[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Next Issue
Volume 15, May
Previous Issue
Volume 15, March
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Pharmaceutics, Volume 15, Issue 4 (April 2023) – 287 articles

Cover Story (view full-size image): Posterior segment eye diseases pose treatment challenges due to ocular barriers limiting drug penetration, residence time, and bioavailability. Current treatments require frequent dosing, including eye drops or intravitreal injections. Biodegradable nano-based drug delivery systems (DDS) offer a solution by staying in ocular tissues longer, passing through barriers for higher bioavailability, and being composed of biodegradable, nanosized polymers. This review examines DDS in ocular disease treatment, therapeutic challenges in posterior segment diseases, and the enhancement of treatment options through various biodegradable nanocarriers. A literature review from 2017 to 2023 highlights the rapid evolution of nano-based DDSs, promising to overcome current clinical challenges. View this paper
  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
21 pages, 8457 KiB  
Review
Natural Compounds: Co-Delivery Strategies with Chemotherapeutic Agents or Nucleic Acids Using Lipid-Based Nanocarriers
by Patrícia V. Teixeira, Eduarda Fernandes, Telma B. Soares, Filomena Adega, Carla M. Lopes and Marlene Lúcio
Pharmaceutics 2023, 15(4), 1317; https://doi.org/10.3390/pharmaceutics15041317 - 21 Apr 2023
Cited by 8 | Viewed by 2392
Abstract
Cancer is one of the leading causes of death, and latest predictions indicate that cancer- related deaths will increase over the next few decades. Despite significant advances in conventional therapies, treatments remain far from ideal due to limitations such as lack of selectivity, [...] Read more.
Cancer is one of the leading causes of death, and latest predictions indicate that cancer- related deaths will increase over the next few decades. Despite significant advances in conventional therapies, treatments remain far from ideal due to limitations such as lack of selectivity, non-specific distribution, and multidrug resistance. Current research is focusing on the development of several strategies to improve the efficiency of chemotherapeutic agents and, as a result, overcome the challenges associated with conventional therapies. In this regard, combined therapy with natural compounds and other therapeutic agents, such as chemotherapeutics or nucleic acids, has recently emerged as a new strategy for tackling the drawbacks of conventional therapies. Taking this strategy into consideration, the co-delivery of the above-mentioned agents in lipid-based nanocarriers provides some advantages by improving the potential of the therapeutic agents carried. In this review, we present an analysis of the synergistic anticancer outcomes resulting from the combination of natural compounds and chemotherapeutics or nucleic acids. We also emphasize the importance of these co-delivery strategies when reducing multidrug resistance and adverse toxic effects. Furthermore, the review delves into the challenges and opportunities surrounding the application of these co-delivery strategies towards tangible clinical translation for cancer treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Problems associated with the classical single-delivery therapy (i.e., administration of each of the therapeutic agents in their free form). Nucleic acids, if delivered in the free form, would face different pharmacokinetic challenges, including inactivation by nucleases (A), lack of serum stability due to the immune system (B) and serum proteins (C), extravasation difficulties (D), non-specific distribution in target cells (E), difficulties entering the cell (F), and degradation if not able to escape endosomes (G). Chemotherapeutic drugs, when delivered in the free form, have a nonspecific distribution in cancer cells and healthy cells causing serious adverse side effects, commonly affecting hair follicles, the digestive tract, blood cells and nerves. Furthermore, several MDR mechanisms, such as drug efflux by multidrug resistance protein 1 (MRP1), P-glycoprotein (P-gp), and breast cancer resistance protein (BCRP), or inactivation of apoptotic pathways by B cell leukemia protein (Bcl2), can impair their efficiency. Natural compounds, when administered in their free form, exhibit a number of pharmacokinetic issues that affect their biodistribution and efficacy (1–6).</p>
Full article ">Figure 2
<p>Schematic illustration of lipid-based nanocarriers and advantages of their use for the co-delivery of natural compounds and chemotherapeutic drugs or nucleic acids. Adapted from [<a href="#B36-pharmaceutics-15-01317" class="html-bibr">36</a>,<a href="#B37-pharmaceutics-15-01317" class="html-bibr">37</a>], and from [<a href="#B38-pharmaceutics-15-01317" class="html-bibr">38</a>,<a href="#B39-pharmaceutics-15-01317" class="html-bibr">39</a>] with permission from Elsevier.</p>
Full article ">Figure 3
<p>Main natural compounds considered chemosensitizing agents, according to their chemical family [<a href="#B47-pharmaceutics-15-01317" class="html-bibr">47</a>].</p>
Full article ">Figure 4
<p>Potential targets associated with Curcumin anticancer activity. This natural compound induces a reduction in its target genes by inhibiting NF-kβ signaling. Bcl-2-B-cell limphoma-2; COX-2-ciclo-oxigenase-2; IL-6-interleukin 6; IL-10-interleukin 10; IL-18-interleukin 18; MMP9-matrix metallopeptidase 9; NF-kβ-nuclear factor kappa B; VEGF-vascular endothelial growth factor; XIAP-X-linked inhibitor of apoptosis protein.</p>
Full article ">
12 pages, 4047 KiB  
Article
In Situ Synthesis of a Tumor-Microenvironment-Responsive Chemotherapy Drug
by Xiupeng Wang, Ayako Oyane, Tomoya Inose and Maki Nakamura
Pharmaceutics 2023, 15(4), 1316; https://doi.org/10.3390/pharmaceutics15041316 - 21 Apr 2023
Cited by 2 | Viewed by 1913
Abstract
Current chemotherapy still suffers from unsatisfactory therapeutic efficacy, multi-drug resistance, and severe adverse effects, thus necessitating the development of techniques to confine chemotherapy drugs in the tumor microenvironment. Herein, we fabricated nanospheres of mesoporous silica (MS) doped with Cu (MS-Cu) and polyethylene glycol [...] Read more.
Current chemotherapy still suffers from unsatisfactory therapeutic efficacy, multi-drug resistance, and severe adverse effects, thus necessitating the development of techniques to confine chemotherapy drugs in the tumor microenvironment. Herein, we fabricated nanospheres of mesoporous silica (MS) doped with Cu (MS-Cu) and polyethylene glycol (PEG)-coated MS-Cu (PEG-MS-Cu) as exogenous copper supply systems to tumors. The synthesized MS-Cu nanospheres showed diameters of 30–150 nm with Cu/Si molar ratios of 0.041–0.069. Only disulfiram (DSF) and only MS-Cu nanospheres showed little cytotoxicity in vitro, whereas the combination of DSF and MS-Cu nanospheres showed significant cytotoxicity against MOC1 and MOC2 cells at concentrations of 0.2–1 μg/mL. Oral DSF administration in combination with MS-Cu nanospheres intratumoral or PEG-MS-Cu nanospheres intravenous administration showed significant antitumor efficacy against MOC2 cells in vivo. In contrast to traditional drug delivery systems, we herein propose a system for the in situ synthesis of chemotherapy drugs by converting nontoxic substances into antitumor chemotherapy drugs in a specific tumor microenvironment. Full article
(This article belongs to the Special Issue Beyond the Platinum in Metal-Based Cancer Therapy, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>TEM (<b>a</b>,<b>c</b>,<b>e</b>) and STEM-EDX (<b>b</b>,<b>d</b>,<b>f</b>) images of MS-Cu nanospheres with different particle size. MS-Cu-1 (<b>a</b>,<b>b</b>), MS-Cu-2 (<b>c</b>,<b>d</b>), MS-Cu-3 (<b>e</b>,<b>f</b>).</p>
Full article ">Figure 2
<p>Physicochemical characterization of MS-Cu nanospheres with different particle size. XRD patterns (<b>a</b>), N<sub>2</sub> adsorption-desorption isotherms (<b>b</b>), pore size distributions (<b>c</b>), BET surface areas (<b>d</b>), and Cu/Si mol ratio (<b>e</b>) of MS-Cu-1, MS-Cu-2, and MS-Cu-3 (*, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Cytotoxicity of only MS-Cu nanospheres (<b>a</b>,<b>c</b>), only DSF (<b>a</b>,<b>c</b>), and combination of MS-Cu nanospheres and DSF (<b>b</b>,<b>d</b>)against MOC1 (<b>a</b>,<b>b</b>) and MOC2 (<b>c</b>,<b>d</b>) cells in vitro. In vivo antitumor efficacy of combined oral administration of DSF and intratumoral administration of MS-Cu nanospheres. Experimental protocol (<b>e</b>), tumor volume (<b>f</b>), and tumor weight at the endpoint (<b>g</b>) (*, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Combination of oral administration of DSF and intravenous administration of PEG-MS-Cu nanospheres inhibited MOC2 cell growth in vivo. STEM-EDX images of PEG-MS-Cu nanospheres (<b>a</b>). In vivo antitumor efficacy of combined oral administration of DSF and intravenous administration of PEG-MS-Cu nanospheres. Experimental protocol (<b>b</b>), tumor volume (<b>c</b>), and tumor weight at the endpoint (<b>d</b>). HE and TUNEL staining of tumor with no treatment (<b>e</b>), after only oral administration of DSF (<b>f</b>), only intravenous administration of PEG-MS-Cu nanospheres (<b>g</b>), and combined oral administration of DSF and intravenous administration of PEG-MS-Cu nanospheres (<b>h</b>) (*, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Combination of oral administration of DSF and intravenous administration of PEG-MS-Cu nanospheres showed no obvious toxicity to normal tissues in vivo. Histological sections of heart, kidney, liver, lung, and spleen of mice without any treatment (<b>a</b>), and with combined oral administration of DSF and intravenous administration of PEG-MS-Cu nanospheres (<b>b</b>).</p>
Full article ">
22 pages, 4678 KiB  
Article
Influence of Solid Oral Dosage Form Characteristics on Swallowability, Visual Perception, and Handling in Older Adults
by Henriette Hummler, Susanne Page, Cordula Stillhart, Lisa Meilicke, Michael Grimm, Marwan Mannaa, Maik Gollasch and Werner Weitschies
Pharmaceutics 2023, 15(4), 1315; https://doi.org/10.3390/pharmaceutics15041315 - 21 Apr 2023
Cited by 4 | Viewed by 3533
Abstract
Swallowability, visual perception, and any handling to be conducted prior to use are all influence factors on the acceptability of an oral dosage form by the patient. Knowing the dosage form preferences of older adults, as the major group of medication end users, [...] Read more.
Swallowability, visual perception, and any handling to be conducted prior to use are all influence factors on the acceptability of an oral dosage form by the patient. Knowing the dosage form preferences of older adults, as the major group of medication end users, is needed for patient-centric drug development. This study aimed at evaluating the ability of older adults to handle tablets as well as to assess the anticipated swallowability of tablets, capsules, and mini tablets based on visual perception. The randomized intervention study included 52 older adults (65 to 94 years) and 52 younger adults (19 to 36 years). Within the tested tablets, ranging from 125 mg up to 1000 mg in weight and being of different shapes, handling was not seen as the limiting factor for the decision on appropriate tablet size. However, the smallest sized tablets were rated worst. According to visual perception, the limit of acceptable tablet size was reached at around 250 mg for older adults. For younger adults, this limit was shifted to higher weights and was dependent on the tablet shape. Differences in anticipated swallowability with respect to tablet shapes were most pronounced for tablets of 500 mg and 750 mg in weight, independent of the age category. Capsules performed worse compared to tablets, while mini tablets appeared as a possible alternative dosage form to tablets of higher weight. Within the deglutition part of this study, swallowability capabilities of the same populations were assessed and have been reported previously. Comparing the present results with the swallowing capabilities of the same populations with respect to tablets, it shows adults’ clear self-underestimation of their ability to swallow tablets independent of their age. Full article
(This article belongs to the Special Issue Advance in Development of Patient-Centric Dosage Form, 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Eight different showcases were shown to the participants consecutively. Participants estimated the swallowability of the different dosage forms.</p>
Full article ">Figure 2
<p>Responsibilities for medication management among the older participants.</p>
Full article ">Figure 3
<p>Handling of tablets differing in weight and shape rated as <span class="html-italic">not</span>, <span class="html-italic">moderately,</span> or <span class="html-italic">well to handle</span>. Data are shown separately for the older participants (<b>left</b> panel) and younger participants (<b>right</b> panel). Each participant gave a rating for every tablet. The value of 80% was used as a cutoff value for the data evaluation as it is a commonly used threshold for acceptability.</p>
Full article ">Figure 4
<p>Number of responses and handling ratings (<span class="html-italic">moderately to handle</span> on the <b>left</b> panels and <span class="html-italic">not to handle</span> on the <b>right</b> panels). Crosses indicate the number of handling ratings (absolute numbers). Data are shown separately for the older participants (<b>top</b> panels) and younger participants (<b>bottom</b> panels).</p>
Full article ">Figure 5
<p>Visual perception of dosage forms’ swallowability by older participants (<b>left</b> panel) and younger participants (<b>right</b> panel). Capsules (size 4 to 00 with anticipated weight of 125 mg to 500 mg), round shaped tablets (125 mg to 750 mg), as well as oval and oblong shaped tablets (125 mg to 1250 mg) were shown to the participants in the shape-showcases.</p>
Full article ">Figure 6
<p>Visual perception of dosage forms’ swallowability by older participants (<b>left</b> panel) and younger participants (<b>right</b> panel). Capsules (250 mg and 500 mg), round shaped tablets (250 mg to 750 mg), oval and oblong shaped tablets (250 mg to 1000 mg), as well as mini tablets (750 mg and 1000 mg) were shown to the participants in showcases dependent on the weight of the dosage forms.</p>
Full article ">Figure 7
<p>Significant differences according to the McNemar test for the data of visual perception. Arrows point to dosage forms showing a worse rating in terms of the anticipated swallowability. Results for the older participants (continuous line) and younger participants (dashed line) are shown. Comparisons between the dosage forms of the same kind were calculated for adjacent weights. For dosage forms of the same weight, all dosage forms were compared to each other. Stars indicate a significant difference between age categories according to Fisher’s exact test, implying better anticipated swallowability ratings for younger compared to older participants. To account for multiple testing, all calculated <span class="html-italic">p</span>-values were corrected according to Benjamini and Hochberg. <span class="html-italic">p</span>-values of &lt;0.05 were deemed as significant.</p>
Full article ">Figure 8
<p>Influence of the showcase style (shape- or weight-showcase) on the visual perception of dosage forms’ swallowability. Data are shown for older participants (<b>upper</b> panel) as well as younger participants (<b>lower</b> panel). Significant differences according to the McNemar test and consequent Benjamini–Hochberg correction (<span class="html-italic">p</span>-value &lt; 0.05) are marked with an asterisk.</p>
Full article ">Figure 9
<p>Influence of sex on the visual perception of the swallowability of the dosage forms presented within the shape-showcases. Data are shown separately for older participants (67.3% female; <b>left</b> panel) and younger participants (55.8% female; <b>right</b> panel).</p>
Full article ">Figure 10
<p>Influence of sex on the visual perception of the swallowability of the dosage forms presented within weight-showcases. Data are shown separately for older participants (67.3% female; <b>left</b> panel) and for younger participants (55.8% female; <b>right</b> panel).</p>
Full article ">Figure 11
<p>Comparison of the swallowability ratings after actual deglutition compared to those according to visual perception. Data of the visual perception only included ratings of those participants that had actually swallowed the particular tablets. Data are shown for older participants (<b>upper</b> panel) and younger participants (<b>lower</b> panel). Significant differences according to the McNemar test using paired data and consequent Benjamini–Hochberg correction (<span class="html-italic">p</span>-value &lt; 0.05) are marked with an asterisk.</p>
Full article ">
18 pages, 22565 KiB  
Article
Highly Cytotoxic Copper(II) Mixed-Ligand Quinolinonato Complexes: Pharmacokinetic Properties and Interactions with Drug Metabolizing Cytochromes P450
by Martina Medvedíková, Václav Ranc, Ján Vančo, Zdeněk Trávníček and Pavel Anzenbacher
Pharmaceutics 2023, 15(4), 1314; https://doi.org/10.3390/pharmaceutics15041314 - 21 Apr 2023
Cited by 1 | Viewed by 1635
Abstract
The effects of two anticancer active copper(II) mixed-ligand complexes of the type [Cu(qui)(mphen)]Y·H2O, where Hqui = 2-phenyl-3-hydroxy- 1H-quinolin-4-one, mphen = bathophenanthroline, and Y = NO3 (complex 1) or BF4 (complex 2) on the activities of different isoenzymes [...] Read more.
The effects of two anticancer active copper(II) mixed-ligand complexes of the type [Cu(qui)(mphen)]Y·H2O, where Hqui = 2-phenyl-3-hydroxy- 1H-quinolin-4-one, mphen = bathophenanthroline, and Y = NO3 (complex 1) or BF4 (complex 2) on the activities of different isoenzymes of cytochrome P450 (CYP) have been evaluated. The screening revealed significant inhibitory effects of the complexes on CYP3A4/5 (IC50 values were 2.46 and 4.88 μM), CYP2C9 (IC50 values were 16.34 and 37.25 μM), and CYP2C19 (IC50 values were 61.21 and 77.07 μM). Further, the analysis of mechanisms of action uncovered a non-competitive type of inhibition for both the studied compounds. Consequent studies of pharmacokinetic properties proved good stability of both the complexes in phosphate buffer saline (>96% stability) and human plasma (>91% stability) after 2 h of incubation. Both compounds are moderately metabolised by human liver microsomes (<30% after 1 h of incubation), and over 90% of the complexes bind to plasma proteins. The obtained results showed the potential of complexes 1 and 2 to interact with major metabolic pathways of drugs and, as a consequence of this finding, their apparent incompatibility in combination therapy with most chemotherapeutic agents. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structural formulas of one example from Casiopeínas<sup>®</sup> family (<b>a</b>) together with the herein studied copper(II) complexes—complex <b>1</b> (<b>b</b>) and complex <b>2</b> (<b>c</b>).</p>
Full article ">Figure 2
<p>The geometry of the [Cu(mphen)(qui)]<sup>+</sup> complex cation optimised at the ϖB97X-D/LACVP** level of theory.</p>
Full article ">Figure 3
<p>Overview of inhibition rates of CYP activities with specific substrates by the copper(II) complexes <b>1</b> and <b>2,</b> free Hqui and mphen ligands, and Cu(NO<sub>3</sub>)<sub>2</sub>·3H<sub>2</sub>O at 100 μM concentration. The results represent the amount of residual activity of CYPs expressed as a percentage of the activity of an uninhibited enzyme.</p>
Full article ">Figure 4
<p>(<b>a</b>) The effect of complex <b>1</b> on the enzymatic activity of CYP2C9, CYP2C19, and CYP3A4/5 with the specific substrates of testosterone and midazolam in HLMs. The inhibition of activity is determined as the mean of the two independent experiments performed in triplicates ± SD and is expressed as a percentage of activity remaining relative to the control (set to 100%, without the addition of the studied compounds). Concentrations of complex <b>1</b> in reaction mixtures were 0, 10, 25, 50, 75, and 100 μM. (<b>b</b>) Dixon plot for inhibition of CYP3A4/5 (testosterone 6β-hydroxylation) by complex <b>1</b>, and (<b>c</b>) Lineweaver–Burk plot for inhibition of CYP3A4/5 (testosterone 6β-hydroxylation) enzymatic activity by complex <b>1</b> at four substrate concentrations (50, 100, 200, 400 µM) for eight concentrations of complex <b>1</b> (0, 0.8, 1.6, 3.2, 6.4, 12.8, 25 and 50 μM).</p>
Full article ">Figure 5
<p>Difference spectra demonstrating the interaction of human liver microsomal cytochrome P450s with complex <b>1</b> (<b>a</b>), complex <b>2</b> (<b>b</b>), free Hqui ligand (<b>c</b>), free mphen ligand (<b>d</b>), and Cu(NO<sub>3</sub>)<sub>2</sub>·3H<sub>2</sub>O (<b>e</b>). CYPs concentration was 1 μM, and the concentrations of the tested complexes varied from 0.002 to 33.310 μM. Insets: plots of the absorbance changes at 386 nm vs. concentration of the respective compound.</p>
Full article ">Figure 6
<p>Representative ITC data for complex <b>1</b> binding to bactosomes CYP1A2 (<b>a</b>), bactosomes CYP2A6 (<b>b</b>), bactosomes CYP3A4 (<b>c</b>), and recombinant human CYP3A4 (<b>d</b>). (Top) Raw data plot of heat flow against time for the titration of CYP with complex <b>1</b>. (Bottom) Plot of molar enthalpy change against the complex <b>1</b>/cytochrome P450 molar ratio.</p>
Full article ">
69 pages, 17242 KiB  
Review
Chitosan: A Potential Biopolymer in Drug Delivery and Biomedical Applications
by Nimeet Desai, Dhwani Rana, Sagar Salave, Raghav Gupta, Pranav Patel, Bharathi Karunakaran, Amit Sharma, Jyotsnendu Giri, Derajram Benival and Nagavendra Kommineni
Pharmaceutics 2023, 15(4), 1313; https://doi.org/10.3390/pharmaceutics15041313 - 21 Apr 2023
Cited by 172 | Viewed by 14259
Abstract
Chitosan, a biocompatible and biodegradable polysaccharide derived from chitin, has surfaced as a material of promise for drug delivery and biomedical applications. Different chitin and chitosan extraction techniques can produce materials with unique properties, which can be further modified to enhance their bioactivities. [...] Read more.
Chitosan, a biocompatible and biodegradable polysaccharide derived from chitin, has surfaced as a material of promise for drug delivery and biomedical applications. Different chitin and chitosan extraction techniques can produce materials with unique properties, which can be further modified to enhance their bioactivities. Chitosan-based drug delivery systems have been developed for various routes of administration, including oral, ophthalmic, transdermal, nasal, and vaginal, allowing for targeted and sustained release of drugs. Additionally, chitosan has been used in numerous biomedical applications, such as bone regeneration, cartilage tissue regeneration, cardiac tissue regeneration, corneal regeneration, periodontal tissue regeneration, and wound healing. Moreover, chitosan has also been utilized in gene delivery, bioimaging, vaccination, and cosmeceutical applications. Modified chitosan derivatives have been developed to improve their biocompatibility and enhance their properties, resulting in innovative materials with promising potentials in various biomedical applications. This article summarizes the recent findings on chitosan and its application in drug delivery and biomedical science. Full article
Show Figures

Figure 1

Figure 1
<p>Extraction of chitin and chitosan.</p>
Full article ">Figure 2
<p>Functionalized chitosan derivatives. Adapted from reference [<a href="#B88-pharmaceutics-15-01313" class="html-bibr">88</a>].</p>
Full article ">Figure 3
<p>Various applications of chitosan.</p>
Full article ">Figure 4
<p>((<b>A</b>), i) SEM micrographs of isolated drug-free chitosan–arginine–alginate bioparticles after lyophilization, showing globular morphology and smooth to a slightly irregular surface. ((<b>A</b>), ii) SEM micrographs of particle cluster containing 38 μM praziquantel unveiling irregular polygonal-like structures and wrinkled surface. Confocal laser scanning micrographs (epifluorescence of labeled fluorescein isothiocyanate, phase contrast and merge) of anterior (upper panels) and posterior (lower panels) intestine of Carassius auratus after 30 min ((<b>A</b>), iii) and 8 h ((<b>A</b>), iv) (Scale bar: 50 μm). Reproduced with permission from [<a href="#B129-pharmaceutics-15-01313" class="html-bibr">129</a>], copyright Elsevier 2020. ((<b>B</b>), i) SEM images and diameter distributions of electrospun nanofiber (30/70 weight ratio of chitosan to Pullulan) ((<b>B</b>), ii) Presentation of solubility behavior of C/P 0/100 nanofiber film and C/P 30/70 nanofiber film. The photos were taken before the film was in contact with water (a, a’) and after the film was in contact with water for 5 s (b, b’), 30 s (c, c’), and 60 s (d, d’). Reproduced with permission from [<a href="#B132-pharmaceutics-15-01313" class="html-bibr">132</a>], copyright Elsevier 2019.</p>
Full article ">Figure 5
<p>((<b>A</b>), i) Inverted fluorescence microscope micrographs after time-coursed in vivo corneal permeation of the preparations, the eye treated with normal saline was used as control. (Scale bar: 150 μm). ((<b>A</b>), ii) Ex vivo fluorescence imaging of rabbit ocular tissues from rabbits treated with different Coumarin 6-loaded formulations. Here, F1 = chitosan-N-acetyl-L-cysteine-coated NLC, F2 = chitosan oligosaccharide-coated NLC, F3 = carboxymethyl chitosan-coated NLC, F4 = Uncoated NLC, F5 = Coumarin 6 eyedrops. Reproduced with permission from [<a href="#B141-pharmaceutics-15-01313" class="html-bibr">141</a>], copyright Elsevier 2017. ((<b>B</b>), i) SEM images of polyacrylonitrile modified nanofibers at different magnifications: PAN (a) and (b); PANEDA (c) and (d); PAN-EDA-OC (e) and (f); PAN-EDA-OC-ACY (g) and (h). ((<b>B</b>), ii) Cell attachment of HASCs on modified fibrous scaffolds. Reproduced with permission from [<a href="#B150-pharmaceutics-15-01313" class="html-bibr">150</a>], copyright Elsevier 2020.</p>
Full article ">Figure 6
<p>((<b>A</b>), i) Cryo-TEM images of N-O-carboxymethyl chitosan-dopamine amide conjugate nanoparticles (Scale bar: 500 nm). ((<b>A</b>), ii) Epifluorescence microscopy of olfactory ensheathing cells incubated with FITC-loaded N-O-carboxymethyl chitosan-dopamine amide conjugate nanoparticles at dopamine concentrations of 18.75 (a,c) and 75 μM (b,d), and FITC-loaded N-O-carboxymethyl chitosan-dopamine amide conjugate nanoparticles 75 μM (e,f), incubated with olfactory ensheathing cells in the presence or absence of mucin for 2 h and then evaluated by epifluorescence microscopy. Controls (CTRL) were cells incubated with medium only in the presence or absence of mucin (g,h). Arrows indicate nanoparticles in close vicinity of nuclei as dots, while arrowheads point to more diffuse perinuclear staining (Scale bar: 10 μm). Reproduced with permission from [<a href="#B162-pharmaceutics-15-01313" class="html-bibr">162</a>], copyright MDPI 2019. ((<b>B</b>), i) TEM image of luteolin-loaded chitosomes (150,000× magnification). Arrows point to the chitosan coating layer. ((<b>B</b>), ii) Photomicrograph immunohistochemistry of GFAP expression in brain tissue. Group 2 (disease control) shows a marked expression of GFAP; however, the morphological difference between the two treated groups was not observed. Here, significant difference was considered at <span class="html-italic">p</span> &lt; 0.05, *. Statistically significant difference from the normal group at <span class="html-italic">p</span> &lt; 0.05, **. Reproduced with permission from [<a href="#B163-pharmaceutics-15-01313" class="html-bibr">163</a>], copyright MDPI 2022.</p>
Full article ">Figure 7
<p>((<b>A</b>), i) Vulvovaginal histological sections from the experimental groups of animals. Female rats were intravaginally infected or not with <span class="html-italic">C. albicans</span> and treated or not with MFM-chitosan gel or clotrimazole. H&amp;E-stained vulvovaginal tissue sections were analyzed by light microscopy. ((<b>A</b>), ii) SEM views of vulvovaginal epithelium in vulvovaginal candidiasis rat models after vaginal topical treatment with MFM-chitosan gel. (A–C) shows the well-preserved ultrastructure of vaginal epithelium treated with chitosan-gel containing 2.5% (A), 5.0% (B), and 10.0% (C) MFM [fungal cells are highlighted in green]. Reproduced with permission from [<a href="#B170-pharmaceutics-15-01313" class="html-bibr">170</a>], copyright Elsevier 2020. ((<b>B</b>), i) SEM images of (A) Unloaded 5:5 chitosan/sodium alginate PEC-based insert, (B) Fluconazole 5:5 chitosan/sodium alginate PEC, (C) Unloaded 5:5 chitosan/xanthan gum PEC-based insert, (D) Fluconazole 5:5 chitosan/xanthan gum PEC, (E) Unloaded 5:5 chitosan/carpobol PEC-based insert, (F) Fluconazole 5:5 chitosan/carpobol PEC ((<b>B</b>), ii) Histological examination of Candida infected vaginal tissue treated by unloaded vaginal insert, fluconazole PEC based vaginal insert and fluconazole solution. (A) Control normal vaginal tissue; (B) Control Candida infected, non-treated vaginal tissue; (C) Candida infected vaginal tissue treated by unloaded vaginal insert; (D) Candida infected vaginal tissue treated by fluconazole solution; (E) Candida infected vaginal tissue treated by fluconazole vaginal insert. Stars represent inflammatory cells; Black arrows represent normal epithelium; Dotted arrows represent hyperplastic or damaged epithelium. Reproduced with permission from [<a href="#B176-pharmaceutics-15-01313" class="html-bibr">176</a>], copyright MDPI 2018.</p>
Full article ">Figure 7 Cont.
<p>((<b>A</b>), i) Vulvovaginal histological sections from the experimental groups of animals. Female rats were intravaginally infected or not with <span class="html-italic">C. albicans</span> and treated or not with MFM-chitosan gel or clotrimazole. H&amp;E-stained vulvovaginal tissue sections were analyzed by light microscopy. ((<b>A</b>), ii) SEM views of vulvovaginal epithelium in vulvovaginal candidiasis rat models after vaginal topical treatment with MFM-chitosan gel. (A–C) shows the well-preserved ultrastructure of vaginal epithelium treated with chitosan-gel containing 2.5% (A), 5.0% (B), and 10.0% (C) MFM [fungal cells are highlighted in green]. Reproduced with permission from [<a href="#B170-pharmaceutics-15-01313" class="html-bibr">170</a>], copyright Elsevier 2020. ((<b>B</b>), i) SEM images of (A) Unloaded 5:5 chitosan/sodium alginate PEC-based insert, (B) Fluconazole 5:5 chitosan/sodium alginate PEC, (C) Unloaded 5:5 chitosan/xanthan gum PEC-based insert, (D) Fluconazole 5:5 chitosan/xanthan gum PEC, (E) Unloaded 5:5 chitosan/carpobol PEC-based insert, (F) Fluconazole 5:5 chitosan/carpobol PEC ((<b>B</b>), ii) Histological examination of Candida infected vaginal tissue treated by unloaded vaginal insert, fluconazole PEC based vaginal insert and fluconazole solution. (A) Control normal vaginal tissue; (B) Control Candida infected, non-treated vaginal tissue; (C) Candida infected vaginal tissue treated by unloaded vaginal insert; (D) Candida infected vaginal tissue treated by fluconazole solution; (E) Candida infected vaginal tissue treated by fluconazole vaginal insert. Stars represent inflammatory cells; Black arrows represent normal epithelium; Dotted arrows represent hyperplastic or damaged epithelium. Reproduced with permission from [<a href="#B176-pharmaceutics-15-01313" class="html-bibr">176</a>], copyright MDPI 2018.</p>
Full article ">Figure 8
<p>(<b>A</b>) Synthesis of the dual-responsive hydrogel composite by incorporating NIR-responsive polydopamine-coated magnesium–calcium carbonate microspheres into a thermo-responsive hydroxy butyl chitosan hydrogel and its application for sequential Aspirin/bone morphogenetic protein-2 delivery. Reproduced with permission from [<a href="#B181-pharmaceutics-15-01313" class="html-bibr">181</a>], copyright Elsevier 2022. ((<b>B</b>), i) Photographic and SEM images of porous hybrid calcium phosphate/chitosan membranes, scale bar: 10 μm. ((<b>B</b>), ii) SEM image showing the osteoblast cell growth and formation of mineral-surrounded clusters, indicated by arrows. Scale bar: 10 μm. ((<b>B</b>), iii) X-ray and micro-CT imaging in Sprague-Dawley rats (21 days post-surgery). All rat skulls were punched with two holes having 4 mm diameter and then covered with different membranes listed in the top panel. B–F panel are X-ray images while B’–F’ are micro-CT images as explained in figure. B–F and B’–F’ correspond to materials 1–5 as shown in panel above X-ray images. Reproduced with permission from [<a href="#B182-pharmaceutics-15-01313" class="html-bibr">182</a>], copyright Elsevier 2019.</p>
Full article ">Figure 9
<p>(<b>i</b>) Graphical illustration for the mechanism of the self-assembled microspheres. (<b>ii</b>) Bright-field [a], florescent [b], mix CLSM [c], and SEM images [d, e] of MC3T3-E1 cells co-cultured with microspheres after 3 days. The graph shows the adhesion-related gene expression of MC3T3-E1 cells on chitosan microspheres without nanofibers and nanofibrous chitosan microspheres (Here, **** <span class="html-italic">p</span> &lt; 0.0001) [f]. (<b>iii</b>) Micro CT reconstruction images of the bone defect. Reproduced with permission from [<a href="#B184-pharmaceutics-15-01313" class="html-bibr">184</a>], copyright Elsevier 2022.</p>
Full article ">Figure 10
<p>((<b>A</b>), i) Schematic image of the scaffold showing mean pore size in each phase with corresponding SEM images showing internal pore structure of the composite scaffolds in the cartilage and bone phases. ((<b>A</b>), ii) Comparison of compressive modulus of chitosan-only and chitosan-nano-hydroxyapatite composite scaffolds. ((<b>A</b>), iii) Sulphated glycosaminoglycans measured in mesenchymal stem cells-seeded chitosan scaffolds exposed to chondrogenic culture conditions. (Here, SC = chitosan scaffold in standard culture, CC = chitosan scaffold in chondrogenic medium, SnHA = chitosan-nHA scaffold in standard culture, CnHA = chitosan-nHA scaffold in chondrogenic medium; <span class="html-italic">p</span> values ** ≤ 0.01, *** ≤ 0.001, **** ≤ 0.0001). ((<b>A</b>), iv) Fluorescent microscopy showing MSCs seeded onto chitosan-nHA composite scaffolds after 14 days in osteogenic medium. Reproduced with permission from [<a href="#B188-pharmaceutics-15-01313" class="html-bibr">188</a>], copyright Elsevier 2022. (<b>B</b>) Immunofluorescent staining of bone marrow mesenchymal stem cells encapsulated in hyaluronic acid/chitosan coacervate-based scaffolds at different days of chondrogenic differentiation. Blue represents cell nuclei, green represents COL2A1, red represents ACAN and Phalloidin. Reproduced with permission from [<a href="#B190-pharmaceutics-15-01313" class="html-bibr">190</a>], copyright Elsevier 2021.</p>
Full article ">Figure 11
<p>((<b>A</b>), i) Schematic depicting the synthesis route and UCMSCs encapsulated in CS/Dex/β-GP hydrogel for use in cardiac repair applications. ((<b>A</b>), ii) Gelation time of CS/Dex/β-GP hydrogel with varying concentrations of dextran, here significant differences were defined as <span class="html-italic">p</span> values * ≤ 0.05, *** ≤ 0.001. ((<b>A</b>), iii) Confocal microscope images showing the morphology of UCMSCs in hydrogels before and after injection. ((<b>A</b>), iv) Representative Western blot assay for detecting the levels of p-ERK and p-ERK1/2 of UCMSCs cultured in hydrogels for 2 days. ((<b>A</b>), v) cTnI (green) and Cx43 (red) expression of UCMSCs in cultured hydrogels, cell nuclei were stained by Hoechst (blue). Reproduced with permission from [<a href="#B196-pharmaceutics-15-01313" class="html-bibr">196</a>], copyright Elsevier 2020. ((<b>B</b>), i) SEM image of polypyrrole/chitosan/collagen electrospun nanofiber scaffold. ((<b>B</b>), ii) The electrical conductivity and stress–strain curve of different nanofibrous scaffolds. Reproduced with permission from [<a href="#B197-pharmaceutics-15-01313" class="html-bibr">197</a>], copyright Elsevier 2019.</p>
Full article ">Figure 12
<p>((<b>A</b>), i) Macroscopic photographs of dried composite membranes showing a decrease in visual transparency with increasing PCL content. ((<b>A</b>), ii) The effect of PCL content on light transmittance of the composite membranes (significant differences were defined as <span class="html-italic">p</span> values **** ≤ 0.0001). ((<b>A</b>), iii) Representative SEM images of corneal epithelial cells cultured on CSNP/PCL 50/25 for 5 days. Reproduced with permission from [<a href="#B204-pharmaceutics-15-01313" class="html-bibr">204</a>], copyright Springer Nature 2021. ((<b>B</b>), i) Transmittance of DC hydrogel in water and in PBS (2 wt%) between 25 and 40 °C (λ = 700 nm). ((<b>B</b>), ii) AFM images of self-assembled DC hydrogel at different temperatures. ((<b>B</b>), iii) Representative micrographs of DC hydrogel stimulating corneal stromal cell migration after 12 h in the scratching assay (100× magnification, Scale bar: 100 μm). ((<b>B</b>), iv) Confocal laser scanning microscopy graphs of rabbit corneal stromal cells cultured in hydrogel at day 7 (Scale bar: 20 μm). ((<b>B</b>), v) H&amp;E and Masson staining images of corneal stroma defect with and without DC hydrogel at 4 weeks after surgery (Scale bar: 500 μm for 2×, 20 μm for 40×; Black arrows point to keratocytes). Reproduced with permission from [<a href="#B205-pharmaceutics-15-01313" class="html-bibr">205</a>], copyright American Chemical Society.</p>
Full article ">Figure 13
<p>((<b>A</b>), i) Image showing the one-walled defects created surgically at mesial and distal sides of maxillary first premolar ((<b>A</b>), ii) New bone formation seen in defects of trilayer functional chitosan membrane (left)- and Biomend<sup>®</sup> (right)-treated groups, the rectangular frame was chosen for bone density analysis. ((<b>A</b>), iii) Graph depicting the percentage of new bone formation (Here, significant difference was labeled as * <span class="html-italic">p</span> &lt; 0.05). Reproduced with permission from [<a href="#B213-pharmaceutics-15-01313" class="html-bibr">213</a>], copyright Elsevier 2016. ((<b>B</b>), i) SEM image of fibrin with ε-aminocaproic acid loaded chitosan-tripolyphosphate nanoparticles (Scale bar: 2 μm). ((<b>B</b>), ii) Micro-computed topography-based measurement of the linear distance of vertical alveolar bone regeneration (compared with enamel matrix derivative, EMD). ((<b>B</b>), iii) Histological and immune-histological analyses of cementum formation and Sharpey’s fiber insertions to bone and newly formed cementum tissues. Upon comparison with fibrin-only (unmodified fibrin hydrogel) and EMD groups, the fibrin-ACP group facilitated the regeneration of periodontal tissues such as cementum on tooth-root surfaces, periodontal ligament, and the alveolar bone. More critically, the fibrin-ACP promoted Sharpey’s fiber formations and insertions into the cementum layers and alveolar bone surfaces, indicated by white arrows. Reproduced with permission from [<a href="#B215-pharmaceutics-15-01313" class="html-bibr">215</a>], copyright Elsevier 2017.</p>
Full article ">Figure 14
<p>((<b>A</b>), i) Schematic illustration of the fabrication process of the MACSs. (Here, F: PLA microfiber; C: Chitosan; M: Microchannel; AC: Alkylated chitosan). ((<b>A</b>), ii) Micro-CT images depicting the macro and microstructure of the alkylated chitosan sponge (without microchannel structure) ACS and different preparations of MACS. ((<b>A</b>), iii) Macro photographs of the blood-triggered shape recovery of MACS. ((<b>A</b>), iv) Photographs of the hemostatic effect of CELOXTM and MACS in the normal rat liver perforation wound model (yellow arrow and dotted line represented the bleeding site and liver boundary, respectively) along with quantitative data of total blood loss and hemostatic time for different treatment groups. Reproduced with permission from [<a href="#B222-pharmaceutics-15-01313" class="html-bibr">222</a>], copyright Springer Nature 2021. ((<b>B</b>), i) Photographs (upper), SEM images (middle), and fluorescent images (bottom) of electrospinning membranes (scale bar: 1 cm, black; 10 μm, Re; 100 μm, white). ((<b>B</b>), ii) Representative images showing full-thickness skin defects treated with the petrolatum gauze (Control) and nBG-TFM at a predetermined time post-surgery (scale bar: 10 mm). Reproduced with permission from [<a href="#B225-pharmaceutics-15-01313" class="html-bibr">225</a>], copyright Elsevier 2019.</p>
Full article ">Figure 15
<p>(<b>i</b>) Schematic illustrations for the preparation of PEI/PAA/QCS powder and the formation of PEI/PAA/QCS powder-derived hydrogel by adding anticoagulated blood. The photos are of the PEI/PAA/QCS powder and a pentagram PEI/PAA/QCS hydrogel formed by adding anticoagulated blood. (<b>ii</b>) SEM images of red blood cells (red arrow) and activated platelets (blue arrow) on the surface of PEI/PAA/QCS hydrogel. (<b>iii</b>) Schematic and photos of creating acute bleeding and stopping bleeding by applying PEI/PAA/QCS powder femoral artery and tail vein bleeding models. Reproduced with permission from [<a href="#B226-pharmaceutics-15-01313" class="html-bibr">226</a>], copyright Wiley-VCH 2021.</p>
Full article ">Figure 16
<p>(<b>i</b>) SEM images of BP/CS-bFGF hydrogels. (Scale bar: 200 μm). (<b>ii</b>) Photographs of the self-healing performance of the BP/CS-bFGF hydrogel. (<b>iii</b>) Images of cell migration at different times with corresponding values of wound area closure treated with various samples and migration rate of hGFs cells upon the prepared hydrogels (Here, significant differences were defined as <span class="html-italic">p</span> values * ≤ 0.05, *** ≤ 0.001). Reproduced with permission from [<a href="#B228-pharmaceutics-15-01313" class="html-bibr">228</a>], copyright Elsevier 2022.</p>
Full article ">Figure 17
<p>(<b>i</b>) Schematic mechanism of RCP/pJUN-PSPF@PGA scaffold on the nerve regeneration via the located gene transfection of c-JUN: (a) preparation of RCP/pJUN and RCP/pDNA-PSPF@PGA; (b) bridging surgery in sciatic nerve defect of rat; (c) located delivery of RCP/pJUN nanoparticles and nerve repair; (d) transfection of c-Jun via RCP/pJUN in cells and three factors secretion; (e) Bungner bands formation and axon regeneration. (<b>ii</b>) Nerve growth factor and brain-derived neurotrophic factor expression level in transfected RSC96s cell line (Here, ## <span class="html-italic">p</span> &lt; 0.01, compared with control; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). (<b>iii</b>) Evaluation of nerve regeneration: (a) H&amp;E-stained tissue section images and (b) TB-stained tissue sections images at 12 weeks postoperatively. Reproduced with permission from [<a href="#B241-pharmaceutics-15-01313" class="html-bibr">241</a>], copyright Elsevier 2022.</p>
Full article ">Figure 18
<p>(<b>i</b>) Schematic illustration of NS-GAM as a gene delivery system for deep second-degree burn wound. (<b>ii</b>) In vitro release of pDNA from NS-GAM: (a) Cumulative amount of pDNA released in vitro from NS-GAM and the agarose gel electrophoresis of the plasmids; (b) SEM of the surface of NS-GAM. (<b>iii</b>) Gross examination and healing rate: (a) Observation of the deep second-degree burn wounds. A: Control group; B: NS-GAM group; (b) The calculated wound size reduction (Significant difference was considered at <span class="html-italic">p</span> &lt; 0.05 *). Reproduced with permission from [<a href="#B243-pharmaceutics-15-01313" class="html-bibr">243</a>], copyright Elsevier 2022.</p>
Full article ">Figure 19
<p>((<b>A</b>), i) The fluorescence intensities of tumor-bearing mice were monitored after 4 h of the last injection to detect the targeting effect. ((<b>A</b>), ii) The protein expression of CLl-2 was analyzed by western blotting (quantification of the protein level was normalized to GAPDH, significant differences were defined as <span class="html-italic">p</span> values * ≤ 0.05 and ns represents non-significance). ((<b>A</b>), iii) Immunofluorescence observation under the same exposure for each fluorescent channel after siRNA@chitosan-HAD nanoparticles (b), naked siRNA (c), and siRNA@chitosan nanoparticles (d) treatment compared with the control group (a). Reproduced with permission from [<a href="#B247-pharmaceutics-15-01313" class="html-bibr">247</a>], copyright Elsevier 2021. ((<b>B</b>), i) Confocal laser scanning microscopy images showing uptake in A549 cells after incubation with AA-CS/pDNA (2 μg/mL pDNA-rho) for 4 h. Hoechst 33342 (blue) and Lyso-Tracker Green were used to stain cell nuclei and lysosome, respectively, (Scale bar: 10 μm) ((<b>B</b>), ii) Confocal laser scanning microscopy images showing the endosomal escape of DMAPAPA-chitosan/pDNA-rho, PEI-chitosan/pDNA-rho, or PEI-25 kDa/pDNA-rho in A549 cells. The cells were stained with Lyso-Tracker Green and Hoechst 33342 (Scale bar: 10 μm). Reproduced with permission from [<a href="#B248-pharmaceutics-15-01313" class="html-bibr">248</a>], copyright Elsevier 2020.</p>
Full article ">Figure 20
<p>((<b>A</b>), i) Fluorescence image excited at 488 nm and NIR image excited at 808 nm of HeLa cells incubated with Ag<sub>2</sub>S(DOX)@CS nanospheres for 12 h. The fluorescence image was acquired in a wavelength window between 560 and 600 nm (Scale bar: 25 μm) ((<b>A</b>), ii) Viability of HeLa cells incubated with different concentrations of Ag<sub>2</sub>S@CS nanospheres ((<b>A</b>), iii) ICP-MS analysis of tumor and five major organs of the mice sacrificed at different time points (statistical significance: * <span class="html-italic">p</span> &lt; 0.05). ((<b>A</b>), iv) In vivo NIR images of a nude mouse at 6 h (i), 12 h (ii), and 24 h (iii) after injection of the Ag<sub>2</sub>S(DOX)@CS nanospheres; ex vivo NIR image of the tumor (iv) and the organs (v) harvested from the sacrificed nude mouse. Reproduced with permission from [<a href="#B140-pharmaceutics-15-01313" class="html-bibr">140</a>], copyright Elsevier 2017. ((<b>B</b>), i) TEM images of blank TPE-bi(SS-CS-Bio) micelles (Scale bar: 100 nm). ((<b>B</b>), ii) Confocal laser scanning microscopy images of MCF-7 cells after incubation with TPE-bi(SS-CS-Bio) for 4 h (Scale bar: 10 μm); and after incubation at different time points (1, 2, 3, and 4 h). Reproduced with permission from [<a href="#B255-pharmaceutics-15-01313" class="html-bibr">255</a>], copyright Elsevier 2021.</p>
Full article ">
23 pages, 3204 KiB  
Review
Local Delivery and Controlled Release Drugs Systems: A New Approach for the Clinical Treatment of Periodontitis Therapy
by Mariacristina Amato, Simona Santonocito, Alessandro Polizzi, Gianluca Martino Tartaglia, Vincenzo Ronsivalle, Gaia Viglianisi, Cristina Grippaudo and Gaetano Isola
Pharmaceutics 2023, 15(4), 1312; https://doi.org/10.3390/pharmaceutics15041312 - 21 Apr 2023
Cited by 25 | Viewed by 5033
Abstract
Periodontitis is an inflammatory disease of the gums characterized by the degeneration of periodontal ligaments, the formation of periodontal pockets, and the resorption of the alveolar bone, which results in the destruction of the teeth’s supporting structure. Periodontitis is caused by the growth [...] Read more.
Periodontitis is an inflammatory disease of the gums characterized by the degeneration of periodontal ligaments, the formation of periodontal pockets, and the resorption of the alveolar bone, which results in the destruction of the teeth’s supporting structure. Periodontitis is caused by the growth of diverse microflora (particularly anaerobes) in the pockets, releasing toxins and enzymes and stimulating the immune system. Various approaches, both local and systemic, have been used to treat periodontitis effectively. Successful treatment depends on reducing bacterial biofilm, bleeding on probing (BOP), and reducing or eliminating pockets. Currently, the use of local drug delivery systems (LDDSs) as an adjunctive therapy to scaling and root planing (SRP) in periodontitis is a promising strategy, resulting in greater efficacy and fewer adverse effects by controlling drug release. Selecting an appropriate bioactive agent and route of administration is the cornerstone of a successful periodontitis treatment plan. In this context, this review focuses on applications of LDDSs with varying properties in treating periodontitis with or without systemic diseases to identify current challenges and future research directions. Full article
Show Figures

Figure 1

Figure 1
<p>The available LDDSs for periodontal treatment.</p>
Full article ">Figure 2
<p>Fibers and their placement. Reproduced with the permission from Rajeshwari et al. [<a href="#B32-pharmaceutics-15-01312" class="html-bibr">32</a>].</p>
Full article ">Figure 3
<p>The procedure of electrospinning to make nanofibers. Each nanofiber has antibacterial and antiphlogistic activity and promotes tissue growth. Nanofibers are assembled in membranes and then applied to the periodontitis-affected site.</p>
Full article ">Figure 4
<p>Strips and Films and their placement. Reproduced with the permission from Rajeshwari et al. [<a href="#B32-pharmaceutics-15-01312" class="html-bibr">32</a>].</p>
Full article ">Figure 5
<p>Microparticles and nanosystems and their placement, reproduced with permission from Rajeshwari et al. [<a href="#B32-pharmaceutics-15-01312" class="html-bibr">32</a>].</p>
Full article ">Figure 6
<p>Gels and their placement. Reproduced with the permission from Rajeshwari et al. [<a href="#B32-pharmaceutics-15-01312" class="html-bibr">32</a>].</p>
Full article ">Figure 7
<p>Membrane and its placement in order to act as a barrier.</p>
Full article ">Figure 8
<p>Scaffold and its placement.</p>
Full article ">Figure 9
<p>A flow chart of the classification of LDDSs and their modalities of application [<a href="#B32-pharmaceutics-15-01312" class="html-bibr">32</a>,<a href="#B122-pharmaceutics-15-01312" class="html-bibr">122</a>].</p>
Full article ">
10 pages, 4537 KiB  
Communication
Activation of Insulin Gene Expression via Transfection of a CRISPR/dCas9a System Using Magnetic Peptide-Imprinted Nanoparticles
by Mei-Hwa Lee, James L. Thomas, Chien-Yu Lin, Yi-Chen Ethan Li and Hung-Yin Lin
Pharmaceutics 2023, 15(4), 1311; https://doi.org/10.3390/pharmaceutics15041311 - 21 Apr 2023
Cited by 1 | Viewed by 2166
Abstract
A CRISPRa transcription activation system was used to upregulate insulin expression in HEK293T cells. To increase the delivery of the targeted CRISPR/dCas9a, magnetic chitosan nanoparticles, imprinted with a peptide from the Cas9 protein, were developed, characterized, and then bound to dCas9a that was [...] Read more.
A CRISPRa transcription activation system was used to upregulate insulin expression in HEK293T cells. To increase the delivery of the targeted CRISPR/dCas9a, magnetic chitosan nanoparticles, imprinted with a peptide from the Cas9 protein, were developed, characterized, and then bound to dCas9a that was complexed with a guide RNA (gRNA). The adsorption of dCas9 proteins conjugated with activators (SunTag, VPR, and p300) to the nanoparticles was monitored using both ELISA kits and Cas9 staining. Finally, the nanoparticles were used to deliver dCas9a that was complexed with a synthetic gRNA into HEK293T cells to activate their insulin gene expression. Delivery and gene expression were examined using quantitative real-time polymerase chain reaction (qRT-PCR) and staining of insulin. Finally, the long-term release of insulin and the cellular pathway related to stimulation by glucose were also investigated. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Size distribution of the magnetic nanoparticles (MNPs), magnetic non-imprinted chitosan nanoparticles (MNIPs), and peptide-imprinted chitosan nanoparticles (MPIPs). (<b>b</b>) Magnetization curves of MNPs, MNIPs, and MPIPs after template removal. (<b>c</b>) Immunostaining images of MPIPs without and with adsorption of dCas9a proteins. (<b>d</b>) Adsorption capacities of MPIPs with the same concentration of dCas9a proteins. All experiments were carried out in triplicate, and data are expressed as means ± standard deviation. Standard deviation is based on at least three measurements, and **: <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 2
<p>Immunohistochemistry (IHC) images of (<b>a</b>) optical, (<b>b</b>) DAPI-staining, (<b>c</b>) anti-insulin staining, and (<b>d</b>) merge of (<b>b</b>) and (<b>c</b>) in HEK-293T cells treated with MPIPs/dCas9-VPR and crRNA.</p>
Full article ">Figure 3
<p>IHC images of optical, DAPI-staining, and anti-insulin staining in HEK-293T cells without RNPs/MPIPs.</p>
Full article ">Figure 4
<p>(<b>a</b>) Relative gene expression of key cellular differentiation genes of β-cells for activation of insulin with MPIPs/RNPs in HEK-293T cells. The administration of RNPs had no effect on the expression levels of these genes. (<b>b</b>) Insulin release from transfected HEK293T cells during an hour of glucose stimulation. (<b>c</b>) Insulin secretion of controls, RNPs, with MNIP, MPIPs, or Lipofectamine™ CRISPRMAX™-treated HEK293T cells after high glucose stimulation. (<b>d</b>) Insulin secretion 30 min after glucose re-stimulation on the 3rd, 6th, 9th 16th, 23rd, and 30th day. (*: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.005, and ***: <span class="html-italic">p</span> &lt; 0.0005) The image in (<b>b</b>) has been published elsewhere [<a href="#B34-pharmaceutics-15-01311" class="html-bibr">34</a>], where it was used only for comparison purposes. Reproduced from Ref. [<a href="#B34-pharmaceutics-15-01311" class="html-bibr">34</a>] with permission from the Royal Society of Chemistry.</p>
Full article ">Figure 4 Cont.
<p>(<b>a</b>) Relative gene expression of key cellular differentiation genes of β-cells for activation of insulin with MPIPs/RNPs in HEK-293T cells. The administration of RNPs had no effect on the expression levels of these genes. (<b>b</b>) Insulin release from transfected HEK293T cells during an hour of glucose stimulation. (<b>c</b>) Insulin secretion of controls, RNPs, with MNIP, MPIPs, or Lipofectamine™ CRISPRMAX™-treated HEK293T cells after high glucose stimulation. (<b>d</b>) Insulin secretion 30 min after glucose re-stimulation on the 3rd, 6th, 9th 16th, 23rd, and 30th day. (*: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.005, and ***: <span class="html-italic">p</span> &lt; 0.0005) The image in (<b>b</b>) has been published elsewhere [<a href="#B34-pharmaceutics-15-01311" class="html-bibr">34</a>], where it was used only for comparison purposes. Reproduced from Ref. [<a href="#B34-pharmaceutics-15-01311" class="html-bibr">34</a>] with permission from the Royal Society of Chemistry.</p>
Full article ">Figure 5
<p>Immunohistochemistry (IHC) images of (<b>a</b>,<b>e</b>) optical, (<b>b</b>,<b>f</b>) DAPI, (<b>c</b>) anti-PDX1, (<b>d</b>) anti-MAFA, (<b>g</b>) anti-NKX6.1, and (<b>h</b>) anti-Ngn3 staining in HEK293T cells treated with MPIPs/RNPs.</p>
Full article ">Figure 6
<p>Relative gene expression of proteins of insulin-activated HEK-293T cells with MPIPs/RNPs before and after glucose stimulation. Many genes responsible for cellular proliferation are concomitantly upregulated with insulin. All experiments were carried out in triplicate, and data are expressed as means ± standard deviation; *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.005, and ***: <span class="html-italic">p</span> &lt; 0.0005.</p>
Full article ">Scheme 1
<p>Activation of insulin gene expression via transfection of a CRISPR/dCas9a system using magnetic peptide-imprinted nanoparticles.</p>
Full article ">
18 pages, 3742 KiB  
Article
Linker-Free Synthesis of Antimicrobial Peptides Using a Novel Cleavage Reagent: Characterisation of the Molecular and Ionic Composition by nanoESI-HR MS
by Roser Segovia, Mireia Díaz-Lobo, Yolanda Cajal, Marta Vilaseca and Francesc Rabanal
Pharmaceutics 2023, 15(4), 1310; https://doi.org/10.3390/pharmaceutics15041310 - 21 Apr 2023
Cited by 1 | Viewed by 3244
Abstract
The efficient preparation of novel bioactive peptide drugs requires the availability of reliable and accessible chemical methodologies together with suitable analytical techniques for the full characterisation of the synthesised compounds. Herein, we describe a novel acidolytic method with application to the synthesis of [...] Read more.
The efficient preparation of novel bioactive peptide drugs requires the availability of reliable and accessible chemical methodologies together with suitable analytical techniques for the full characterisation of the synthesised compounds. Herein, we describe a novel acidolytic method with application to the synthesis of cyclic and linear peptides involving benzyl-type protection. The process consists of the in situ generation of anhydrous hydrogen bromide and a trialkylsilyl bromide that acts as protic and Lewis acid reagents. This method proved to be useful to effectively remove benzyl-type protecting groups and cleave Fmoc/tBu assembled peptides directly attached to 4-methylbenzhydrylamine (MBHA) resins with no need for using mild trifluoroacetic acid labile linkers. The novel methodology was successful in synthesising three antimicrobial peptides, including the cyclic compound polymyxin B3, dusquetide, and RR4 heptapeptide. Furthermore, electrospray mass spectrometry (ESI-MS) is successfully used for the full characterisation of both the molecular and ionic composition of the synthetic peptides. Full article
(This article belongs to the Special Issue Chemically Enhanced Peptide and Protein Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Scheme of the synthesis of polymyxin B<sub>3</sub> (<span class="html-italic">n</span>-octyl-Dab-Thr-Dab-<span class="html-italic">cyclo</span>[Dab-Dab-DPhe-Leu-Dab-Dab-Thr]. (i) 1.2 eq of Fmoc-Thr(Bn)-OH and 4 eq of DIPEA in CH<sub>2</sub>Cl<sub>2</sub> at room temperature, overnight; (ii) 20% of piperidine in DMF (1 × 1 min, 2 × 10 min); (iii) repetitive cycles of coupling and deprotection using a 3-fold molar excess of protected Fmoc-AA-OH (or octanoic acid at the end) and DIC/HOBt; (iv) CH<sub>2</sub>Cl<sub>2</sub>/TFA/TES/H<sub>2</sub>O (55:40:3:2) for 30 min at room temperature (ŋ<sub>cleavage</sub> = 82%); (v) HATU/HOAt/DIPEA (2:2:4 eq) for 2–4 h, monitored by TLC stained with ninhydrin (ŋ<sub>cyclization</sub> = 81%); and (vi) HBr/Et<sub>3</sub>Si-Br/TES/TFA, in situ prepared with TFA/TES/Br<sub>2</sub> (82.5:15:2.5) for 45 min at room temperature (ŋ<sub>acidolysis</sub> = 92%). Final yield: 21%, purity &gt; 95%. R = C<sub>7</sub>H<sub>15</sub> (corresponding to octanoyl).</p>
Full article ">Figure 2
<p>PxB<sub>3</sub> crude HPLC chromatograms obtained by elution using a linear gradient from 5% to 95% of B over 30 min; eluants, A: 0.045% TFA in H<sub>2</sub>O and B: 0.036% TFA in acetonitrile. Crudes were obtained after treatment of #1, #2, #3, #4, and #5 (see <a href="#pharmaceutics-15-01310-t001" class="html-table">Table 1</a> for detailed reaction conditions). ▲ = PxB<sub>3</sub>, C<sub>55</sub>H<sub>96</sub>N<sub>16</sub>O<sub>13</sub>, exact mass: 1188.73, found 1190.11 [M + H]<sup>+</sup>; ● = PxB<sub>3</sub> + Bn, C<sub>62</sub>H<sub>102</sub>N<sub>16</sub>O<sub>13</sub>, exact mass: 1278.78, found 1279.72 [M + H]<sup>+</sup>; ■ = PxB<sub>3</sub> + 2Bn, C<sub>69</sub>H<sub>108</sub>N<sub>16</sub>O<sub>13</sub>, exact mass: 1368.82, found 1370.10 [M + H]<sup>+</sup>; ◆ = PxB<sub>3</sub> − 2H<sub>2</sub>O, C<sub>55</sub>H<sub>92</sub>N<sub>16</sub>O<sub>11</sub>, exact mass: 1152.71, found 1153.86 [M + H]<sup>+</sup>.</p>
Full article ">Figure 3
<p>HPLC chromatogram of purified PxB<sub>3</sub>, using a linear gradient from 5% to 95% of B in 30 min; eluants, A: 0.045% TFA in H<sub>2</sub>O and B: 0.036% TFA in acetonitrile, detection at 220 nm.</p>
Full article ">Figure 4
<p>Complete scheme of the synthetic route of (<b>A</b>) dusquetide; (<b>B</b>) segments 8–14 of RR4. (i) Repetitive cycles of coupling and deprotection using a 3-fold molar excess of Fmoc-AA-OH/DIC/HOBt and 20% of piperidine in DMF (1 × 1 min, 2 × 10 min); (ii) TFA/HBr/Et<sub>3</sub>SiBr/TES, obtained by reaction at 0 °C of TFA:TES:Br<sub>2</sub> (82.5:15:2.5) for 90 min at room temperature; (ii*) TFA/HBr/iP<sub>3</sub>SiBr/TIS, obtained by reaction at 0 °C of TFA:TIS:Br<sub>2</sub> (82.5:15:2.5) for 90 min at room temperature.</p>
Full article ">Figure 5
<p>HPLC chromatograms of (<b>A</b>) dusquetide, (<b>B</b>) segment 8–14 of RR4 using a linear gradient from 5% to 95% of B over 30 min with an elution system of (<b>A</b>) 0.045% TFA in H<sub>2</sub>O and (<b>B</b>) 0.036% TFA in acetonitrile and processed at 220 nm.</p>
Full article ">Figure 6
<p>HPLC chromatogram of commercially available polymyxin B run at 1 mL/min using a linear gradient from 20% to 35% of B over 30 min with an elution system of A: 0.045% TFA in H<sub>2</sub>O and B: 0.036% TFA in acetonitrile and processed at 220 nm.</p>
Full article ">Figure 7
<p>Spectra obtained by nanoESI-HR MS in the negative mode for samples: (<b>A</b>) PxB<sub>3</sub> after the in situ-generated HBr acidolysis method; (<b>B</b>) purified PxB<sub>3</sub>; and (<b>C</b>) PxB<sub>3</sub>, obtained after the aqueous HCl exchange by lyophilization. “z” indicates the charge of the non-covalent ion complexes detected.</p>
Full article ">Figure 8
<p>Spectra obtained by ESI-MS in the negative mode for samples: (<b>A</b>) reaction crude of synthetic PxB<sub>3</sub> after HBr deprotection treatment; (<b>B</b>) purified PxB<sub>3</sub>; and (<b>C</b>) PxB<sub>3</sub> hydrochloride salt.</p>
Full article ">Figure 9
<p>Spectra obtained by ESI-MS in the negative mode for samples: (<b>A</b>) crude RR4; (<b>B</b>) crude dusquetide. The MS spectra correspond to samples whose HPLC profile can be found in <a href="#pharmaceutics-15-01310-f005" class="html-fig">Figure 5</a>.</p>
Full article ">Scheme 1
<p>In situ hydrogen bromide generation by the reaction of triethylsilane with bromine.</p>
Full article ">Scheme 2
<p>In situ hydrogen bromide generation by reaction of triisopropylsilane with bromine.</p>
Full article ">
17 pages, 2294 KiB  
Article
Cerium End-Deposited Gold Nanorods-Based Photoimmunotherapy for Boosting Tumor Immunogenicity
by Yanlin Feng, Yumei Xu, Zhaoyang Wen, Xin Ning, Jianlin Wang, Deping Wang, Jimin Cao and Xin Zhou
Pharmaceutics 2023, 15(4), 1309; https://doi.org/10.3390/pharmaceutics15041309 - 21 Apr 2023
Cited by 2 | Viewed by 2393
Abstract
Background: Triple-negative breast cancer (TNBC) was closely related to high metastatic risk and mortality and has not yet found a targeted receptor for targeted therapy. Cancer immunotherapy, especially photoimmunotherapy, shows promising potential in TNBC treatment because of great spatiotemporal controllability and non-trauma. However, [...] Read more.
Background: Triple-negative breast cancer (TNBC) was closely related to high metastatic risk and mortality and has not yet found a targeted receptor for targeted therapy. Cancer immunotherapy, especially photoimmunotherapy, shows promising potential in TNBC treatment because of great spatiotemporal controllability and non-trauma. However, the therapeutic effectiveness was limited by insufficient tumor antigen generation and the immunosuppressive microenvironment. Methods: We report on the design of cerium oxide (CeO2) end-deposited gold nanorods (CEG) to achieve excellent near-infrared photoimmunotherapy. CEG was synthesized through hydrolyzing of ceria precursor (cerium acetate, Ce(AC)3) on the surface of Au nanorods (NRs) for cancer therapy. The therapeutic response was first verified in murine mammary carcinoma (4T1) cells and then monitored by analysis of the anti-tumor effect in xenograft mouse models. Results: Under near-infrared (NIR) light irradiation, CEG can efficiently generate hot electrons and avoid hot-electron recombination to release heat and form reactive oxygen species (ROS), triggering immunogenic cell death (ICD) and activating part of the immune response. Simultaneously, combining with PD-1 antibody could further enhance cytotoxic T lymphocyte infiltration. Conclusions: Compared with CBG NRs, CEG NRs showed strong photothermal and photodynamic effects to destroy tumors and activate a part of the immune response. Combining with PD-1 antibody could reverse the immunosuppressive microenvironment and thoroughly activate the immune response. This platform demonstrates the superiority of combination therapy of photoimmunotherapy and PD-1 blockade in TNBC therapy. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the combination therapy of photoimmunotherapy synergizes with PD-1 blockade in triple-negative breast cancer using CEG NRs. (<b>A</b>) Illustration of the synthesis process of CEG and CBG [<a href="#B14-pharmaceutics-15-01309" class="html-bibr">14</a>]. (<b>B</b>) NIR laser-activated charge carrier spatial separation to release heat and promote more ROS production for CEG than CBG. (<b>C</b>) CEG NRs displayed significant PDT and PTT effects to activate systemic immunity to destroy tumor cells together with α-PD-1 after intravenous administration to breast cancer-bearing mice.</p>
Full article ">Figure 2
<p>Physicochemical characterization of CEG and CBG NRs. (<b>A</b>–<b>C</b>) TEM images of GNRs, CEG, and CBG NRs, respectively. (<b>D</b>,<b>E</b>) STEM and EDS elemental mapping images of CEG and CBG NRs. (<b>F</b>) The XRD pattern of CEG and CBG NRs. (<b>G</b>) The UV-Vis-NIR absorption spectra of GNRs, CEG, and CBG.</p>
Full article ">Figure 3
<p>The photothermal and photodynamic performance of GNRs, CEG, and CBG NRs. (<b>A</b>) Heating and cooling curves and (<b>B</b>) infrared thermal images of GNRs, CEG, CBG NRs, and H<sub>2</sub>O. Total ROS, abiotic <sup>•</sup>OH, <sup>1</sup>O<sub>2</sub>, and O<sub>2</sub><sup>•−</sup> assessments of GNRs, CEG, and CBG NRs (OD = 1) upon 808 nm laser irradiation (1 W/cm<sup>2</sup>, 10 min) assessed by DCF (<b>C</b>), TA (<b>D</b>), SOSG (<b>E</b>) and superoxide anion assay (<b>F</b>), respectively.</p>
Full article ">Figure 4
<p>In vitro phototherapeutic efficacy of CEG in 4T1 cells. (<b>A</b>) Result of CCK-8 assay showing the cell viability of 4T1 cells following the treatment of CEG (OD = 0, 0.25, 0.5, 1, and 2) without or with 808 nm laser irradiation (1 W/cm<sup>2</sup>, 10 min). (<b>B</b>) Live/dead staining of 4T1 cells treated with CEG (OD = 0, 0.25, 0.5, 1, and 2) without or with 808 nm laser irradiation (1 W/cm<sup>2</sup>, 10 min). (<b>C</b>) Results of western blotting showing the expression levels of HSP-70 and HO-1 without or with NIR laser irradiation. (<b>D</b>) Flow cytometric analysis of intracellular ROS levels in 4T1 cells after the treatment of CEG upon 808 nm laser irradiation or not. CLSM images of (<b>E</b>) mitochondrial membrane depolarization (JC-1, green) and (<b>F</b>) superoxide production (Mitosox Red, red). DAPI staining for Cell nuclei (blue). (<b>G</b>) Analysis of flow cytometry for the apoptotic 4T1 cells induced by CEG upon 808 nm laser irradiation or not. Statistical differences were determined by Student’s t-test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>In vivo photoimmunotherapy of CEG synergized with α-PD-1 in 4T1 tumor-bearing mice. (<b>A</b>) Therapeutic protocol illustration of 4T1 tumor mice. (<b>B</b>) Fluorescence images of mice after intravenous injection of CEG-Cy7 at 1, 12, and 24 h. (<b>C</b>) Infrared thermal images of 4T1 tumor-bearing mice at 24 h post-injection of PBS or CEG with 808 nm laser radiation (1 W/cm<sup>2</sup>, 10 min). (<b>D</b>) Tumor growth curves of different groups for 14 days after intravenous injection of CEG. (<b>E</b>) Tumor images of different groups at the end of treatment. (<b>F</b>,<b>G</b>) H&amp;E and TUNEL staining for the tumor tissues in each group. (<b>H</b>) Immunofluorescence staining for CD3 T cells and CD8 T cells in tumors after different treatments. (<b>I</b>) Serum IgG, (<b>J</b>) TNF-α, and (<b>K</b>) IFN-γ levels of six groups after different treatments measured by ELISA. Data are presented as means ± s.d. (<span class="html-italic">n</span> = 3). Statistical differences were determined by Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
29 pages, 6477 KiB  
Article
Amphiphilic Polypeptides Obtained by Post-Polymerization Modification of Poly-l-Lysine as Systems for Combined Delivery of Paclitaxel and siRNA
by Apollinariia Dzhuzha, Erik Gandalipov, Viktor Korzhikov-Vlakh, Elena Katernyuk, Natalia Zakharova, Sergey Silonov, Tatiana Tennikova and Evgenia Korzhikova-Vlakh
Pharmaceutics 2023, 15(4), 1308; https://doi.org/10.3390/pharmaceutics15041308 - 21 Apr 2023
Cited by 4 | Viewed by 2174
Abstract
The development of effective anti-cancer therapeutics remains one of the current pharmaceutical challenges. The joint delivery of chemotherapeutic agents and biopharmaceuticals is a cutting-edge approach to creating therapeutic agents of enhanced efficacy. In this study, amphiphilic polypeptide delivery systems capable of loading both [...] Read more.
The development of effective anti-cancer therapeutics remains one of the current pharmaceutical challenges. The joint delivery of chemotherapeutic agents and biopharmaceuticals is a cutting-edge approach to creating therapeutic agents of enhanced efficacy. In this study, amphiphilic polypeptide delivery systems capable of loading both hydrophobic drug and small interfering RNA (siRNA) were developed. The synthesis of amphiphilic polypeptides included two steps: (i) synthesis of poly-αl-lysine by ring-opening polymerization and (ii) its post-polymerization modification with hydrophobic l-amino acid and l-arginine/l-histidine. The obtained polymers were used for the preparation of single and dual delivery systems of PTX and short double-stranded nucleic acid. The obtained double component systems were quite compact and had a hydrodynamic diameter in the range of 90–200 nm depending on the polypeptide. The release of PTX from the formulations was studied, and the release profiles were approximated using a number of mathematical dissolution models to establish the most probable release mechanism. A determination of the cytotoxicity in normal (HEK 293T) and cancer (HeLa and A549) cells revealed the higher toxicity of the polypeptide particles to cancer cells. The separate evaluation of the biological activity of PTX and anti-GFP siRNA formulations testified the inhibitory efficiency of PTX formulations based on all polypeptides (IC50 4.5–6.2 ng/mL), while gene silencing was effective only for the Tyr-Arg-containing polypeptide (56–70% GFP knockdown). Full article
(This article belongs to the Special Issue Self-Assembled Amphiphilic Copolymers in Drug Delivery)
Show Figures

Figure 1

Figure 1
<p>Scheme of the synthesis of cationic amphiphilic polypeptides: (<b>A</b>) synthesis of P[K] by ROP; (<b>B</b>) post-polymerization modification of P[K] by hydrophobic and basic amino acids; (<b>C</b>) deprotection of side chain functional groups.</p>
Full article ">Figure 2
<p>Effect of PTX loading into polypeptide nanoparticles on the hydrodynamic diameters of the delivery systems.</p>
Full article ">Figure 3
<p>Effect of polypeptide/oligo-dT-dA ratio on <span class="html-italic">D<sub>H</sub></span> and ζ-potential (above the bars) for the complexes based on P[KK(Y)K(R)] (<b>A</b>) and P[KK(Y)K(H)] (<b>B</b>) (PP is non-loaded polypeptide particles).</p>
Full article ">Figure 4
<p>Agarose gel electrophorese images: Arg-containing polypeptide/oligo-dT-dA ratio = 8 (<b>A</b>), Arg-containing polypeptide/oligo-dT-dA ratio = 12 (<b>B</b>), His-containing polypeptide/oligo-dT-dA ratio = 12 (<b>C</b>), His-containing polypeptide/oligo-dT-dA ratio = 16 (<b>D</b>). Lanes: 1—markers; 2—free oligo-dT-dA; (<b>A</b>,<b>B</b>) 3—P[KK(F)K(R)]@oligo-dT-dA; 4—P[KK(V)K(R)]@oligo-dT-dA; 5—P[KK(I)K(R)]@oligo-dT-dA; 6—P[KK(Y)K(R)]@oligo-dT-dA; 7—P[KK(W)K(R)]@oligo-dT-dA; (<b>C</b>,<b>D</b>) 3—P[KK(F)K(H)]@oligo-dT-dA; 4—P[KK(V)K(H)]@oligo-dT-dA; 5—P[KK(I)K(H)]@oligo-dT-dA; 6—P[KK(Y)K(H)]@oligo-dT-dA; 7—P[KK(W)K(H)]@oligo-dT-dA.</p>
Full article ">Figure 5
<p>Comparison of hydrodynamic dimeters of empty polypeptide particles (PP), single-laded particles containing PTX (PP@PTX) or oligo-dT-dA (PP@oligo-dT-dA), as well as dual-component systems (PP@PTX + oligo-dT-dA). The dual-component delivery systems were prepared using 25 μg of PTX per mg of polymer and at the optimal polypeptide/oligo-dT-dA ratios: 12 for Arg-containing polypeptides and 16 for His-containing ones. For Ile-containing polypeptides, the polymer/oligo-dT-dA ratios were 16 and 20 for P[KK(I)K(R)] and P[KK(I)K(H)], respectively.</p>
Full article ">Figure 6
<p>TEM images of empty PP (<b>A</b>), PP@PTX (<b>B</b>), and PP@PTX + oligo-dT-dA (<b>C</b>) based on the P[KK(Y)K(R)] polypeptide. Scale bar is 200 nm; staining with uranyl acetate. In (<b>B</b>,<b>C</b>), the PTX load was 50 μg/mg of polymer; the ratio of polypeptide/oligo-dT-dA in (<b>C</b>) was 12 (<span class="html-italic">w</span>/<span class="html-italic">w</span>).</p>
Full article ">Figure 7
<p>Storage stability of empty nanoparticles, as well as single- and dual-component systems based on P[KK(Y)K(R)] (<b>A</b>) and P[KK(Y)K(H)] (<b>B</b>) polypeptides, within 3 weeks at room temperature (20 °C, pH 7.4).</p>
Full article ">Figure 8
<p>Heparin displacement test (agarose gel electrophoresis): (<b>A</b>) PP@oligo-dT-dA based on Arg-containing polypeptides; (<b>B</b>) PP@oligo-dT-dA based on His-containing polypeptides; (<b>C</b>) PP@PTX+oligo-dT-dA based on Arg-containing polypeptides; (<b>D</b>) PP@PTX+oligo-dT-dA based on His-containing polypeptides. The concentration of heparin was varied from 0 to 40 IU. In the case of Arg-containing polypeptides besides P[KK(I)K(R)], the polypeptide/oligo-dT-dA ratio was 12; for P[KK(I)K(R)], it was 16. In the case of His-containing polypeptides besides P[KK(I)K(H)], the polypeptide/oligo-dT-dA ratio was 16; for P[KK(I)K(H)], it was 20.</p>
Full article ">Figure 9
<p>PTX release profiles from single (<b>A</b>) and double (<b>B</b>) component formulations (0.01 M PBS, pH 7.4, 37 °C).</p>
Full article ">Figure 10
<p>Effect of co-encapsulation of PTX and oligo-dT-dA on the mechanism of PTX release from P[KK(Y)K(R)]-based delivery systems, evaluated with the application of various mathematical modeling: (<b>A</b>) comparison of correlation coefficients of the regressions obtained with different models for the release of PTX from single- and dual-component systems during 240 h; (<b>B</b>) effect of oligo-dT-dA co-encapsulation on the n parameter evaluated from the Korsmeyer–Peppas model; (<b>C</b>) results obtained by application of the Peppas–Sahlin model, where <span class="html-italic">K</span><sub>1</sub> is the impact of diffusion and <span class="html-italic">K</span><sub>2</sub> is the impact of relaxation on the release mechanism.</p>
Full article ">Figure 11
<p>GFP silencing in K562/GFP cells after transfection with PP@anti-GFP siRNA (48 h): (<b>A</b>) the results of analysis by flow cytometry; (<b>B</b>) total GFP knockdown (flow cytometry); (<b>C</b>) ubnormal cells (flow cytometry); (<b>D</b>) GFP mRNA expression (RT PCR). The amount of anti-GFP siRNA used for the study was 100 and 200 nM. The polypeptide/siRNA ratio was 12 for Arg-containing polypeptides and 16 for His-containing ones. Complexes of GenJect-39 and GenJect-40 with anti-GFP siRNA were prepared according to the protocol of the manufacturer. The differences with the positive control (GenJect-39/GenJect-40) (<b>B</b>,<b>D</b>) and negative control (non-treated cells) (<b>C</b>) were significant with <span class="html-italic">p</span> &lt; 0.05 (*) and <span class="html-italic">p</span> &lt; 0.005 (**).</p>
Full article ">
15 pages, 7315 KiB  
Article
Poly(l-Ornithine)-Based Polymeric Micelles as pH-Responsive Macromolecular Anticancer Agents
by Miao Pan, Chao Lu, Wancong Zhang, Huan Huang, Xingyu Shi, Shijie Tang and Daojun Liu
Pharmaceutics 2023, 15(4), 1307; https://doi.org/10.3390/pharmaceutics15041307 - 21 Apr 2023
Cited by 4 | Viewed by 1943
Abstract
Anticancer peptides and polymers represent an emerging field of tumor treatment and can physically interact with tumor cells to address the problem of multidrug resistance. In the present study, poly(l-ornithine)-b-poly(l-phenylalanine) (PLO-b-PLF) block copolypeptides were prepared [...] Read more.
Anticancer peptides and polymers represent an emerging field of tumor treatment and can physically interact with tumor cells to address the problem of multidrug resistance. In the present study, poly(l-ornithine)-b-poly(l-phenylalanine) (PLO-b-PLF) block copolypeptides were prepared and evaluated as macromolecular anticancer agents. Amphiphilic PLO-b-PLF self-assembles into nanosized polymeric micelles in aqueous solution. Cationic PLO-b-PLF micelles interact steadily with the negatively charged surfaces of cancer cells via electrostatic interactions and kill the cancer cells via membrane lysis. To alleviate the cytotoxicity of PLO-b-PLF, 1,2-dicarboxylic-cyclohexene anhydride (DCA) was anchored to the side chains of PLO via an acid-labile β-amide bond to fabricate PLO(DCA)-b-PLF. Anionic PLO(DCA)-b-PLF showed negligible hemolysis and cytotoxicity under neutral physiological conditions but recovered cytotoxicity (anticancer activity) upon charge reversal in the weakly acidic microenvironment of the tumor. PLO-based polypeptides might have potential applications in the emerging field of drug-free tumor treatment. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Hydrodynamic diameter (<b>a</b>) and zeta potential (<b>b</b>) of block polypeptide micelles in PBS as determined by dynamic light scattering.</p>
Full article ">Figure 2
<p>Zeta potentials of <b>PD4</b> as a function of incubation time at pH 6.5 or 7.4.</p>
Full article ">Figure 3
<p>Fluorescence images of HepG2 cells costained with PI (red, dead cells) and calcein AM (green, live cells) after exposure to <b>PD4</b> for 24 h at different pH values.</p>
Full article ">Figure 4
<p>(<b>A</b>) Zeta potential of HepG2 cells treated with different concentrations of <b>P1</b> and <b>P4</b>, as well as HK-2 cells treated with different concentrations of <b>PD4</b>, for 30 min. (<b>B</b>) LDH release by HepG2 cells treated with different concentrations of <b>P1</b> and <b>P4</b>. (<b>C</b>) Schematic illustration of polymer interaction with cells.</p>
Full article ">Figure 5
<p>Flow cytometry analysis of untreated HepG2 cells and HepG2 cells treated with different <b>P4</b> concentrations for 60 min. FITC<sup>low</sup> PI<sup>low</sup>: live cells; FITC<sup>high</sup> PI<sup>low</sup>: apoptotic cells; FITC<sup>high/low</sup> PI<sup>high</sup>: necrotic cells.</p>
Full article ">Figure 6
<p>SEM images of HepG2 cells before and after treatment with different concentrations of <b>P4</b> for 60 min.</p>
Full article ">Figure 7
<p>CLSM images of HepG2 cells before and after incubation with different concentrations of <b>P4</b> for 30 min. Nuclei were stained with Hoechst (blue) while the cell membrane was stained with DiO (green).</p>
Full article ">Scheme 1
<p>Synthetic route of PLO-based polypeptides.</p>
Full article ">
22 pages, 1043 KiB  
Review
Stability of Oral Liquid Dosage Forms in Pediatric Cardiology: A Prerequisite for Patient’s Safety—A Narrative Review
by Carmen-Maria Jîtcă, George Jîtcă, Bianca-Eugenia Ősz, Amalia Pușcaș and Silvia Imre
Pharmaceutics 2023, 15(4), 1306; https://doi.org/10.3390/pharmaceutics15041306 - 21 Apr 2023
Cited by 1 | Viewed by 6680
Abstract
The development of safe and effective pediatric formulations is essential, especially in therapeutic areas such as pediatric cardiology, where the treatment requires multiple dosing or outpatient care. Although liquid oral dosage forms are considered the formulation of choice given the dose flexibility and [...] Read more.
The development of safe and effective pediatric formulations is essential, especially in therapeutic areas such as pediatric cardiology, where the treatment requires multiple dosing or outpatient care. Although liquid oral dosage forms are considered the formulation of choice given the dose flexibility and acceptability, the compounding practices are not endorsed by the health authorities, and achieving stability can be problematic. The purpose of this study is to provide a comprehensive overview of the stability of liquid oral dosage forms used in pediatric cardiology. An extensive review of the literature has been performed, with a particular focus on cardiovascular pharmacotherapy, by consulting the current studies indexed in PubMed, ScienceDirect, PLoS One, and Google Scholar databases. Regulations and guidelines have been considered against the studies found in the literature. Overall, the stability study is well-designed, and the critical quality attributes (CQAs) have been selected for testing. Several approaches have been identified as innovative in order to optimize stability, but opportunities to improve have been also identified, such as in-use studies and achieving dose standardization. Consequently, the information gathering and the results of the studies can be translated into clinical practice in order to achieve the desired stability of liquid oral dosage forms. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart describing literature search.</p>
Full article ">Figure 2
<p>Left-hand side: stability issues for liquid dosage forms depending on the formulation (solutions or suspensions) and the compounding technique, respectively; right-hand side: common stability issues for liquid oral dosage forms.</p>
Full article ">
18 pages, 2028 KiB  
Article
Design, Evaluation and Comparison of Nanostructured Lipid Carriers and Chitosan Nanoparticles as Carriers of Poorly Soluble Drugs to Develop Oral Liquid Formulations Suitable for Pediatric Use
by Giulia Nerli, Lídia M. D. Gonçalves, Marzia Cirri, António J. Almeida, Francesca Maestrelli, Natascia Mennini and Paola A. Mura
Pharmaceutics 2023, 15(4), 1305; https://doi.org/10.3390/pharmaceutics15041305 - 21 Apr 2023
Cited by 10 | Viewed by 2252
Abstract
There is a serious need of pediatric drug formulations, whose lack causes the frequent use of extemporaneous preparations obtained from adult dosage forms, with consequent safety and quality risks. Oral solutions are the best choice for pediatric patients, due to administration ease and [...] Read more.
There is a serious need of pediatric drug formulations, whose lack causes the frequent use of extemporaneous preparations obtained from adult dosage forms, with consequent safety and quality risks. Oral solutions are the best choice for pediatric patients, due to administration ease and dosage-adaptability, but their development is challenging, particularly for poorly soluble drugs. In this work, chitosan nanoparticles (CSNPs) and nanostructured lipid carriers (NLCs) were developed and evaluated as potential nanocarriers for preparing oral pediatric solutions of cefixime (poorly soluble model drug). The selected CSNPs and NLCs showed a size around 390 nm, Zeta-potential > 30 mV, and comparable entrapment efficiency (31–36%), but CSNPs had higher loading efficiency (5.2 vs. 1.4%). CSNPs maintained an almost unchanged size, homogeneity, and Zeta-potential during storage, while NLCs exhibited a marked progressive Zeta-potential decrease. Drug release from CSNPs formulations (differently from NLCs) was poorly affected by gastric pH variations, and gave rise to a more reproducible and controlled profile. This was related to their behavior in simulated gastric conditions, where CSNPs were stable, while NLCs suffered a rapid size increase, up to micrometric dimensions. Cytotoxicity studies confirmed CSNPs as the best nanocarrier, proving their complete biocompatibility, while NLCs formulations needed 1:1 dilution to obtain acceptable cell viability values. Full article
(This article belongs to the Special Issue Development of Chitosan/Cyclodextrins in Drug Delivery Field)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Release profiles of CEF from NLC4<sub>DL4</sub> (○) and CS NP<sub>DL4</sub> (□) formulations: (<b>A</b>) 2 h in SGF pH 1.2 and 4 h in SIF pH 6.8; (<b>B</b>) 2 h in SGF pH 4.5 and 4 h in SIF pH 6.8.</p>
Full article ">Figure 2
<p>Stability of the selected NLC4<sub>DL4</sub> and CS NP<sub>DL4</sub> formulations in SGF at pH 1.2 (<b>A</b>) or at pH 4.5 (<b>B</b>).</p>
Full article ">Figure 3
<p>Effect of storage on mean particle size, PDI, and Zeta potential of the selected NLC4<sub>DL4</sub> and CS NP<sub>DL4</sub> formulations.</p>
Full article ">Figure 4
<p>Propidium iodide (PI) uptake by Caco-2 cell line after 24 h incubation with empty and CEF-loaded NLCs (orange bars) or CS NPs (blue bars) and their components. Fresh culture medium and SDS (1 mg/mL) were used as negative and positive control. Results are expressed as relative fluorescence intensity (RFI) with respect to cells incubated with culture medium (mean ± SD, 4 replicates per plate in 3 independent plates).</p>
Full article ">Figure 5
<p>The percent cell viability of Caco-2 cell line measured by MTT test after 24 h incubation with empty and CEF-loaded NLCs (orange bars) or CS NPs (blue bars) and their components. Fresh culture medium and SDS (1 mg/mL) were used as negative and positive control, respectively (mean ± SD, 4 replicates per plate in 3 independent plates).</p>
Full article ">
14 pages, 474 KiB  
Article
A Multi-Criteria Decision Aid Tool for Radiopharmaceutical Selection in Tau PET Imaging
by Ilker Ozsahin, Efe Precious Onakpojeruo, Berna Uzun, Dilber Uzun Ozsahin and Tracy A. Butler
Pharmaceutics 2023, 15(4), 1304; https://doi.org/10.3390/pharmaceutics15041304 - 21 Apr 2023
Cited by 5 | Viewed by 2111
Abstract
The accumulation of pathologically misfolded tau is a feature shared by a group of neurodegenerative disorders collectively referred to as tauopathies. Alzheimer’s disease (AD) is the most prevalent of these tauopathies. Immunohistochemical evaluation allows neuropathologists to visualize paired-helical filaments (PHFs)—tau pathological lesions, but [...] Read more.
The accumulation of pathologically misfolded tau is a feature shared by a group of neurodegenerative disorders collectively referred to as tauopathies. Alzheimer’s disease (AD) is the most prevalent of these tauopathies. Immunohistochemical evaluation allows neuropathologists to visualize paired-helical filaments (PHFs)—tau pathological lesions, but this is possible only after death and only shows tau in the portion of brain sampled. Positron emission tomography (PET) imaging allows both the quantitative and qualitative analysis of pathology over the whole brain of a living subject. The ability to detect and quantify tau pathology in vivo using PET can aid in the early diagnosis of AD, provide a way to monitor disease progression, and determine the effectiveness of therapeutic interventions aimed at reducing tau pathology. Several tau-specific PET radiotracers are now available for research purposes, and one is approved for clinical use. This study aims to analyze, compare, and rank currently available tau PET radiotracers using the fuzzy preference ranking organization method for enrichment of evaluations (PROMETHEE), which is a multi-criteria decision-making (MCDM) tool. The evaluation is based on relatively weighted criteria, such as specificity, target binding affinity, brain uptake, brain penetration, and rates of adverse reactions. Based on the selected criteria and assigned weights, this study shows that a second-generation tau tracer, [18F]RO-948, may be the most favorable. This flexible method can be extended and updated to include new tracers, additional criteria, and modified weights to help researchers and clinicians select the optimal tau PET tracer for specific purposes. Additional work is needed to confirm these results, including a systematic approach to defining and weighting criteria and clinical validation of tracers in different diseases and patient populations. Full article
(This article belongs to the Special Issue Radiopharmaceuticals: From Design to Applications)
Show Figures

Figure 1

Figure 1
<p>Positive and negative aspects of the alternatives. c1 is specificity, c2 is target binding affinity, c3 is brain uptake and penetration, and c4 is adverse reaction. The criteria that stand on the upper side of the plot are considered positive aspects, while the ones below zero mean that they exhibit undesired properties.</p>
Full article ">
16 pages, 2864 KiB  
Article
Release of TGF-β3 from Surface-Modified PCL Fiber Mats Triggers a Dose-Dependent Chondrogenic Differentiation of Human Mesenchymal Stromal Cells
by Leonie Berten-Schunk, Yvonne Roger, Heike Bunjes and Andrea Hoffmann
Pharmaceutics 2023, 15(4), 1303; https://doi.org/10.3390/pharmaceutics15041303 - 21 Apr 2023
Cited by 5 | Viewed by 1955
Abstract
The design of implants for tissue transitions remains a major scientific challenge. This is due to gradients in characteristics that need to be restored. The rotator cuff in the shoulder, with its direct osteo-tendinous junction (enthesis), is a prime example of such a [...] Read more.
The design of implants for tissue transitions remains a major scientific challenge. This is due to gradients in characteristics that need to be restored. The rotator cuff in the shoulder, with its direct osteo-tendinous junction (enthesis), is a prime example of such a transition. Our approach towards an optimized implant for entheses is based on electrospun fiber mats of poly(ε-caprolactone) (PCL) as biodegradable scaffold material, loaded with biologically active factors. Chitosan/tripolyphosphate (CS/TPP) nanoparticles were used to load transforming growth factor-β3 (TGF-β3) with increasing loading concentrations for the regeneration of the cartilage zone within direct entheses. Release experiments were performed, and the concentration of TGF-β3 in the release medium was determined by ELISA. Chondrogenic differentiation of human mesenchymal stromal cells (MSCs) was analyzed in the presence of released TGF-β3. The amount of released TGF-β3 increased with the use of higher loading concentrations. This correlated with larger cell pellets and an increase in chondrogenic marker genes (SOX9, COL2A1, COMP). These data were further supported by an increase in the glycosaminoglycan (GAG)-to-DNA ratio of the cell pellets. The results demonstrate an increase in the total release of TGF-β3 by loading higher concentrations to the implant, which led to the desired biological effect. Full article
(This article belongs to the Special Issue Innovative Drug Delivery Systems for Regenerative Medicine)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Procedure of fiber mat loading and release experiments for ELISA/cell culture testing. During release experiments, loaded fiber mat samples were transferred to fresh release medium at each sampling time point. Furthermore, the release samples were used to determine the TGF-β<sub>3</sub> concentration by ELISA and to examine the chondrogenic effect in the cell culture.</p>
Full article ">Figure 2
<p>Cumulative release and release rate of TGF-β<sub>3</sub> from fiber mats (1.28 cm<sup>2</sup>) loaded with various protein concentrations. The graphs show the results of (<b>a</b>) release experiment “A” (ELISA analysis) and (<b>b</b>) the corresponding release experiment “C” for cell culture (sodium azide-free medium, ELISA analysis, later use for chondrogenic induction). The cumulated amount of TGF-β<sub>3</sub> released until the end of the experiment increased with the use of higher loading concentrations, as indicated in the two upper graphs. (<b>c</b>) The release rate of TGF-β<sub>3</sub> from fiber mats (1.28 cm<sup>2</sup>) loaded with various protein concentrations (release experiment “A”) and (<b>d</b>) the release rate in the corresponding release experiment “C” for the cell culture. With the use of higher loading concentrations, release doses above the minimal effective dose for cell culture (MED<sub>cell culture</sub>, marked in <b>c</b>,<b>d</b>) of 0.5 ng/d could be maintained for a longer period of time.</p>
Full article ">Figure 3
<p>Correlation between TGF-β<sub>3</sub> loading concentration and total release from implant prototype. The adjusted coefficient of determination for a linear correlation was 0.881, while variations increased with higher loading concentrations.</p>
Full article ">Figure 4
<p>Chondrogenic differentiation of MSCs from donor A was enhanced with increasing loading concentrations. w/: with TGF-β<sub>3</sub>, w/o: without TGF-β<sub>3</sub>. (<b>a</b>) Chondrogenic cell pellets. In the presence of TGF-β<sub>3</sub> (0 µg/mL w/ TGF-β<sub>3</sub> (i.e., release medium of fiber mats w/o TGF-β<sub>3</sub> loading, but with external addition of TGF-β<sub>3</sub>, positive control), 1 µg/mL, 10 µg/mL, 20 µg/mL), the morphology of the pellets was round and compact, and the size increased with increasing loading concentrations. In contrast, without TGF-β<sub>3</sub> (0 µg/mL w/o TGF-β<sub>3</sub>), the pellets showed considerably less compact growth and were smaller in size. Scale bar: 500 µm. (<b>b</b>) Ratio of glycosaminoglycan (GAG) to DNA. The increasing loading concentration led to an increase in GAG/DNA. Cell pellets, which were not treated with TGF-β<sub>3</sub> (0 µg/mL w/o TGF-β<sub>3</sub>) or were treated with release medium from the lowest loading concentration (1 µg/mL), showed a very low GAG/DNA ratio. (<b>c</b>) Gene expression analysis of the chondrogenic marker genes <span class="html-italic">SOX9</span>, <span class="html-italic">COL2A1</span> and <span class="html-italic">COMP</span>. Shown is the fold change against 0 µg/mL w/o TGF-β<sub>3</sub>. All three chondrogenic marker genes resulted in an increase in expression with the treatment of TGF-β<sub>3</sub>, and almost no expression without TGF-β<sub>3</sub> treatment.</p>
Full article ">Figure 5
<p>MSCs seeded as positive and negative controls in a 2D manner (<b>left panel</b>) as well as on modified fiber mats loaded with empty chitosan nanoparticles (<b>right panel</b>). Cells were stained with phalloidin (red) and DAPI (blue) to visualize the actin cytoskeleton and the nuclei, respectively. Both sets were analyzed in the presence and absence of TGF-β<sub>3</sub> (upper and lower panel, respectively). The control with TGF-β<sub>3</sub> under 2D conditions showed rounded cells (<b>left</b>, upper panel), with morphology similar to that of chondrocytes and in striking contrast to the elongated fibroblast-like cells observed in the absence of TGF-β<sub>3</sub> (<b>left</b>, lower panel). The right panel shows MSCs seeded on modified fiber mats loaded with empty chitosan nanoparticles. The right upper panel shows the condition with TGF-β<sub>3</sub> with some rounded cells (white arrow) and the lower panel without TGF-β<sub>3</sub>.</p>
Full article ">Figure 6
<p>Chondrogenic differentiation on modified fiber mats. (<b>a</b>) Modified fiber mats loaded with different TGF-β<sub>3</sub> concentrations (1 µg/mL (<b>upper panel</b>), 10 µg/mL (<b>middle panel</b>) and 20 µg/mL (<b>lower panel</b>)), were seeded with MSCs and cultivated for 27 days. Cells were stained with phalloidin for the actin cytoskeleton and with DAPI for the DNA. Chondrocyte-like cells are marked with white arrows in the merge panel. Scale bar: 100 µm. (<b>b</b>) Gene expression analyses of chondrogenic marker genes <span class="html-italic">SOX9</span>, <span class="html-italic">COL2A1</span> and <span class="html-italic">COMP</span>, normalized to the housekeeper <span class="html-italic">RPS29</span> and shown in fold change to the negative control 0 µg/mL w/o TGF-β<sub>3</sub>.</p>
Full article ">
14 pages, 2153 KiB  
Article
Improved Tumor Control Following Radiosensitization with Ultrasound-Sensitive Oxygen Microbubbles and Tumor Mitochondrial Respiration Inhibitors in a Preclinical Model of Head and Neck Cancer
by Quezia Lacerda, Hebah Falatah, Ji-Bin Liu, Corinne E. Wessner, Brian Oeffinger, Ankit Rochani, Dennis B. Leeper, Flemming Forsberg, Joseph M. Curry, Gagan Kaushal, Scott W. Keith, Patrick O’Kane, Margaret A. Wheatley and John R. Eisenbrey
Pharmaceutics 2023, 15(4), 1302; https://doi.org/10.3390/pharmaceutics15041302 - 21 Apr 2023
Cited by 8 | Viewed by 3084
Abstract
Tumor hypoxia (oxygen deficiency) is a major contributor to radiotherapy resistance. Ultrasound-sensitive microbubbles containing oxygen have been explored as a mechanism for overcoming tumor hypoxia locally prior to radiotherapy. Previously, our group demonstrated the ability to encapsulate and deliver a pharmacological inhibitor of [...] Read more.
Tumor hypoxia (oxygen deficiency) is a major contributor to radiotherapy resistance. Ultrasound-sensitive microbubbles containing oxygen have been explored as a mechanism for overcoming tumor hypoxia locally prior to radiotherapy. Previously, our group demonstrated the ability to encapsulate and deliver a pharmacological inhibitor of tumor mitochondrial respiration (lonidamine (LND)), which resulted in ultrasound-sensitive microbubbles loaded with O2 and LND providing prolonged oxygenation relative to oxygenated microbubbles alone. This follow-up study aimed to evaluate the therapeutic response to radiation following the administration of oxygen microbubbles combined with tumor mitochondrial respiration inhibitors in a head and neck squamous cell carcinoma (HNSCC) tumor model. The influences of different radiation dose rates and treatment combinations were also explored. The results demonstrated that the co-delivery of O2 and LND successfully sensitized HNSCC tumors to radiation, and this was also enhanced with oral metformin, significantly slowing tumor growth relative to unsensitized controls (p < 0.01). Microbubble sensitization was also shown to improve overall animal survival. Importantly, effects were found to be radiation dose-rate-dependent, reflecting the transient nature of tumor oxygenation. Full article
(This article belongs to the Special Issue Cavitation-Enhanced Drug Delivery and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Light microscopy images of (<b>A</b>) SE61O<sub>2</sub> and (<b>B</b>) SE61O<sub>2</sub>/LND.</p>
Full article ">Figure 2
<p>(<b>A</b>) Illustrative ultrasound images of SE61O<sub>2</sub>/LND in a closed-loop flow phantom showing contrast mode (<b>left</b>) and fundamental (B-mode) (<b>right</b>). Image depth markers correspond to 0.3 cm increments with the flow direction from right to left. (<b>B</b>) Microbubble stability curves of SE61O<sub>2</sub>/LND (green), SE61O<sub>2</sub> (yellow), and SE61N<sub>2</sub>/LND (blue) at a non-destructive MI (MI = 0.12). No statistically significant differences among formulations were observed in vitro (<span class="html-italic">p</span> = 0.99).</p>
Full article ">Figure 3
<p>Illustrative ultrasound images of SE61O<sub>2</sub>/LND in an HNSCC tumor model generated in immunocompromised mice in dual B-mode/cadence pulse sequencing mode. Images show pre-injection of microbubbles (MB), start of MB perfusion, peak enhancement post-injection, start of MB destruction, complete MB destruction, and start of reperfusion (image depth corresponds to 0.3 cm increments). The left side of the display is the nonlinear imaging mode (contrast), and the right is the conventional B-mode (grayscale imaging).</p>
Full article ">Figure 4
<p>Tumor growth over time per animal for each group showing raw tumor volumes (mm<sup>3</sup>) from the day of treatment until sacrifice was required. Results show the influence of ultrasound (US), SE61 microbubbles, radiation therapy (5 Gy), gas (oxygen or nitrogen), and tumor mitochondrial respiration inhibitors (metformin (OM) and lonidamine (LND)) on an HNSCC tumor model.</p>
Full article ">Figure 5
<p>Tumor volumes plotted against time with fitted exponential growth curves showing tumoral response to therapy, with 95% confidence bands shown as dashed lines. Tumor volumes are plotted to the day of treatment to show the influence of ultrasound (US), SE61 microbubbles, radiation therapy (5 Gy), gas (oxygen or nitrogen), and tumor mitochondrial respiration inhibitors (metformin (OM) and lonidamine (LND)).</p>
Full article ">Figure 6
<p>Survival proportion of animals following treatment until day of sacrifice. (<b>A</b>) Animal survival of all groups; treated groups received radiation with Filter 1 (F1; 3.59 cGy/min). (<b>B</b>) A comparison of the experimental group (SE61O<sub>2</sub>/LND) with ultrasound triggering (+ US) and metformin pre-treatment (OM) that received radiation with Filter 1 (dark green plot) and animals treated with the same treatment but that received radiation with Filter 2 (F2; 1.36 cGy/min) (brown plot).</p>
Full article ">
14 pages, 1516 KiB  
Article
Controlled Release of Flurbiprofen from 3D-Printed and Supercritical Carbon Dioxide Processed Methacrylate-Based Polymer
by Truc T. Ngo and Jae D. Kim
Pharmaceutics 2023, 15(4), 1301; https://doi.org/10.3390/pharmaceutics15041301 - 21 Apr 2023
Viewed by 1525
Abstract
The ability to engineer and predict drug release behavior during treatment is critical to the design and implementation of effective drug delivery systems. In this study, a drug delivery system consisting of a methacrylate-based polymer and flurbiprofen was studied, and its release profile [...] Read more.
The ability to engineer and predict drug release behavior during treatment is critical to the design and implementation of effective drug delivery systems. In this study, a drug delivery system consisting of a methacrylate-based polymer and flurbiprofen was studied, and its release profile in a controlled phosphate-buffered saline solution was characterized. The polymer, which was 3D printed and processed in supercritical carbon dioxide under different temperature and pressure settings, showed sustained drug release over a prolonged period. A computer algorithm was used to determine the drug release time duration before reaching steady state and the maximum drug release at steady state. Several empirical models were applied to fit the release kinetic data to gain information about the drug release mechanism. The diffusion coefficients for each system were also estimated using Fick’s law. Based on the results, the influence of supercritical carbon dioxide processing conditions on the diffusion behavior is interpreted, providing insights into the effective and tunable design of drug delivery systems for targeted treatment specifications. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Example of algorithm execution outcome for steady state determination.</p>
Full article ">Figure 2
<p>Examples of diffusion coefficient estimations using Fick’s law and drug release data.</p>
Full article ">Figure 3
<p>Release time to steady state for samples treated under different scCO<sub>2</sub> conditions.</p>
Full article ">Figure 4
<p>Steady state drug release level for samples treated under different scCO<sub>2</sub> conditions.</p>
Full article ">
18 pages, 2526 KiB  
Review
Microfluidic Liver-on-a-Chip for Preclinical Drug Discovery
by Jingyu Fu, Hailong Qiu and Cherie S. Tan
Pharmaceutics 2023, 15(4), 1300; https://doi.org/10.3390/pharmaceutics15041300 - 21 Apr 2023
Cited by 4 | Viewed by 3347
Abstract
Drug discovery is an expensive, long, and complex process, usually with a high degree of uncertainty. In order to improve the efficiency of drug development, effective methods are demanded to screen lead molecules and eliminate toxic compounds in the preclinical pipeline. Drug metabolism [...] Read more.
Drug discovery is an expensive, long, and complex process, usually with a high degree of uncertainty. In order to improve the efficiency of drug development, effective methods are demanded to screen lead molecules and eliminate toxic compounds in the preclinical pipeline. Drug metabolism is crucial in determining the efficacy and potential side effects, mainly in the liver. Recently, the liver-on-a-chip (LoC) platform based on microfluidic technology has attracted widespread attention. LoC systems can be applied to predict drug metabolism and hepatotoxicity or to investigate PK/PD (pharmacokinetics/pharmacodynamics) performance when combined with other artificial organ-on-chips. This review discusses the liver physiological microenvironment simulated by LoC, especially the cell compositions and roles. We summarize the current methods of constructing LoC and the pharmacological and toxicological application of LoC in preclinical research. In conclusion, we also discussed the limitations of LoC in drug discovery and proposed a direction for improvement, which may provide an agenda for further research. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Cell distribution structure in a liver lobule. Double blood supply of hepatic arteriole and portal vein enters the liver’s portal triad region and the central vein. (<b>B</b>) Three-dimensional liver tissue diagram in LoC. The fluid flows through the pores on the scaffold, providing the biochemical environment and mechanical stimulation. (<b>C</b>) Microstructure of liver. The liver lobule is hexagonal, with a 1 mm diameter and a 2 mm thickness, functionally divided into three regions. (<b>D</b>) Example of zonal heterogeneity of hepatotoxicity. The yellow arrow shows the flow direction. (<b>A</b>,<b>B</b>) reproduced with permission of [<a href="#B34-pharmaceutics-15-01300" class="html-bibr">34</a>], Copyright © 2017. Published by Elsevier. (<b>C</b>,<b>D</b>) reproduced with permission of [<a href="#B29-pharmaceutics-15-01300" class="html-bibr">29</a>], Copyright © 2019. Published by MDPI.</p>
Full article ">Figure 2
<p>Different microfluidic models are used to construct liver-on-a-chip. (<b>A</b>) A 24-well PDMS mold with through holes at the bottom. Hepatocytes (labeled green) selectively adhered to the collagen structure, clustered together, and then surrounded by mouse 3T3-J2 fibroblasts (labeled orange). (<b>B</b>) Microfluidic endothelial-like barrier. The barrier consists of parallel channels connecting the cell culture zone with the external flow channel. (<b>C</b>) Simulation of liver lobules in Christmas tree structure, with gradient generator. (<b>D</b>) Spheroid culture. Cultured spheroids in an ultra-low-attachment plate, transferred them to a 96-well plate or liver chip and compared their performance. (<b>A</b>) reproduced with permission of [<a href="#B11-pharmaceutics-15-01300" class="html-bibr">11</a>], Copyright © 2008. Published by Nature Publishing Group. (<b>B</b>) reproduced with permission of [<a href="#B78-pharmaceutics-15-01300" class="html-bibr">78</a>], Copyright © 2007. Published by Wiley Periodicals, Inc. (<b>C</b>) reproduced with permission of [<a href="#B79-pharmaceutics-15-01300" class="html-bibr">79</a>], Copyright © 2018. Published by Nature Publishing Group. (<b>D</b>) reproduced with permission of [<a href="#B80-pharmaceutics-15-01300" class="html-bibr">80</a>], Copyright © 2022 Published by Elsevier.</p>
Full article ">Figure 3
<p>Example of 3D perfusion platforms. (<b>A</b>) Twelve fluid-isolated bioreactors with connected cell culture plate (yellow) and pneumatic plate (gray). (<b>B</b>) Cross section diagram of the bioreactor, the membrane separates the upper cell culture plate from the lower pneumatic plate, and the micropump provides power for medium circulation in the channel. (<b>C</b>) OrganoPlate LiverTox model. The iHeps differentiated in 2D culture are harvested and seeded in OrganoPlate 2-lane through a liquid processor. Perfusion and organ channels were separated by phase guidance (PhG). (<b>A</b>,<b>B</b>) reproduced with permission of [<a href="#B16-pharmaceutics-15-01300" class="html-bibr">16</a>], Copyright © 2016. Published by The American Society for Pharmacology and Experimental Therapeutics. (<b>C</b>) reproduced with permission of [<a href="#B20-pharmaceutics-15-01300" class="html-bibr">20</a>], Copyright © 2021. Published by Elsevier.</p>
Full article ">Figure 4
<p>Example of multi-organ chips platforms. (<b>A</b>) Multi-organ chips based on liver, intestinal, vascular, and kidney chips. Caco-2, HUVEC, HepG2, and HK-2 occupy one floor from top to bottom (<b>B</b>) Taking a two-layer chip assembly as an example, the system was built in the order of inoculating cells to a porous membrane, providing a cell culture medium and assembling chips. The peristaltic pump provides power for fluid. (<b>C</b>) The liver and lung cancer parts on chips are connected through a microchannel. Channel is also equipped with a stirrer-based micropump to provide power. The value in the arrow is the flow ratio. (<b>D</b>) Concept diagram of PK model based on the multi-organ chip. (<b>A</b>,<b>B</b>), reproduced with permission of [<a href="#B23-pharmaceutics-15-01300" class="html-bibr">23</a>], Copyright © 2020. Published by Elsevier. (<b>C</b>,<b>D</b>) reproduced with permission of [<a href="#B91-pharmaceutics-15-01300" class="html-bibr">91</a>], Copyright © 2020. Published by AIP Publishing.</p>
Full article ">
16 pages, 3575 KiB  
Article
mTOR Inhibition Impairs the Activation and Function of Belatacept-Resistant CD4+CD57+ T Cells In Vivo and In Vitro
by Florence Herr, Manon Dekeyser, Jerome Le Pavec, Christophe Desterke, Andrada-Silvana Chiron, Karen Bargiel, Olaf Mercier, Amelia Vernochet, Elie Fadel and Antoine Durrbach
Pharmaceutics 2023, 15(4), 1299; https://doi.org/10.3390/pharmaceutics15041299 - 20 Apr 2023
Cited by 1 | Viewed by 1321
Abstract
Calcineurin inhibitors have improved graft survival in solid-organ transplantation but their use is limited by toxicity, requiring a switch to another immunosuppressor in some cases. Belatacept is one option that has been shown to improve graft and patient survival despite being associated with [...] Read more.
Calcineurin inhibitors have improved graft survival in solid-organ transplantation but their use is limited by toxicity, requiring a switch to another immunosuppressor in some cases. Belatacept is one option that has been shown to improve graft and patient survival despite being associated with a higher risk of acute cellular rejection. This risk of acute cellular rejection is correlated with the presence of belatacept-resistant T cells. We performed a transcriptomic analysis of in vitro-activated cells to identify pathways affected by belatacept in belatacept-sensitive cells (CD4+CD57) but not in belatacept-resistant CD4+CD57+ T cells. mTOR was significantly downregulated in belatacept-sensitive but not belatacept-resistant T cells. The inhibition of mTOR strongly decreases the activation and cytotoxicity of CD4+CD57+ cells. In humans, the use of a combination of mTOR inhibitor and belatacept prevents graft rejection and decreases the expression of activation markers on CD4 and CD8 T cells. mTOR inhibition decreases the functioning of belatacept-resistant CD4+CD57+ T cells in vitro and in vivo. It could potentially be used in association with belatacept to prevent acute cellular rejection in cases of calcineurin intolerance. Full article
Show Figures

Figure 1

Figure 1
<p>CD4<sup>+</sup> T-cell phenotypes, proliferation and activation. (<b>A</b>) The CD57 and PD1 expression phenotypes of T cells collected from renal transplant recipients before transplantation were assessed by flow cytometry. Naïve (CCR7<sup>+</sup>CD45RO<sup>−</sup>), central memory (CM; CCR7<sup>+</sup>CD45RO<sup>+</sup>), effector memory re-expressing CD45RA (EMRA; CCR7<sup>−</sup>CD45RO<sup>−</sup>), and effector memory cells (EM; CCR7<sup>−</sup>CD45RO<sup>+</sup>) were characterized according to their expression of CD45R0 and CCR7. Mean (bar chart) and SEM of six independent experiments. (<b>B</b>) Proliferation of CD4<sup>+</sup> T-cell populations (VPD450 dilution) from healthy donor T cells assessed by flow cytometry after 5 days of MLR in the presence and absence of belatacept. Data for six (CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>+</sup>) and eight (CD4<sup>+</sup>CD57<sup>−</sup> and CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>−</sup>) independent experiments (pictograms) are shown, with the mean (bar chart) and SEM. (<b>C</b>) The expression of CD38 and CD25, as markers of activation, in the various CD4<sup>+</sup> T-cell populations from healthy donors were assessed, after 5 days of MLR, by flow cytometry in six independent experiments (pictograms). The mean (bar chart) and SEM are shown.</p>
Full article ">Figure 2
<p>Transcriptomic analysis of the effect of belatacept on CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>−</sup> and CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>+</sup> allogeneic T cells after 6 h of culture with activated dendritic cells. (<b>A</b>) Heatmap of normalized enrichment score (NES) of untreated versus belatacept-treated CD4 subpopulations analyzed by gene-set enrichment analysis (GSEA). (<b>B</b>) Network representation of gene sets MYC target V1, mTORC1 signaling and IFN alpha response profiles of untreated CD4<sup>+</sup>CD57<sup>−</sup> versus belatacept-treated. (<b>C</b>) mTORC1 signaling enrichment plot for CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>+</sup> and CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>−</sup>cells activated in the presence and absence of belatacept. Normalized enrichment score (NES), false discovery rate (FDR-<span class="html-italic">q</span> value) and family-wise error rate (FWER <span class="html-italic">p</span>-value) of the hallmark mTORC1 signaling gene set for the various CD4<sup>+</sup> populations, in the presence and absence of belatacept. Belatacept significantly impaired the transcription of mTORC1 pathway genes only in CD4<sup>+</sup>CD57<sup>−</sup> cells. (<b>D</b>) Heatmap of the first 30 genes of the leading-edge subset of the mTORC1 signaling enrichment plot ranked for CD4<sup>+</sup>CD57<sup>−</sup> cells untreated versus belatacept-treated. The leading-edge subset is the core of the gene set accounting for the enrichment signal. Enrichment score at the position in the ranked list of genes (RUNNING ES) of CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>+</sup> and CD4<sup>+</sup>CD57<sup>+</sup>PD1<sup>−</sup> have been ordered as a CD4<sup>+</sup>CD57<sup>−</sup> list and plotted on the heat map allowing comparisons.</p>
Full article ">Figure 3
<p>mTOR expression and activation of mTORC1 pathway in CD57<sup>+</sup> and CD57<sup>−</sup> T cells. (<b>A</b>) mTOR expression by CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>, CD8<sup>+</sup>CD57<sup>−</sup> and CD8<sup>+</sup>CD57<sup>+</sup> T cells was analyzed by flow cytometry. Left panel, mean of fluorescence intensity (MFI) of cells of six healthy donors, the mean (bar chart) and SEM are shown. Right panel, representative flow cytometry profile. (<b>B</b>) Phosphorylation of S6 in CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>, CD8<sup>+</sup>CD57<sup>−</sup> and CD8<sup>+</sup>CD57<sup>+</sup> T cells were analyzed with and without anti-CD3 activation by flow cytometry. Left panel, percentage of positive cells of six healthy donors, the mean (bar chart) and SEM are shown. Right panel, representative flow cytometry profile. (<b>C</b>) Percentage of S6-phosphorylation induced by anti-CD3 activation calculated for each experiment was depicted.</p>
Full article ">Figure 4
<p>Activation and proliferation of CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>, CD8<sup>+</sup>CD57<sup>−</sup> and CD8<sup>+</sup>CD57<sup>+</sup> T cells in the presence of mTOR inhibitor: (<b>A</b>) Proliferation and (<b>B</b>) CD38 expression of CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>, CD8<sup>+</sup>CD57<sup>−</sup> and CD8<sup>+</sup>CD57<sup>+</sup> T cells after 5 days of culture with allogeneic aDCs without (DMSO) or with everolimus, or everolimus + belatacept. Eight (CD57−) or ten (CD57+) independent experiments (pictograms) were performed, and the mean (bar chart) and SEM are shown.</p>
Full article ">Figure 5
<p>Granzyme B expression and cytotoxicity in CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>, CD8<sup>+</sup>CD57<sup>−</sup> and CD8<sup>+</sup>CD57<sup>+</sup> T cells treated with mTOR inhibitor: (<b>A</b>) Expression of granzyme B and (<b>B</b>) CD107a by CD4<sup>+</sup>CD57<sup>−</sup>, CD4<sup>+</sup>CD57<sup>+</sup>, CD8<sup>+</sup>CD57<sup>−</sup> and CD8<sup>+</sup>CD57<sup>+</sup> T cells after 5 days of culture with allogeneic aDCs without (DMSO) or with everolimus, followed by incubation with target cells for 4 h. Nine (CD57<sup>−</sup>) and 14 (CD57<sup>+</sup>) (<b>A</b>) or 9 (CD57<sup>−</sup>) and 11 (CD57+) (<b>B</b>) independent experiments (pictograms) were performed. The mean (bar chart) and SEM are shown.</p>
Full article ">Figure 6
<p>CD38 expression on CD4<sup>+</sup> or CD8<sup>+</sup> T cells from patients treated with belatacept, before (T0) or 3 months after (T3m) the introduction of everolimus (<span class="html-italic">n</span> = 3 patients).</p>
Full article ">
12 pages, 2783 KiB  
Article
Imageable AuNP-ECM Hydrogel Tissue Implants for Regenerative Medicine
by Malka Shilo, Ester-Sapir Baruch, Lior Wertheim, Hadas Oved, Assaf Shapira and Tal Dvir
Pharmaceutics 2023, 15(4), 1298; https://doi.org/10.3390/pharmaceutics15041298 - 20 Apr 2023
Cited by 1 | Viewed by 1841
Abstract
In myocardial infarction, a blockage in one of the coronary arteries leads to ischemic conditions in the left ventricle of the myocardium and, therefore, to significant death of contractile cardiac cells. This process leads to the formation of scar tissue, which reduces heart [...] Read more.
In myocardial infarction, a blockage in one of the coronary arteries leads to ischemic conditions in the left ventricle of the myocardium and, therefore, to significant death of contractile cardiac cells. This process leads to the formation of scar tissue, which reduces heart functionality. Cardiac tissue engineering is an interdisciplinary technology that treats the injured myocardium and improves its functionality. However, in many cases, mainly when employing injectable hydrogels, the treatment may be partial because it does not fully cover the diseased area and, therefore, may not be effective and even cause conduction disorders. Here, we report a hybrid nanocomposite material composed of gold nanoparticles and an extracellular matrix-based hydrogel. Such a hybrid hydrogel could support cardiac cell growth and promote cardiac tissue assembly. After injection of the hybrid material into the diseased area of the heart, it could be efficiently imaged by magnetic resonance imaging (MRI). Furthermore, as the scar tissue could also be detected by MRI, a distinction between the diseased area and the treatment could be made, providing information about the ability of the hydrogel to cover the scar. We envision that such a nanocomposite hydrogel may improve the accuracy of tissue engineering treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the technology. AuNPs are conjugated with Gd and encapsulated within an ECM-based hydrogel. The hydrogel is injected into the left ventricle of mice post ischemia–reperfusion injury, allowing to determine the location of the treatment in relation to scar tissue in the heart.</p>
Full article ">Figure 2
<p>Gd-AuNP characterization. (<b>a</b>) TEM image of the pristine AuNPs; scale bar = 200 nm. (<b>b</b>) Size distribution of the AuNPs. (<b>c</b>) Absorbance spectrum of the AuNPs. (<b>d</b>) Scheme of the Gd-AuNPs. (<b>e</b>) FTIR analysis of the different steps in Gd-AuNP synthesis: pristine AuNPs in orange, DTPA complex in green, GdL complex in pink, and Gd-AuNPs in purple. (<b>f</b>) Diameter of the conjugated AuNPs as indicated by DLS. (<b>g</b>) Charge of the conjugated AuNPs as indicated by zeta potential measurements. (<b>h</b>–<b>j</b>) TEM images of (<b>h</b>) pristine AuNPs and (<b>i</b>,<b>j</b>) Gd-AuNPs after negative staining; scale bars = 100, 50, and 25 nm, respectively.</p>
Full article ">Figure 3
<p>Gd-AuNP nanocomposite hydrogel characterization. (<b>a</b>) An image of the nanocomposite hydrogel after gelation at 37 °C. (<b>b</b>,<b>c</b>) HRSEM images of the nanocomposite hydrogel. The NPs appear in white. Scale bars (<b>b</b>) = 1 μm, (<b>c</b>) = 300 nm. (<b>d</b>) EDX analysis of the composite hydrogel. (<b>e</b>,<b>f</b>) Rheological properties of the composite hydrogel over time (<b>e</b>) and 15 min post gelation in 37 °C (<b>f</b>).</p>
Full article ">Figure 4
<p>Engineered cardiac implants within the nanocomposite hydrogel. (<b>a</b>,<b>b</b>) Viability test within the nanocomposite (<b>a</b>) and pristine hydrogels (<b>b</b>), 1 week post encapsulation; scale bar = 100 μm. (<b>c</b>,<b>d</b>) Cardiac sarcomeric actinin immunostaining of the nanocomposite implants on day 7 (<b>c</b>) and day 14 (<b>d</b>). Actinin in pink, nuclei in blue; scale bar = 25 μm. (<b>e</b>) Contraction rate of the nanocomposite implant before and after the addition of 1 µM noradrenaline.</p>
Full article ">Figure 5
<p>Detection of the nanocomposite hydrogel by MRI. (<b>a</b>) MRI imaging of AuNP-Gd, AuNP, and water solutions. (<b>b</b>) MRI imaging of droplets of the nanocomposite hydrogel, pristine hydrogel, and water. (<b>c</b>–<b>f</b>) Images of the heart slice (<b>c</b>,<b>e</b>) and ex vivo MRI imaging (<b>d</b>,<b>f</b>) of the treated (<b>c</b>,<b>d</b>) and untreated (<b>e</b>,<b>f</b>) hearts, 6 weeks post IRI surgery; scale bar = 1 mm.</p>
Full article ">Figure 6
<p>MRI imaging of the heart. (<b>a</b>–<b>d</b>) MRI images for (<b>a</b>) untreated, (<b>b</b>) pristine hydrogel-, (<b>c</b>) AuNP composite hydrogel-, and (<b>d</b>) Gd-AuNP composite hydrogel-treated mice, 6 weeks post IRI. The white arrow in (<b>d</b>) indicates the location of the Gd-AuNP composite hydrogel (black area), which was not observed in all other treatments. (<b>e</b>,<b>f</b>) MRI analysis monitoring the location of Gd-AuNP composite hydrogel (dashed area) over time. (<b>e</b>) SAX (short axis slices) and (<b>f</b>) LAX (long axis slices) at (<b>I</b>) 1 week, (<b>II</b>) 4 weeks, and (<b>III</b>) 6 weeks post IRI; scale bar = 1 mm.</p>
Full article ">Figure 7
<p>Imaging the coverage area of the treatment in relation to scar area. (<b>a</b>–<b>d</b>) Segmentation process for detection of the Gd-AuNP nanocomposite hydrogel (yellow). (<b>a</b>) LV wall in MRI imaging. (<b>b</b>) K-means algorithm, k = 4. (<b>c</b>) The location of the Gd-AuNPs composite hydrogel as detected by the algorithm. (<b>d</b>) Final MRI image with the location of the hydrogel. (<b>e</b>–<b>h</b>) Subtraction process for detection of the scar tissue (red). (<b>e</b>) LV wall post systemic injection of Gd. (<b>f</b>) LV wall before systemic injection of Gd. (<b>g</b>) Subtraction MRI image. (<b>h</b>) Final MRI image with the location of the scar tissue after noise filtering. (<b>i</b>) Merged image of (<b>d</b>,<b>h</b>). The nanocomposite hydrogel (yellow) with respect to the scar tissue (red), 45 min post systemic injection of Gd-DTPA solution; scale bars = 1 mm.</p>
Full article ">
17 pages, 9050 KiB  
Article
Proliferative and Osteogenic Supportive Effect of VEGF-Loaded Collagen-Chitosan Hydrogel System in Bone Marrow Derived Mesenchymal Stem Cells
by Jeevithan Elango
Pharmaceutics 2023, 15(4), 1297; https://doi.org/10.3390/pharmaceutics15041297 - 20 Apr 2023
Cited by 7 | Viewed by 2051
Abstract
The use of hydrogel (HG) in regenerative medicine is an emerging field and thus several approaches have been proposed recently to find an appropriate hydrogel system. In this sense, this study developed a novel HG system using collagen, chitosan, and VEGF composites for [...] Read more.
The use of hydrogel (HG) in regenerative medicine is an emerging field and thus several approaches have been proposed recently to find an appropriate hydrogel system. In this sense, this study developed a novel HG system using collagen, chitosan, and VEGF composites for culturing mesenchymal stem cells (MSCs), and investigated their ability for osteogenic differentiation and mineral deposition. Our results showed that the HG loaded with 100 ng/mL VEGF (HG-100) significantly supported the proliferation of undifferentiated MSCs, the fibrillary filament structure (HE stain), mineralization (alizarin red S and von Kossa stain), alkaline phosphatase, and the osteogenesis of differentiated MSCs compared to other hydrogels (loaded with 25 and 50 ng/mL VEGF) and control (without hydrogel). HG-100 showed a higher VEGF releasing rate from day 3 to day 7 than other HGs, which substantially supports the proliferative and osteogenic properties of HG-100. However, the HGs did not increase the cell growth in differentiated MSCs on days 14 and 21 due to the confluence state (reach stationary phase) and cell loading ability, regardless of the VEGF content. Similarly, the HGs alone did not stimulate the osteogenesis of MSCs; however, they increased the osteogenic ability of MSCs in presence of osteogenic supplements. Accordingly, a fabricated HG with VEGF could be used as an appropriate system to culture stem cells for bone and dental regeneration. Full article
(This article belongs to the Special Issue Hydrogels in Drug Delivery: Progress and Challenges)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of collagen–chitosan hydrogel fabrication with different contents of VEGF.</p>
Full article ">Figure 2
<p>The release pattern of VEGF from hydrogels at different incubation times. (<b>A</b>) Actual release in ng/mL and (<b>B</b>) cumulative release in percentage. HG-25, HG-50, and HG-100—hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively.</p>
Full article ">Figure 3
<p>Proliferative (<b>A</b>), cytotoxic (<b>B</b>), and cell-seeding ability (<b>C</b>,<b>D</b>) of VEGF-loaded collagen–chitosan hydrogels. Control: cells without hydrogels, HG: hydrogels without VEGF, HG−25, HG−50, and HG−100: hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively. * and different alphabets denote statistical significance, * vs. day 1 and different alphabets vs. control group, respectively, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Morphological analysis of MSCs cultured on hydrogels by HE staining. Control—cells without hydrogels, HG—hydrogels without VEGF, HG-25, -50, and -100—hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively. Scale bar 10×—100 μm. The asterisk indicates cellular aggregation and matrix deposition.</p>
Full article ">Figure 5
<p>The cell growth of osteogenic cells differentiated from MSCs. Control—cells without hydrogels, HG—hydrogels without VEGF, HG-25, -50, and -100—hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively.</p>
Full article ">Figure 6
<p>Von Kossa staining of osteogenic cells differentiated from MSC cultured on hydrogels for 14 and 21 days. Cells without osteogenic supplement (<b>Left</b>) and cells with osteogenic supplement (<b>Right</b>). (<b>A</b>) Cells cultured on hydrogels and stained, (<b>B</b>) only hydrogels with von Kossa stain without cells and supplement. NC—cells without hydrogels, HG—hydrogels without VEGF, HG-25, -50, and -100—hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively. Scale bar 10×—100 μm.</p>
Full article ">Figure 7
<p>Alizarin red-S staining of osteogenic cells differentiated from MSC cultured on hydrogels for 14 and 21 days. Cells without osteogenic supplement (<b>Left</b>) and cells with osteogenic supplement (<b>Right</b>). (<b>A</b>) Cells cultured on hydrogels and stained, (<b>B</b>) only hydrogels with alizarin red S stain without cells and supplement. NC—cells without hydrogels, HG—hydrogels without VEGF, HG-25, -50, and -100—hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively. Scale bar 10×—100 μm.</p>
Full article ">Figure 8
<p>Quantification of the stained area of differentiated bone marrow mesenchymal stem cells cultured on hydrogels. The percentage of stained area in osteogenic cells was quantified using ImageJ software (Version 1.52n). Control—cells without hydrogel, HG—hydrogels without VEGF, HG-25, HG-50, and HG-100—hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively. ALP-alkaline phosphatase. * <span class="html-italic">p</span> &lt; 0.05 vs. control, ** <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">Figure 9
<p>Alkaline phosphatase staining of osteogenic cells differentiated from MSC cultured on hydrogels for 14 and 21 days. Cells without osteogenic supplement (<b>Left</b>) and cells with osteogenic supplement (<b>Right</b>). (<b>A</b>) Cells cultured on hydrogels and stained, (<b>B</b>) only hydrogels with ALP stain without cells and supplement. NC—cells without hydrogels, HG—hydrogels without VEGF, HG-25, -50, and -100—hydrogels with 25 ng/mL, 50 ng/mL, and 100 ng/mL of VEGF, respectively. Scale bar 10×—100 μm.</p>
Full article ">
17 pages, 4364 KiB  
Article
Formulation and Characterization of Electrospun Nanofibers for Melatonin Ocular Delivery
by Alessia Romeo, Adrienn Kazsoki, Safaa Omer, Balázs Pinke, László Mészáros, Teresa Musumeci and Romána Zelkó
Pharmaceutics 2023, 15(4), 1296; https://doi.org/10.3390/pharmaceutics15041296 - 20 Apr 2023
Cited by 4 | Viewed by 2463
Abstract
The poor ocular bioavailability of melatonin (MEL) limits the therapeutic action the molecule could exert in the treatment of ocular diseases. To date, no study has explored the use of nanofiber-based inserts to prolong ocular surface contact time and improve MEL delivery. Here, [...] Read more.
The poor ocular bioavailability of melatonin (MEL) limits the therapeutic action the molecule could exert in the treatment of ocular diseases. To date, no study has explored the use of nanofiber-based inserts to prolong ocular surface contact time and improve MEL delivery. Here, the electrospinning technique was proposed to prepare poly (vinyl alcohol) (PVA) and poly (lactic acid) (PLA) nanofiber inserts. Both nanofibers were produced with different concentrations of MEL and with or without the addition of Tween® 80. Nanofibers morphology was evaluated by scanning electron microscopy. Thermal and spectroscopic analyses were performed to characterize the state of MEL in the scaffolds. MEL release profiles were observed under simulated physiological conditions (pH 7.4, 37 °C). The swelling behavior was evaluated by a gravimetric method. The results confirmed that submicron-sized nanofibrous structures were obtained with MEL in the amorphous state. Different MEL release rates were achieved depending on the nature of the polymer. Fast (20 min) and complete release was observed for the PVA-based samples, unlike the PLA polymer, which provided slow and controlled MEL release. The addition of Tween® 80 affected the swelling properties of the fibrous structures. Overall, the results suggest that membranes could be an attractive vehicle as a potential alternative to liquid formulations for ocular administration of MEL. Full article
(This article belongs to the Special Issue Recent Development of Electrospinning for Drug Delivery, 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the production of melatonin-loaded PLA nanofibers.</p>
Full article ">Figure 2
<p>Schematic illustration of the production of melatonin-loaded PVA nanofibers.</p>
Full article ">Figure 3
<p>SEM images (magnification: 500× and 3500×) and diameter distribution of Melatonin-loaded PLA nanofibers with different concentration of drug 0.1% <span class="html-italic">w</span>/<span class="html-italic">w</span> (<b>A</b>), 0.3% <span class="html-italic">w</span>/<span class="html-italic">w</span> (<b>B</b>), 0.5% <span class="html-italic">w</span>/<span class="html-italic">w</span> (<b>C</b>), and Melatonin-loaded PLA nanofibers with Tween<sup>®</sup> 80 and different concentrations of drug 0.1% <span class="html-italic">w</span>/<span class="html-italic">w</span> (<b>D</b>), 0.3% <span class="html-italic">w</span>/<span class="html-italic">w</span> (<b>E</b>), 0.5% <span class="html-italic">w</span>/<span class="html-italic">w</span> (<b>F</b>).</p>
Full article ">Figure 4
<p>SEM images (magnification: 500× and 3500×) and diameter distribution of Melatonin-loaded PVA nanofibers with different concentration of drug 0.1% <span class="html-italic">w/w</span> (<b>A</b>), 0.3% <span class="html-italic">w/w</span> (<b>B</b>), 0.5% <span class="html-italic">w/w</span> (<b>C</b>), and Melatonin-loaded PVA nanofibers with Tween<sup>®</sup> 80 and different concentrations of drug 0.1% <span class="html-italic">w/w</span> (<b>D</b>), 0.3% <span class="html-italic">w/w</span> (<b>E</b>), 0.5% <span class="html-italic">w/w</span> (<b>F</b>).</p>
Full article ">Figure 5
<p>DSC curves of (<b>A</b>) Melatonin (a), PLA polymer (b), physical mixture MEL + PLA polymer (c), empty-PLA nanofibers (d), Melatonin-loaded PLA nanofibers with different concentration of drug 0.1% <span class="html-italic">w/w</span> (e), 0.3% <span class="html-italic">w/w</span> (f), 0.5% <span class="html-italic">w/w</span> (g), and Melatonin-loaded PLA nanofibers with Tween<sup>®</sup> 80 and different concentration of drug 0.1% <span class="html-italic">w/w</span> (h), 0.3% <span class="html-italic">w/w</span> (i), 0.5% <span class="html-italic">w/w</span> (l); and (<b>B</b>) Melatonin (a), PVA polymer (b), physical mixture MEL+PVA polymer (c), empty-PVA nanofibers (d), Melatonin-loaded PVA nanofibers with different concentration of drug 0.1% <span class="html-italic">w/w</span> (e), 0.3% <span class="html-italic">w/w</span> (f), 0.5% <span class="html-italic">w/w</span> (g), and Melatonin-loaded PVA nanofibers with Tween<sup>®</sup> 80 and different concentration of drug 0.1% <span class="html-italic">w/w</span> (h), 0.3% <span class="html-italic">w/w</span> (i), 0.5% <span class="html-italic">w/w</span> (l).</p>
Full article ">Figure 6
<p>FT-IR curves of (<b>A</b>) Melatonin (a), PLA polymer (b), Tween<sup>®</sup> 80 (c), physical mixture MEL+PLA polymer (d), empty-PLA nanofibers (e), Melatonin-loaded PLA nanofibers with different concentration of drug 0.1% <span class="html-italic">w/w</span> (f), 0.3% <span class="html-italic">w/w</span> (g), 0.5% <span class="html-italic">w/w</span> (h), and Melatonin-loaded PLA nanofibers with Tween<sup>®</sup> 80 and different concentration of drug 0.1% <span class="html-italic">w/w</span> (i), 0.3% <span class="html-italic">w/w</span> (l), 0.5% <span class="html-italic">w/w</span> (m); and (<b>B</b>) Melatonin (a), PVA polymer (b), Tween<sup>®</sup> 80 (c), physical mixture MEL+PVA polymer (d), empty-PVA nanofibers (e), Melatonin-loaded PVA nanofibers with different concentration of drug 0.1% <span class="html-italic">w/w</span> (f), 0.3% <span class="html-italic">w/w</span> (g), 0.5% <span class="html-italic">w/w</span> (h), and Melatonin-loaded PVA nanofibers with Tween<sup>®</sup> 80 and different concentration of drug 0.1% <span class="html-italic">w/w</span> (i), 0.3% <span class="html-italic">w/w</span> (l), 0.5% <span class="html-italic">w/w</span> (m).</p>
Full article ">Figure 7
<p>In vitro release profiles of MEL-loaded PLA nanofibers Melatonin-loaded PLA nanofibers with different concentration of drug 0.1% <span class="html-italic">w/w</span> (LM1), 0.3% <span class="html-italic">w/w</span> (LM2), 0.5% <span class="html-italic">w/w</span> (LM3), and Melatonin-loaded PLA nanofibers with Tween<sup>®</sup> 80 and different concentration of drug 0.1% <span class="html-italic">w/w</span> (LMT1), 0.3% <span class="html-italic">w/w</span> (LMT2), 0.5% <span class="html-italic">w/w</span> (LMT3) in phosphate buffered solution (pH 7.4) at 37 °C. Each point represents the mean value of three different experiments ±S.D.</p>
Full article ">Figure 8
<p>In vitro release profiles of MEL-loaded PVA nanofibers Melatonin-loaded PVA nanofibers with different concentration of drug 0.1% <span class="html-italic">w/w</span> (PM1), 0.3% <span class="html-italic">w/w</span> (PM2), 0.5% <span class="html-italic">w/w</span> (PM3), and Melatonin-loaded PVA nanofibers with Tween<sup>®</sup> 80 and different concentration of drug 0.1% <span class="html-italic">w/w</span> (PMT1), 0.3% <span class="html-italic">w/w</span> (PMT2), 0.5% <span class="html-italic">w/w</span> (PMT3) in phosphate buffered solution (pH 7.4) at 37 °C. Each point represents the mean value of three different experiments ± S.D.</p>
Full article ">Figure 9
<p>Images of the PVA (top) and PLA-based nanofibres (bottom) in the dry state, after addition of the medium (time 0), after 30 min and at the end of the experiment (24 h) at the two pH conditions tested (5.5 and 7.4).</p>
Full article ">Figure 10
<p>Swelling behavior of electrospun PLA-based nanofibers at pH 7.4 and 5.5.</p>
Full article ">
13 pages, 2720 KiB  
Article
Adoptive Transfer of Photosensitizer-Loaded Cytotoxic T Cells for Combinational Photodynamic Therapy and Cancer Immuno-Therapy
by André-René Blaudszun, Woo Jun Kim, Wooram Um, Hong Yeol Yoon, Man Kyu Shim and Kwangmeyung Kim
Pharmaceutics 2023, 15(4), 1295; https://doi.org/10.3390/pharmaceutics15041295 - 20 Apr 2023
Cited by 3 | Viewed by 2459
Abstract
Adoptive cell transfer (ACT) has shown remarkable therapeutic efficacy against blood cancers such as leukemia and lymphomas, but its effect is still limited due to the lack of well-defined antigens expressed by aberrant cells within tumors, the insufficient trafficking of administered T cells [...] Read more.
Adoptive cell transfer (ACT) has shown remarkable therapeutic efficacy against blood cancers such as leukemia and lymphomas, but its effect is still limited due to the lack of well-defined antigens expressed by aberrant cells within tumors, the insufficient trafficking of administered T cells to the tumor sites, as well as immunosuppression induced by the tumor microenvironment (TME). In this study, we propose the adoptive transfer of photosensitizer (PS)-loaded cytotoxic T cells for a combinational photodynamic and cancer immunotherapy. Temoporfin (Foscan®), a clinically applicable porphyrin derivative, was loaded into OT-1 cells (PS-OT-1 cells). The PS-OT-1 cells efficiently produced a large amount of reactive oxygen species (ROS) under visible light irradiation in a culture; importantly, the combinational photodynamic therapy (PDT) and ACT with PS-OT-1 cells induced significant cytotoxicity compared to ACT alone with unloaded OT-1 cells. In murine lymphoma models, intravenously injected PS-OT-1 cells significantly inhibited tumor growth compared to unloaded OT-1 cells when the tumor tissues were locally irradiated with visible light. Collectively, this study suggests that combinational PDT and ACT mediated by PS-OT-1 cells provides a new approach for effective cancer immunotherapy. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

Figure 1
<p>Adoptive transfer of photosensitizer (PS)-loaded cytotoxic T cells for combinational photodynamic therapy (PDT) and cancer immunotherapy. (<b>a</b>) Photosensitizer (PS) is loaded into cytotoxic T cells (OT-1 cells), resulting in PS-OT-1 cells. (<b>b</b>) When PS-OT-1 cells are intravenously injected into the tail vein, they accumulate within the targeted tumor tissues owing to T cells’ tumor homing effect. (<b>c</b>) Upon light irradiation, PS-OT-1 cells can effectively eradicate the tumor cells that are directly attacked by T cells, as well as neighboring non-target cancer cells that are not immediately recognized by T cells in the tumor tissues by combinational effect of PDT and ACT.</p>
Full article ">Figure 2
<p>Preparation of temoporfin-loaded OT-1 cells (PS-OT-1 cells). (<b>a</b>) Fluorescence images of OT-1 cells after incubation with different concentrations of temoporfin from 12.5–100 μg/mL. (<b>b</b>) Cell sediment of PS-OT-1 cells or unloaded OT-1 cells that are incubated with 25 μg/mL for 2 h or unloaded OT-1 cells. (<b>c</b>) Flow cytometric analysis of OT-1 cells after incubation with different concentrations of temoporfin. (<b>d</b>) Cell viability of OT-1 cells after incubation with temoporfin. (<b>e</b>) Flow cytometric analysis of OT-1 cells after incubation with temoporfin at 4 or 37 °C. (<b>f</b>) GMFI of OT-1 cells after incubation with temoporfin at 4 or 37 °C. (<b>g</b>) Fluorescence images of PS-OT-1 cells or unloaded OT-1 cells. The asterisk in figures indicate a statistical significance with <span class="html-italic">p</span> value of &lt;0.001 ***.</p>
Full article ">Figure 3
<p>In vitro combinational antitumor effect of PDT and ACT by PS-OT-1 cells. (<b>a</b>) Fluorescence images of PS-OT-1 cells after 0, 1 or 2 days of incubation. (<b>b</b>) Flow cytometric analysis of PS-OT-1 cells after 0, 1 or 2 days of incubation. (<b>c</b>) Extracellular ROS generation of PS-OT-1 cells that were incubated in cell culture media at 37 °C for 2 days. After 6 h of incubation of PS-OT-1 cells at day 1, 2, each cell media was isolated and the extracellular ROS was measured using DPBF bleaching assays. (<b>d</b>) Cell viability of EG.7-OVA after incubation with PS-OT-1 cells or unloaded OT-1 cells in presence or absence of the light irradiation. (<b>e</b>) Cell viability of EG.7-OVA after incubation with PS-OT-1 cells or unloaded OT-1 cells after 8 or 24 h of incubation. (<b>f</b>) Cell viability of EL-4 cells after incubation with PS-OT-1 cells or unloaded OT-1 cells. The asterisks in figures indicate a statistical significance with <span class="html-italic">p</span> value of &lt;0.01 ** and &lt;0.001 ***. N.S. in figures indicate a statistically not significant.</p>
Full article ">Figure 4
<p>Biodistribution of PS-OT-1 cells. (<b>a</b>) In vivo fluorescence images of a tumor treated with PS-OT-1 cells. (<b>b</b>) Quantitative analysis of fluorescence intensity in the tumor region.</p>
Full article ">Figure 5
<p>Antitumor efficacy of PS-OT-1 cells. (<b>a</b>) Tumor growth of mice treated with saline, unloaded OT-1 cells or PS-OT-1 cells. (<b>b</b>) Optical images of mice treated with saline, unloaded OT-1 cells or PS-OT-1 cells. (<b>c</b>) Tumor tissues stained with TUNEL of mice treated with saline, unloaded OT-1 cells or PS-OT-1 cells. (<b>d</b>) Normal organs stained with H&amp;E. The asterisk in figures indicate a statistical significance with <span class="html-italic">p</span> value of &lt;0.001 ***.</p>
Full article ">
26 pages, 11693 KiB  
Article
Hydroxyapatite Thin Films of Marine Origin as Sustainable Candidates for Dental Implants
by Gabriela Dorcioman, Valentina Grumezescu, George E. Stan, Mariana Carmen Chifiriuc, Gratiela Pircalabioru Gradisteanu, Florin Miculescu, Elena Matei, Gianina Popescu-Pelin, Irina Zgura, Valentin Craciun, Faik Nüzhet Oktar and Liviu Duta
Pharmaceutics 2023, 15(4), 1294; https://doi.org/10.3390/pharmaceutics15041294 - 20 Apr 2023
Cited by 15 | Viewed by 2916
Abstract
Novel biomaterials with promising bone regeneration potential, derived from rich, renewable, and cheap sources, are reported. Thus, thin films were synthesized from marine-derived (i.e., from fish bones and seashells) hydroxyapatite (MdHA) by pulsed laser deposition (PLD) technique. Besides the physical–chemical and mechanical investigations, [...] Read more.
Novel biomaterials with promising bone regeneration potential, derived from rich, renewable, and cheap sources, are reported. Thus, thin films were synthesized from marine-derived (i.e., from fish bones and seashells) hydroxyapatite (MdHA) by pulsed laser deposition (PLD) technique. Besides the physical–chemical and mechanical investigations, the deposited thin films were also evaluated in vitro using dedicated cytocompatibility and antimicrobial assays. The morphological examination of MdHA films revealed the fabrication of rough surfaces, which were shown to favor good cell adhesion, and furthermore could foster the in-situ anchorage of implants. The strong hydrophilic behavior of the thin films was evidenced by contact angle (CA) measurements, with values in the range of 15–18°. The inferred bonding strength adherence values were superior (i.e., ~49 MPa) to the threshold established by ISO regulation for high-load implant coatings. After immersion in biological fluids, the growth of an apatite-based layer was noted, which indicated the good mineralization capacity of the MdHA films. All PLD films exhibited low cytotoxicity on osteoblast, fibroblast, and epithelial cells. Moreover, a persistent protective effect against bacterial and fungal colonization (i.e., 1- to 3-log reduction of E. coli, E. faecalis, and C. albicans growth) was demonstrated after 48 h of incubation, with respect to the Ti control. The good cytocompatibility and effective antimicrobial activity, along with the reduced fabrication costs from sustainable sources (available in large quantities), should, therefore, recommend the MdHA materials proposed herein as innovative and viable solutions for the development of novel coatings for metallic dental implants. Full article
Show Figures

Figure 1

Figure 1
<p>Top-view and cross-sectional SEM micrographs of simple and doped fish bone (FB) and seashell (SS)-derived hydroxyapatite thin films. Magnification bars: 2 µm (top-view) and 200 nm (cross-section).</p>
Full article ">Figure 2
<p>The value of the Ca/P ratio in the case of simple and doped FB (<b>a</b>) and SS (<b>b</b>) thin films.</p>
Full article ">Figure 3
<p>Comparison of the X-ray diffractograms recorded for the FB and SS source target materials (collected in Bragg–Brentano geometry) and of the simple and doped FB and SS thin films (collected in grazing incidence geometry). Symbols: <span class="html-fig-inline" id="pharmaceutics-15-01294-i001"><img alt="Pharmaceutics 15 01294 i001" src="/pharmaceutics/pharmaceutics-15-01294/article_deploy/html/images/pharmaceutics-15-01294-i001.png"/></span>—hydroxyapatite; <span style="color:blue">▼</span>—β-tricalcium phosphate; ■—titanium; ◯—TiO).</p>
Full article ">Figure 4
<p>FTIR-ATR spectra of simple and doped FB (<b>a</b>) and SS (<b>b</b>) thin films.</p>
Full article ">Figure 5
<p>The contact angle values inferred in the case of Ti and simple and doped FB (<b>a</b>,<b>b</b>) and SS (<b>c</b>,<b>d</b>) thin films. Test liquids were water (<b>a</b>,<b>c</b>), and diiodomethane (<b>b</b>,<b>d</b>).</p>
Full article ">Figure 6
<p>The values of the surface free energy calculated for Ti and simple and doped FB (<b>a</b>) and SS (<b>b</b>) thin films (γ<sup>d</sup>—hatched region, γ<sup>p</sup>—grey region).</p>
Full article ">Figure 7
<p>The adherence values at the film–Ti substrate interface for simple and doped FB (<b>a</b>) and SS (<b>b</b>) thin films.</p>
Full article ">Figure 8
<p>SEM images of the surface of the simple and doped FB- and SS-based thin films before and after testing in the complete DMEM-FBS solution for different time intervals (3, 7, and 28 days). Magnification bar: 500 nm.</p>
Full article ">Figure 9
<p>SEM images of the simple and doped FB- and SS-based thin films immersed in SBF for 30 days. Magnification bar: 500 nm.</p>
Full article ">Figure 10
<p>FTIR-ATR spectra of simple and doped FB and SS thin films immersed in SBF for 30 days.</p>
Full article ">Figure 11
<p>MTT (<b>a</b>,<b>c</b>,<b>e</b>) and LDH (<b>b</b>,<b>d</b>,<b>f</b>) biocompatibility tests (24 h) in the case of simple and doped FB and SS thin films, using osteoblast (<b>a</b>,<b>b</b>), fibroblast (<b>c</b>,<b>d</b>), and HeLa (<b>e</b>,<b>f</b>) cells.</p>
Full article ">Figure 12
<p>The effect of simple and doped FB and SS thin films on the alkaline phosphatase (ALP), 48 h after incubation.</p>
Full article ">Figure 13
<p>The effect of simple and doped FB and SS thin films on the osteocalcin concentration, 48 h after incubation (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
19 pages, 1953 KiB  
Review
The Science of Selecting Excipients for Dermal Self-Emulsifying Drug Delivery Systems
by Daniélle van Staden, Richard K. Haynes and Joe M. Viljoen
Pharmaceutics 2023, 15(4), 1293; https://doi.org/10.3390/pharmaceutics15041293 - 20 Apr 2023
Cited by 4 | Viewed by 3009
Abstract
Self-emulsification is considered a formulation technique that has proven capacity to improve oral drug delivery of poorly soluble drugs by advancing both solubility and bioavailability. The capacity of these formulations to produce emulsions after moderate agitation and dilution by means of water phase [...] Read more.
Self-emulsification is considered a formulation technique that has proven capacity to improve oral drug delivery of poorly soluble drugs by advancing both solubility and bioavailability. The capacity of these formulations to produce emulsions after moderate agitation and dilution by means of water phase addition provides a simplified method to improve delivery of lipophilic drugs, where prolonged drug dissolution in the aqueous environment of the gastro-intestinal (GI) tract is known as the rate-limiting step rendering decreased drug absorption. Additionally, spontaneous emulsification has been reported as an innovative topical drug delivery system that enables successful crossing of mucus membranes as well as skin. The ease of formulation generated by the spontaneous emulsification technique itself is intriguing due to the simplified production procedure and unlimited upscaling possibilities. However, spontaneous emulsification depends solely on selecting excipients that complement each other in order to create a vehicle aimed at optimizing drug delivery. If excipients are not compatible or unable to spontaneously transpire into emulsions once exposed to mild agitation, no self-emulsification will be achieved. Therefore, the generalized view of excipients as inert bystanders facilitating delivery of an active compound cannot be accepted when selecting excipients needed to produce self-emulsifying drug delivery systems (SEDDSs). Hence, this review describes the excipients needed to generate dermal SEDDSs as well as self-double-emulsifying drug delivery systems (SDEDDSs); how to consider combinations that complement the incorporated drug(s); and an overview of using natural excipients as thickening agents and skin penetration enhancers. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Biopharmaceutical Classification System (BCS) [<a href="#B23-pharmaceutics-15-01293" class="html-bibr">23</a>].</p>
Full article ">Figure 2
<p>Topical Drug Classification System (TCS) [<a href="#B22-pharmaceutics-15-01293" class="html-bibr">22</a>].</p>
Full article ">Figure 3
<p>Structure of the lymphatic network of the skin.</p>
Full article ">Figure 4
<p>Hydrophilic–Lipophilic Balance (HLB) Scale [<a href="#B42-pharmaceutics-15-01293" class="html-bibr">42</a>,<a href="#B43-pharmaceutics-15-01293" class="html-bibr">43</a>].</p>
Full article ">Figure 5
<p>Formulation recommendation system (FRS) when considering drugs belonging to different biopharmaceutical classifications for incorporation into dermal spontaneous emulsions.</p>
Full article ">
25 pages, 5444 KiB  
Article
In Vivo Acute Toxicity and Immunomodulation Assessment of a Novel Nutraceutical in Mice
by Tatiana Onisei, Bianca-Maria Tihăuan, Georgiana Dolete, Mădălina Axinie (Bucos), Manuela Răscol and Gheorghița Isvoranu
Pharmaceutics 2023, 15(4), 1292; https://doi.org/10.3390/pharmaceutics15041292 - 20 Apr 2023
Cited by 3 | Viewed by 1527
Abstract
Achieving and maintaining a well-balanced immune system has righteously become an insightful task for the general population and an even more fundamental goal for those affected by immune-related diseases. Since our immune functions are indispensable in defending the body against pathogens, diseases and [...] Read more.
Achieving and maintaining a well-balanced immune system has righteously become an insightful task for the general population and an even more fundamental goal for those affected by immune-related diseases. Since our immune functions are indispensable in defending the body against pathogens, diseases and other external attacks, while playing a vital role in maintaining health and modulating the immune response, we require an on-point grasp of their shortcoming as a foundation for the development of functional foods and novel nutraceuticals. Seeing that immunoceuticals are considered effective in improving immune functions and reducing the incidence of immunological disorders, the main focus of this study was to assess the immunomodulatory properties and possible acute toxicity of a novel nutraceutical with active substances of natural origin on C57BL/6 mice for 21 days. We evaluated the potential hazards (microbial contamination and heavy metals) of the novel nutraceutical and addressed the acute toxicity according to OECD guidelines of a 2000 mg/kg dose on mice for 21 days. The immunomodulatory effect was assessed at three concentrations (50 mg/kg, 100 mg/kg and 200 mg/kg) by determining body and organ indexes through a leukocyte analysis; flow cytometry immunophenotyping of lymphocytes populations and their subpopulations (T lymphocytes (LyCD3+), cytotoxic suppressor T lymphocytes (CD3+CD8+), helper T lymphocytes (CD3+CD4+), B lymphocytes (CD3−CD19+) and NK cells (CD3−NK1.1.+); and the expression of the CD69 activation marker. The results obtained for the novel nutraceutical referred to as ImunoBoost indicated no acute toxicity, an increased number of lymphocytes and the stimulation of lymphocyte activation and proliferation, demonstrating its immunomodulatory effect. The safe human consumption dose was established at 30 mg/day. Full article
(This article belongs to the Special Issue Recent Advances in Oral Solid Dosages)
Show Figures

Figure 1

Figure 1
<p>FSC/SSC dot-plot with isolated cells of interest (P1).</p>
Full article ">Figure 2
<p>Unlabeled cell suspension (no specific fluorescence signals)—blue laser stimulation.</p>
Full article ">Figure 3
<p>Unlabeled cell suspension (no specific fluorescence signals)—red laser stimulation.</p>
Full article ">Figure 4
<p>Labeled cell suspension—blue laser stimulation.</p>
Full article ">Figure 5
<p>Labeled cell suspension—red laser stimulation.</p>
Full article ">Figure 6
<p>(<b>a</b>) Selection of singlet events. In an FSC-H/SSC-A dot-plot, the Singleti gate was constructed in which siglet events were included. The introduction of cellular aggregates into the analysis was avoided. (<b>b</b>) Selection of the lymphocyte population. From the gate containing the singlet events, the Ly gate (FSC-A/SSC-A) was constructed in which lymphocytes were isolated. (<b>c</b>) Selection of CD3ε+ lymphocytes. Using a histogram (CD3ε/Count), the population of CD3ε+ lymphocytes (total T lymphocytes) was isolated. The CD3ε- lymphocytes were virtually isolated using the ”invert gate” function. (<b>d</b>) Selection of Th and Ts lymphocytes. From the T lymphocytes (CD3ε+), using a CD4/CD8a dot-plot with a quadrant, we isolated the Th (helper) lymphocyte subpopulations with the phenotype CD3ɛ+CD4+CD8a− and Ts subpopulations (suppressor/cytotoxic) with the phenotype CD3ɛ+CD8a+CD4−. (<b>e</b>) Selection of B lymphocytes and NK cells. From CD3ε-negative lymphocytes, B lymphocytes (phenotype CD3ɛ−CD19+NK1.1−) and NK cells (phenotype CD3ɛ−CD19−NK1.1+) were isolated.</p>
Full article ">Figure 7
<p>(<b>a</b>) Selection of singlet events. (<b>b</b>) Selection of the lymphocyte population. (<b>c</b>) Selection of lymphocytes of CD3ε+. (<b>d</b>) Selection of lymphocytes of CD8a+. (<b>e</b>) Selection of B lymphocytes. (<b>f</b>) Selection of NK cells. (<b>g</b>) Selection of lymphocytes of CD69+. (<b>h</b>) Selection of B lymphocytes and NK cells.</p>
Full article ">Figure 7 Cont.
<p>(<b>a</b>) Selection of singlet events. (<b>b</b>) Selection of the lymphocyte population. (<b>c</b>) Selection of lymphocytes of CD3ε+. (<b>d</b>) Selection of lymphocytes of CD8a+. (<b>e</b>) Selection of B lymphocytes. (<b>f</b>) Selection of NK cells. (<b>g</b>) Selection of lymphocytes of CD69+. (<b>h</b>) Selection of B lymphocytes and NK cells.</p>
Full article ">Figure 8
<p>Body weight assessment—acute toxicity testing.</p>
Full article ">Figure 9
<p>Assessment of leukocyte numbers in acute toxicity testing for the total numbers of leukocytes (WBC), lymphocytes (LY), neutrophils (NE) and monocytes (MO) in mice; * = <span class="html-italic">p</span> value &lt; 0.05.</p>
Full article ">Figure 10
<p>Body weight assessment of animals that received the product for 21 days.</p>
Full article ">Figure 11
<p>Evaluation of the number of leukocytes in the groups that received the novel nutraceutical compared to the control group: total numbers of leukocytes (WBC), lymphocytes (LY) and neutrophils (NE) in mice.</p>
Full article ">Figure 12
<p>CD69 expression on lymphocytes in the groups that received ImunoBoost compared to the control group—stimulation with LPS; * = <span class="html-italic">p</span> value &lt; 0.05.</p>
Full article ">Figure 13
<p>CD69 expression on lymphocytes in the groups that received ImunoBoost compared to the control group—stimulation with conA; * = <span class="html-italic">p</span> value &lt; 0.05.</p>
Full article ">Figure 14
<p>Lymphocyte proliferation capacity in groups that received the novel nutraceutical compared to the control group—stimulation with LPS; * = <span class="html-italic">p</span> value &lt; 0.05.</p>
Full article ">Figure 15
<p>Lymphocyte proliferation capacity levels in the groups that received the novel nutraceutical compared to the control group—stimulation with conA; * = <span class="html-italic">p</span> value &lt; 0.05.</p>
Full article ">
22 pages, 4927 KiB  
Article
In Vitro Biotransformation and Anti-Inflammatory Activity of Constituents and Metabolites of Filipendula ulmaria
by Anastasia Van der Auwera, Laura Peeters, Kenn Foubert, Stefano Piazza, Wim Vanden Berghe, Nina Hermans and Luc Pieters
Pharmaceutics 2023, 15(4), 1291; https://doi.org/10.3390/pharmaceutics15041291 - 20 Apr 2023
Cited by 4 | Viewed by 1966
Abstract
(1) Background: Filipendula ulmaria (L.) Maxim. (Rosaceae) (meadowsweet) is widely used in phytotherapy against inflammatory diseases. However, its active constituents are not exactly known. Moreover, it contains many constituents, such as flavonoid glycosides, which are not absorbed, but metabolized in the colon by [...] Read more.
(1) Background: Filipendula ulmaria (L.) Maxim. (Rosaceae) (meadowsweet) is widely used in phytotherapy against inflammatory diseases. However, its active constituents are not exactly known. Moreover, it contains many constituents, such as flavonoid glycosides, which are not absorbed, but metabolized in the colon by gut microbiota, producing potentially active metabolites that can be absorbed. The aim of this study was to characterize the active constituents or metabolites. (2) Methods: A F. ulmaria extract was processed in an in vitro gastrointestinal biotransformation model, and the metabolites were characterized using UHPLC-ESI-QTOF-MS analysis. In vitro anti-inflammatory activity was evaluated by testing the inhibition of NF-κB activation, COX-1 and COX-2 enzyme inhibition. (3) Results: The simulation of gastrointestinal biotransformation showed a decrease in the relative abundance of glycosylated flavonoids such as rutin, spiraeoside and isoquercitrin in the colon compartment, and an increase in aglycons such as quercetin, apigenin, naringenin and kaempferol. The genuine as well as the metabolized extract showed a better inhibition of the COX-1 enzyme as compared to COX-2. A mix of aglycons present after biotransformation showed a significant inhibition of COX-1. (4) Conclusions: The anti-inflammatory activity of F. ulmaria may be explained by an additive or synergistic effect of genuine constituents and metabolites. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Total ion chromatogram of the <span class="html-italic">F. ulmaria</span> extract before (<b>A</b>) and after (<b>B</b>) in vitro gastrointestinal biotransformation.</p>
Full article ">Figure 2
<p>Time profiles (time in hours) of rutin (<b>A</b>), isoquercitrin (<b>B</b>) and quercetin (<b>C</b>) during gastrointestinal biotransformation. The gastric phase continued for 1 h, followed by the small intestinal phase for 1.5 h and a 72 h colonic phase. FEX (i.e., <span class="html-italic">F. ulmaria</span> extract) in red, NCFEX (i.e., negative control) in blue and MB (i.e., method blank) in green.</p>
Full article ">Figure 3
<p>Time profiles (time in hours) of astragalin (<b>A</b>) and isorhamnetin-O-hexoside (<b>B</b>) during gastrointestinal biotransformation. See also legend of <a href="#pharmaceutics-15-01291-f002" class="html-fig">Figure 2</a>.</p>
Full article ">Figure 4
<p>Metabolic pathway of rutin. Detected metabolites are in black; non-detected metabolites are in grey.</p>
Full article ">Figure 5
<p>Time profile (time in hours) of luteolin (<b>A</b>), kaempferol (<b>B</b>), isorhamnetin (<b>C</b>), apigenin (<b>D</b>), naringenin (<b>E</b>), phloretin (<b>F</b>), chrysoeriol (<b>G</b>) and 3′-methoxy-5,7-dihydroxyflavone (<b>H</b>) during gastrointestinal biotransformation. See also legend of <a href="#pharmaceutics-15-01291-f002" class="html-fig">Figure 2</a>.</p>
Full article ">Figure 6
<p>Time profiles (time in hours) of monotropitin (<b>A</b>) and salicylic acid (<b>B</b>) during gastrointestinal biotransformation. See also legend of <a href="#pharmaceutics-15-01291-f002" class="html-fig">Figure 2</a>.</p>
Full article ">Figure 7
<p>Time profiles (time in hours) of tellimagrandin II (<b>A</b>) and ellagic acid (<b>B</b>) during gastrointestinal biotransformation. See also legend of <a href="#pharmaceutics-15-01291-f002" class="html-fig">Figure 2</a>.</p>
Full article ">Figure 8
<p>The effect of the <span class="html-italic">F. ulmaria</span> extract and the mix (20 µM of gallic acid and salicylic acid, 6 µM of quercetin and 4 µM of syringic acid) on COX-1 enzyme inhibition. Indomethacin (1.25 µM), in yellow, served as positive control. The graph depicts compiled data of three independent experiments (mean ± SD). <span class="html-italic">p</span>-values are expressed as * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.0001 compared to control.</p>
Full article ">Figure 9
<p>The effect of the <span class="html-italic">F. ulmaria</span> extract and the mix (20 µM of gallic acid and salicylic acid, 6 µM of quercetin and 4 µM of syringic acid) on COX-2 enzyme inhibition. Celecoxib (2.5 µM), in yellow, served as positive control. The graph depicts compiled data of three independent experiments (mean ± SD). <span class="html-italic">p</span>-values are expressed as *** <span class="html-italic">p</span> &lt; 0.0001 compared to control.</p>
Full article ">Figure 10
<p>The effect of the digested <span class="html-italic">F. ulmaria</span> extract (FEX 72 h) and different extraction procedures of FEX 72 h on COX-1 enzyme inhibition. Indomethacin (1.25 µM), in yellow, served as positive control. The graph depicts compiled data of three independent experiments (mean ± SD). <span class="html-italic">p</span>-values are expressed as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.0001 compared to control.</p>
Full article ">Figure 11
<p>The effect of the digested <span class="html-italic">F. ulmaria extract</span> (FEX 72 h) and different extraction procedures of FEX 72 h on COX-2 enzyme inhibition. Celecoxib (2.5 µM), in yellow, served as positive control. The graph depicts compiled data of three independent experiments (mean ± SD). <span class="html-italic">p</span>-values are expressed as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.0001 compared to control.</p>
Full article ">Figure 12
<p>The effect of the <span class="html-italic">F. ulmaria</span> extract and the mix (20 µM of gallic acid and salicylic acid, 6 µM of quercetin and 4 µM of syringic acid) on NF-κB-driven transcription in L929 cells. Dexamethasone (Dexa, 1 µM), in yellow, served as positive control. The graph depicts compiled data of three independent experiments (mean ± SD). <span class="html-italic">p</span>-values are expressed as *** <span class="html-italic">p</span> &lt; 0.0001 compared to control stimulated with TNF-α.</p>
Full article ">
21 pages, 5257 KiB  
Article
Extracellular Vesicles and Their Mimetics: A Comparative Study of Their Pharmacological Activities and Immunogenicity Profiles
by Wei Heng Chng, Ram Pravin Kumar Muthuramalingam, Charles Kang Liang Lou, Silas New, Yub Raj Neupane, Choon Keong Lee, Ayca Altay Benetti, Chenyuan Huang, Praveen Thoniyot, Wei Seong Toh, Jiong-Wei Wang and Giorgia Pastorin
Pharmaceutics 2023, 15(4), 1290; https://doi.org/10.3390/pharmaceutics15041290 - 20 Apr 2023
Cited by 3 | Viewed by 2232
Abstract
Extracellular vesicles (EVs), which are miniaturised carriers loaded with functional proteins, lipids, and nucleic acid material, are naturally secreted by cells and show intrinsic pharmacological effects in several conditions. As such, they have the potential to be used for the treatment of various [...] Read more.
Extracellular vesicles (EVs), which are miniaturised carriers loaded with functional proteins, lipids, and nucleic acid material, are naturally secreted by cells and show intrinsic pharmacological effects in several conditions. As such, they have the potential to be used for the treatment of various human diseases. However, the low isolation yield and laborious purification process are obstacles to their translation for clinical use. To overcome this problem, our lab developed cell-derived nanovesicles (CDNs), which are EV mimetics produced by shearing cells through membrane-fitted spin cups. To evaluate the similarities between EVs and CDNs, we compare the physical properties and biochemical composition of monocytic U937 EVs and U937 CDNs. Besides having similar hydrodynamic diameters, the produced CDNs had proteomic, lipidomic, and miRNA profiles with key communalities compared to those of natural EVs. Further characterisation was conducted to examine if CDNs could exhibit similar pharmacological activities and immunogenicity when administered in vivo. Consistently, CDNs and EVs modulated inflammation and displayed antioxidant activities. EVs and CDNs both did not exert immunogenicity when administered in vivo. Overall, CDNs could serve as a scalable and efficient alternative to EVs for further translation into clinical use. Full article
(This article belongs to the Special Issue Exosome-Based Drug Delivery: Translation from Bench to Clinic)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) Isolation of U937 EVs with sucrose cushion differential ultracentrifugation. The isolated EV pellet was resuspended in PBS. (<b>B</b>) Production of U937 CDNs by shearing cells through spin cups fitted with membrane filters. Spin cups were centrifuged at 14,000× <span class="html-italic">g</span> for 10 min at 4 °C. The resultant product was purified using a Sephadex G-50 size exclusion column. Image created using BioRender.</p>
Full article ">Figure 2
<p>Physical properties of U937 EVs and U937 CDNs. (<b>A</b>) Hydrodynamic diameters and polydispersity index of U937 EVs and U937 CDNs; <span class="html-italic">n</span> = 3. Data presented as mean ± SEM. (<b>B</b>) Zeta potentials of U937 EVs and U937 CDNs; <span class="html-italic">n</span> = 3. Data presented as mean ± SEM. Transmission electron microscopy images of (<b>C</b>) U937 EVs and (<b>D</b>) U937 CDNs. The red arrow indicates nanovesicles. The scale bar is 200 nm.</p>
Full article ">Figure 3
<p>Comparison of the biochemical composition of U937 EVs and U937 CDNs. (<b>A</b>) Venn diagram comparing the protein profiles of U937 EVs and U937 CDNs. (<b>B</b>) Gene ontology of U937 EVs and U937 CDNs with respect to the cellular component. (<b>C</b>) Comparison of the lipid profiles of U937 EVs and U937 CDNs; <span class="html-italic">n</span> = 3. (<b>D</b>) Heat map comparing the miRNA profiles of (left) U937 EVs and (right) U937 CDNs; <span class="html-italic">n</span> = 2. Red indicates high abundance (low Ct values), blue indicates low abundance (high Ct values), and black indicates a lack of detection in at least one of the two replicates.</p>
Full article ">Figure 3 Cont.
<p>Comparison of the biochemical composition of U937 EVs and U937 CDNs. (<b>A</b>) Venn diagram comparing the protein profiles of U937 EVs and U937 CDNs. (<b>B</b>) Gene ontology of U937 EVs and U937 CDNs with respect to the cellular component. (<b>C</b>) Comparison of the lipid profiles of U937 EVs and U937 CDNs; <span class="html-italic">n</span> = 3. (<b>D</b>) Heat map comparing the miRNA profiles of (left) U937 EVs and (right) U937 CDNs; <span class="html-italic">n</span> = 2. Red indicates high abundance (low Ct values), blue indicates low abundance (high Ct values), and black indicates a lack of detection in at least one of the two replicates.</p>
Full article ">Figure 3 Cont.
<p>Comparison of the biochemical composition of U937 EVs and U937 CDNs. (<b>A</b>) Venn diagram comparing the protein profiles of U937 EVs and U937 CDNs. (<b>B</b>) Gene ontology of U937 EVs and U937 CDNs with respect to the cellular component. (<b>C</b>) Comparison of the lipid profiles of U937 EVs and U937 CDNs; <span class="html-italic">n</span> = 3. (<b>D</b>) Heat map comparing the miRNA profiles of (left) U937 EVs and (right) U937 CDNs; <span class="html-italic">n</span> = 2. Red indicates high abundance (low Ct values), blue indicates low abundance (high Ct values), and black indicates a lack of detection in at least one of the two replicates.</p>
Full article ">Figure 4
<p>Nitrite concentrations in cell culture supernatant after RAW264.7 macrophages had been treated with increasing concentrations of (<b>A</b>) U937 EVs and (<b>B</b>) CDNs for 24 h. TNF-α concentrations in cell culture supernatant after RAW264.7 macrophages were treated with increasing concentrations of (<b>C</b>) U937 EVs and (<b>D</b>) CDNs for 24 h. IL-6 concentrations in the cell culture supernatant after RAW264.7 macrophages were treated with increasing concentrations of (<b>E</b>) U937 EVs and (<b>F</b>) CDNs for 24 h. IL-1β and IL-10 concentrations were below the detection limit of the ELISA kits (&lt;31.3 pg/mL). The LPS concentration was 10 ng/mL. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 3). n.s., no statistical difference; *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4 Cont.
<p>Nitrite concentrations in cell culture supernatant after RAW264.7 macrophages had been treated with increasing concentrations of (<b>A</b>) U937 EVs and (<b>B</b>) CDNs for 24 h. TNF-α concentrations in cell culture supernatant after RAW264.7 macrophages were treated with increasing concentrations of (<b>C</b>) U937 EVs and (<b>D</b>) CDNs for 24 h. IL-6 concentrations in the cell culture supernatant after RAW264.7 macrophages were treated with increasing concentrations of (<b>E</b>) U937 EVs and (<b>F</b>) CDNs for 24 h. IL-1β and IL-10 concentrations were below the detection limit of the ELISA kits (&lt;31.3 pg/mL). The LPS concentration was 10 ng/mL. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 3). n.s., no statistical difference; *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>(<b>A</b>) Total antioxidant capacity of U937 EVs and U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 5). (<b>B</b>) Detoxification of ROS by cellular antioxidant enzymes. In the first line of defence, antioxidant enzymes such as SOD and CAT can convert ROS into oxygen and water. In the third line of defence, antioxidant enzymes such as GST could detoxify oxidised biomolecules and repair damage caused by free radicals. (<b>C</b>) Catalase activity of U937 EVs and U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 4). (<b>D</b>) Glutathione S-transferase activity of U937 EVs and U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 3). ns, no statistical difference; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>(<b>A</b>) Total antioxidant capacity of U937 EVs and U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 5). (<b>B</b>) Detoxification of ROS by cellular antioxidant enzymes. In the first line of defence, antioxidant enzymes such as SOD and CAT can convert ROS into oxygen and water. In the third line of defence, antioxidant enzymes such as GST could detoxify oxidised biomolecules and repair damage caused by free radicals. (<b>C</b>) Catalase activity of U937 EVs and U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 4). (<b>D</b>) Glutathione S-transferase activity of U937 EVs and U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 3). ns, no statistical difference; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Immune cell profiles of (<b>A</b>) blood and (<b>B</b>) spleen after intravenous administration of saline, U937 EVs or U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 4 for saline, <span class="html-italic">n</span> = 5 for U937 EVs and CDNs). ns indicates no statistical difference, * indicates <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6 Cont.
<p>Immune cell profiles of (<b>A</b>) blood and (<b>B</b>) spleen after intravenous administration of saline, U937 EVs or U937 CDNs. Data presented as mean ± SEM (<span class="html-italic">n</span> = 4 for saline, <span class="html-italic">n</span> = 5 for U937 EVs and CDNs). ns indicates no statistical difference, * indicates <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
27 pages, 7036 KiB  
Article
Multicomponent Lipid Nanoparticles for RNA Transfection
by Nataliya Gretskaya, Mikhail Akimov, Dmitry Andreev, Anton Zalygin, Ekaterina Belitskaya, Galina Zinchenko, Elena Fomina-Ageeva, Ilya Mikhalyov, Elena Vodovozova and Vladimir Bezuglov
Pharmaceutics 2023, 15(4), 1289; https://doi.org/10.3390/pharmaceutics15041289 - 20 Apr 2023
Cited by 7 | Viewed by 3101
Abstract
Despite the wide variety of available cationic lipid platforms for the delivery of nucleic acids into cells, the optimization of their composition has not lost its relevance. The purpose of this work was to develop multi-component cationic lipid nanoparticles (LNPs) with or without [...] Read more.
Despite the wide variety of available cationic lipid platforms for the delivery of nucleic acids into cells, the optimization of their composition has not lost its relevance. The purpose of this work was to develop multi-component cationic lipid nanoparticles (LNPs) with or without a hydrophobic core from natural lipids in order to evaluate the efficiency of LNPs with the widely used cationic lipoid DOTAP (1,2-dioleoyloxy-3-[trimethylammonium]-propane) and the previously unstudied oleoylcholine (Ol-Ch), as well as the ability of LNPs containing GM3 gangliosides to transfect cells with mRNA and siRNA. LNPs containing cationic lipids, phospholipids and cholesterol, and surfactants were prepared according to a three-stage procedure. The average size of the resulting LNPs was 176 nm (PDI 0.18). LNPs with DOTAP mesylate were more effective than those with Ol-Ch. Core LNPs demonstrated low transfection activity compared with bilayer LNPs. The type of phospholipid in LNPs was significant for the transfection of MDA-MB-231 and SW 620 cancer cells but not HEK 293T cells. LNPs with GM3 gangliosides were the most efficient for the delivery of mRNA to MDA-MB-231 cells and siRNA to SW620 cells. Thus, we developed a new lipid platform for the efficient delivery of RNA of various sizes to mammalian cells. Full article
(This article belongs to the Special Issue Liposomal and Lipid-Based Drug Delivery Systems and Vaccines)
Show Figures

Figure 1

Figure 1
<p>Structural formulas of the LNP and cLNP components. Cationic lipids: <b>1</b>, dioleoyltrimethylaminopropane (DOTAP); <b>2</b>, oleoylcholine (Ol-Ch). Structural lipids: <b>3</b>, dioleoylphosphatidylethanolamine (DOPE); <b>4</b>, dioleoylphosphatidylcholine (DOPC); <b>5</b>, cholesterol (Ch). Additional components: <b>6</b>, stearoylmonosorbitan (SPAN60); <b>7</b>, polyethylene glycol sorbitan monooleate (polysorbate 80 (PS80)), w + x + y = 20. Core lipids: <b>8</b>, coconut oil triglycerides (COTs); <b>9</b>, squalene (Sq); <b>10</b>, cholesterol acetate (ChA). Functional lipids: <b>11</b>, GM3 ganglioside (GM3).</p>
Full article ">Figure 2
<p>AFM images of LNPs and lipoplexes with mRNA formed in water. Experimental conditions: mi-ca, scanning area of 4 × 4 µm (512 × 512 pixels). (<b>a</b>) LNP sample S1 diluted to 1:20 with water; (<b>b</b>,<b>c</b>) LNP lipoplex (sample S1) and mRNA (N/P = 15) diluted to 1:20 with water. (<b>a</b>), the cross-sections from top to bottom, half-height: 16, 10.5 and 4.6 nm, half-width at half maximum: 115, 90 and 50 nm, (<b>b</b>), half-height 3.3 nm, half-width at half maximum 59 nm, (<b>c</b>), half-height 1.8, half-width at half maximum 180 nm. Representative samples from multiple scan fields are shown.</p>
Full article ">Figure 3
<p>Electropherogram and kinetics of mRNA hydrolysis and mRNA–LNP lipoplex by RNase A. (<b>a</b>) Electropherogram of LNP (sample S1b) lipoplex sample: 1—markers; 2—mRNA; 3—LNP S1b and mRNA. (<b>b</b>) Kinetics of mRNA and lipoplex with LNP (sample S1b) hydrolysis. Values are given as percentages relative to the starting point of each sample. Tris–borate–EDTA buffer, 8.0 pH, 200 ng of mRNA, and 0.05 units of RNase A (Thermo). LNP sample S1 composition is given in <a href="#pharmaceutics-15-01289-t002" class="html-table">Table 2</a>, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 4
<p>Quantity of RNA (relative to free RNA control, %) calculated from the average fluorescence intensity of SYBR Green I during the incubation of lipoplex samples for 30 min. Incubation of samples with RNase (index r) and without enzyme (no index); all samples were prepared in water. See <a href="#pharmaceutics-15-01289-t001" class="html-table">Table 1</a> for LNP composition, mean ± SE. **, A statistically significant difference, <span class="html-italic">p</span> &lt; 0.001; ****, a statistically significant difference, <span class="html-italic">p</span> &lt; 0.0001; ns, not significant.</p>
Full article ">Figure 5
<p>Viability assessment of HEK 293T cells. Microscopy of lipoplexes prepared from LNP (sample S1), labelled with BODIPY-C3 dye (S1Flu + mRNA, (<b>a</b>)) and BODIPY-C3 labelled LNPs (S1Flu, (<b>b</b>)); incubation time of 24 h and scale bar of 100 nm. Light, brightfield channel, Flu + Light, pseudocolor combined image, gray, brightfield channel; green, green fluorescence, (<b>c</b>) MTT test of HEK 293T cells after incubation with lipoplexes prepared from LNP samples for 24 h. See <a href="#pharmaceutics-15-01289-t001" class="html-table">Table 1</a> for LNP composition; control—HEK 293T cells without particles, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 6
<p>Transfection efficiency of HEK 293T cells with mRNA–LNP (sample S1) lipoplexes depending on various counterions of DOTAP: chloride (Cl), iodide (I) or mesylate (Mes). *, A statistically significant difference from samples with Cl and I counterions, <span class="html-italic">p</span> ≤ 0.05. Incubation time was 24 h, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 7
<p>Transfection efficiency of HEK 293T cells with lipoplexes with various cationic lipids. Sample S1 contained 100% DOTAP, S3 contained 100% oleoylcholine, and samples S15–S13 contained a mixture of DOTAP and oleoylcholine in amounts of 11, 22, and 44%, respectively, of the total amount of cationic lipids (see LNP composition in <a href="#pharmaceutics-15-01289-t001" class="html-table">Table 1</a>). The untreated controls did not differ from the background signal (700 counts per 10 s). Incubation time was 24 h, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 8
<p>Transfection efficiency of HEK 293T cells with lipoplexes with different N/P ratios. *, A statistically significant difference from samples with N/P ratios of 7.5 and 30, <span class="html-italic">p</span> &lt; 0.05. Incubation time was 24 h, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 9
<p>The efficiency of cell transfection with lipoplexes prepared from LNPs with different structural lipids. Phospholipid composition: 100% DOPE (sample S1), 100% DOPC (sample S4), and 50:50 DOPE/DOPC (sample S5). Incubation time was 24 h. (<b>a</b>) HEK 293T cells, (<b>b</b>) MDA-MB-231 cells, (<b>c</b>) SW 620 cells. The untreated controls did not differ from the background signal (700 counts per 10 s) and are not displayed. ****, A statistically significant difference, <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">n</span> = 3. ns, not significant.</p>
Full article ">Figure 10
<p>The efficiency of the transfection of HEK 293T cells with lipoplexes prepared from nanoparticles of the base composition (sample S1) stored at +4–8 °C. Incubation time was 24 h. ns. not significant, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 11
<p>The efficiency of the transfection of HEK 293T cells with lipoplexes formed from LNPs with hydrophobic cores of various compositions. (<b>a</b>), Comparison of lipoplexes with different cLNP compositions. Samples S11 and S12 contained cholesterol acetate and coconut oil triglycerides in ratios of 7:7 and 4:4 mole %, respectively; samples S9 and S10 contained squalene and coconut oil triglycerides in ratios of 7:7 and 4:4 mole %, respectively (<a href="#pharmaceutics-15-01289-t001" class="html-table">Table 1</a>). (<b>b</b>) Comparison of the efficacy of lipoplexes prepared from LNPs (sample S1) and cLNPs (samples S11 and S12). The untreated controls did not differ from the background signal (700 counts per 10 s). Incubation time was 24 h. ns—not significant. ns. not significant, n = 3.</p>
Full article ">Figure 12
<p>The efficiency of cell transfection with lipoplexes prepared from LNPs with GM3 gangliosides. Ganglioside content: 0.5% (sample S6), 1% (sample S7), and 1.7% (sample S8); control—sample S1 (<a href="#pharmaceutics-15-01289-t001" class="html-table">Table 1</a>). Incubation time was 24 h. (<b>a</b>) HEK 293T cells, (<b>b</b>) MDA-MB-231 cells, (<b>c</b>) SW 620 cells. ns—not significant. ns. not significant, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 13
<p>Cell transfection efficiency with lipoplexes prepared from LNPs with GM3 gangliosides and siRNA against the GPR55 receptor gene. Control, native MDA-MB-231 cells; LF, siRNA transfection with lipofectamine; S8, siRNA–LNPs S8 lipoplex transfection (1.7% GM3; see <a href="#pharmaceutics-15-01289-t001" class="html-table">Table 1</a>). Genes: rpii, POLR2A housekeeping gene; gpr55, GPR55 receptor gene. Incubation time was 72 h. RT-qPCR data and expression results are presented as values of 2 to the degree minus the number of the quantification cycle, <span class="html-italic">n</span> = 2.</p>
Full article ">
13 pages, 2200 KiB  
Article
Experimental Elucidation of Templated Crystallization and Secondary Processing of Peptides
by Vivek Verma, Isha Bade, Vikram Karde and Jerry Y. Y. Heng
Pharmaceutics 2023, 15(4), 1288; https://doi.org/10.3390/pharmaceutics15041288 - 20 Apr 2023
Cited by 3 | Viewed by 2047
Abstract
The crystallization of peptides offers a sustainable and inexpensive alternative to the purification process. In this study, diglycine was crystallised in porous silica, showing the porous templates’ positive yet discriminating effect. The diglycine induction time was reduced by five-fold and three-fold upon crystallising [...] Read more.
The crystallization of peptides offers a sustainable and inexpensive alternative to the purification process. In this study, diglycine was crystallised in porous silica, showing the porous templates’ positive yet discriminating effect. The diglycine induction time was reduced by five-fold and three-fold upon crystallising in the presence of silica with pore sizes of 6 nm and 10 nm, respectively. The diglycine induction time had a direct relationship with the silica pore size. The stable form (α-form) of diglycine was crystallised in the presence of porous silica, with the diglycine crystals obtained associated with the silica particles. Further, we studied the mechanical properties of diglycine tablets for their tabletability, compactability, and compressibility. The mechanical properties of the diglycine tablets were similar to those of pure MCC, even with the presence of diglycine crystals in the tablets. The diffusion studies of the tablets using the dialysis membrane presented an extended release of diglycine through the dialysis membrane, confirming that the peptide crystal can be used for oral formulation. Hence, the crystallization of peptides preserved their mechanical and pharmacological properties. More data on different peptides can help us produce oral formulation peptides faster than usual. Full article
(This article belongs to the Special Issue Recent Advances in Secondary Processing of Pharmaceutical Powders)
Show Figures

Figure 1

Figure 1
<p>Comparison of % desupersaturation curves of diglycine in the absence and presence of porous silica at S = 1.20; volume = 40 mL; T<sub>sat</sub> = 40 °C; T<sub>cry</sub> = 32.7 °C.</p>
Full article ">Figure 2
<p>(<b>A</b>) Powder X-ray diffraction spectra of the diglycine–silica composite solids isolated upon complete desupersaturation of diglycine in the presence of porous silica at S = 1.20, along with the diglycine patterns; (<b>B</b>) Scanning electron microscopy images of diglycine, porous silica, and isolated solids after the desupersaturation experiments in the presence of porous silica.</p>
Full article ">Figure 3
<p>(<b>Top</b>) Tabletability, (<b>Middle</b>) compactability, and (<b>Bottom</b>) compressibility profiles of the 25% loading blend of diglycine–silica–MCC composite tablets prepared along with MCC alone (blue squares, PH101). Red circles, blue upwards triangles, green downwards triangles, and pink rhombuses represent diglycine crystallised using silica with pore sizes of 6 nm, 10 nm, 30 nm, and 50 nm as templates, respectively, with n ≥ 3 (n is the number of experiments).</p>
Full article ">Figure 4
<p>%diffusion of diglycine from the diglycine, silica, and MCC composite tablet compressed at 2.5 kN. The diffusion medium was water; volume = 100 mL; and stirring = 100 rpm.</p>
Full article ">
Previous Issue
Next Issue
Back to TopTop