101
|
Kapoor B, Gupta R, Gulati M, Singh SK, Khursheed R, Gupta M. The Why, Where, Who, How, and What of the vesicular delivery systems. Adv Colloid Interface Sci 2019; 271:101985. [PMID: 31351415 DOI: 10.1016/j.cis.2019.07.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/06/2019] [Accepted: 07/06/2019] [Indexed: 12/14/2022]
Abstract
Though vesicular delivery systems have been widely explored and reviewed, no comprehensive review exists that covers their development from the inception of the concept to its culmination in the form of regulated marketed formulations. With the advancement of scientific research in the field of nanomedicine, certain category of vesicular delivery systems have successfully reached the global market. Despite extensive research and highly encouraging results in a plethora of pathological conditions in the preclinical studies, translation of these nanomedicines from laboratory to market has been very limited. Aim of this review is to describe comprehensively the various colloidal delivery systems, focusing mainly on their conventional and advanced methods of preparation, different characterization techniques and main success stories of their journey from bench to bedside of the patient. The review also touches the finer nuances of the use of modern formulation approach of DoE (Design of Experiments) in their formulation and the status of regulatory guidelines for the approval of these nanomedicines.
Collapse
|
102
|
Bnyan R, Khan I, Ehtezazi T, Saleem I, Gordon S, O’Neill F, Roberts M. Formulation and optimisation of novel transfersomes for sustained release of local anaesthetic. J Pharm Pharmacol 2019; 71:1508-1519. [DOI: 10.1111/jphp.13149] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/26/2019] [Accepted: 07/06/2019] [Indexed: 01/28/2023]
Abstract
Abstract
Objective
To investigate the effect of formulation parameters on the preparation of transfersomes as sustained-release delivery systems for lidocaine and to develop and validate a new high-performance liquid chromatography (HPLC) method for analysis.
Method
Taguchi design of experiment (DOE) was used to optimise lidocaine-loaded transfersomes in terms of phospholipid, edge activator (EA) and phospholipid : EA ratio. Transfersomes were characterised for size, polydispersity index (PDI), charge and entrapment efficiency (%EE). A HPLC method for lidocaine quantification was optimised and validated using a mobile phase of 30%v/v PBS (0.01 m) : 70%v/v Acetonitrile at a flow rate of 1 ml/min, detected at 255 nm with retention time of 2.84 min. The release of lidocaine from selected samples was assessed in vitro.
Key findings
Transfersomes were 200 nm in size, with PDI ~ 0.3. HPLC method was valid for linearity (0.1–2 mg/ml, R2 0.9999), accuracy, intermediate precision and repeatability according to ICH guidelines. The %EE was between 44% and 56% and dependent on the formulation parameters. Taguchi DOE showed the effect of factors was in the rank order : lipid : EA ratio ˃ EA type ˃ lipid type. Optimised transfersomes sustained the release of lidocaine over 24 h.
Conclusion
Sustained-release, lidocaine-loaded transfersomes were successfully formulated and optimised using a DOE approach, and a new HPLC method for lidocaine analysis was developed and validated.
Collapse
Affiliation(s)
- Ruba Bnyan
- Formulation and Drug Delivery Research Group, School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Iftikhar Khan
- Formulation and Drug Delivery Research Group, School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Touraj Ehtezazi
- Formulation and Drug Delivery Research Group, School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Imran Saleem
- Formulation and Drug Delivery Research Group, School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Sarah Gordon
- Formulation and Drug Delivery Research Group, School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Francis O’Neill
- Institute of Clinical Sciences, University of Liverpool Dental School, Liverpool, UK
| | - Matthew Roberts
- Formulation and Drug Delivery Research Group, School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
103
|
White DT, Saxena MT, Mumm JS. Let's get small (and smaller): Combining zebrafish and nanomedicine to advance neuroregenerative therapeutics. Adv Drug Deliv Rev 2019; 148:344-359. [PMID: 30769046 PMCID: PMC6937731 DOI: 10.1016/j.addr.2019.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/21/2018] [Accepted: 01/28/2019] [Indexed: 01/18/2023]
Abstract
Several key attributes of zebrafish make them an ideal model system for the discovery and development of regeneration promoting therapeutics; most notably their robust capacity for self-repair which extends to the central nervous system. Further, by enabling large-scale drug discovery directly in living vertebrate disease models, zebrafish circumvent critical bottlenecks which have driven drug development costs up. This review summarizes currently available zebrafish phenotypic screening platforms, HTS-ready neurodegenerative disease modeling strategies, zebrafish small molecule screens which have succeeded in identifying regeneration promoting compounds and explores how intravital imaging in zebrafish can facilitate comprehensive analysis of nanocarrier biodistribution and pharmacokinetics. Finally, we discuss the benefits and challenges attending the combination of zebrafish and nanoparticle-based drug optimization, highlighting inspiring proof-of-concept studies and looking toward implementation across the drug development community.
Collapse
Affiliation(s)
- David T White
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Meera T Saxena
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA; Luminomics Inc., Baltimore, MD 21286, USA
| | - Jeff S Mumm
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
104
|
Hill LK, Meleties M, Katyal P, Xie X, Delgado-Fukushima E, Jihad T, Liu CF, O’Neill S, Tu RS, Renfrew PD, Bonneau R, Wadghiri YZ, Montclare JK. Thermoresponsive Protein-Engineered Coiled-Coil Hydrogel for Sustained Small Molecule Release. Biomacromolecules 2019; 20:3340-3351. [DOI: 10.1021/acs.biomac.9b00107] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lindsay K. Hill
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
- Department of Biomedical Engineering, SUNY Downstate Medical Center, Brooklyn, New York 11203, United States
| | - Michael Meleties
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Priya Katyal
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Xuan Xie
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Erika Delgado-Fukushima
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Teeba Jihad
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Che-Fu Liu
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Sean O’Neill
- Chemical Engineering Department, The City College of New York, 160 Convent Avenue, New York, New York 10031, United States
| | - Raymond S. Tu
- Chemical Engineering Department, The City College of New York, 160 Convent Avenue, New York, New York 10031, United States
| | - P. Douglas Renfrew
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, New York 10010, United States
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, New York 10010, United States
- Center for Genomics and Systems Biology, New York University, New York, New York 10003, United States
- Computer Science Department, Courant Institute of Mathematical Sciences, New York University, New York, New York 10009, United States
| | | | - Jin Kim Montclare
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
- Department of Chemistry, New York University, New York, New York 10012, United States
- Department of Biomaterials, New York University College of Dentistry, New York, New York 10010, United States
| |
Collapse
|
105
|
Creighton RL, Suydam IT, Ebner ME, Afunugo WE, Bever AM, Cao S, Jiang Y, Woodrow KA. Sustained Intracellular Raltegravir Depots Generated with Prodrugs Designed for Nanoparticle Delivery. ACS Biomater Sci Eng 2019; 5:4013-4022. [PMID: 33117884 DOI: 10.1021/acsbiomaterials.9b00658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Polymeric nanocarriers have been extensively used to improve the delivery of hydrophobic drugs, but often provide low encapsulation efficiency and percent loading for hydrophilic compounds. In particular, insufficient loading of hydrophilic antiretroviral drugs such as the integrase inhibitor raltegravir (RAL) has limited the development of sustained-release therapeutics or prevention strategies against HIV. To address this, we developed a generalizable prodrug strategy using RAL as a model where loading, release and subsequent hydrolysis can be tuned by promoiety selection. Prodrugs with large partition coefficients increased the encapsulation efficiency up to 25-fold relative to RAL, leading to significant dose reductions in antiviral activity assays. The differential hydrolysis rates of these prodrugs led to distinct patterns of RAL availability and observed antiviral activity. We also developed a method to monitor the temporal distribution of both prodrug and RAL in cells treated with free prodrug or prodrug-NPs. Results of these studies indicated that prodrug-NPs create an intracellular drug reservoir capable of sustained intracellular drug release. Overall, our results suggest that the design of prodrugs for specific polymeric nanocarrier systems could provide a more generalized strategy to formulate physicochemically diverse hydrophilic drugs with a number of biomedical applications.
Collapse
Affiliation(s)
- Rachel L Creighton
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195-5061, United States
| | - Ian T Suydam
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195-5061, United States.,Department of Chemistry, Seattle University, 901 12th Ave, Seattle, Washington 98122, United States
| | - Mikaela E Ebner
- Department of Chemistry, Seattle University, 901 12th Ave, Seattle, Washington 98122, United States
| | - Wilma E Afunugo
- Department of Chemistry, Seattle University, 901 12th Ave, Seattle, Washington 98122, United States
| | - Alaina M Bever
- Department of Chemistry, Seattle University, 901 12th Ave, Seattle, Washington 98122, United States
| | - Shijie Cao
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195-5061, United States
| | - Yonghou Jiang
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195-5061, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, Washington 98195-5061, United States
| |
Collapse
|
106
|
Zhang W, Tang G, Dong H, Geng Q, Niu J, Tang J, Yang J, Huo H, Cao Y. Targeted release mechanism of λ-cyhalothrin nanocapsules using dopamine-conjugated silica as carrier materials. Colloids Surf B Biointerfaces 2019; 178:153-162. [DOI: 10.1016/j.colsurfb.2019.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 12/27/2022]
|
107
|
Reduction responsive liposomes based on paclitaxel-ss-lysophospholipid with high drug loading for intracellular delivery. Int J Pharm 2019; 564:244-255. [DOI: 10.1016/j.ijpharm.2019.04.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/02/2019] [Accepted: 04/20/2019] [Indexed: 11/20/2022]
|
108
|
Lin CY, Hsu CY, Elzoghby AO, Alalaiwe A, Hwang TL, Fang JY. Oleic acid as the active agent and lipid matrix in cilomilast-loaded nanocarriers to assist PDE4 inhibition of activated neutrophils for mitigating psoriasis-like lesions. Acta Biomater 2019; 90:350-361. [PMID: 30951898 DOI: 10.1016/j.actbio.2019.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/16/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
Both phosphodiesterase (PDE4) inhibitors and omega-9 fatty acids show anti-inflammatory activity for treating inflamed skin diseases, but their efficacy remains low. Combinatorial agents are anticipated to offer an advanced strategy for efficient therapy. We prepared cilomilast-loaded oleic acid (OA) nanocarriers to test the inhibitory capability against human neutrophil stimulation and a murine psoriasis model. OA played dual roles in the nanocarriers as both the active ingredient and lipid matrix in the nanoparticulate core. OA nanoparticles but not free OA could restrain calcium mobilization in activated neutrophils. The inhibition level of superoxide anion and elastase by cilomilast-loaded OA nanocarriers approximated that of free forms. In the mouse model, the intradermal nanosystems reduced imiquimod-induced epidermal thickening from 230.4 to 63.1 μm. Transepidermal water loss was decreased from 30.2 to 11.3 g/m2/h by integrated nanocarriers. The nanosystems mitigated neutrophil infiltration and hyperproliferation in the psoriasiform lesion via decreased expression of cytokines and chemokines. STATEMENT OF SIGNIFICANCE: The long-term therapy for psoriasis is unsatisfactory due to the possible adverse effects and inefficiency after prolonged use. Both phosphodiesterase (PDE4) inhibitors and omega-9 fatty acids such as oleic acid (OA) show anti-inflammatory activity for treating inflamed skin diseases. Combinatorial agents are anticipated to offer an advanced strategy for efficient therapy. OA is also ideal for incorporation into nanoparticles to enhance particulate emulsification, drug entrapment, and biocompatibility. We prepared cilomilast-loaded oleic acid (OA) nanocarriers to test the inhibitory capability against human neutrophil stimulation and a murine psoriasis lesion. OA nanocarriers are indigenous to prevent neutrophil activation and the deterioration of psoriatic lesion. Cilomilast incorporation in OA nanocarriers could further mitigate the clinical score and suppressing proinflammatory mediators.
Collapse
|
109
|
Jones SJ, Taylor AF, Beales PA. Towards feedback-controlled nanomedicines for smart, adaptive delivery. Exp Biol Med (Maywood) 2019; 244:283-293. [PMID: 30205721 PMCID: PMC6435888 DOI: 10.1177/1535370218800456] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
IMPACT STATEMENT The timing and rate of release of pharmaceuticals from advanced drug delivery systems is an important property that has received considerable attention in the scientific literature. Broadly, these mostly fall into two classes: controlled release with a prolonged release rate or triggered release where the drug is rapidly released in response to an environmental stimulus. This review aims to highlight the potential for developing adaptive release systems that more subtlety modulate the drug release profile through continuous communication with its environment facilitated through feedback control. By reviewing the key elements of this approach in one place (fundamental principles of nanomedicine, enzymatic nanoreactors for medical therapies and feedback-controlled chemical systems) and providing additional motivating case studies in the context of chronobiology, we hope to inspire innovative development of novel "chrononanomedicines."
Collapse
Affiliation(s)
- Stephen J. Jones
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Annette F. Taylor
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Paul A Beales
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
110
|
Rizvi SZH, Shah FA, Khan N, Muhammad I, Ali KH, Ansari MM, Din FU, Qureshi OS, Kim KW, Choe YH, Kim JK, Zeb A. Simvastatin-loaded solid lipid nanoparticles for enhanced anti-hyperlipidemic activity in hyperlipidemia animal model. Int J Pharm 2019; 560:136-143. [PMID: 30753932 DOI: 10.1016/j.ijpharm.2019.02.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/24/2019] [Accepted: 02/06/2019] [Indexed: 02/07/2023]
Abstract
The objective of current study was to develop solid lipid nanoparticles-loaded with simvastatin (SIM-SLNs) and investigate their in vivo anti-hyperlipidemic activity in poloxamer-induced hyperlipidemia model. Nano-template engineering technique was used to prepare SIM-SLNs with palmityl alcohol as lipid core and a mixture of Tween 40/Span 40/Myrj 52 to stabilize the core. The prepared SIM-SLNs were evaluated for physicochemical parameters including particle diameter, surface charge, morphology, incorporation efficiency, thermal behaviour and crystallinity. In vitro release profile of SIM-SLNs in simulated gastric and intestinal fluids was evaluated by using dialysis bag technique and anti-hyperlipidemic activity was assessed in hyperlipidemia rat model. SIM-SLNs revealed uniform particle size with spherical morphology, zeta potential of -24.9 mV and high incorporation efficiency (∼85%). Thermal behaviour and crystallinity studies demonstrated successful incorporation of SIM in the lipid core and its conversion to amorphous form. SIM-SLNs demonstrated a sustained SIM release from the lipid core of nanoparticles. SIM-SLNs significantly reduced the elevated serum lipids as indicated by ∼3.9 and ∼1.5-times decreased total cholesterol compared to those of untreated control and SIM dispersion treated hyperlipidemic rats. In conclusion, SIM-SLNs showed a great promise for improving the therapeutic outcomes of SIM via its effective oral delivery.
Collapse
Affiliation(s)
- Syed Zaki Husain Rizvi
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Namrah Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Iftikhar Muhammad
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Khan Hashim Ali
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Muhammad Mohsin Ansari
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Fakhar Ud Din
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | | | - Kyoung-Won Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi, Republic of Korea
| | - Yeong-Hwan Choe
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi, Republic of Korea
| | - Jin-Ki Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi, Republic of Korea.
| | - Alam Zeb
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan.
| |
Collapse
|
111
|
Chen Z, Zhang Z, Chen M, Xie S, Wang T, Li X. Synergistic antitumor efficacy of hybrid micelles with mitochondrial targeting and stimuli-responsive drug release behavior. J Mater Chem B 2019; 7:1415-1426. [PMID: 32255012 DOI: 10.1039/c8tb02843e] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The term synergism means that the overall therapeutic benefits should be greater than the sum of the effects of individual agents and that the optimal therapeutic efficacy can be achieved at reduced doses. Micellar systems usually fail to deliver multiple drugs to target sites at synergistic doses and thus are not able to maximize the antitumor efficacy. In the current study, we demonstrate a strategy to coordinate the release of camptothecin (CPT) and α-tocopheryl succinate (TOS) from hybrid micelles for nucleus and mitochondrion interferences. TOS is decorated with cationic triphenylphosphonium (TPP) to promote the targeting capability of TOS-TPP to mitochondria. The combination of CPT and TOS-TPP shows strong synergistism with a combination index of 0.186. Hyaluronic acid (HA) is conjugated with CPT or TOS-TPP via disulfide linkages for tumor cell targeting and intracellular reduction-triggered release. Both conjugates either separately self-assemble into MC and MT micelles, or are blended at different ratios to form MC-T hybrid micelles. In response to elevated intracellular glutathione levels, the coordinated release of CPT and TOS-TPP from MC-T results in a combination index of 0.26 and the dose-reduction indexes of CPT and TOS are 7.7 and 3.4, respectively. Compared with MC and MT, MC-T micelles with 5 fold lower doses exhibit higher intracellular reactive oxygen species (ROS) levels, comparable tumor growth inhibition and animal survival, indicating no hematologic and intestinal toxicities. Moreover, the HA conjugates of MC-T are linked to polylactide via acid-labile linkages and electrospun into short fibers (MC-T@SF) as an injectable depot to release MC-T in response to the acidic tumor microenvironment. At a predetermined synergistic ratio, MC-T@SF with 5 fold lower doses achieves antitumor profiles comparable to those of individual micelle-loaded short fibers. Therefore, the hybrid micelles and micelle-releasing short fibers represent a feasible strategy to synergistically enhance the therapeutic efficacy and enable significant reduction in effective doses of chemotherapeutic agents.
Collapse
Affiliation(s)
- Zhoujiang Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China.
| | | | | | | | | | | |
Collapse
|
112
|
Novel biosurfactant and lipid core-shell type nanocapsular sustained release system for intravenous application of methotrexate. Int J Pharm 2019; 557:86-96. [DOI: 10.1016/j.ijpharm.2018.12.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 01/08/2023]
|
113
|
Nanocarrier-mediated Delivery of CORM-2 Enhances Anti-allodynic and Anti-hyperalgesic Effects of CORM-2. Mol Neurobiol 2019; 56:5539-5554. [PMID: 30637664 DOI: 10.1007/s12035-019-1468-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/04/2019] [Indexed: 12/16/2022]
Abstract
Neuropathic pain is a devastating chronic condition and effective treatments are still lacking. Carbon monoxide-releasing molecule-2 (CORM-2) as a carbon monoxide (CO) carrier, exerts potent anti-neuropathic pain effects; however, its poor water solubility and short half-life hinder its clinical utility. Therefore, the aim of this study was to investigate whether CORM-2-loaded solid lipid nanoparticles (CORM-2-SLNs) enhance the anti-allodynic and anti-hyperalgesic effects of CORM-2 in a rat chronic constriction injury (CCI) model. CORM-2-SLNs were prepared using a nanotemplate engineering technique with slight modifications. The physiochemical properties of CORM-2-SLNs were characterized and CO release from CORM-2-SLNs was assessed using a myoglobin assay. CO was slowly released from CORM-2-SLNs, was observed, and the half-life of CO release was 50 times longer than that of CORM-2. In vivo results demonstrate that intraperitoneal administration of CORM-2-SLNs (5 and 10 mg/kg/day, ip) once daily for seven consecutive days significantly reduced the mechanical allodynia and mechanical hyperalgesia compared with CORM-2 (10 mg/kg/day, ip). RT-PCR and Western blot analyses on days 7 and 14, revealed that treatment with CORM-2-SLNs resulted in greater reductions in the CCI-elevated levels of heme-oxygenase-2 (HO-2); inducible nitric oxide synthase (iNOS); neuronal NOS (nNOS); and inflammatory mediators (TNF-α, IBA-1, and GFAP) in the spinal cord and dorsal root ganglions compared with treatment with CORM-2. In contrast, HO-1 and IL-10 were significantly increased in the CORM-2-SLN-treated group compared with the group treated with CORM-2. These data indicate that CORM-2-SLNs are superior to CORM-2-S in alleviating mechanical allodynia and mechanical hyperalgesia.
Collapse
|
114
|
Surface biofunctionalization of the decellularized porcine aortic valve with VEGF-loaded nanoparticles for accelerating endothelialization. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 97:632-643. [PMID: 30678950 DOI: 10.1016/j.msec.2018.12.079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/09/2018] [Accepted: 12/24/2018] [Indexed: 12/15/2022]
Abstract
The original intention for building a tissue-engineered heart valve (TEHV) was to simulate a normal heart valve and overcome the insufficiency of the commonly used heart valve replacement in the clinic. The endothelialization of the TEHV is very important as the endothelialized TEHV can decrease platelet adhesion and delay the valvular calcification decline process. In this work, we encapsulated vascular endothelial growth factor (VEGF) into polycaprolactone (PCL) nanoparticles. Then, through the Michael addition reaction, PCL nanoparticles were introduced onto the decellularized aortic valve to prepare a hybrid valve. The encapsulation efficiency of the PCL nanoparticles for VEGF was up to 82%, and the in vitro accumulated release rate was slow without an evident initial burst release. In addition, the hybrid valve had a decreased hemolysis ratio and possessed antiplatelet adhesion capacity, and it was able to promote the adhesion and proliferation of endothelial cells, covering the surface with a dense cell layer to accelerate endothelialization. An experiment involving the subcutaneous implant in SD rats showed that at week 8, lots of blood capillaries were formed in the hybrid valve. Mechanics performance testing indicated that the mechanical property of the hybrid valve was partly improved. Taken together, we applied a nano-drug controlled release system to fabricate TEHV, and provide an approach for the biofunctionalization of the TEHV scaffold for accelerating endothelialization.
Collapse
|
115
|
Enhanced drug delivery, mechanical properties and antimicrobial activities in poly(lactic acid) nanofiber with mesoporous Fe3O4-COOH nanoparticles. Colloids Surf A Physicochem Eng Asp 2018. [DOI: 10.1016/j.colsurfa.2018.09.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
116
|
Katiyar SS, Kushwah V, Dora CP, Jain S. Lipid and TPGS based novel core-shell type nanocapsular sustained release system of methotrexate for intravenous application. Colloids Surf B Biointerfaces 2018; 174:501-510. [PMID: 30497012 DOI: 10.1016/j.colsurfb.2018.11.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/11/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022]
Abstract
Core shell nanocapsules present an interesting system for attaining high loading of drug. In an attempt, lipid and TPGS based novel core-shell nanocapsule were prepared to achieve high drug loading with sustained release of model hydrophilic drug methotrexate (MTX). Antisolvent nanoprecipitation was utilized for the formulation of nanoparticles. Optimized formulation depicted 223.6 ± 24.1 nm particle size, 0.243 ± 0.034 PDI, zeta potential -2.07 ± 0.51 mV and 15.03 ± 1.92%drug loading. In vitro release showed biphasic release for 12 h with initial burst phase followed by sustained release phase. Haemolytic study on RBCs revealed haemocompatible nature of MTX-TPGS nanoparticles compared to Biotrexate® (Zydus). In vitro cell culture studies depicted 3 folds and 2.66 folds increase in cellular uptake of MTX at 10 μg/ml and 15 μg/ml respectively for developed nanoparticles with 3.81 folds decrease in IC50 value as compared to Biotrexate®. Higher apoptosis and increased lysosomal membrane permeability were also depicted by MTX-TPGS nanoparticles. 2.45 folds increase in AUC and 3.68 folds increase in T1/2 was achieved in pharmacokinetic study. Significant reduction in tumor burden and serum biochemical parameters depicted efficacy and safety respectively of the formulation as compared to Biotrexate®. RBCs morphology was retained after MTX-TPGS exposure proving its haemocompatibility in vivo.
Collapse
Affiliation(s)
- Sameer S Katiyar
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, SAS Nagar, Punjab, 160 062, India
| | - Varun Kushwah
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, SAS Nagar, Punjab, 160 062, India
| | - Chander Parkash Dora
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, SAS Nagar, Punjab, 160 062, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, SAS Nagar, Punjab, 160 062, India.
| |
Collapse
|
117
|
Ali I, Rehman JU, Ullah S, Imran M, Javed I, El-Haj BM, Saad Ali H, Arfan M, Shah MR. Preliminary investigation of novel tetra-tailed macrocycle amphiphile based nano-vesicles for amphotericin B improved oral pharmacokinetics. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1204-S1214. [PMID: 30453792 DOI: 10.1080/21691401.2018.1536061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Supramolecular macrocycles-based drug delivery systems are receiving wider recognition due to their self-assembly into nanostructures with unique characteristics. This study reports synthesis of resorcinarene-based novel and biocompatible amphiphilic supramolecular macrocycle that self-assembles into nano-vesicular system for Amphotericin B (Am-B) delivery, a model hydrophobic drug. The macrocycle was synthesized through a two-step reaction and was characterized with 1 H NMR and mass spectrometric techniques. Its biocompatibility was assessed in cancer cell lines, blood and animals. Its critical micelle concentration (CMC) was determined using UV spectrophotometer. Am-B loaded in novel macrocycle-based vesicles were examined according to their shape, size, surface charge, drug entrapment efficiency and excepients compatibility using atomic force microscope (AFM), Zetasizer, HPLC and FT-IR spectroscopy. Drug-loaded vesicles were also investigated for their in-vitro release, stability and in-vivo oral bioavailability in rabbits. The macrocycle was found to be nontoxic against cancer cells, haemo-compatible and safe in mice and revealed lower CMC. It formed mono-dispersed spherical shape vesicles of 174.4 ± 3.78 nm in mean size. Vesicles entrapped 92.05 ± 4.39% drug and were stable upon storage with gastric-simulated fluid and increased the drug oral bioavailability in rabbits. Results confirmed novel macrocycle as biocompatible vesicular nanocarrier for enhancing the oral bioavailability of lipophilic drugs.
Collapse
Affiliation(s)
- Imdad Ali
- a H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences , Karachi University , Karachi , Pakistan
| | - Jawad Ur Rehman
- a H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences , Karachi University , Karachi , Pakistan
| | - Shafi Ullah
- a H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences , Karachi University , Karachi , Pakistan
| | - Muhammad Imran
- a H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences , Karachi University , Karachi , Pakistan
| | - Ibrahim Javed
- b ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Department of Drug Delivery , Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville , Australia
| | - Babiker M El-Haj
- c Department of Pharmaceutical Sciences , College of Pharmacy, Ajman University , Ajman , UAE
| | - Heyam Saad Ali
- d Department of Pharmaceutics , Dubai Pharmacy College , Dubai , UAE
| | - Muhammad Arfan
- a H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences , Karachi University , Karachi , Pakistan
| | - Muhammad Raza Shah
- a H.E.J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences , Karachi University , Karachi , Pakistan
| |
Collapse
|
118
|
Lipid-bilayer-coated nanogels allow for sustained release and enhanced internalization. Int J Pharm 2018; 551:8-13. [DOI: 10.1016/j.ijpharm.2018.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
|
119
|
Maher S, Kumeria T, Aw MS, Losic D. Diatom Silica for Biomedical Applications: Recent Progress and Advances. Adv Healthc Mater 2018; 7:e1800552. [PMID: 30118185 DOI: 10.1002/adhm.201800552] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/12/2018] [Indexed: 12/30/2022]
Abstract
Diatoms are unicellular photosynthetic algae enclosed in porous 3D nanopatterned silica enclosures called "frustules." The diatom frustules are made from biosilica self-assembled into intricate porous shells that feature unique properties including high specific surface area, biocompatibility, tailorable surface chemistry, thermal stability, and high mechanical and chemical resistance. The ability to cultivate diatoms in artificial environments and their abundant availability of diatom frustules as mineable fossilized mineral deposits (diatomite or diatomaceous earth; DE) make diatom silica a promising natural alternative to synthetic porous silica for a broad range of biomedical, environmental, agricultural, and energy applications. This review article provides a comprehensive and current account of the use of natural DE silica materials in biomedical applications focused mainly on drug delivery with some highlights on biosensing, tissue engineering, and clotting agents. The article also covers some basic physical and chemical aspects of DE material such as purification, surface chemical functionalization, biocompatibility, and cellular uptake that are critical for the development of an efficient drug carrier.
Collapse
Affiliation(s)
- Shaheer Maher
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
| | - Tushar Kumeria
- School of Pharmacy The University of Queensland Pharmacy Australia Center of Excellence Building Woolloongabba Queensland 4102 Australia
| | - Moom Sin Aw
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
| | - Dusan Losic
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
| |
Collapse
|
120
|
Wang Q, Newby BMZ. Layer-by-layer Polyelectrolytes Coating of Alginate Microgels for Sustained Release of Sodium Benzoate and Zosteric Acid. J Drug Deliv Sci Technol 2018; 46:46-54. [PMID: 30555539 PMCID: PMC6289541 DOI: 10.1016/j.jddst.2018.04.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The potential of sustaining release of very small (Mw < 250 g/mol) hydrophilic drugs up to several days from layer-by-layer (LbL) polyelectrolyte coated alginate microgels (Alg-Ms) was investigated. One purpose is to minimize post-surgical adhesions, which develop in 12 h to 3 days after surgery. The LbL polyelectrolyte layer would serve as a diffusion barrier for their release. The LbL polyelectrolyte bilayers were prepared using poly(allylamine) (PAH) and poly(styrene sulfonate) (PSS). Sodium benzoate (NaB, Mw = 144 g/mol) and zosteric acid (ZA, Mw = 244 g/mol), two anti-inflammatory and anti-microbial compounds, were used as model drugs. A higher number of PAH/PSS bilayer lead to a greater sustained release of both drugs, and with 4 bilayers, the release of NaB and ZA was prolonged from 24 h to 72 h and 120 h, respectively. Fitting the data to the Ritger-Peppas' equation showed that as the bilayer number increased, the release constant and/or exponent decreased, indicating the LbL PAH/PSS bilayer effectively reduced the permeability of these two very small hydrophilic drugs. The ability to prolong the release of such small hydrophilic molecules, which has rarely been investigated previously, would find broad applications in fields such as anti-adhesion treatment and antifouling coatings.
Collapse
Affiliation(s)
- Qing Wang
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH, 44325-3906, United States
| | - Bi-min Zhang Newby
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH, 44325-3906, United States
| |
Collapse
|
121
|
Formulation and in vitro evaluation of magnetoliposomes as a potential nanotool in colorectal cancer therapy. Colloids Surf B Biointerfaces 2018; 171:553-565. [PMID: 30096477 DOI: 10.1016/j.colsurfb.2018.07.070] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/27/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022]
Abstract
Magnetoliposomes (MLPs) offer many new possibilities in cancer therapy and diagnosis, including the transport of antitumor drugs, hyperthermia treatment, detection using imaging techniques, and even cell migration. However, high biocompatibility and functionality after cell internalization are essential to their successful application. We synthesized maghemite nanoparticles (γ-Fe2O3) by oxidizing magnetite cores (Fe3O4) and coating them with phosphatidylcholine (PC) liposomes, obtained using the thin film hydration method, to generate MLPs. The MLPs were tested in vitro, using human tumor and non-tumor colon cell lines, for cytotoxicity, cell uptake and cellular distribution, and magnetically-induced cell mobility. In addition, blood cells biocompatibility studies were performed. The mean size of the MLPs, with a core of γ-Fe2O3 completely surrounded by PC liposomes, was 90 ± 20 nm, showing a soft magnetic character and a great biocompatibility in all the cell lines assayed including blood cells. Prussian blue staining showed a high MLP cell uptake with maximum internalization at 24 h. TEM analysis showed the MLPs surrounded by the cell membrane and in the cell periphery, suggesting internalization by endocytosis and/or macropinocytosis. Interestingly, the mitochondria presented MLP accumulations, particularly in tumor cells. Finally, MLPs within colon cancer cells were able to induce cell migration when a magnetic field was applied in vitro, indicating the functionality of our nanoformulation. A promising biomedical application of these MLPs is anticipated based on their physical, chemical and biological properties.
Collapse
|
122
|
Gatti THH, Eloy JO, Ferreira LMB, Silva ICD, Pavan FR, Gremião MPD, Chorilli M. Insulin-loaded polymeric mucoadhesive nanoparticles: development, characterization and cytotoxicity evaluation. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000117314] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
123
|
Chen G, Svirskis D, Lu W, Ying M, Huang Y, Wen J. N-trimethyl chitosan nanoparticles and CSKSSDYQC peptide: N-trimethyl chitosan conjugates enhance the oral bioavailability of gemcitabine to treat breast cancer. J Control Release 2018; 277:142-153. [PMID: 29548985 DOI: 10.1016/j.jconrel.2018.03.013] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/10/2018] [Accepted: 03/12/2018] [Indexed: 10/17/2022]
Abstract
Gemcitabine is a nucleoside analogue effective against a number of cancers. However, the full potential of this drug has not been realised, in part due to low oral bioavailability and frequent dosing requirements. This study reports the synthesis, in-vitro, ex-vivo and in-vivo evaluation of trimethyl chitosan (TMC) - CSKSSDYQC (CSK) peptide conjugates capable of enhancing the oral bioavailability of gemcitabine due to the ability to target intestinal goblet cells and promote intestinal cellular uptake. TMC was synthesized by a novel two-step methylation method to improve quanternization and yield. The CSK-TMC conjugates were prepared by ionic gelation to achieve particles sized at 173.6 ± 6.8 nm, zeta potential of +18.5 ± 0.2 mV and entrapment efficiency of 66.4 ± 0.1%, capable of sustained drug release. By encapsulating gemcitabine into CSK-TMC conjugates, an increased amount of drug permeated through porcine intestinal epithelial membranes compared with the unconjugated TMC nanoparticles (NPs). The rate of cellular uptake of drug loaded conjugates into HT29-MTX-E12 intestinal goblet cells, was time- and concentration-dependant. The conjugates underwent active transport associated with adsorptive mediated, clathrin and caveolae mediated endocytosis. In cellular transport studies, drug loaded conjugates had greater drug transport capability compared with drug solution and TMC NPs over the co-cultured Caco-2/HT29-MTX-E12 cell monolayer. The drug loaded conjugates exhibited electrostatic interaction with the intestinal epithelial cells. Both P-glycoprotein (P-gp) and multiple resistance protein-2 (MRP2) efflux affected the cellular transport of the conjugates. Importantly, during the pharmacokinetic studies, the orally administrated drug loaded into TMC NPs showed an improved oral bioavailability of 54.0%, compared with gemcitabine solution of 9.9%. Notable, the CSK-TMC conjugates further improved oral bioavailability to 60.1% and reduced the tumour growth rate in a BALB/c nude mouse model, with a 5.1-fold and 3.3-fold reduction compare with the non-treated group and gemcitabine solution group. Furthermore, no major evidence of toxicity was discernible on histologic studies of selected organs. In conclusion, the presented CSK-TMC conjugates and TMC nanoparticles both significantly improve the oral bioavailability of gemcitabine and have the potential for the treatment of breast cancer.
Collapse
Affiliation(s)
- Guanyu Chen
- School of Pharmacy, Faculty of Medical and Health Science, University of Auckland, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Science, University of Auckland, New Zealand
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, China
| | - Man Ying
- Department of Pharmaceutics, School of Pharmacy, Fudan University, China
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, China
| | - Jingyuan Wen
- School of Pharmacy, Faculty of Medical and Health Science, University of Auckland, New Zealand.
| |
Collapse
|
124
|
Caldera F, Argenziano M, Trotta F, Dianzani C, Gigliotti L, Tannous M, Pastero L, Aquilano D, Nishimoto T, Higashiyama T, Cavalli R. Cyclic nigerosyl-1,6-nigerose-based nanosponges: An innovative pH and time-controlled nanocarrier for improving cancer treatment. Carbohydr Polym 2018; 194:111-121. [PMID: 29801818 DOI: 10.1016/j.carbpol.2018.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/05/2018] [Accepted: 04/05/2018] [Indexed: 11/26/2022]
Abstract
The design and structural optimisation of a novel polysaccharide-based nanomaterial for the controlled and sustained release of doxorubicin are here reported. A cross-linked polymer was obtained by reacting a tetraglucose, named cyclic nigerosyl-1-6-nigerose (CNN), with pyromellitic dianhydride. The cross-linking reaction formed solid nanoparticles, named nanosponges, able to swell as a function of the pH. Nanoparticle sizes were reduced using High Pressure Homogenization, to obtain uniform nanosuspensions. Doxorubicin was incorporated into the CNN-nanosponges in a good extent. DSC and solid state NMR analyses proved the drug interaction with the polymer matrix. In vitro studies demonstrated pH-dependent slow and prolonged release kinetics of the drug from the nanoformulation. Doxorubicin-loaded CNN-nanosponges were easily internalized in A2780 cell line. They might considered an intracellular doxorubicin reservoir, able to slowly release the drug over time. CNN-nanosponges may be promising biocompatible nanocarriers for the sustained delivery of doxorubicin with potential localised application in cancer treatments.
Collapse
Affiliation(s)
- F Caldera
- Dipartimento di Chimica-Università di Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - M Argenziano
- Dipartimento di Scienza e Tecnologia del Farmaco-Università di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - F Trotta
- Dipartimento di Chimica-Università di Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - C Dianzani
- Dipartimento di Scienza e Tecnologia del Farmaco-Università di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - L Gigliotti
- Dipartimento di Scienze della Salute, UPO, Via Solaroli 17, 28100 Novara, Italy
| | - M Tannous
- Dipartimento di Chimica-Università di Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - L Pastero
- Dipartimento di Scienze della Terra, Università di Torino, Via V. Caluso 35, 10125 Torino, Italy
| | - D Aquilano
- Dipartimento di Scienze della Terra, Università di Torino, Via V. Caluso 35, 10125 Torino, Italy
| | - T Nishimoto
- Division Manager New Material Development Division R&D Center Hayashibara Co., Ltd. 675-1 Fujisaki, Naka-ku, Okayama 702-8006, Japan
| | - T Higashiyama
- NAGASE (EUROPA) GmbH/Hayashibara-Immermannstrasse 65c, 40210 Düsseldorf, Germany
| | - R Cavalli
- Dipartimento di Scienza e Tecnologia del Farmaco-Università di Torino, Via P. Giuria 9, 10125 Torino, Italy.
| |
Collapse
|
125
|
Takalkar D, Desai N. Nanolipid Gel of an Antimycotic Drug for Treating Vulvovaginal Candidiasis-Development and Evaluation. AAPS PharmSciTech 2018; 19:1297-1307. [PMID: 29340981 DOI: 10.1208/s12249-017-0918-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/03/2017] [Indexed: 01/05/2023] Open
Abstract
This paper focuses on the development and evaluation of mucoadhesive vaginal gel of fluconazole using nanolipid carriers to enhance tissue deposition in treating vulvovaginal candidiasis. Treatment of vulvovaginal candidiasis includes antimycotic agents prescribed for 1 to 7 days or longer, in relapse either orally or topically. The delivery of fluconazole as nanolipid carriers in vaginal gel can be proposed as suitable alternative to the existing conventional formulations to improve the patient acceptability, compliance and localized drug action. The nanolipid carriers of fluconazole were prepared by phase inversion temperature technique and incorporated into Carbopol 974P as gelling polymer. GRAS excipients selected and optimized were Precirol ATO 5, oleic acid and Kolliphor RH 40 to produce nanolipid dispersions. Stable nanolipid dispersions were developed using sodium dodecyl sulfate as the charge inducer. The optimized nanolipid dispersion of fluconazole had particle size, polydispersity index and zeta potential value of 158.33 ± 2.55 nm, 0.278 ± 0.003 and - 27.33 ± 0.40 mV, respectively and the average entrapment of fluconazole in the lipid carriers was found to be 67.24 ± 0.87%. The optimized vaginal gel had satisfactory mucoadhesive strength and rheological properties to facilitate vaginal application. The fluconazole release from the gel was sustained showing 30.69 ± 1.02% drug deposition in the porcine vaginal mucosa at the end of 8 h with improved antifungal activity against Candida albicans during well diffusion studies. The optimized gel was non-irritant to the vaginal mucosa of female Wistar rats with no signs of erythema or edema.
Collapse
|
126
|
Fizir M, Dramou P, Zhang K, Sun C, Pham-Huy C, He H. Polymer grafted-magnetic halloysite nanotube for controlled and sustained release of cationic drug. J Colloid Interface Sci 2017. [DOI: 10.1016/j.jcis.2017.04.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
127
|
Ni XL, Chen LX, Zhang H, Yang B, Xu S, Wu M, Liu J, Yang LL, Chen Y, Fu SZ, Wu JB. In vitro and in vivo antitumor effect of gefitinib nanoparticles on human lung cancer. Drug Deliv 2017; 24:1501-1512. [PMID: 28961023 PMCID: PMC8241075 DOI: 10.1080/10717544.2017.1384862] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 01/31/2023] Open
Abstract
Gefitinib (GEF) is the first epidermal growth factor receptor (EGFR)-targeting agent launched as an anticancer drug. It is an accepted opinion that modifying GEF strong hydrophobicity and poor bioavailability would not only enhance its antitumor effects, but also reduce its side effects. In this study, GEF-loadedpoly(ε-caprolactone)-poly(ethyleneglycol)-poly(ε-caprolactone) (PCEC) -bearing nanoparticles (GEF-NPs) were prepared by a solid dispersion method and characterized. The particle sizes increased with the increase in GEF/PCEC mass ratio in feed. GEF-NPs (10%) were mono-dispersed, smaller than 24 nm, zeta potential was approximately -18 mV, percentage encapsulation and loading, were more than 9% and 92%, respectively, and drug was slowly released but without a biphasic pattern. Microscopy studies of the optimized formulation confirmed that the prepared nanoparticles are spherical in nature. Cytotoxicity results indicated that cell growth inhibition induced by free GEF and GEF-NPs were dose and time dependent. Compared with free GEF, GEF-NPs enhanced antitumor effects, reduced side effects and significantly prolonged survival time in vivo. CD31, ki-67 and EGFR expression were significantly lower in the GEF-NPs group compared with other groups (p< .05). These findings demonstrated that GEF-NPs have the potential to attain superior outcomes and to overcome complications such as organs toxicity, therapeutic resistance and disease relapse.
Collapse
Affiliation(s)
- Xiao Ling Ni
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Long Xia Chen
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Heng Zhang
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Bo Yang
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Shan Xu
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Min Wu
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Jing Liu
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Ling Lin Yang
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Yue Chen
- Department of Nuclear Medicine, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Shao Zhi Fu
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| | - Jing Bo Wu
- Department of Oncology, the Affiliated hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
128
|
Silva KCG, Sato ACK. Biopolymer gels containing fructooligosaccharides. Food Res Int 2017; 101:88-95. [DOI: 10.1016/j.foodres.2017.08.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 12/17/2022]
|
129
|
Meng F, Wang S, Wang Y, Liu H, Huo X, Ma H, Ma Z, Xiong H. Microencapsulation of oxalic acid via oil-in-oil (O/O) emulsion solvent evaporation. POWDER TECHNOL 2017. [DOI: 10.1016/j.powtec.2017.07.073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
130
|
He N, Chen Z, Yuan J, Zhao L, Chen M, Wang T, Li X. Tumor pH-Responsive Release of Drug-Conjugated Micelles from Fiber Fragments for Intratumoral Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:32534-32544. [PMID: 28876891 DOI: 10.1021/acsami.7b09519] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The tumor accumulation of micelles is essential to enhance the cellular uptake and extend the release of chemotherapeutic agents. In the previous study camptothecin (CPT)-conjugated micelles (MCPT) were constructed with disulfide linkages and folate moieties for reduction-sensitive release and cell-selective uptake. This study proposes a strategy to integrate the promicelle polymers (PMCPT) into fiber fragments for intratumoral injection, realizing acid-liable release of PMCPT in response to acidic tumor microenvironment and spontaneous self-assembly into MCPT. Acid-liable 2-propionic-3-methylmaleic anhydride (CDM)-linked poly(ethylene glycol) initiates the ring-opening polymerization of dl-lactide as the fiber matrix. There is no apparent burst release of MCPT from fiber fragments and around 80% of accumulated releases after incubation in pH 6.5 buffers for 40 days. Compared to MCPT freshly prepared via solvent evaporation, the micelles released from fiber fragments reveal similar profiles, such as folate-mediated cellular uptake and glutathione-sensitive drug release. Taking advantage of the aggregation-induced emission (AIE) effect of tetraphenylethylene (TPE) derivatives, TPE-conjugated micelles (MTPE) have been successfully been used to track the self-assembly into micelles after release from fibers and subsequent cell internalization into cytosol. The self-assembly induced fluorescence light-up was also detected after intratumoral injection of fiber fragments. Compared with CPT-loaded fiber fragments and intratumoral or intravenous injection of free MCPT, the sustained release from fiber fragments and high accumulation of micelles in tumors result in significantly higher inhibition of tumor growths, prolongation of animal survival, and induction of tumor cell apoptosis. Thus, the integration of double targeting and double stimuli responsiveness into fragmented fibers provides a feasible strategy to realize the sustained micelle release from fibers and promote the therapeutic efficacy.
Collapse
Affiliation(s)
- Nan He
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| | - Zhoujiang Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| | - Jiang Yuan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| | - Long Zhao
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| | - Maohua Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| | - Tao Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| | - Xiaohong Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| |
Collapse
|
131
|
Jurj A, Braicu C, Pop LA, Tomuleasa C, Gherman CD, Berindan-Neagoe I. The new era of nanotechnology, an alternative to change cancer treatment. Drug Des Devel Ther 2017; 11:2871-2890. [PMID: 29033548 PMCID: PMC5628667 DOI: 10.2147/dddt.s142337] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In the last few years, nanostructures have gained considerable interest for the safe delivery of therapeutic agents. Several therapeutic approaches have been reported, such as molecular diagnosis, disease detection, nanoscale immunotherapy and anticancer drug delivery that could be integrated into clinical use. The current paper aims to highlight the background that supports the use of nanoparticles conjugated with different types of therapeutic agents, applicable in targeted therapy and cancer research, with a special emphasis on hematological malignancies. A particular key point is the functional characterization of nonviral delivery systems, such as gold nanoparticles, liposomes and dendrimers. The paper also presents relevant published data related to microRNA and RNA interference delivery using nanoparticles in cancer therapy.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy “Iuliu-Hatieganu”, Cluj-Napoca, Romania
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy “Iuliu-Hatieganu”, Cluj-Napoca, Romania
| | - Laura-Ancuta Pop
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy “Iuliu-Hatieganu”, Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy “Iuliu-Hatieganu”, Cluj-Napoca, Romania
- Department of Hematology, The Oncology Institute “Prof Dr Ion Chiricuta”, Cluj-Napoca, Romania
| | - Claudia Diana Gherman
- Practical Abilities, Department of Medical Education, University of Medicine and Pharmacy “Iuliu-Hatieganu”, Cluj-Napoca, Romania
- Department of Medical Education, University of Medicine and Pharmacy “Iuliu Hatieganu”, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy “Iuliu-Hatieganu”, Cluj-Napoca, Romania
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof Dr Ion Chiricuta”, Cluj-Napoca, Romania
- MedFuture Research Center for Advanced Medicine, University of Medicine and Pharmacy “Iuliu-Hatieganu”, Cluj-Napoca, Romania
| |
Collapse
|
132
|
Evaluation of in vitro efficacy of docetaxel-loaded calcium carbonate aragonite nanoparticles (DTX-CaCO 3NP) on 4T1 mouse breast cancer cell line. In Vitro Cell Dev Biol Anim 2017; 53:896-907. [PMID: 28916966 DOI: 10.1007/s11626-017-0197-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 08/09/2017] [Indexed: 10/18/2022]
Abstract
Cockle shell-derived calcium carbonate nanoparticles have shown promising potentials as slow drug-releasing compounds in cancer chemotherapy. In this study, we evaluated the in vitro efficacy of docetaxel (DTX)-loaded CaCO3NP on 4T1 cell line. This was achieved by evaluating the following: cytotoxicity using MTT assay, fluorescence imaging, apoptosis with Annexin V assay, cell cycle analysis, scanning (SEM) and transmission electron microscopy (TEM), and scratch assay. Based on the results, DTX-CaCO3NP with a DTX concentration of 0.5 μg/mL and above had comparable cytotoxic effects with free DTX at 24 h, while all concentrations had similar cytotoxic effect on 4T1 cells at 48 and 72 h. Fluorescence and apoptosis assay showed a higher (p < 0.05) number of apoptotic cells in both free DTX and DTX-CaCO3NP groups. Cell cycle analysis showed cycle arrest at subG0 and G2/M phases in both treatment groups. SEM showed presence of cellular blebbing, while TEM showed nuclear fragmentation, apoptosis, and vacuolation in the treatment groups. Scratch assay showed lower (p < 0.05) closure in both free DTX and DTX-CaCO3NP groups. The results from this study showed that DTX-CaCO3NP has similar anticancer effects on 4T1 cells as free DTX, and since it has a slow release rate, it is a more preferred substitute for free DTX.
Collapse
|
133
|
Schwarzl R, Du F, Haag R, Netz RR. General method for the quantification of drug loading and release kinetics of nanocarriers. Eur J Pharm Biopharm 2017; 116:131-137. [DOI: 10.1016/j.ejpb.2016.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/20/2016] [Accepted: 12/20/2016] [Indexed: 10/20/2022]
|
134
|
Menagen B, Pedahzur R, Avnir D. Sustained release from a metal - Analgesics entrapped within biocidal silver. Sci Rep 2017; 7:4161. [PMID: 28646180 PMCID: PMC5482836 DOI: 10.1038/s41598-017-03195-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/27/2017] [Indexed: 11/25/2022] Open
Abstract
Matrices for sustained release of drugs have been based on polymers, biomaterials and oxides. The use of the major family of metals as matrices for sustained release is, to the best of our knowledge, unknown. In this context we describe a new family of bio-composites for sustained release of drugs, namely analgesic drugs entrapped within metallic silver. Synthetic methodologies were developed for the preparation of ibuprofen@Ag, naproxen@Ag, tramadol@Ag and bupivacaine@Ag composites. Detailed kinetic analysis of the release of the drugs from within the metal, is provided, demonstrating that metals can indeed serve as reservoirs for drug release. The metal in our case acts not only as a drug releasing source, but also as an antibacterial agent and this property of the composites was studied. Unexpectedly, it was found that the entrapment of the analgesics within silver, dramatically enhances the growth inhibition activity of wild type Pseudomonas aeruginosa, exceeding by far the inhibition activity of the separate components. A mechanism for this interesting observation is provided. The strong antimicrobial activity combined with the analgesic activity open the road for future applications of these materials as dual-purpose components in wound treatment.
Collapse
Affiliation(s)
- Barak Menagen
- Institute of Chemistry and the Center for Nanoscience and Nanotechnology, the Hebrew University of Jerusalem, Jerusalem, 9190402, Israel
| | - Rami Pedahzur
- Department of Environmental Health, Hadassah Academic College, Jerusalem, 91010, Israel
| | - David Avnir
- Institute of Chemistry and the Center for Nanoscience and Nanotechnology, the Hebrew University of Jerusalem, Jerusalem, 9190402, Israel.
| |
Collapse
|
135
|
Qureshi OS, Kim HS, Zeb A, Choi JS, Kim HS, Kwon JE, Kim MS, Kang JH, Ryou C, Park JS, Kim JK. Sustained release docetaxel-incorporated lipid nanoparticles with improved pharmacokinetics for oral and parenteral administration. J Microencapsul 2017; 34:250-261. [PMID: 28557649 DOI: 10.1080/02652048.2017.1337247] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of this study was to develop docetaxel-incorporated lipid nanoparticles (DTX-NPs) to improve the pharmacokinetic behaviour of docetaxel (DTX) after oral and parenteral administration via sustained release. DTX-NPs were prepared by nanotemplate engineering technique with palmityl alcohol as a solid lipid and Tween-40/Span-40/Myrj S40 as a surfactants mixture. Spherical DTX-NPs below 100 nm were successfully prepared with a narrow particle size distribution, 96% of incorporation efficiency and 686 times increase in DTX solubility. DTX-NPs showed a sustained release over 24 h in phosphate-buffered saline and simulated gastric and intestinal fluids, while DTX-micelles released DTX completely within 12 h. The half-maximal inhibitory concentration (IC50) of DTX-NPs against human breast cancer MCF-7 cells was 1.9 times lower than that of DTX-micelles and DTX solution. DTX-NPs demonstrated 3.7- and 2.8-fold increase in the area under the plasma concentration-time curve compared with DTX-micelles after oral and parenteral administration, respectively.
Collapse
Affiliation(s)
- Omer Salman Qureshi
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea.,b Faculty of Pharmacy , The University of Lahore , Lahore , Punjab , Pakistan
| | - Hyung-Seo Kim
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea
| | - Alam Zeb
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea.,c Riphah Institute of Pharmaceutical Sciences , Riphah International University , Islamabad , Pakistan
| | - Jin-Seok Choi
- d College of Pharmacy , Chungnam National University , Daejeon , Republic of Korea
| | - Hoo-Seong Kim
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea
| | - Jung-Eun Kwon
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea
| | - Myung-Sic Kim
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea
| | - Jong-Ho Kang
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea
| | - Chongsuk Ryou
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea
| | - Jeong-Sook Park
- d College of Pharmacy , Chungnam National University , Daejeon , Republic of Korea
| | - Jin-Ki Kim
- a College of Pharmacy, Institute of Pharmaceutical Science and Technology , Hanyang University , Ansan , Gyeonggi , Republic of Korea
| |
Collapse
|
136
|
Freire MCLC, Alexandrino F, Marcelino HR, Picciani PHDS, Silva KGDHE, Genre J, Oliveira AGD, Egito ESTD. Understanding Drug Release Data through Thermodynamic Analysis. MATERIALS (BASEL, SWITZERLAND) 2017; 10:E651. [PMID: 28773009 PMCID: PMC5554032 DOI: 10.3390/ma10060651] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/13/2017] [Accepted: 05/18/2017] [Indexed: 11/29/2022]
Abstract
Understanding the factors that can modify the drug release profile of a drug from a Drug-Delivery-System (DDS) is a mandatory step to determine the effectiveness of new therapies. The aim of this study was to assess the Amphotericin-B (AmB) kinetic release profiles from polymeric systems with different compositions and geometries and to correlate these profiles with the thermodynamic parameters through mathematical modeling. Film casting and electrospinning techniques were used to compare behavior of films and fibers, respectively. Release profiles from the DDSs were performed, and the mathematical modeling of the data was carried out. Activation energy, enthalpy, entropy and Gibbs free energy of the drug release process were determined. AmB release profiles showed that the relationship to overcome the enthalpic barrier was PVA-fiber > PVA-film > PLA-fiber > PLA-film. Drug release kinetics from the fibers and the films were better fitted on the Peppas-Sahlin and Higuchi models, respectively. The thermodynamic parameters corroborate these findings, revealing that the AmB release from the evaluated systems was an endothermic and non-spontaneous process. Thermodynamic parameters can be used to explain the drug kinetic release profiles. Such an approach is of utmost importance for DDS containing insoluble compounds, such as AmB, which is associated with an erratic bioavailability.
Collapse
Affiliation(s)
| | - Francisco Alexandrino
- Programa de Pós-graduaçãoem Nanotecnologia Farmacêutica, Universidade Federal do Rio Grande do Norte, Natal-RN 59012-570, Brazil.
| | - Henrique Rodrigues Marcelino
- Programa de Pós-graduaçãoem Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal-RN 59012-570, Brazil.
| | | | | | - Julieta Genre
- Programa de Pós-graduaçãoem Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal-RN 59012-570, Brazil.
| | - Anselmo Gomes de Oliveira
- Departamento de Fármacos e Medicamentos, Faculdade de Ciências Farmacêuticas, Universidade Estadual Paulista, Araraquara-SP 14800-903, Brazil.
| | - Eryvaldo Sócrates Tabosa do Egito
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Norte, Natal-RN 59012-570, Brazil.
- Programa de Pós-graduaçãoem Nanotecnologia Farmacêutica, Universidade Federal do Rio Grande do Norte, Natal-RN 59012-570, Brazil.
- Programa de Pós-graduaçãoem Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal-RN 59012-570, Brazil.
| |
Collapse
|
137
|
Nanotechnological strategies for nerve growth factor delivery: Therapeutic implications in Alzheimer’s disease. Pharmacol Res 2017; 120:68-87. [DOI: 10.1016/j.phrs.2017.03.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 02/23/2017] [Accepted: 03/22/2017] [Indexed: 12/30/2022]
|
138
|
Mouhid L, Corzo-Martínez M, Torres C, Vázquez L, Reglero G, Fornari T, Ramírez de Molina A. Improving In Vivo Efficacy of Bioactive Molecules: An Overview of Potentially Antitumor Phytochemicals and Currently Available Lipid-Based Delivery Systems. JOURNAL OF ONCOLOGY 2017; 2017:7351976. [PMID: 28555156 PMCID: PMC5438845 DOI: 10.1155/2017/7351976] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/06/2017] [Indexed: 02/07/2023]
Abstract
Cancer is among the leading causes of morbidity and mortality worldwide. Many of the chemotherapeutic agents used in cancer treatment exhibit cell toxicity and display teratogenic effect on nontumor cells. Therefore, the search for alternative compounds which are effective against tumor cells but reduce toxicity against nontumor ones is of great importance in the progress or development of cancer treatments. In this sense, scientific knowledge about relevant aspects of nutrition intimately involved in the development and progression of cancer progresses rapidly. Phytochemicals, considered as bioactive ingredients present in plant products, have shown promising effects as potential therapeutic/preventive agents on cancer in several in vitro and in vivo assays. However, despite their bioactive properties, phytochemicals are still not commonly used in clinical practice due to several reasons, mainly attributed to their poor bioavailability. In this sense, new formulation strategies are proposed as carriers to improve their bioefficacy, highlighting the use of lipid-based delivery systems. Here, we review the potential antitumoral activity of the bioactive compounds derived from plants and the current studies carried out in animal and human models. Furthermore, their association with lipids as a formulation strategy to enhance their efficacy in vivo is also reported. The development of high effective bioactive supplements for cancer treatment based on the improvement of their bioavailability goes through this association.
Collapse
Affiliation(s)
- Lamia Mouhid
- Molecular Oncology and Nutritional Genomics of Cancer, IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | - Marta Corzo-Martínez
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - Carlos Torres
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - Luis Vázquez
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - Guillermo Reglero
- Molecular Oncology and Nutritional Genomics of Cancer, IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - Tiziana Fornari
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology and Nutritional Genomics of Cancer, IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| |
Collapse
|
139
|
Hayashi K, Iwai H, Kamei T, Iwamoto K, Shimanouchi T, Fujita S, Nakamura H, Umakoshi H. Tailor-made drug carrier: Comparison of formation-dependent physicochemical properties within self-assembled aggregates for an optimal drug carrier. Colloids Surf B Biointerfaces 2017; 152:269-276. [DOI: 10.1016/j.colsurfb.2017.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/30/2016] [Accepted: 01/07/2017] [Indexed: 10/20/2022]
|
140
|
Zhang Z, Qian H, Yang M, Li R, Hu J, Li L, Yu L, Liu B, Qian X. Gambogic acid-loaded biomimetic nanoparticles in colorectal cancer treatment. Int J Nanomedicine 2017; 12:1593-1605. [PMID: 28280328 PMCID: PMC5339001 DOI: 10.2147/ijn.s127256] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Gambogic acid (GA) is expected to be a potential new antitumor drug, but its poor aqueous solubility and inevitable side effects limit its clinical application. Despite these inhe rent defects, various nanocarriers can be used to promote the solubility and tumor targeting of GA, improving antitumor efficiency. In addition, a cell membrane-coated nanoparticle platform that was reported recently, unites the customizability and flexibility of a synthetic copolymer, as well as the functionality and complexity of natural membrane, and is a new synthetic biomimetic nanocarrier with improved stability and biocompatibility. Here, we combined poly(lactic-co-glycolic acid) (PLGA) with red blood-cell membrane (RBCm), and evaluated whether GA-loaded RBCm nanoparticles can retain and improve the antitumor efficacy of GA with relatively lower toxicity in colorectal cancer treatment compared with free GA. We also confirmed the stability, biocompatibility, passive targeting, and few side effects of RBCm-GA/PLGA nanoparticles. We expect to provide a new drug carrier in the treatment of colorectal cancer, which has strong clinical application prospects. In addition, the potential antitumor drug GA and other similar drugs could achieve broader clinical applications via this biomimetic nanocarrier.
Collapse
Affiliation(s)
- Zhen Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine
| | - Hanqing Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Mi Yang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Rutian Li
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Jing Hu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine
| | - Li Li
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine
| | - Lixia Yu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Baorui Liu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine; Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Xiaoping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine; Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| |
Collapse
|
141
|
Plourde K, Derbali RM, Desrosiers A, Dubath C, Vallée-Bélisle A, Leblond J. Aptamer-based liposomes improve specific drug loading and release. J Control Release 2017; 251:82-91. [PMID: 28238787 DOI: 10.1016/j.jconrel.2017.02.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/27/2017] [Accepted: 02/07/2017] [Indexed: 01/02/2023]
Abstract
Aptamer technology has shown much promise in cancer therapeutics for its targeting abilities. However, its potential to improve drug loading and release from nanocarriers has not been thoroughly explored. In this study, we employed drug-binding aptamers to actively load drugs into liposomes. We designed a series of DNA aptamer sequences specific to doxorubicin, displaying multiple binding sites and various binding affinities. The binding ability of aptamers was preserved when incorporated into cationic liposomes, binding up to 15equivalents of doxorubicin per aptamer, therefore drawing the drug into liposomes. Optimization of the charge and drug/aptamer ratios resulted in ≥80% encapsulation efficiency of doxorubicin, ten times higher than classical passively-encapsulating liposomal formulations and similar to a pH-gradient active loading strategy. In addition, kinetic release profiles and cytotoxicity assay on HeLa cells demonstrated that the release and therapeutic efficacy of liposomal doxorubicin could be controlled by the aptamer's structure. Our results suggest that the aptamer exhibiting a specific intermediate affinity is the best suited to achieve high drug loading while maintaining efficient drug release and therapeutic activity. This strategy was successfully applied to tobramycin, a hydrophilic drug suffering from low encapsulation into liposomes, where its loading was improved six-fold using aptamers. Overall, we demonstrate that aptamers could act, in addition to their targeting properties, as multifunctional excipients for liposomal formulations.
Collapse
Affiliation(s)
- Kevin Plourde
- Faculty of Pharmacy, University of Montreal, QC H3T 1J4, Canada
| | | | | | - Céline Dubath
- Faculty of Pharmacy, University of Montreal, QC H3T 1J4, Canada
| | | | - Jeanne Leblond
- Faculty of Pharmacy, University of Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
142
|
Bazylińska U, Pietkiewicz J, Rossowska J, Chodaczek G, Gamian A, Wilk KA. Polyelectrolyte Oil-Core Nanocarriers for Localized and Sustained Delivery of Daunorubicin to Colon Carcinoma MC38 Cells: The Case of Polysaccharide Multilayer Film in Relation to PEG-ylated Shell. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201600356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/04/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Urszula Bazylińska
- Department of Organic and Pharmaceutical Technology; Faculty of Chemistry; Wroclaw University of Science and Technology; Wybrzeze Wyspianskiego 27 50-370 Wroclaw Poland
| | - Jadwiga Pietkiewicz
- Department of Medical Biochemistry; Medical University of Wroclaw; Chalubinskiego 10 50-368 Wroclaw Poland
| | - Joanna Rossowska
- Institute of Immunology and Experimental Therapy Polish Academy of Sciences; Rudolfa Weigla 12 53-114 Wroclaw Poland
| | - Grzegorz Chodaczek
- Wroclaw Research Centre EIT+; Confocal Microscopy Laboratory; Stablowicka 147 54-066 Wroclaw Poland
| | - Andrzej Gamian
- Department of Medical Biochemistry; Medical University of Wroclaw; Chalubinskiego 10 50-368 Wroclaw Poland
| | - Kazimiera A. Wilk
- Department of Organic and Pharmaceutical Technology; Faculty of Chemistry; Wroclaw University of Science and Technology; Wybrzeze Wyspianskiego 27 50-370 Wroclaw Poland
| |
Collapse
|
143
|
Stagnoli S, Luna MA, Villa CC, Alustiza F, Niebylski A, Moyano F, Correa NM, Falcone RD. Unique catanionic vesicles as a potential “Nano-Taxi” for drug delivery systems. In vitro and in vivo biocompatibility evaluation. RSC Adv 2017. [DOI: 10.1039/c6ra27020d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We evaluatein vitroandin vivotoxicity and stability in an acidic environment of new vesicles formed by the catanionic surfactant AOT-BHD in order to investigate their potential application as an oral drug delivery system.
Collapse
Affiliation(s)
- Soledad Stagnoli
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - M. Alejandra Luna
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - Cristian C. Villa
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - Fabrisio Alustiza
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - Ana Niebylski
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - Fernando Moyano
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - N. Mariano Correa
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| | - R. Darío Falcone
- Departamento de Química
- Universidad Nacional de Río Cuarto
- C.P. X5804BYA Río Cuarto
- Argentina
| |
Collapse
|
144
|
Bazylińska U, Frąckowiak R, Brzózka Z, Wilk KA. The effect of anionic dicephalic surfactants on fabrication of varied-core nanocarriers for sustained release of porphyrin photosensitizers. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 166:169-179. [PMID: 27915030 DOI: 10.1016/j.jphotobiol.2016.11.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/05/2016] [Accepted: 11/11/2016] [Indexed: 12/15/2022]
Abstract
Double-headed anionic surfactants could provide a profound group of efficient stabilizers of new template-mediated nanocarriers for effective encapsulation and sustained release of highly hydrophobic photosensitizers, and therefore their improved therapeutic activity in photodynamic therapy (PDT) protocols. We have thus encapsulated porphyrin-origin dyes, i.e., verteporfin (VP) and meso-tetraphenylporphyrin (TPP) in different types of sodium alkyliminobisacetates, Cn(COONa)2-stabilized nanosystems including biocompatible poly(l-glutamic acid)/poly(l-lysine) - PGA/PLL, multilayer nanocapsules (NCs). The latter were prepared via a layer-by-layer (LbL) approach with either solid (nanoprecipitated), or liquid (nanoemulsion-templated) oil core while zeta potential measurements enabled to evaluate progress of the polyelectrolytes LbL deposition on both cores and the NCs' stability. Backscattering profiles (BS) confirmed the long-lasting stability of the optimized nanosystems, which size (<200nm), polidyspersity and morphology were examined by dynamic light scattering (DLS) and atomic force microscopy (AFM) techniques. Our studies indicated that the encapsulation of VP and TPP in the both type of multilayer NCs increases their solubility in aqueous solution and protects them from the surrounding medium. Mainly, it reduces the photobleaching rate of these porphyrin-type photosensitizers and improves their photochemical properties during irradiation in regards to the free (non-encapsulated) molecules. As far as the core-type is considered, both nanoemulsion-loaded porphyrins, photobleached ca. 15-20% faster than the solid nanoparticle analogs. By using 9,10-anthracenediyl-bis(methylene)dimalonic acid (ABMDMA) as a singlet oxygen (1O2) scavenger molecule, the enhanced generation of reactive species was evaluated for the both encapsulated photosensitizers in comparison to their native form. In vitro sustained release under physiological conditions or in the presence of human serum albumin (HSA) was achieved in favor of the solid core NCs for VP and TPP. The designed NCs - offering better chemical and physical stability, high loading capacity for the cargo and ability to release it in a controlled and continuous manner - can be considered as efficacious nanocarriers for PDT.
Collapse
Affiliation(s)
- Urszula Bazylińska
- Departament of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland.
| | - Renata Frąckowiak
- Departament of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Zbigniew Brzózka
- Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Kazimiera A Wilk
- Departament of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| |
Collapse
|
145
|
Enhanced acute anti-inflammatory effects of CORM-2-loaded nanoparticles via sustained carbon monoxide delivery. Eur J Pharm Biopharm 2016; 108:187-195. [DOI: 10.1016/j.ejpb.2016.09.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/09/2016] [Accepted: 09/10/2016] [Indexed: 11/20/2022]
|
146
|
Maher S, Kumeria T, Wang Y, Kaur G, Fathalla D, Fetih G, Santos A, Habib F, Evdokiou A, Losic D. From The Mine to Cancer Therapy: Natural and Biodegradable Theranostic Silicon Nanocarriers from Diatoms for Sustained Delivery of Chemotherapeutics. Adv Healthc Mater 2016; 5:2667-2678. [PMID: 27594524 DOI: 10.1002/adhm.201600688] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 07/31/2016] [Indexed: 01/24/2023]
Abstract
Drug delivery using synthetic nanoparticles including porous silicon has been extensively used to overcome the limitations of chemotherapy. However, their synthesis has many challenges such as lack of scalability, high cost, and the use of toxic materials with concerning environmental impact. Nanoscale materials obtained from natural resources are an attractive option to address some of these disadvantages. In this paper, a new mesoporous biodegradable silicon nanoparticle (SiNP) drug carrier obtained from natural diatom silica mineral available from the mining industry is presented. Diatom silica structures are mechanically fragmented and converted into SiNPs by simple and scalable magnesiothermic reduction process. Results show that SiNPs have many desirable properties including high surface area, high drug loading capacity, strong luminescence, biodegradability, and no cytotoxicity. The in-vitro release results from SiNPs loaded with anticancer drugs (doxorubicin) demonstrate a pH-dependent and sustained drug release with enhanced cytotoxicity against cancer cells. The cells study using doxorubicin loaded SiNPs shows a significantly enhanced cytotoxicity against cancer cells compared with free drug, suggesting their considerable potential as theranostic nanocarriers for chemotherapy. Their low-cost manufacturing using abundant natural materials and outstanding chemotherapeutic performance has made them as a promising alternative to synthetic nanoparticles for drug delivery applications.
Collapse
Affiliation(s)
- Shaheer Maher
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
- Faculty of Pharmacy Assiut University 71526 Assiut Egypt
| | - Tushar Kumeria
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
| | - Ye Wang
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
- Discipline of Surgery Basil Hetzel Institute The University of Adelaide 5005 Adelaide SA Australia
| | - Gagandeep Kaur
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
- Discipline of Surgery Basil Hetzel Institute The University of Adelaide 5005 Adelaide SA Australia
| | - Dina Fathalla
- Faculty of Pharmacy Assiut University 71526 Assiut Egypt
| | - Gihan Fetih
- Faculty of Pharmacy Assiut University 71526 Assiut Egypt
| | - Abel Santos
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
| | - Fawzia Habib
- Faculty of Pharmacy Assiut University 71526 Assiut Egypt
| | - Andreas Evdokiou
- Discipline of Surgery Basil Hetzel Institute The University of Adelaide 5005 Adelaide SA Australia
| | - Dusan Losic
- School of Chemical Engineering The University of Adelaide Engineering North Building 5005 Adelaide Australia
| |
Collapse
|
147
|
De Matteis L, Alleva M, Serrano-Sevilla I, García-Embid S, Stepien G, Moros M, de la Fuente JM. Controlling Properties and Cytotoxicity of Chitosan Nanocapsules by Chemical Grafting. Mar Drugs 2016; 14:E175. [PMID: 27706041 PMCID: PMC5082323 DOI: 10.3390/md14100175] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/14/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022] Open
Abstract
The tunability of the properties of chitosan-based carriers opens new ways for the application of drugs with low water-stability or high adverse effects. In this work, the combination of a nanoemulsion with a chitosan hydrogel coating and the following poly (ethylene glycol) (PEG) grafting is proven to be a promising strategy to obtain a flexible and versatile nanocarrier with an improved stability. Thanks to chitosan amino groups, a new easy and reproducible method to obtain nanocapsule grafting with PEG has been developed in this work, allowing a very good control and tunability of the properties of nanocapsule surface. Two different PEG densities of coverage are studied and the nanocapsule systems obtained are characterized at all steps of the optimization in terms of diameter, Z potential and surface charge (amino group analysis). Results obtained are compatible with a conformation of PEG molecules laying adsorbed on nanoparticle surface after covalent linking through their amino terminal moiety. An improvement in nanocapsule stability in physiological medium is observed with the highest PEG coverage density obtained. Cytotoxicity tests also demonstrate that grafting with PEG is an effective strategy to modulate the cytotoxicity of developed nanocapsules. Such results indicate the suitability of chitosan as protective coating for future studies oriented toward drug delivery.
Collapse
Affiliation(s)
- Laura De Matteis
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza, Edificio I+D, calle Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| | - Maria Alleva
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza, Edificio I+D, calle Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| | - Inés Serrano-Sevilla
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza, Edificio I+D, calle Mariano Esquillor s/n, 50018 Zaragoza, Spain.
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC-Universidad de Zaragoza, Edificio I+D, calle Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| | - Sonia García-Embid
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza, Edificio I+D, calle Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| | - Grazyna Stepien
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza, Edificio I+D, calle Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| | - María Moros
- Istituto di Scienze Applicate e Sistemi Intelligenti "E. Caianiello", Consiglio Nazionale delle Ricerche, Pozzuoli 80078, Italy.
| | - Jesús M de la Fuente
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC-Universidad de Zaragoza, Edificio I+D, calle Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| |
Collapse
|
148
|
Chen H, Woods A, Forbes B, Jones S. Controlled drug release from lung-targeted nanocarriers via chemically mediated shell permeabilisation. Int J Pharm 2016; 511:1033-41. [PMID: 27506512 DOI: 10.1016/j.ijpharm.2016.08.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/03/2016] [Accepted: 08/06/2016] [Indexed: 01/26/2023]
Abstract
Nanocarriers can aid therapeutic agent administration to the lung, but controlling drug delivery from these systems after deposition in the airways can be problematic. The aim of this study was to evaluate if chemically mediated shell permeabilisation could help manipulate the rate and extent of nanocarrier drug release. Rifampicin was loaded into lipid shell (loading efficiency 41.0±11.4%, size 50nm) and polymer shell nanocarriers (loading efficiency 25.9±2.3%, size 250nm). The drug release at pH 7.4 (lung epithelial pH) and 4.2 (macrophage endosomal pH) with and without the chemical permeabilisers (Pluronic L62D - lipid nanocarriers; H(+)- polymer nanocarriers) was then tested. At pH 7.4 the presence of the permeabilisers increased nanocarrier drug release rate (from 3.2μg/h to 6.8μg/h for lipid shell nanocarriers, 2.3μg/h to 3.4μg/h for polymer shell nanocarriers) and drug release extent (from 50% to 80% for lipid shell nanocarriers, from 45% to 76% for polymer shell nanocarriers). These effects were accompanied by lipid nanocarrier distension (from 50 to 240nm) and polymer shell hydrolysis. At pH 4.2 the polymer nanocarriers did not respond to the permeabiliser, but the lipid nanocarrier maintained a robust drug release enhancement response and hence they demonstrated that the manipulation of controlled drug release from lung-targeted nanocarriers was possible through chemically mediated shell permeabilisation.
Collapse
Affiliation(s)
- Hanpeng Chen
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, Franklin-Wilkins Building, King's College London, 150 Stamford Street, London SE1 9NH, UK
| | - Arcadia Woods
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, Franklin-Wilkins Building, King's College London, 150 Stamford Street, London SE1 9NH, UK
| | - Ben Forbes
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, Franklin-Wilkins Building, King's College London, 150 Stamford Street, London SE1 9NH, UK
| | - Stuart Jones
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, Franklin-Wilkins Building, King's College London, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
149
|
Lamch Ł, Kulbacka J, Pietkiewicz J, Rossowska J, Dubińska-Magiera M, Choromańska A, Wilk KA. Preparation and characterization of new zinc(II) phthalocyanine — Containing poly(l-lactide)-b-poly(ethylene glycol) copolymer micelles for photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 160:185-97. [DOI: 10.1016/j.jphotobiol.2016.04.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 11/26/2022]
|
150
|
Rancan F, Asadian-Birjand M, Dogan S, Graf C, Cuellar L, Lommatzsch S, Blume-Peytavi U, Calderón M, Vogt A. Effects of thermoresponsivity and softness on skin penetration and cellular uptake of polyglycerol-based nanogels. J Control Release 2016; 228:159-169. [DOI: 10.1016/j.jconrel.2016.02.047] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/08/2016] [Accepted: 02/29/2016] [Indexed: 12/19/2022]
|