liver. J Drug Target 2008; 13:359-66. [PMID: 16278155 DOI: 10.1080/10611860500230294] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The intrinsic liver tropism of liposomes can be augmented by the addition of targeting features such as the incorporation of hepatotropic elements of the hepatitis viruses. Hepatitis B virus is known to infect hepatocytes after viremia by asialoglycoprotein receptor mediated uptake. However, the specificity of hepatitis B virus surface protein (HBsAg) towards hepatocytes has confronting reports. In the present study, we evaluated the functional ability of HBsAg to be employed as a ligand for targeting hepatocytes. We prepared (14)C labeled small unilamellar vesicles (SUVs) composed of egg PC/Cholesterol/N-glutarylphosphatidylethanolamine (NGPE) in a 60:30:10 molar ratio. HBsAg was covalently linked to SUVs using a water-soluble carbodiimide (EDC) mediated conjugation with NGPE. In vitro cell binding and uptake studies revealed that bioprotein docked carrier system was efficiently taken up by HepG2 cells by the receptor mediated endocytosis. The biodistribution behaviour of plain and HBsAg coated liposomes was also examined followed by intravenous injection. The study revealed that almost 75% of the radioactivity was recovered in the liver after 4 h of injection that was nearly three-fold greater in magnitude than the plain liposomes. Further, fractionation of liver into liver parenchymal cells (PC) and non-parenchymal cells confirmed the preferential localization of the HBsAg coated liposomal carrier in the parenchymal cells.
Collapse
Affiliation(s)
- Kapil Khatri
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Hari Singh Gour Vishwavidyalaya, Sagar, India
| | | | | | | | | |
Collapse
|
39
|
Biotinylated thermoresponsive micelle self-assembled from double-hydrophilic block copolymer for drug delivery and tumor target. Biomaterials 2008; 29:497-505. [DOI: 10.1016/j.biomaterials.2007.10.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Accepted: 10/01/2007] [Indexed: 11/18/2022]
|
40
|
Wang X, Guo Z. Towards the rational design of platinum(ii) and gold(iii) complexes as antitumour agents. Dalton Trans 2008:1521-32. [DOI: 10.1039/b715903j] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
41
|
Pristensky DV, Staroverov SA, Ermilov DN, Shchyogolev SY, Dykman LA. Analysis of effectiveness of intracellular penetration of ivermectin immobilized onto corpuscular carriers. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2007. [DOI: 10.1134/s1990750807030146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Béduneau A, Saulnier P, Benoit JP. Active targeting of brain tumors using nanocarriers. Biomaterials 2007; 28:4947-67. [PMID: 17716726 DOI: 10.1016/j.biomaterials.2007.06.011] [Citation(s) in RCA: 282] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Accepted: 06/06/2007] [Indexed: 02/06/2023]
Abstract
The delivery of drugs to brain tumors is limited by the presence of the blood-brain barrier (BBB) separating the blood from the cerebral parenchyma. An understanding of the specific mechanisms of the brain capillary endothelium has led to the development of various strategies to enhance the penetration of drugs into the brain tissue. Active targeting is a non-invasive approach, which consists in transporting drugs to target organs using site-specific ligands. Drug-loaded nanocarriers capable of recognizing brain capillary endothelial cells and cerebral tumoral cells have shown promising potential in oncology. Endogenous and chimeric ligands binding to carriers or receptors of the BBB have been directly or indirectly conjugated to nanocarriers. This review indexes the main targeted colloidal systems used for drug delivery to the brain. Their pharmacological behavior and their therapeutic effect are discussed.
Collapse
|
43
|
Yang Z, Wang X, Diao H, Zhang J, Li H, Sun H, Guo Z. Encapsulation of platinum anticancer drugs by apoferritin. Chem Commun (Camb) 2007:3453-5. [PMID: 17700879 DOI: 10.1039/b705326f] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Apoferritin derived from the native protein ferritin was employed to encapsulate anticancer drugs cisplatin and carboplatin.
Collapse
Affiliation(s)
- Zhen Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, PR China
| | | | | | | | | | | | | |
Collapse
|
44
|
Li YQ, Liu B, Zhao CG, Zhang W, Yang BS. Characterization of transferrin receptor-dependent GaC–Tf–FeN transport in human leukemic HL60 cells. Clin Chim Acta 2006; 366:225-32. [PMID: 16360136 DOI: 10.1016/j.cca.2005.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2005] [Revised: 10/11/2005] [Accepted: 10/12/2005] [Indexed: 11/29/2022]
Abstract
BACKGROUND Understanding the uptake of GaC-Tf-FeN by cells will provide key insights into studies on transferrin-mediated drug delivery. METHODS The mechanism of GaC-Tf-FeN transporting into and out of HL60 cells has been investigated by comparing transports between GaC-Tf-FeN and apoTf by means of 125I-labeled transferrin. RESULTS An association constant for GaC-Tf-FeN was 2 times that for apoTf. GaC-Tf-FeN and apoTf of cell surface-bound displayed similar kinetics during the uptake, but the release rates of internalized GaC-Tf-FeN and apoTf from cells were different which showed characteristic disparate. The release continued to occur during the incubation of GaC-Tf-FeN in the presence of nonradioactive apoTf. Neither NaN3 nor NH4Cl could completely block internalization of GaC-Tf-FeN, but they prevented the release of GaC-Tf-FeN from the cells. Excess cold unlabeled apoTf could overcome the block in the release due to NH4Cl but not NaN3. The binding and internalization of GaC-Tf-FeN could be competitively inhibited by nonradioactive apoTf. It implies that both bind to the same receptor on the membrane and the localization of GaC-Tf-FeN resembles that of apoTf inside cells. Pretreated cells with pronase abolished the binding of GaC-Tf-FeN significantly. CONCLUSION On the basis of these findings, we proposed the "transferrin receptor" for the mechanism of GaC-Tf-FeN transport by HL60 cells.
Collapse
Affiliation(s)
- Ying-Qi Li
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan, Shanxi 030006, China
| | | | | | | | | |
Collapse
|
45
|
Arnedo A, Irache JM, Merodio M, Espuelas Millán MS. Albumin nanoparticles improved the stability, nuclear accumulation and anticytomegaloviral activity of a phosphodiester oligonucleotide. J Control Release 2004; 94:217-27. [PMID: 14684285 DOI: 10.1016/j.jconrel.2003.10.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The goal of this study was to evaluate the potential of albumin nanoparticles as a delivery system for antisense oligonucleotides. Nanoparticles were prepared by a coacervation process and cross-linkage with glutaraldehyde. Phosphodiester (PO) and phosphorotioate (PS) oligonucleotides were either adsorbed on the surface of nanoparticles (PO-NPA and PS-NPA) or incorporated in the nanoparticle matrix (PO-NPB and PS-NPB). When PO-loaded nanoparticles were incubated with phosphodiesterase, only NPB was able to keep the oligonucleotide hybridization capability for at least 60 min. The antiviral activity was evaluated in MRC-5 fibroblasts infected with human cytomegalovirus at a MOI of 0.0035. Both PO nanoparticle formulations significantly increased the antiviral activity of free PO (P<0.001) and NPB showed slightly higher efficacies than NPA (P<0.05). On the other hand, PS exhibited significant higher activity than free PO (P<0.001), however, no significant differences were found between PS-nanoparticle and PO-nanoparticle formulations. These findings were well correlated with the intracellular distribution observed for fluorescent oligonucleotide-loaded albumin nanoparticles. Even these carriers delayed and decreased the uptake of PO by MRC-5 cells, they finally induced a diffused cytoplasmic distribution and major nuclear accumulation. In summary, albumin nanoparticles partially protected a PO against enzymatic degradation and improved their presence in the nucleus and thus, increased its efficiency.
Collapse
Affiliation(s)
- A Arnedo
- Centro Galénico, Departamento de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Navarra, Irunlarrea 1, Ap. 177, 31080 Pamplona, Spain
| | | | | | | |
Collapse
|
46
|
Xiu-Lian D, Kui W, Ya K, Lan Y, Rong-Chang L, Yan Zhong C, Kwok Ping H, Zhong Ming Q. Apotransferrin is internalized and distributed in the same way as holotransferrin in K562 cells. J Cell Physiol 2004; 201:45-54. [PMID: 15281088 DOI: 10.1002/jcp.20051] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Transferrin (Tf), a naturally existing protein, has received considerable attention in the area of drug targeting since it is biodegradable, non-toxic, and non-immunogenic. The efficient cellular uptake of Tf shows it has potential in the delivery of anti-cancer drugs, proteins, and therapeutic genes into proliferating malignant cells that overexpress transferrin receptor (TfR). In human serum, about 30% of Tf exists in the iron-saturated form (Fe(2)-Tf) and the remainder exists as apotransferrin (apo-Tf). Understanding the uptake of apo-Tf by cells will provide key insights into studies on Tf-mediated drug delivery. In the present study, we investigated visually the transport of apo-Tf into K562 cells and its intracellular localization by laser-scanning confocal microscopy (LSCM) and flow cytometry analysis (FCA). It was found that, like Fe(2)-Tf, apo-Tf can be taken up into the cells. The process is time- and temperature-dependent, competitively inhibited by Fe(2)-Tf, and significantly abolished by pronase pretreatment. Visual evidence showed that the transport of apo-Tf into K562 cells is a TfR-mediated process. Furthermore, the investigations using optical-slicing technique demonstrated that the distribution of apo-Tf is similar to that of Fe(2)-Tf, both appearing in the perinuclear region in ball-in-bowl shape.
Collapse
Affiliation(s)
- Du Xiu-Lian
- School of Pharmaceutical Science and Joint Laboratory of Perking University & The Hong Kong Polytechnic University, Perking University, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Macromolecular therapeutics provide numerous benefits for the delivery of cytotoxic or poorly soluble drugs in vivo. However, these constructs often encounter barriers for drug delivery on both the systemic and subcellular level. Many soluble polymer carriers have been designed to surmount specific physiological barriers individually, but less work has been dedicated to designing an all-encompassing construct that addresses multiple therapeutic barriers at once. Incorporation of multiple agents already individually known to increase effectiveness into one carrier could further improve current drug delivery technology. Recent developments in subcellular delivery of therapeutic agents in soluble macromolecular carriers are discussed in the context of the future possibility for the design of an all-encompassing soluble multi-functional drug delivery vehicle.
Collapse
Affiliation(s)
- R J Christie
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523-1872, USA
| | | |
Collapse
|
48
|
Abstract
Since transferrin was discovered more than half a century ago, a considerable effort has been made towards understanding tranferrin-mediated iron uptake. However, it was not until recently with the identification and characterization of several new genes related to iron homeostasis, such as the hemochromatosis protein HFE and the iron transporter DMT1, that our knowledge has been advanced dramatically. A major pathway for cellular iron uptake is through internalization of the complex of iron-bound transferrin and the transferrin receptor, which is negatively modulated by HFE, a protein related to hereditary hemochromatosis. Iron is released from transferrin as the result of the acidic pH in endosome and then is transported to the cytosol by DMT1. The iron is then utilized as a cofactor by heme and ribonucleotide reductase or stored in ferritin. Apart from iron, many other metal ions of therapeutic and diagnostic interests can also bind to transferrin at the iron sites and their transferrin complexes can be recognized by many cells. Therefore, transferrin has been thought as a "delivery system" for many beneficial and harmful metal ions into the cells. Transferrin has also be widely applied as a targeting ligand in the active targeting of anticancer agents, proteins, and genes to primary proliferating malignant cells that overexpress transferrin receptors. This is achieved by conjugation of transferrin with drugs, proteins, hybride systems with marcomolecules and as liposomal-coated systems. Conjugates of anticancer drugs with transferrin can significantly improve the selectivity and toxicity and overcome drug resistance, thereby leading to a better treatment. The coupling of DNA to transferrin via a polycation such as polylysine or via cationic liposomes can target and transfer of the extrogenous DNA particularly into proliferating cells through receptor-mediated endocytosis. These kinds of non-viral vectors are potential alternatives to viral vectors for gene therapy, if the transfection efficiency can be improved. Moreover, transferrin receptors have shown potentials in delivery of therapeutic drugs or genes into the brain across blood-brain barrier.
Collapse
Affiliation(s)
- Hongyan Li
- Laboratory of Iron Metabolism, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | | |
Collapse
|
49
|
Li H, Sun H, Qian ZM. The role of the transferrin-transferrin-receptor system in drug delivery and targeting. Trends Pharmacol Sci 2002; 23:206-9. [PMID: 12007993 DOI: 10.1016/s0165-6147(02)01989-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Exploration of the potential of site-specific and target-oriented drug delivery systems has gained interest recently. Indeed, the efficient cellular mechanism of transferrin uptake has been exploited for the delivery not only of anticancer drugs and proteins, but also of therapeutic genes into proliferating malignant cells that overexpress transferrin receptors. In particular, the transferrin receptor offers great promise in the delivery of therapeutic agents across the blood-brain barrier to the brain.
Collapse
Affiliation(s)
- Hongyan Li
- Dept of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | | | | |
Collapse
|