[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (216)

Search Parameters:
Keywords = transdermal penetration

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 1620 KiB  
Article
Application of a Validated HPLC Method for the Determination of Resveratrol, Ferulic Acid, Quercetin, Retinol, and α-Tocopherol in a Cold Cream—Permeability Study
by Athanasia Karavalasi, Sofia Almpani, Panagiota Tserkezou, Konstantina Chachlioutaki, Georgios Kamaris and Catherine K. Markopoulou
Appl. Sci. 2024, 14(24), 11843; https://doi.org/10.3390/app142411843 - 18 Dec 2024
Viewed by 309
Abstract
Due to the rapid increase in the use of anti-aging cosmetic products, there is a need to develop valid analytical methods to control their quality. The present work deals with the development and validation of a new chromatographic method for the quantitative determination [...] Read more.
Due to the rapid increase in the use of anti-aging cosmetic products, there is a need to develop valid analytical methods to control their quality. The present work deals with the development and validation of a new chromatographic method for the quantitative determination of five lipophilic components (resveratrol, ferulic acid, quercetin, retinol, and α-tocopherol), with anti-aging properties, in a cold cream (w/o). For the HPLC-UV separation of the active ingredients, an HS, Discovery® Supelco (Supelco Inc., Bellefonte, PA, USA), C18 column (25 cm × 4.6 mm), 5 μm (at 40 °C) was used as a stationary phase while a binary system of A: Acetonitrile with formic acid 0.2% and B: H2O with formic acid 0.2%, in gradient elution (flow 1.5 mL·min−1), was used as mobile. The analytical method was validated according to ICH guidelines Q2(R2), where linearity (r2 ≥ 0.998), selectivity, precision (% recovery 97.1–101.9), and accuracy (%RSD < 2) were evaluated. The processing of the samples for the recovery of the five analytes from the cream was investigated by experimental design methodology and the cross D-optimal technique (% recovery 98.5–102.9, %RSD < 2%, n = 5). Finally, the same analysis was applied to study the transdermal penetration of the active ingredients incorporated in cold cream (over a period of 8 h). Their behavior was compared with the corresponding one in suspension using Franz cells in a vertical arrangement. The new method is considered reliable for the analysis of the anti-aging product. Full article
(This article belongs to the Special Issue Research on Organic and Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of RSV, FERA, QR, RTN, and α-ΤΟC.</p>
Full article ">Figure 2
<p>Chromatogram at 290 nm of (1) standard solution, (2) sample, (3) blank of diluent, and (4) substrate blank.</p>
Full article ">Figure 3
<p>Stability study of substances at the temperature of 45 °C.</p>
Full article ">Figure 4
<p>Suggested optimal values of factors (<b>A</b>) MeOH (mL), (<b>B</b>) ACN (mL), (<b>C</b>) sonication time (min) and (<b>D</b>) freezing time (min) and predicted % recovery values of (<b>a</b>): vitamin A, (<b>b</b>): vitamin E, (<b>c</b>): resveratrol, (<b>d</b>): ferulic acid, (<b>e</b>): quercetin.</p>
Full article ">Figure 5
<p>Permeability study of ferulic acid, resveratrol, and quercetin (<b>a</b>) cream, (<b>b</b>) suspension.</p>
Full article ">
21 pages, 5755 KiB  
Article
Unlocking the Stratum Corneum Barrier to Skin Penetration for the Transdermal Delivery of Cyclovirobuxine D
by Yun-Hao Ren, Feng-Yuan Song, Jing-Yu Zhao, Bing-Wen Liang and Li-Hua Peng
Pharmaceutics 2024, 16(12), 1600; https://doi.org/10.3390/pharmaceutics16121600 - 16 Dec 2024
Viewed by 417
Abstract
Background/Objectives: Cyclovirobuxine D, a natural compound derived from the medicinal plant Buxus sinica, demonstrates a diverse array of therapeutic benefits, encompassing anti-arrhythmic properties, blood pressure regulation, neuronal protection, and anti-ischemic activity. However, its limited solubility hinders the bioavailability of current oral and injectable [...] Read more.
Background/Objectives: Cyclovirobuxine D, a natural compound derived from the medicinal plant Buxus sinica, demonstrates a diverse array of therapeutic benefits, encompassing anti-arrhythmic properties, blood pressure regulation, neuronal protection, and anti-ischemic activity. However, its limited solubility hinders the bioavailability of current oral and injectable formulations, causing considerable adverse reactions and toxicity. Methods: In this investigation, we embarked on an unprecedented exploration of the skin penetration potential of cyclovirobuxine D utilizing chemical penetration enhancers and niosomes as innovative strategies to enhance its dermal absorption. These strategies were rigorously tested and optimized. Results: Among the tested chemical penetration enhancers, azone emerged as the most potent, achieving a 4.55-fold increase in skin penetration compared to the untreated group. Additionally, when encapsulated within niosomes, primarily composed of Span60 and cholesterol, the skin penetration of cyclovirobuxine D was notably enhanced by 1.50-fold. Furthermore, when both cyclovirobuxine D and azone were co-encapsulated within the niosomes, the skin penetration of cyclovirobuxine D was remarkably elevated by 8.10-fold compared to the solvent-dispersed group. This enhancement was corroborated through rigorous in vitro and in vivo experiments. Notably, the combination of other chemical penetration enhancers with niosome encapsulation also exhibited synergistic effects in enhancing the skin penetration of cyclovirobuxine D. Conclusions: These findings provide a compelling rationale for the administration of cyclovirobuxine D via skin-mediated transdermal delivery, offering superior safety, efficacy, and convenience. The innovative combination of niosomes and chemical penetration enhancers represents a novel system for the transdermal delivery of cyclovirobuxine D, holding immense promise for clinical applications in the treatment of brain, neuronal, and cardiovascular disorders. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

Figure 1
<p>Extraction and separation, capability for the transdermal delivery and disease treatment of cyclovirobuxine D.</p>
Full article ">Figure 2
<p>(<b>a</b>) standard curves obtained from different concentrations of cyclovirobuxine D samples by LC–MS; (<b>b</b>) peak time of cyclovirobuxine D samples under liquid quality conditions by LC–MS; (<b>c</b>) cumulative skin penetration and (<b>d</b>) cumulative skin penetration percentage (%) of cyclovirobuxine D under different receiving liquids A, B, C, and D. Among these, PEG400: ethanol: saline was selected as the receiving liquid, with volume ratios of 1:3:6, 2:2:6, and 2.5:2.5:5, respectively, represented by B, C, and D, where A is saline as the receiving liquid. * <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; 0.001.</p>
Full article ">Figure 3
<p>(<b>a</b>) cumulative skin penetration; and (<b>b</b>) skin penetration percentage (%) of cyclovirobuxine D under the condition of V (ethanol: saline) = 3:7 as the transdermal receiving liquid for different chemical penetration enhancers. * <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; 0.001.</p>
Full article ">Figure 4
<p>(<b>a</b>) pressure was reduced and evaporation was rotated at 40 °C until a yellow viscous solid film appeared at the bottom of the round bottomed flask; (<b>b</b>) after magnetic stirring for 1 h in a constant temperature environment of 55 °C, the white homogeneous liquid appears viscous; (<b>c</b>) Cy-Nio observed under a 15k transmission electron microscope; (<b>d</b>) particle size; and (<b>e</b>) potential distribution of niosomes. Evaluation of: (<b>f</b>) the photostability; (<b>g</b>) storage stability; (<b>h</b>) thermal stability; and (<b>i</b>) acid-base stability of Cy-Nio in the range of 1, 5, 10, and 15 d.</p>
Full article ">Figure 5
<p>(<b>a<sub>1</sub></b>) photostability; (<b>a<sub>2</sub></b>) storage stability; (<b>a<sub>3</sub></b>) thermal stability; and (<b>a<sub>4</sub></b>) acid-base stability of 1 mg/mL cyclovirobuxine D solution. (<b>b<sub>1</sub></b>) Photostability; (<b>b<sub>2</sub></b>) storage stability; (<b>b<sub>3</sub></b>) thermal stability; and (<b>b<sub>4</sub></b>) acid-base stability of 1 mg/mL Cy-Nio. Among these, the statistical time for photostability and storage stability was 10 d, and the statistical time for thermal stability and acid-base stability was 1 h.</p>
Full article ">Figure 6
<p>(<b>a</b>) Cumulative skin penetration; and (<b>b</b>) skin penetration percentage (%) of Cy-Nio under the condition of V (ethanol: saline) = 3:7 as the transdermal receiving liquid for different chemical penetration enhancers. (<b>c</b>) Pharmacokinetic curves of cyclovirobuxine D at different time points in the oral group, skin application group, and combination group of cyclovirobuxine D niosomes and azone. (<b>d</b>) The pH of cyclovirobuxine D niosome was detected using pH test strips. ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>At 24 h after treatment with cyclovirobuxine D and Cy-Nio, the residual amount of cyclovirobuxine D in various organs of mice was detected, including: (<b>a</b>) heart; (<b>b</b>) liver; (<b>c</b>) spleen; (<b>d</b>) lungs; (<b>e</b>) brain; (<b>f</b>) blood; (<b>g</b>) left kidney; (<b>h</b>) right kidney; (<b>i</b>) large intestine; (<b>j</b>) small intestine; (<b>k</b>) stomach; (<b>l</b>) feces; and (<b>m</b>) oral method. (<b>n</b>) Cy-Nio; and (<b>o</b>) Cy-Nio were combined with azone to determine the percentage of residual cyclovirobuxine D in each organ. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8
<p>The effects of different concentrations of: (<b>a</b>) cyclovirobuxine D; (<b>b</b>) Cy-Nio; (<b>c</b>) azonel (<b>d</b>) borneol; (<b>e</b>) oleic acid; (<b>f</b>) retinol; and (<b>g</b>) glabridin on cell proliferation of RSC96, HACAT, and BJ cells. <sup>ns</sup> &gt; 0.05, * <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, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>Skin irritation intensity of intact and scratched back skin of New Zealand white rabbit treated with Cy-Nio (contain azone) for 1 h and 3 d. Among them, (<b>a<sub>1</sub></b>,<b>a<sub>2</sub></b>) are the intact blank group skin, (<b>b<sub>1</sub></b>,<b>b<sub>1</sub></b>) are the scratched blank group skin, (<b>c<sub>1</sub></b>,<b>c<sub>2</sub></b>) are the intact Cy-Nio group skin, and (<b>d<sub>1</sub></b>,<b>d<sub>2</sub></b>) are the scratched Cy-Nio group skin.</p>
Full article ">
20 pages, 3731 KiB  
Review
Advances in Research of Hydrogel Microneedle-Based Delivery Systems for Disease Treatment
by Juan Cao, Bo Wu, Ping Yuan, Yeqi Liu and Cheng Hu
Pharmaceutics 2024, 16(12), 1571; https://doi.org/10.3390/pharmaceutics16121571 - 9 Dec 2024
Viewed by 561
Abstract
Microneedles (MNs), composed of multiple micron-scale needle-like structures attached to a base, offer a minimally invasive approach for transdermal drug delivery by penetrating the stratum corneum and delivering therapeutic agents directly to the epidermis or dermis. Hydrogel microneedles (HMNs) stand out among various [...] Read more.
Microneedles (MNs), composed of multiple micron-scale needle-like structures attached to a base, offer a minimally invasive approach for transdermal drug delivery by penetrating the stratum corneum and delivering therapeutic agents directly to the epidermis or dermis. Hydrogel microneedles (HMNs) stand out among various MN types due to their excellent biocompatibility, high drug-loading capacity, and tunable drug-release properties. This review systematically examines the matrix materials and fabrication methods of HMN systems, highlighting advancements in natural and synthetic polymers, and explores their applications in treating conditions such as wound healing, hair loss, cardiovascular diseases, and cancer. Furthermore, the potential of HMNs for disease diagnostics is discussed. The review identifies key challenges, including limited mechanical strength, drug-loading efficiency, and lack of standardization, while proposing strategies to overcome these issues. With the integration of intelligent design and enhanced control over drug dosage and safety, HMNs are poised to revolutionize transdermal drug delivery and expand their applications in personalized medicine. Full article
(This article belongs to the Special Issue Multifunctional Hydrogels for Controlled Drug Delivery)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The structure and drug-release mechanisms of these different MN types.</p>
Full article ">Figure 2
<p>The preparation methods and therapeutic applications of HMNs.</p>
Full article ">Figure 3
<p>Schematics of the adhesive microneedle patch containing gemcitabine-loaded GelMA for pancreatic cancer treatment. Copyright permission from Fu et al. [<a href="#B33-pharmaceutics-16-01571" class="html-bibr">33</a>], <span class="html-italic">Chemical Engineering Journal</span>, 2022.</p>
Full article ">Figure 4
<p>Schematic overview of the design and function of the detachable polymer microneedles. Copyright permission from Ye et al. [<a href="#B39-pharmaceutics-16-01571" class="html-bibr">39</a>], <span class="html-italic">ACS Materials Letters</span>, 2023.</p>
Full article ">Figure 5
<p>Schematic overview of the design and function of the sc-TDDS. (<b>a</b>) The sc-TDDS consists of PENG and MNP; (<b>b</b>) MNP includes PLA-Au MNs and PLA-Au-PPy MNs loaded with Dex; (<b>c</b>) The mechanism of loading and releasing Dex with PPy. Copyright permission from Yang et al. [<a href="#B45-pharmaceutics-16-01571" class="html-bibr">45</a>], <span class="html-italic">Advanced Functional Materials</span>, 2021.</p>
Full article ">Figure 6
<p>Schematic illustration of the fabrication process of self-healing PMN patch. Copyright permission from Ling et al. [<a href="#B56-pharmaceutics-16-01571" class="html-bibr">56</a>], <span class="html-italic">Small</span>, 2023.</p>
Full article ">Figure 7
<p>The manufacturing process of HepMi-PCL. Copyright permission from Liu et al. [<a href="#B14-pharmaceutics-16-01571" class="html-bibr">14</a>], <span class="html-italic">Advanced Functional Materials</span>, 2024.</p>
Full article ">Figure 8
<p>Schematic of microneedle (MN) patch (denoted as Bi/Vp@MN) for scarless wound healing. Copyright permission from Wei et al. [<a href="#B71-pharmaceutics-16-01571" class="html-bibr">71</a>], <span class="html-italic">Chemical Engineering Journal</span>, 2023.</p>
Full article ">Figure 9
<p>Schematic of a codelivery NLPs-integrated MNs (KK-NLPs@MNs) delivery platform for the treatment of AGA. Copyright permission from Zhang et al. [<a href="#B77-pharmaceutics-16-01571" class="html-bibr">77</a>], <span class="html-italic">ACS Applied Materials and Interfaces</span>, 2024.</p>
Full article ">Figure 10
<p>A conductive microneedle patch and miR-30d nano delivery system are integrated to alleviate myocardial ischemia-reperfusion injury (I/RI). Copyright permission from Chen et al. [<a href="#B89-pharmaceutics-16-01571" class="html-bibr">89</a>], <span class="html-italic">ACS Nano</span>. 2024.</p>
Full article ">Figure 11
<p>Schematic outline of the fabrication and application of CryoMNs-R.r-Au. (<b>a</b>) CryoMNs-R.r-Au utilizes cryomicro needles for transdermal delivery of nanogold-engineered <span class="html-italic">Rhodospirillum rubrum</span>, aiming to remodel the tumor microenvironment through optical biotherapy. (<b>b</b>) Under laser irradiation, R.r-Au can effectively enhance lactate consumption and hydrogen production via photochemical transformation by transferring electrons into the photosynthetic system of R. <span class="html-italic">rubrum</span>, thereby improving antitumor immune activation. Copyright permission from Shi et al. [<a href="#B105-pharmaceutics-16-01571" class="html-bibr">105</a>], <span class="html-italic">Bioactive Materials</span>, 2024.</p>
Full article ">
13 pages, 4506 KiB  
Article
Prescription Design of Sinomenine Gel Based on Molecular Dynamics Simulations
by Yiran Kang, Wei Shen, Shili Pan, Haiying Lian, Xuehui Ding, Jingying Li, Jiaoyue Zhu, Lin Wang and Wei Xu
Int. J. Mol. Sci. 2024, 25(23), 12863; https://doi.org/10.3390/ijms252312863 - 29 Nov 2024
Viewed by 481
Abstract
Sinomenine (SIN) is a drug for the treatment of rheumatoid arthritis, most of which is administered orally, but it is prone to adverse gastrointestinal effects. Gel can overcome the gastrointestinal adverse effects caused by oral administration. In this paper, a multiscale computational pharmaceutics [...] Read more.
Sinomenine (SIN) is a drug for the treatment of rheumatoid arthritis, most of which is administered orally, but it is prone to adverse gastrointestinal effects. Gel can overcome the gastrointestinal adverse effects caused by oral administration. In this paper, a multiscale computational pharmaceutics strategy was developed to guide the systematic study of formulation factors of a SIN gel and, further, to guide the formulation design. A molecular dynamics (MD) simulations method was utilized to successfully screen the optimal prescription of SIN gel and to elucidate the molecular mechanisms affecting the quality of SIN gel. The optimal prescription was 3.0% of SIN, 1.0% of Carbopol (Cp), 30% of Ethanol (Eth), 5.0% of Glycerine (Gly) and 10.0% of Menthol (Men). The influence mechanism can be explained by the combination of multiple parameters, such as the microstructure diagram, the radius of gyration (Rg) and the radial distribution function (RDF). In vitro transdermal studies were carried out using a modified Franz diffusion cell method to evaluate the quality of the screened and reference prescriptions. The results showed that the cumulative penetration and penetration rate of the screening of prescription were better than the reference formulation. Most important of all, the simulation results are in good agreement with the in vitro release experiment, indicating that the strategy has good applicability. This study was able to accurately optimize the formulation and elucidate the molecular mechanism, which would provide a reference for further research on SIN and gel. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Rg variation curves for Cp with different degree of aggregations. (<b>a</b>) n = 15; (<b>b</b>) n = 30; (<b>c</b>) n = 40; (<b>d</b>) n = 50; (<b>e</b>) n = 60; (<b>f</b>) n = 70.</p>
Full article ">Figure 2
<p>Effect of Cp with different degree of aggregations on SIN gel. (<b>a</b>) Rg and RMSD variation curves for Cp with different degree of aggregations; (<b>b</b>) density distribution in the z-axis direction for Cp with different degree of aggregations; (<b>c</b>) diffusion coefficient for Cp with different degree of aggregations; (<b>d</b>) density distribution of solvent water in the z-axis direction for Cp with different degree of aggregations.</p>
Full article ">Figure 3
<p>Effect of different SIN concentrations on the quality of SIN gel. (<b>a</b>) Rg and RMSD variation curves for different SIN concentrations; (<b>b</b>) diffusion coefficient for different SIN concentrations; (<b>c</b>) density profiles in the z-axis direction for different SIN concentrations.</p>
Full article ">Figure 4
<p>Effect of different polymer matrix concentrations on the quality of SIN gel. (<b>a</b>) Rg and RMSD curves variation for different Cp concentrations; (<b>b</b>) diffusion coefficient for different Cp concentrations; (<b>c</b>) density profiles in the z-axis direction for different Cp concentrations.</p>
Full article ">Figure 5
<p>Effect of different Eth concentrations on the quality of SIN gel. (<b>a</b>) Diffusion coefficient for Eth concentrations; (<b>b</b>) RDF values for different Eth concentrations.</p>
Full article ">Figure 6
<p>Effect of different Gly concentrations on the quality of SIN gel. (<b>a</b>) Diffusion coefficient for different Gly concentrations; (<b>b</b>) RDF values for different Gly concentrations.</p>
Full article ">Figure 7
<p>Effect of osmotic promoters on the quality of SIN gel. (<b>a</b>) Conformations of Proy and Lau systems with different concentrations at 0 ns and 200 ns (purple for SIN, blue for lipid bilayer); (<b>b</b>) conformations of Men systems with different concentrations at 0 ns and 200 ns (purple for SIN, green for Men).</p>
Full article ">Figure 8
<p>Traditional experimental validation. (<b>a</b>) Microscopic images at the same magnification (magnification: 4 × 10), in order of SIN and Cp matrix mixture, SIN and CMC mixture, and SIN and CHI mixture; (<b>b</b>) DSC thermogram; (<b>c</b>) in vitro transdermal penetration of the screening prescription and reference formulation.</p>
Full article ">Figure 9
<p>Molecular structure for MD simulations. (<b>a</b>) Molecular structural formulae of each prescriptive factor affecting the quality of the gel; (<b>b</b>) 3D structural diagrams of the molecules that make up the keratin bilayer of the skin: ceramide-neurosphingosine (CER), cholesterol (CHOL) and free fatty acids (FFA).</p>
Full article ">Figure 10
<p>Skin bilayer model. The dark red area is aqueous solvent, and the head group oxygen and nitrogen of CER and FFA chains are shown in red, blue, and the main chains of CER, FFA and CHOL are uniformly shown in light blue.</p>
Full article ">
20 pages, 10346 KiB  
Article
Preparation and In Vitro/In Vivo Characterization of Mixed-Micelles-Loaded Dissolving Microneedles for Sustained Release of Indomethacin
by Baojie Wang, Langkun Liao, Huihui Liang, Jiaxin Chen and Yuqin Qiu
Pharmaceutics 2024, 16(12), 1505; https://doi.org/10.3390/pharmaceutics16121505 - 22 Nov 2024
Viewed by 407
Abstract
Background/Objectives: Indomethacin (IDM) is commonly used to treat chronic inflammatory diseases such as rheumatoid arthritis and osteoarthritis. However, long-term oral IDM treatment can harm the gastrointestinal tract. This study presents a design for encapsulating IDM within mixed micelles (MMs)-loaded dissolving microneedles (DMNs) to [...] Read more.
Background/Objectives: Indomethacin (IDM) is commonly used to treat chronic inflammatory diseases such as rheumatoid arthritis and osteoarthritis. However, long-term oral IDM treatment can harm the gastrointestinal tract. This study presents a design for encapsulating IDM within mixed micelles (MMs)-loaded dissolving microneedles (DMNs) to improve and sustain transdermal drug delivery. Methods: Indomethacin-loaded mixed micelles (IDM-MMs) were prepared from Soluplus® and Poloxamer F127 by means of a thin-film hydration method. The MMs-loaded DMNs were fabricated using a two-step molding method and evaluated for storage stability, insertion ability, in vitro release, in vitro transdermal penetration, and in vivo PK/PD studies. Results: The obtained MMs were stable at 4 °C and 30 °C for 60 days. The in vitro IDM transdermal penetration was remarkably improved by the MMs-loaded DMNs compared to a commercial patch. A pharmacokinetic study demonstrated that the MMs-loaded DMNs had a relative bioavailability of 4.1 in comparison with the commercial patch. Furthermore, the MMs-loaded DMNs showed a significantly shorter lag time than the commercial patch, as well as a more stable plasma concentration than the DMNs without MMs. The therapeutic efficacy of the IDM DMNs was examined in Complete Freund’s Adjuvant-induced arthritis mice. The IDM DMN treatment effectively reduced arthritis severity, resulting in decreased paw swelling, arthritis index, spleen hyperplasia, and serum IL-1β and TNF-α levels. Conclusions: Our findings demonstrated that the novel MMs-loaded DMNs were an effective strategy for sustained IDM release, providing an alternate route of anti-inflammatory drug delivery. Full article
(This article belongs to the Special Issue Microarray Patches for Transdermal Drug Delivery)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic illustration of the fabrication of IDM-MMs-loaded DMNs.</p>
Full article ">Figure 2
<p>TEM image of IDM-MMs (<b>A</b>). TEM image of IDM-MMs (close-up) (<b>B</b>).</p>
Full article ">Figure 3
<p>The morphology of IDM-MMs-loaded DMNs. Photograph of IDM-MMs-loaded DMNs (<b>A</b>); microscope image of the tips of IDM-MMs-loaded DMNs (<b>B</b>); SEM image of IDM-MMs-loaded DMNs (close-up) (<b>C</b>); SEM image of IDM-MMs-loaded DMNs (<b>D</b>).</p>
Full article ">Figure 4
<p>The storage stability of IDM-MMs and IDM-MMs-loaded DMNs. Changes in particle size and PDI of IDM-MMs (<b>A</b>); changes in EE% of IDM-MMs (<b>B</b>); changes in particle size of IDM-MMs redissolved from the IDM-MMs-loaded DMNs (<b>C</b>) (mean ± SD, n = 3) (** <span class="html-italic">p</span> &lt; 0.01 compared with day 0, *** <span class="html-italic">p</span> &lt; 0.001 compared with day 0).</p>
Full article ">Figure 5
<p>The micrographs of different layers of parafilm<sup>®</sup> M after insertion by IDM-MMs-loaded DMNs (<b>A</b>). The first layer (<b>a</b>). The second layer (<b>b</b>). The third layer (<b>c</b>). The fourth layer (<b>d</b>). The fifth layer (<b>e</b>). The sixth layer (<b>f</b>); percentage of holes created in each parafilm layer by the IDM or IDM-MMs-loaded DMNs (mean ± SD, n = 3) (<b>B</b>); H&amp;E-staining image after insertion of IDM-MMs-loaded DMNs into rat skin (<b>C</b>).</p>
Full article ">Figure 5 Cont.
<p>The micrographs of different layers of parafilm<sup>®</sup> M after insertion by IDM-MMs-loaded DMNs (<b>A</b>). The first layer (<b>a</b>). The second layer (<b>b</b>). The third layer (<b>c</b>). The fourth layer (<b>d</b>). The fifth layer (<b>e</b>). The sixth layer (<b>f</b>); percentage of holes created in each parafilm layer by the IDM or IDM-MMs-loaded DMNs (mean ± SD, n = 3) (<b>B</b>); H&amp;E-staining image after insertion of IDM-MMs-loaded DMNs into rat skin (<b>C</b>).</p>
Full article ">Figure 6
<p>In vitro release profile of IDM-MMs (<b>A</b>); in vitro release profiles of IDM DMNs and IDM-MMs-loaded DMNs (<b>B</b>) (mean ± SD, n = 3).</p>
Full article ">Figure 7
<p>In vitro penetration results of IDM across full-thickness porcine skin (<b>A</b>); skin retention of IDM after transdermal penetration (<b>B</b>) (mean ± SD, n = 3. ** <span class="html-italic">p</span> &lt; 0.01 compared with the IDM-MMs-loaded DMNs.</p>
Full article ">Figure 8
<p>The mean plasma concentrations and time profiles of IDM DMNs, IDM MMs-loaded DMNs, and commercial patch (mean ± SD, n = 5).</p>
Full article ">Figure 9
<p>Representative photographs of mouse paw and joint before CFA induction (the picture above) and on day 7 after CFA induction (the picture below) (<b>A</b>); effect of IDM treatment on swelling ratio in AIA mice (<b>B</b>); arthritis index (<b>C</b>); spleen index (<b>D</b>); serum IL-1β level (<b>E</b>); serum TNF-α levels (<b>F</b>) (mean ± SD, n = 6. ** <span class="html-italic">p</span> &lt; 0.01 compared with the AIA model group. *** <span class="html-italic">p</span> &lt; 0.001 compared to the AIA. model group).</p>
Full article ">Figure 9 Cont.
<p>Representative photographs of mouse paw and joint before CFA induction (the picture above) and on day 7 after CFA induction (the picture below) (<b>A</b>); effect of IDM treatment on swelling ratio in AIA mice (<b>B</b>); arthritis index (<b>C</b>); spleen index (<b>D</b>); serum IL-1β level (<b>E</b>); serum TNF-α levels (<b>F</b>) (mean ± SD, n = 6. ** <span class="html-italic">p</span> &lt; 0.01 compared with the AIA model group. *** <span class="html-italic">p</span> &lt; 0.001 compared to the AIA. model group).</p>
Full article ">
18 pages, 1098 KiB  
Review
Liposomes and Ethosomes: Comparative Potential in Enhancing Skin Permeability for Therapeutic and Cosmetic Applications
by Ewelina Musielak and Violetta Krajka-Kuźniak
Cosmetics 2024, 11(6), 191; https://doi.org/10.3390/cosmetics11060191 - 5 Nov 2024
Viewed by 1699
Abstract
Skin diseases are a common health problem affecting millions of people worldwide. Effective treatment often depends on the precise delivery of drugs to the affected areas. One promising approach is currently the transdermal drug delivery system (TDDS), whose significant challenge is the poor [...] Read more.
Skin diseases are a common health problem affecting millions of people worldwide. Effective treatment often depends on the precise delivery of drugs to the affected areas. One promising approach is currently the transdermal drug delivery system (TDDS), whose significant challenge is the poor penetration of many compounds into the skin due to the stratum corneum (SC), which acts as a formidable barrier. To overcome this limitation, nanocarriers have emerged as a highly effective alternative. This review discusses the use of liposomes and ethosomes for transdermal drug delivery. Liposomes are micro- or nanostructures consisting of a lipid bilayer surrounding an aqueous core. They facilitate transdermal drug penetration and may be advantageous for site-specific targeting. Some methods of treating skin diseases involve incorporating drugs such as acyclovir, dithranol, and tretinoin or bioactive compounds such as fluconazole, melanin, glycolic acid, kojic acid, and CoQ10 into nanocarriers. The inability of liposomes to pass through the narrowed intercellular channels of the stratum corneum led to the invention of lipid-based vesicular systems such as ethosomes. They are structurally similar to conventional liposomes, as they are prepared from phospholipids, but they contain a high ethanol concentration. Ethosomes are noninvasive carriers that allow drugs to reach the deep layers of the skin. Examples of commonly used substances and drugs combined with ethosomes in cosmetics include methotrexate, ascorbic acid, vitamin A and E, and colchicine. A significant development in this area is the use of rutin-loaded ethosomes. Encapsulating rutin in ethosomes significantly improves its stability and enhances skin penetration, allowing more efficient delivery to deeper skin layers. In cosmetics, rutin–ethosome formulations are used to protect the skin from oxidative stress, reduce redness, and improve capillary strength, making it a valuable formulation in anti-aging and anti-inflammatory products. The results of the first clinical trial of the acyclovir–ethosome formulation confirm that ethosomes require further investigation. The work provides an update on recent advances in pharmaceutical and cosmetic applications, mentioning the essential points of commercially available formulations, clinical trials, and patents in the recent past. Full article
Show Figures

Figure 1

Figure 1
<p>Diagram of the structure of human skin [<a href="#B11-cosmetics-11-00191" class="html-bibr">11</a>].</p>
Full article ">Figure 2
<p>Schematic representation of the structure of the liposome and ethosome.</p>
Full article ">
29 pages, 3207 KiB  
Review
Skin Structure, Physiology, and Pathology in Topical and Transdermal Drug Delivery
by Sofia Brito, Moonki Baek and Bum-Ho Bin
Pharmaceutics 2024, 16(11), 1403; https://doi.org/10.3390/pharmaceutics16111403 - 31 Oct 2024
Viewed by 2080
Abstract
Several industries are increasingly focused on enhancing the delivery of active ingredients through the skin to optimize therapeutic outcomes. By facilitating the penetration of active ingredients through the skin barrier, these enhancers can significantly improve the efficacy of various formulations, ranging from skincare [...] Read more.
Several industries are increasingly focused on enhancing the delivery of active ingredients through the skin to optimize therapeutic outcomes. By facilitating the penetration of active ingredients through the skin barrier, these enhancers can significantly improve the efficacy of various formulations, ranging from skincare products to therapeutic agents targeting systemic circulation. As the understanding of skin physiology and the mechanisms of drug absorption deepen, these industries are adopting permeation enhancers more widely, ultimately leading to better patient outcomes and expanded treatment options. However, the structure and physiological function of the skin can vary according to different factors, such as the area of the body and between individuals. These variations, along with external environmental exposures, aging and pathological conditions, introduce complexities that must be carefully considered when designing effective delivery systems. Considering the intricacies of skin structure and physiology, tailoring systems to account for regional differences, individual variability, and changes induced by environmental factors or disease is critical to optimizing therapeutic outcomes. This review discusses the features of skin structure, physiology, and pathologies, as well as the application of permeation enhancers in these contexts. Furthermore, it addresses the use of animal skin models in transdermal delivery and dermatological studies, along with the latest developments in this field. Full article
(This article belongs to the Special Issue Transdermal Delivery: Challenges and Opportunities)
Show Figures

Figure 1

Figure 1
<p>Scheme depicting the basic skin structure with particular focus on the epidermis (Not labeled in figure: purple cell (top) = Langerhans cell; brown cell (bottom left) = Melanocyte; yellow cell (bottom right) = Merkel cell).</p>
Full article ">Figure 2
<p>Scheme depicting the routes of drug diffusion through the skin. Each pathway is represented by a purple arrow below each label.</p>
Full article ">Figure 3
<p>Dermal anchoring structures in facial skin (Adapted from Ezure et al., 2015 [<a href="#B65-pharmaceutics-16-01403" class="html-bibr">65</a>]).</p>
Full article ">Figure 4
<p>Scheme depicting the stratum lucidum typically present in the skin of hands and feet.</p>
Full article ">Figure 5
<p>Graphical comparison of the various characteristics of young and aged skin.</p>
Full article ">Figure 6
<p>Graphical comparison of the characteristics of various skin conditions.</p>
Full article ">Figure 7
<p>Graphical comparison of the characteristics of various pigmentary skin conditions. The asterisk indicates a representative illustration of pendulous melanocytes.</p>
Full article ">Figure 8
<p>Histological comparison of young, aged, and psoriatic human and mouse skin. Scale bar = 50 μm. Human forearm skin: Young, 24 years old; Aged: 74 years old; Psoriasis, 34 years old. Mouse dorsal skin: Young, 10 weeks old; Aged, 80 weeks old; Psoriatic model, 10 weeks old. Patient biopsies were obtained after approval by the Institutional Review Board of the Dongguk University Ilsan Hospital and were conducted according to the Declaration of Helsinki principles. Mouse experiments were approved by the Institutional Animal Care and Use Committee of Semyung University (IACUC; Approval No. smecae 20-04-01) and were performed according to the animal testing guidelines. Mouse psoriatic model was induced with 10% hydrogen peroxide treatment for 24 h.</p>
Full article ">
35 pages, 8900 KiB  
Review
Nanocarrier-Based Transdermal Drug Delivery Systems for Dermatological Therapy
by Yunxiang Kang, Sunxin Zhang, Guoqi Wang, Ziwei Yan, Guyuan Wu, Lu Tang and Wei Wang
Pharmaceutics 2024, 16(11), 1384; https://doi.org/10.3390/pharmaceutics16111384 - 28 Oct 2024
Viewed by 1490
Abstract
Dermatoses are among the most prevalent non-fatal conditions worldwide. Given this context, it is imperative to introduce safe and effective dermatological treatments to address the diverse needs and concerns of individuals. Transdermal delivery technology offers a promising alternative compared to traditional administration methods [...] Read more.
Dermatoses are among the most prevalent non-fatal conditions worldwide. Given this context, it is imperative to introduce safe and effective dermatological treatments to address the diverse needs and concerns of individuals. Transdermal delivery technology offers a promising alternative compared to traditional administration methods such as oral or injection routes. Therefore, this review focuses on the recent achievements of nanocarrier-based transdermal delivery technology for dermatological therapy, which summarizes diverse delivery strategies to enhance skin penetration using various nanocarriers including vesicular nanocarriers, lipid-based nanocarriers, emulsion-based nanocarriers, and polymeric nanocarrier according to the pathogenesis of common dermatoses. The fundamentals of transdermal delivery including skin physiology structure and routes of penetration are introduced. Moreover, mechanisms to enhance skin penetration due to the utilization of nanocarriers such as skin hydration, system deformability, disruption of the stratum corneum, surface charge, and tunable particle size are outlined as well. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of drug permeation routes across the skin.</p>
Full article ">Figure 2
<p>Schematic demonstration of various nanocarriers.</p>
Full article ">Figure 3
<p>Transdermal drug delivery technologies for dermatoses.</p>
Full article ">Figure 4
<p>Schematic illustration and characterization of nanoparticles for psoriasis treatment. (<b>A</b>) CLSM images of HaCaT cells treated with TCeO<sub>2</sub>-FNL/CeO<sub>2</sub>-FNL at predetermined time points (red: mitochondria; green: TCeO<sub>2</sub>-FNL/CeO<sub>2</sub>-FNL). Reprinted from Ref. [<a href="#B110-pharmaceutics-16-01384" class="html-bibr">110</a>]. (<b>B</b>) Scheme of the nanoparticle PLN-TPPS2a-TNF siRNA and PCI mechanism. Reprinted with permission from Ref. [<a href="#B111-pharmaceutics-16-01384" class="html-bibr">111</a>]. Copyright 2021 Elsevier. (<b>C</b>) H&amp;E sections and immunohistochemical micrographs of skin of psoriasis mice model treated with HA-MTX-Lipo MN (Scare bar: 50 μm). Reprinted with permission from Ref. [<a href="#B112-pharmaceutics-16-01384" class="html-bibr">112</a>]. Copyright 2024 Elsevier.</p>
Full article ">Figure 5
<p>Schematic illustration and characterization of nanoparticles for vitiligo therapy. (<b>A</b>) Using Fontana–Masson silver staining, the B16F10 cell line’s melanin pigment is visible. Dark black staining of the melanin in various groups is shown by yellow arrows: (<b>A1</b>) Control, (<b>A2</b>) PSR-UDL, (<b>A3</b>) RSV-UDL, and (<b>A4</b>) PSR + RSV-UDL (Scare bar: 1000 μm). Reprinted with permission from Ref. [<a href="#B117-pharmaceutics-16-01384" class="html-bibr">117</a>]. Copyright 2017 Elsevier. (<b>B</b>) Schematic illustration of 5-MOP/8-MOP sol and Fontana–Masson silver staining to visualize melanin pigment in B16F10 cells where melanin is stained dark black in different groups: (<b>B1</b>) PSR Sol, (<b>B2</b>) 5-MOP Sol, (<b>B3</b>) 8-MOP Sol, (<b>B4</b>) PSR UDL, (<b>B5</b>) 5-MOP UDL, and (<b>B6</b>) 8-MOP UDL. Reprinted with permission from Ref. [<a href="#B119-pharmaceutics-16-01384" class="html-bibr">119</a>]. Copyright 2019 Elsevier. (<b>C</b>) The uptake, transport, and accumulation of MelNPs in HEka cells were analyzed using transmission electron microscopy and confocal optical microscopy. Reprinted from Ref. [<a href="#B120-pharmaceutics-16-01384" class="html-bibr">120</a>].</p>
Full article ">Figure 6
<p>(<b>A</b>) Representative images of the wound healing trace after the treatment of OBPG&amp;MP NPs (Scar bar: 5 mm). Reprinted with permission from Ref. [<a href="#B121-pharmaceutics-16-01384" class="html-bibr">121</a>]. Copyright 2024 Wiley. (<b>B</b>) Histological evaluation of the regenerated skin via Masson staining treated with OBPG%MP NPs (Scar bar: 2 and 200 µm). Reprinted with permission from Ref. [<a href="#B121-pharmaceutics-16-01384" class="html-bibr">121</a>]. Copyright 2024 Wiley. (<b>C</b>) Representative images of immunohistochemistry staining of cytokeratin of wound tissues after the treatment of KGF-2/ aFGF-NPs MNs on Day 3 and Day 14. Reprinted with permission from Ref. [<a href="#B124-pharmaceutics-16-01384" class="html-bibr">124</a>]. Copyright 2024 Wiley.</p>
Full article ">Figure 7
<p>(<b>A</b>) Preparation and in vivo antitumor evaluation of Vem-TD-Lip. Reprinted with permission from Ref. [<a href="#B130-pharmaceutics-16-01384" class="html-bibr">130</a>]. Copyright 2018 Elsevier. (<b>B</b>) Design principle of the NIR light-activatable dissolving MN system (MN-pB/I) for multimodal theragnostic application in melanoma. Reprinted with permission from Ref. [<a href="#B131-pharmaceutics-16-01384" class="html-bibr">131</a>]. Copyright 2023 Springer Nature. (<b>C</b>) Scheme and in vivo antitumor evaluation of Cu-PDA-based synergistic comprehensive treatment for melanoma tumor model in Balb/c mice. Reprinted with permission from Ref. [<a href="#B132-pharmaceutics-16-01384" class="html-bibr">132</a>]. Copyright 2021 Elsevier.</p>
Full article ">
34 pages, 2913 KiB  
Review
The Design Features, Quality by Design Approach, Characterization, Therapeutic Applications, and Clinical Considerations of Transdermal Drug Delivery Systems—A Comprehensive Review
by Durgaramani Sivadasan and Osama A. Madkhali
Pharmaceuticals 2024, 17(10), 1346; https://doi.org/10.3390/ph17101346 - 9 Oct 2024
Viewed by 3098
Abstract
Transdermal drug delivery systems (TDDSs) are designed to administer a consistent and effective dose of an active pharmaceutical ingredient (API) through the patient’s skin. These pharmaceutical preparations are self-contained, discrete dosage forms designed to be placed topically on intact skin to release the [...] Read more.
Transdermal drug delivery systems (TDDSs) are designed to administer a consistent and effective dose of an active pharmaceutical ingredient (API) through the patient’s skin. These pharmaceutical preparations are self-contained, discrete dosage forms designed to be placed topically on intact skin to release the active component at a controlled rate by penetrating the skin barriers. The API provides the continuous and prolonged administration of a substance at a consistent rate. TDDSs, or transdermal drug delivery systems, have gained significant attention as a non-invasive method of administering APIs to vulnerable patient populations, such as pediatric and geriatric patients. This approach is considered easy to administer and helps overcome the bioavailability issues associated with conventional drug delivery, which can be hindered by poor absorption and metabolism. A TDDS has various advantages compared to conventional methods of drug administration. It is less intrusive, more patient-friendly, and can circumvent first pass metabolism, as well as the corrosive acidic environment of the stomach, that happens when drugs are taken orally. Various approaches have been developed to enhance the transdermal permeability of different medicinal compounds. Recent improvements in TDDSs have enabled the accurate administration of APIs to their target sites by enhancing their penetration through the stratum corneum (SC), hence boosting the bioavailability of drugs throughout the body. Popular physical penetration augmentation methods covered in this review article include thermophoresis, iontophoresis, magnetophoresis, sonophoresis, needle-free injections, and microneedles. This review seeks to provide a concise overview of several methods employed in the production of TDDSs, as well as their evaluation, therapeutic uses, clinical considerations, and the current advancements intended to enhance the transdermal administration of drugs. These advancements have resulted in the development of intelligent, biodegradable, and highly efficient TDDSs. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Layers of the skin—a schematic sectional view. (<b>b</b>) The epidermis layer of the human skin. Adapted from [<a href="#B20-pharmaceuticals-17-01346" class="html-bibr">20</a>], Springer, 2021.</p>
Full article ">Figure 2
<p>Schematic representation of the relationship between the rate of drug release (Rr) from a transdermal drug delivery system (TDDS) and the rate of drug absorption (Ra) by the skin. <math display="inline"><semantics> <mrow> <msub> <mi>C</mi> <mi>r</mi> </msub> </mrow> </semantics></math> is the drug concentration in the body. Adapted from [<a href="#B35-pharmaceuticals-17-01346" class="html-bibr">35</a>], CBS Publishers, 2005.</p>
Full article ">Figure 3
<p>Approaches used in the development of TDDSs.</p>
Full article ">Figure 4
<p>Enhancement methods of TDDSs.</p>
Full article ">Figure 5
<p>Ishikawa (fishbone) diagram to assess critical quality attributes (CQAs) of TDDSs.</p>
Full article ">
20 pages, 8504 KiB  
Article
Ionic Liquid-Based Grapeseed Oil Emulsion for Enhanced Anti-Wrinkle Treatment
by Bo Yang, Xu Zhang, Liguo Zhang, Jinjin Wu, Wei Wang, Qiaomei Huang, Zhenyuan Wang, Jichuan Zhang, Tongjie Xu, Chengyu Wu and Jiaheng Zhang
Pharmaceuticals 2024, 17(10), 1273; https://doi.org/10.3390/ph17101273 - 26 Sep 2024
Viewed by 806
Abstract
Objectives: To address the poor efficacy and percutaneous penetration of grape seed oil, ionic liquids and nanotechnology were combined to prepare a grape seed oil emulsion. Methods: A novel Menthol-CoQ10 ionic liquid and ionic liquid based grapeseed oil emulsion were prepared and confirmed. [...] Read more.
Objectives: To address the poor efficacy and percutaneous penetration of grape seed oil, ionic liquids and nanotechnology were combined to prepare a grape seed oil emulsion. Methods: A novel Menthol-CoQ10 ionic liquid and ionic liquid based grapeseed oil emulsion were prepared and confirmed. Results: The average size of the grapeseed oil emulsion was 218 nm, and its zeta potential was −33.5 mV. The ionic liquid-based grape seed oil emulsion exhibited a transdermal penetration effect 4.63-fold higher than that of ordinary grape seed oil emulsion. Ionic liquid also displayed enhanced efficiency both in vitro and in vivo. It significantly inhibited the production of DPPH free radicals and tyrosinase, inhibited melanin and matrix metalloproteinase-1 (MMP-1) produced by cells, and promoted type I collagen expression in fibroblasts. After 28 days of continuous use, the grapeseed oil emulsion improved the water content of the stratum corneum and the rate of transepidermal water loss, enhanced the firmness and elasticity of the skin, and significantly improved the total number and length of under-eye lines, tail lines, nasolabial folds, and marionette lines on the face. Conclusions: Menthol-CoQ10 ionic liquid is a promising functional excipient for both transdermal delivery increase and efficient enhancement. Ionic liquid and nanotechnology for grape seed oil facial mask displayed significantly enhanced efficacy and permeability. Full article
(This article belongs to the Special Issue Pharmaceutical Excipients in Formulation Design and Drug Delivery)
Show Figures

Figure 1

Figure 1
<p>Molecular simulation of bonding in Menthol-CoQ10 ionic liquids, weak interaction (<b>a</b>) and electrostatic potential (<b>b</b>).</p>
Full article ">Figure 2
<p>Image of Methol-CoQ10 ionic liquid (<b>a</b>), DSC curve (<b>b</b>), and FTIR spectrum (<b>c</b>) of the ionic liquid Menthol-CoQ10.</p>
Full article ">Figure 3
<p>GSO-ILE appearance (<b>a</b>), particle size (<b>b</b>), zeta potential (<b>c</b>), and TEM view (<b>d</b>).</p>
Full article ">Figure 4
<p>Penetration of Coenzyme Q10 in the 3D cell skin model of GSO-ILE and emulsion of grapeseed oil (<b>left</b>) and fluorescence distribution (<b>right</b>).</p>
Full article ">Figure 5
<p>(<b>a</b>) DPPH free radical scavenging rate and (<b>b</b>) tyrosinase inhibition rate of GSO-ILE.</p>
Full article ">Figure 6
<p>Survival rate of GSO-ILE on various of cell types, human melanoma cells (<b>a</b>), mouse melanoma cells (<b>b</b>), keratinocytes (<b>c</b>), and fibroblasts (<b>d</b>).</p>
Full article ">Figure 7
<p>Inhibition of melanin production by GSO-ILE in melanoma cells (B16). Statistical analysis was performed using the <span class="html-italic">t</span>-test. Compared to the blank group, <span class="html-italic">p</span>-value &lt; 0.01 is expressed as “**”.</p>
Full article ">Figure 8
<p>Collagen type I production (<b>a</b>) and inhibition of MMP-1 (<b>b</b>) by GSO-ILE in fibroblasts. Statistical analysis was performed using the <span class="html-italic">t</span>-test. NC was denoted by “#” compared to the blank group, with a <span class="html-italic">p</span>-value of &lt;0.05 as “#”. The significance of the PC and sample groups was denoted by “**”—<span class="html-italic">p</span>-value of &lt;0.01, compared to the blank group.</p>
Full article ">Figure 9
<p>Clinical trials of GSO-ILE masks and grapeseed oil masks improved outcomes after 28 days of use. All statistics were expressed as mean ± SD (<span class="html-italic">n</span> = 30). Statistical analysis was performed using the <span class="html-italic">t</span>-test. “#” means no statistical difference, <span class="html-italic">p</span> value is greater than or equal to 0.05, <span class="html-italic">p</span> value less than 0.05 means significant difference (“*” means 0.01 ≤ <span class="html-italic">p</span> &lt; 0.05, “**” means 0.001 ≤ <span class="html-italic">p</span> &lt; 0.01, “***” means <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 10
<p>Results of clinical trials using the GSO-ILE mask at different times: cuticle water content (<b>a</b>), percutaneous water loss (<b>b</b>), elasticity R2 (<b>c</b>), and compactness F4 (<b>d</b>). All statistical data are represented as mean ± SD (<span class="html-italic">n</span> = 30). Statistical analysis was performed using the <span class="html-italic">t</span>-test. “n.s.” means no statistical difference, <span class="html-italic">p</span> value is greater than or equal to 0.05, <span class="html-italic">p</span> value less than 0.05 means significant difference (“*” means 0.01 ≤ <span class="html-italic">p</span> &lt; 0.05, “**” means 0.001 ≤ <span class="html-italic">p</span> &lt; 0.01, “***” means <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 11
<p>Clinical trial results of facial wrinkles at different times: eye (<b>a</b>), tail (<b>b</b>), decree (<b>c</b>), puppet (<b>d</b>). All statistical data are represented as mean ± SD (<span class="html-italic">n</span> = 30). Statistical analysis was performed using the <span class="html-italic">t</span>-test. “n.s.” means no statistical difference, <span class="html-italic">p</span> value is greater than or equal to 0.05, <span class="html-italic">p</span> value less than 0.05 means significant difference (“*” means 0.01 ≤ <span class="html-italic">p</span> &lt; 0.05, “**” means 0.001 ≤ <span class="html-italic">p</span> &lt; 0.01, “***” means <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 12
<p>Representative images of improved facial skin wrinkles after 28 days of using a grapeseed oil microemulsion mask.</p>
Full article ">
37 pages, 5256 KiB  
Review
Emerging Trends in Dissolving-Microneedle Technology for Antimicrobial Skin-Infection Therapies
by Rui Luo, Huihui Xu, Qiaoni Lin, Jiaying Chi, Tingzhi Liu, Bingrui Jin, Jiayu Ou, Zejun Xu, Tingting Peng, Guilan Quan and Chao Lu
Pharmaceutics 2024, 16(9), 1188; https://doi.org/10.3390/pharmaceutics16091188 - 8 Sep 2024
Viewed by 1442
Abstract
Skin and soft-tissue infections require significant consideration because of their prolonged treatment duration and propensity to rapidly progress, resulting in severe complications. The primary challenge in their treatment stems from the involvement of drug-resistant microorganisms that can form impermeable biofilms, as well as [...] Read more.
Skin and soft-tissue infections require significant consideration because of their prolonged treatment duration and propensity to rapidly progress, resulting in severe complications. The primary challenge in their treatment stems from the involvement of drug-resistant microorganisms that can form impermeable biofilms, as well as the possibility of infection extending deep into tissues, thereby complicating drug delivery. Dissolving microneedle patches are an innovative transdermal drug-delivery system that effectively enhances drug penetration through the stratum corneum barrier, thereby increasing drug concentration at the site of infection. They offer highly efficient, safe, and patient-friendly alternatives to conventional topical formulations. This comprehensive review focuses on recent advances and emerging trends in dissolving-microneedle technology for antimicrobial skin-infection therapy. Conventional antibiotic microneedles are compared with those based on emerging antimicrobial agents, such as quorum-sensing inhibitors, antimicrobial peptides, and antimicrobial-matrix materials. The review also highlights the potential of innovative microneedles incorporating chemodynamic, nanoenzyme antimicrobial, photodynamic, and photothermal antibacterial therapies. This review explores the advantages of various antimicrobial therapies and emphasizes the potential of their combined application to improve the efficacy of microneedles. Finally, this review analyzes the druggability of different antimicrobial microneedles and discusses possible future developments. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

Figure 1
<p>Skin structure and comparison of the skin-penetration depths of different drug-delivery systems. Reproduced with permission from [<a href="#B7-pharmaceutics-16-01188" class="html-bibr">7</a>] Copyrights 2020, WILEY.</p>
Full article ">Figure 2
<p>Mechanism of bacterial and biofilm resistance. (<b>A</b>) Molecular mechanisms of antibiotic resistance [<a href="#B25-pharmaceutics-16-01188" class="html-bibr">25</a>]. Reproduced with permission from [<a href="#B25-pharmaceutics-16-01188" class="html-bibr">25</a>] Copyrights 2022, Advanced Drug Delivery Reviews. (<b>B</b>) Mechanisms of antimicrobial resistance of biofilms [<a href="#B26-pharmaceutics-16-01188" class="html-bibr">26</a>]. Reproduced with permission from [<a href="#B26-pharmaceutics-16-01188" class="html-bibr">26</a>] Copyrights 2023, Journal of Controlled Release.</p>
Full article ">Figure 3
<p>Five types of microneedles (solid, coated, hollow, dissolving, and hydrogel) developed to date. Reproduced with permission from [<a href="#B35-pharmaceutics-16-01188" class="html-bibr">35</a>] Copyrights 2022, Materials Today Chemistry.</p>
Full article ">Figure 4
<p>Dissolving microneedles constructed from antimicrobial-matrix materials. (<b>A</b>) Schematic illustration depicted the formation mechanism of physically crosslinked networks and amorphous antibiotics in the DOX-loaded EPL microneedles. (<b>B</b>) Synergistic antibacterial mechanism of DOX and EPL. (<b>C</b>) Drug loading of DOX and EPL in the microneedles. (<b>D</b>) Time–Kill curve of DOX and EPL combination at 0.5 × MIC. Reproduced with permission from [<a href="#B100-pharmaceutics-16-01188" class="html-bibr">100</a>] Copyrights 2024, International Journal of Biological Macromolecules.</p>
Full article ">Figure 5
<p>Structures of various AMP-mimicking antimicrobial polymers. (<b>A</b>) Structures of HDP-mimicking amino acid polymers. Reproduced with permission from [<a href="#B120-pharmaceutics-16-01188" class="html-bibr">120</a>] Copyrights 2023, Progress in Polymer Science. (<b>B</b>) Structures of polymethacrylamide derivatives. Reproduced with permission from [<a href="#B109-pharmaceutics-16-01188" class="html-bibr">109</a>] Copyrights 2014, Biomaterials. (<b>C</b>) Structures of polymethacrylate derivatives. Reproduced with permission [<a href="#B110-pharmaceutics-16-01188" class="html-bibr">110</a>] from Copyrights 2009, Chemistry-A European Journal. (<b>D</b>) Structures of polymaleimides derivatives. (<b>E</b>) Structures of polycarbonate derivatives. (<b>F</b>) Structures of peptidopolysaccharide derivatives. (<b>D</b>–<b>F</b>) Reproduced with permission from [<a href="#B113-pharmaceutics-16-01188" class="html-bibr">113</a>] Copyrights 2018, Biomacromolecules. (<b>G</b>) Structures of poly(isobutylene-<span class="html-italic">alt</span>-maleic anhydride) derivatives. Reproduced with permission from [<a href="#B118-pharmaceutics-16-01188" class="html-bibr">118</a>] Copyrights 2013, Chemical Communications.</p>
Full article ">Figure 6
<p>Dissolving microneedles based on CDT for the treatment of skin infection. (<b>A</b>) Optical and SEM images of the PFG/M microneedle patch. (<b>B</b>) Therapeutic mechanism of PFG/M microneedle for promoting healing of infected wounds. (<b>C</b>) Simulation illustrating the wound-healing process. (<b>D</b>) Masson staining and (<b>E</b>) immunohistochemistry staining of the wound bed. Reproduced with permission from [<a href="#B127-pharmaceutics-16-01188" class="html-bibr">127</a>] Copyrights 2023, Advanced Science.</p>
Full article ">Figure 7
<p>Dissolving microneedles containing nanozyme antibacterial therapy for skin infection. (<b>A</b>) Multienzyme-like nanozymes ocular microneedles for Keratitis Treatment. (<b>B</b>) Multienzyme-like activities of MnO<sub>X</sub>/GDY. (<b>C</b>) Representative photographs of fungal-infected rabbit corneas undergoing various treatment interventions. Reproduced with permission from [<a href="#B151-pharmaceutics-16-01188" class="html-bibr">151</a>] Copyrights 2024, Small.</p>
Full article ">Figure 8
<p>Dissolving microneedles incorporating PDT for the treatment of skin infections. (<b>A</b>) Diagram depicting the TSMN for improved penetration and eradication of bacterial biofilms. (<b>B</b>) Fabrication and characterization of the TSMN. (<b>C</b>) Successful penetration of the <span class="html-italic">S. aureus</span> biofilm by TSMN. (<b>D</b>) Confocal laser scanning microscopy images depicting representative <span class="html-italic">S. aureus</span> biofilms following various treatments. Reproduced with permission from [<a href="#B162-pharmaceutics-16-01188" class="html-bibr">162</a>] Copyrights 2023, ACS Applied Materials &amp; Interfaces.</p>
Full article ">Figure 9
<p>Dissolving microneedles based on PTT for the treatment of skin infection. (<b>A</b>) Illustration depicting the antibacterial mechanism of photothermal microneedles. (<b>B</b>) Thermal images of the microneedles. (<b>C</b>) Assessment of the combined effect of heat and vancomycin on inhibiting the growth of methicillin-resistant <span class="html-italic">S. aureus</span> in vitro. Reproduced with permission from [<a href="#B166-pharmaceutics-16-01188" class="html-bibr">166</a>] Copyrights 2023, Chemical Engineering Journal.</p>
Full article ">Figure 10
<p>Dissolving microneedles based on combination therapy involving multiple drugs for the treatment of skin infection. (<b>A</b>) Combined membranolytic–photothermal–photocatalytic therapy of 4K10@V<sub>2</sub>C microneedle. Reproduced with permission from [<a href="#B168-pharmaceutics-16-01188" class="html-bibr">168</a>] Copyrights 2023, Chemical Engineering Journal. (<b>B</b>) Combination of nanoenzymes and AMPs. Reproduced with permission from [<a href="#B138-pharmaceutics-16-01188" class="html-bibr">138</a>] Copyrights 2023, Nano Letters. (<b>C</b>) Combination of PTT and antibiotics. Reproduced with permission from [<a href="#B172-pharmaceutics-16-01188" class="html-bibr">172</a>] Copyrights 2022, Chemical Engineering Journal.</p>
Full article ">Scheme 1
<p>Schematic of various dissolving microneedles for bacterial-infection treatment.</p>
Full article ">
14 pages, 1684 KiB  
Review
Bioactives in Nutricosmetics: A Focus on Caffeine from Tea to Coffee
by Cristina Blanco-Llamero, Hugo F. Macário, Beatriz N. Guedes, Faezeh Fathi, Maria Beatriz P. P. Oliveira and Eliana B. Souto
Cosmetics 2024, 11(5), 149; https://doi.org/10.3390/cosmetics11050149 - 28 Aug 2024
Viewed by 2232
Abstract
Known for its stimulating effects on the nervous and cardiovascular systems, caffeine has proven remarkable versatile properties. It can be used in a wide range of different products, from anti-aging cosmetics to the pharmaceutical treatment of hair loss. Caffeine is known for its [...] Read more.
Known for its stimulating effects on the nervous and cardiovascular systems, caffeine has proven remarkable versatile properties. It can be used in a wide range of different products, from anti-aging cosmetics to the pharmaceutical treatment of hair loss. Caffeine is known for its antioxidant properties and is commonly found in moisturising creams recommended as anti-aging or anti-cellulite and also for the treatment of different skin disorders, including androgenic alopecia. This bioactive is also described to be able to enhance the sunscreen scattering effect of well-known ultraviolet (UV) blockers. One of the major challenges remains its penetration capacity into deeper skin layers, which may be achieved by the use of nanosized delivery systems, yet without the risk of transdermal delivery. In this review, we discuss the nutraceutical value of caffeine in cosmetic products, so-called nutricosmetics, which grants this bioactive several advantages in several formulations, in comparison to other potential bioactives of nutricosmetic value. Furthermore, the disclosed effects of bioactives commonly found in coffee, tea, and their by-products are reviewed and discussed. The discussion concludes by highlighting the significant benefits of caffeine in the treatment of skin disorders and its potential to enhance and promote skin health. Full article
(This article belongs to the Special Issue Bioactive Compounds From Natural Resources Against Skin Aging)
Show Figures

Figure 1

Figure 1
<p>Illustration of the interplay of fields (foods and beverages, pharmaceutics, and cosmetics) in which caffeine and other bioactive ingredients show a high market value.</p>
Full article ">Figure 2
<p>Industrial steps encountered in the process of transforming coffee fruit into coffee beverages, during which several other bioactives and by-products of nutricosmetic value besides caffeine can be obtained.</p>
Full article ">Figure 3
<p>Bibliometric map obtained by VOSviewer software version 1.6.16 [<a href="#B20-cosmetics-11-00149" class="html-bibr">20</a>,<a href="#B21-cosmetics-11-00149" class="html-bibr">21</a>], from articles indexed in the Scopus database published since 2024, searching for “Caffeine” and “Cosmetics” and “Nanoparticles” as keywords limited to the “Pharmacology, Toxicology, and Pharmaceutics” category, retrieved on 25 June 2024.</p>
Full article ">
17 pages, 2423 KiB  
Article
Cell Penetrating Peptide Enhances the Aphidicidal Activity of Spider Venom-Derived Neurotoxin
by Wenxian Wu, Abid Ali, Jinbo Shen, Maozhi Ren, Yi Cai and Limei He
Toxins 2024, 16(8), 358; https://doi.org/10.3390/toxins16080358 - 14 Aug 2024
Viewed by 864
Abstract
HxTx-Hv1h, a neurotoxic peptide derived from spider venom, has been developed for use in commercial biopesticide formulations. Cell Penetrating Peptides (CPPs) are short peptides that facilitate the translocation of various biomolecules across cellular membranes. Here, we evaluated the aphidicidal efficacy of a conjugated [...] Read more.
HxTx-Hv1h, a neurotoxic peptide derived from spider venom, has been developed for use in commercial biopesticide formulations. Cell Penetrating Peptides (CPPs) are short peptides that facilitate the translocation of various biomolecules across cellular membranes. Here, we evaluated the aphidicidal efficacy of a conjugated peptide, HxTx-Hv1h/CPP-1838, created by fusing HxTx-Hv1h with CPP-1838. Additionally, we aimed to establish a robust recombinant expression system for HxTx-Hv1h/CPP-1838. We successfully achieved the secretory production of HxTx-Hv1h, its fusion with Galanthus nivalis agglutinin (GNA) forming HxTx-Hv1h/GNA and HxTx-Hv1h/CPP-1838 in yeast. Purified HxTx-Hv1h exhibited contact toxicity against Megoura crassicauda, with a 48 h median lethal concentration (LC50) of 860.5 μg/mL. Fusion with GNA or CPP-1838 significantly enhanced its aphidicidal potency, reducing the LC50 to 683.5 μg/mL and 465.2 μg/mL, respectively. The aphidicidal efficacy was further improved with the addition of surfactant, decreasing the LC50 of HxTx-Hv1h/CPP-1838 to 66.7 μg/mL—over four times lower compared to HxTx-Hv1h alone. Furthermore, we engineered HxTx-Hv1h/CPP-1838 multi-copy expression vectors utilizing the BglBrick assembly method and achieved high-level recombinant production in laboratory-scale fermentation. This study is the first to document a CPP fusion strategy that enhances the transdermal aphidicidal activity of a natural toxin like HxTx-Hv1h and opens up the possibility of exploring the recombinant production of HxTx-Hv1h/CPP-1838 for potential applications. Full article
Show Figures

Figure 1

Figure 1
<p>Expression and detection of recombinant proteins. (<b>A</b>) Schematic representation of constructs engineered to encode HxTx-Hv1h, HxTx-Hv1h/GNA, and HxTx-Hv1h/CPP-1838 in yeast. The α-factor leader sequence facilitates targeting of the expressed proteins to the yeast secretory pathway, thus enabling their isolation from the fermentation culture supernatant. The His-tag indicates an incorporated hexahistidine motif, which facilitates protein purification via nickel-nitrilotriacetic acid (Ni-NTA) affinity chromatography and permits detection through Western blot analysis. (<b>B</b>) Resolution of purified recombinant proteins on a 15% SDS-PAGE gel, visualized post-staining with Coomassie ultrafast. ‘M’ designates the molecular weight marker; lane 1 contains 5 μg of HxTx-Hv1h; lane 2 contains 7.5 μg of HxTx-Hv1h/GNA; lane 3 features 10 μg of HxTx-Hv1h/CPP-1838. (<b>C</b>) Immunoblot analysis of the recombinant proteins utilizing an anti-His tag antibody. ‘M’ comprises molecular weight standards, with lanes 1–3 corresponding to the samples detailed in (<b>B</b>), loaded with approximately 50 ng of HxTx-Hv1h, 200 ng of HxTx-Hv1h/GNA, and 100 ng of HxTx-Hv1h/CPP-1838, respectively. (<b>D</b>) Western blot detection of the recombinant proteins using an anti-HxTx-Hv1h antibody. Lanes 1–3 follow the pattern established in (<b>B</b>), with loading quantities at approximately 50 ng for HxTx-Hv1h and HxTx-Hv1h/CPP-1838, while 100 ng of HxTx-Hv1h/GNA is introduced in lane 2.</p>
Full article ">Figure 2
<p>Evaluation of the contact toxicity of recombinant proteins against aphids. (<b>A</b>–<b>C</b>) Dose–response curves depicting the contact toxicity of HxTx-Hv1h, HxTx-Hv1h/GNA, and HxTx-Hv1h/CPP-1838 on aphid populations, in the absence of the surfactant Silwet L-77. The curves illustrate the percentage of mortality across a spectrum of concentrations, highlighting the aphidicidal potency of the recombinant proteins without surfactant assistance. (<b>D</b>–<b>F</b>). Dose–response curves showcasing the augmented contact toxicity of HxTx-Hv1h, HxTx-Hv1h/GNA, and HxTx-Hv1h/CPP-1838 when used in conjunction with the surfactant Silwet L-77. These curves demonstrate the synergy between the bioactive proteins and the surfactant, culminating in enhanced lethality towards the aphids.</p>
Full article ">Figure 3
<p>Immunoblot analysis of protein extracts from insects. ‘C’ denotes the lanes loaded with extracts from control aphids that did not undergo exposure to any recombinant proteins. Above each lane, the identity of the specific recombinant protein that the aphids contacted is labeled. The protein samples were derived from groups of 20 aphids harvested 8 h following contact with a 200 μM concentration of each respective recombinant protein. ‘S1’, ‘S2’, and ‘S3’ correspond to the protein standards for HxTx-Hv1h, HxTx-Hv1h/GNA, and HxTx-Hv1h/CPP-1838, each at a concentration of 100 ng.</p>
Full article ">Figure 4
<p>Confirmation of plasmids harboring single and multiple HxTx-Hv1h/CPP-1838 expression cassettes and copy number assessment in transformed yeast clones. (<b>A</b>) plasmid shift analysis. ‘M’ denotes the molecular weight standards. Lanes 1C through 12C contain plasmids pGAPZαA-HxTx-Hv1h/CPP-1838 with varying copy counts, notationally indicated as 1C (one copy), 2C (two copies), 4C (four copies), 6C (six copies), 8C (eight copies), 10C (ten copies), and 12C (twelve copies), respectively. (<b>B</b>) Analysis of plasmid conformation subsequent to double restriction digestion with <span class="html-italic">Bgl</span> II and <span class="html-italic">Bam</span>H I enzymes, with each lane corresponding to the respective plasmid construct detailed in (<b>A</b>). (<b>C</b>) qPCR evaluation of yeast clones transformed with plasmids encompassing single and assorted copy numbers of the HxTx-Hv1h/CPP-1838 expression cassette. The RQ (Relative Quantification) values signify the comparative determination of copy numbers of the HxTx-Hv1h/CPP-1838 construct in relation to a reference housekeeping gene.</p>
Full article ">Figure 5
<p>Evaluation of protein expression levels in <span class="html-italic">P. pastoris</span> transformants with multiple gene copies. (<b>A</b>) Depiction of cultured supernatant protein profiles from <span class="html-italic">P. pastoris</span> transformants incorporating varying copy numbers of the HxTx-Hv1h/CPP-1838 expression cassettes. ‘C’ represents the expression cassette featuring a GAP promoter, the HxTx-Hv1h/CPP-1838 gene, and an AOX1 terminator. ‘1C’ denotes the yeast transformant harboring a single expression cassette. ‘2C’, ‘4C’, ‘6C’, ‘8C’, ‘10C’, and ‘12C’ indicate transformants encompassing two, four, six, eight, ten, and twelve repeats of the expression cassette, respectively. (<b>B</b>) Chronological analysis of HxTx-Hv1h/CPP-1838 protein expression in yeast transformants across diverse fermentation time frames. ‘M’ represents the molecular weight marker.</p>
Full article ">
12 pages, 2398 KiB  
Article
Mesoporous Silica-Based Membranes in Transdermal Drug Delivery: The Role of Drug Loss in the Skin
by Frank Baumann, Theresa Paul, Susann Ossmann, Dirk Enke and Achim Aigner
Pharmaceutics 2024, 16(8), 995; https://doi.org/10.3390/pharmaceutics16080995 - 26 Jul 2024
Viewed by 757
Abstract
Compared to other forms of drug administration, the use of Transdermal Drug Delivery Systems (TDDSs) offers significant advantages, including uniform drug release profiles that contribute to lower side effects and higher tolerability, avoidance of direct exposure to the gastrointestinal tract, better patient compliance [...] Read more.
Compared to other forms of drug administration, the use of Transdermal Drug Delivery Systems (TDDSs) offers significant advantages, including uniform drug release profiles that contribute to lower side effects and higher tolerability, avoidance of direct exposure to the gastrointestinal tract, better patient compliance due to their non-invasive means of application and others. Mesoporous silica membranes are of particular interest in this regard, due to their chemical stability and their tunable porous system, with adjustable pore sizes, pore volumes and surface chemistries. While this allows for fine-tuning and, thus, the development of optimized TDDSs with high loading capacities and the desired release profile of a given drug, its systemic availability also relies on skin penetration. In this paper, using a TDDS based on mesoporous silica membranes in Franz cell experiments on porcine skin, we demonstrate surprisingly substantial drug loss during skin penetration. Drug passage through porcine skin was found to be dependent on the age and pre-treatment of the skin. pH and temperature were major determinants of drug recovery rates as well, indicating drug loss in the skin by enzymatic metabolization. Regarding the TDDS, higher loading obtained by SO3H surface modification of the mesoporous silica membranes reduced drug loss. Still, high loss rates in the skin were determined for different drugs, including anastrozole, xylazine and imiquimod. We conclude that, beyond the fine-tuned drug release profiles from the mesoporous silica membrane TDDS, remarkably high drug loss in the skin is a major issue for achieving desired skin penetration and, thus, the systemic availability of drugs. This also poses critical requirements for defining an optimal TDDS based on mesoporous silica membranes. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the mesoporous silica membrane and skin preparation and the Franz cell experiments.</p>
Full article ">Figure 2
<p>Determination of anastrozole permeation in Franz cell experiments. (<b>A</b>) Time-dependent increase in the anastrozole concentration in the acceptor medium upon direct application of the drug onto the carrier membrane (black), or with porcine skin obtained from a regional butcher (red). (<b>B</b>) Application of the drug using a mesoporous silica membrane as a TDDS, leading to a sustained release profile. Again, increase rates of anastrozole drug concentration (black) are further reduced upon drug penetration through skin (red). (<b>C</b>) Skin age-dependent barrier function for drug penetration, as determined by slower increase in anastrozole concentration in the acceptor medium over time when using skin from older pigs.</p>
Full article ">Figure 3
<p>Determination of variables affecting anastrozole drug loss upon skin penetration. (<b>A</b>) Comparison between adult porcine skin obtained from a local butcher (with heat pretreatment) vs. native skin from a piglet or an older pig. OeX: a mesoporous silica membrane acting as a TDDS. (<b>B</b>) Comparison of anastrozole loss over two different experimental durations. (<b>C</b>) Arrhenius plot demonstrating the direct correlation between temperature (32 °C, 37 °C and 42 °C in the Franz cell) and the permeation rate. (<b>D</b>) Temperature-dependency of drug loss upon skin permeation, with highest loss observed at a physiological 37 °C. (<b>E</b>,<b>F</b>) pH-dependence of (<b>E</b>) anastrozole or (<b>F</b>) xylazine drug loss. * indicates statistical significance.</p>
Full article ">Figure 4
<p>Anastrozole drug loss dependence on the properties of the mesoporous silica membrane acting as a TDDS. (<b>A</b>) Comparison of drug loss between an unmodified (OeX) and a chemically modified (SO<sub>3</sub>H) mesoporous silica membrane. Right: scheme of the chemical surface structures. (<b>B</b>) Markedly reduced drug loss during skin penetration upon loading of the chemically modified (SO<sub>3</sub>H) mesoporous silica membrane with 5-fold larger anastrozole amounts (50 mg/mL vs. 10 mg/mL, referring to the concentration of the solution in the initial loading process).</p>
Full article ">Figure 5
<p>Comparison of drug loss between different drugs during skin permeation. Similar drug losses upon application of the TDDS containing the aromatase inhibitor anastrozole, the veterinary drug xylazine used for sedation, anesthesia and muscle relaxation or the chemotherapeutic imiquimod for topical application. Right: chemical structures of the drugs employed in the experiments.</p>
Full article ">
14 pages, 2854 KiB  
Article
The Preparation and Evaluation of a Hydrochloride Hydrogel Patch with an Iontophoresis-Assisted Release of Terbinafine for Transdermal Delivery
by Mengfei Li, Xinghao Chen, Xiangxiang Su and Wenyan Gao
Gels 2024, 10(7), 456; https://doi.org/10.3390/gels10070456 - 12 Jul 2024
Viewed by 1267
Abstract
Background: Terbinafine hydrochloride (TEB) is a broad-spectrum antifungal medication commonly used to treat fungal infections of the skin. This study designed a hydrogel patch assisted by an iontophoresis system to enhance the transdermal permeability of TEB, enabling deeper penetration into the skin layers. [...] Read more.
Background: Terbinafine hydrochloride (TEB) is a broad-spectrum antifungal medication commonly used to treat fungal infections of the skin. This study designed a hydrogel patch assisted by an iontophoresis system to enhance the transdermal permeability of TEB, enabling deeper penetration into the skin layers. Methods: The influences of current intensity, pH levels, and drug concentration on the TEB hydrogel patch’s permeability were explored using an adaptive ion electroosmosis system. The pharmacokinetic profile, facilitated by iontophoresis for transdermal permeation, was analyzed through the application of microdialysis technology. Scanning electron microscopy and transmission electron microscopy were employed to assess the impact of ion electroosmotic systems on skin integrity. Results: The cumulative drug accumulation within 8 h of the TEB hydrogel patches, assisted by iontophoresis, was 2.9 and 7.9 times higher than without iontophoresis assistance and TEB cream in the control group, respectively. TEB hydrogel patches assisted by iontophoresis can significantly increase the permeability of TEB, and the AUC(0–8 h) was 3.4 and 5.4 times higher, while the Cmax was 4.2 and 7.3 times higher than the TEB hydrogel patches without iontophoresis, respectively. This system has no significant impact on deep-layer cells. Conclusions: This system may offer a safe and effective clinical strategy for the local treatment of deep antifungal infections. Full article
(This article belongs to the Special Issue Novel Functional Gels for Biomedical Applications)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of terbinafine hydrochloride (<b>A</b>) and terbinafine hydrochloride hydrogel patch (<b>B</b>).</p>
Full article ">Figure 2
<p>HPLC chromatogram for specificity investigation. (<b>A</b>) Blank mobile phase (methanol–water (0.2% triethylamine and 1% acetic acid) = 7:3). (<b>B</b>) 20% PEG 400-20 mM NaH<sub>2</sub>PO<sub>4</sub>. (<b>C</b>) Reference substance of TEB (6.25 μg/mL, Dissolve in NS). (<b>D</b>) Reference substance of TEB (5.85 μg/mL, Dissolve in 20% PEG 400-20 mM NaH<sub>2</sub>PO<sub>4</sub>). (<b>E</b>) Standard curve of TEB transdermal receiver solution.</p>
Full article ">Figure 3
<p>The effect of different factors influencing the iontophoresis-assisted transdermal permeation of TEB in vitro. (<b>A</b>) Schematic diagram of iontophoresis transdermal delivery in vitro. (<b>B</b>) The effect of current density on the iontophoresis penetration of TEB hydrogel patches (n = 4). (<b>C</b>) The effect of pH on the iontophoresis penetration of TEB hydrogel patches (n = 4). (<b>D</b>) The effect of drug concentration on the iontophoresis penetration of TEB hydrogel patches (n = 4). (<b>E</b>) A comparison of different TEB formulations of percutaneous penetration (n = 4). (<b>F</b>) The skin retention of different TEB formulations. * <span class="html-italic">p</span> &lt; 0.05, as compared with the group of TEB hydrogel patches without iontophoresis; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, as compared with the TEB cream group.</p>
Full article ">Figure 4
<p>Microdialysis recovery rate of TEB in vitro and in vivo, and percutaneous pharmacokinetics. (<b>A</b>) Schematic diagram of microdialysis recovery rate in vitro. (<b>B</b>) Schematic diagram of iontophoresis transdermal delivery and microdialysis in vitro. (<b>C</b>) The effect of different flow rates and TEB concentrations on the recoveries of probes in vitro (retrodialysis, n = 4). (<b>D</b>) The effect of different flow rates and TEB concentrations on the recoveries of probes in vivo (retrodialysis, n = 4). (<b>E</b>) Percutaneous pharmacokinetics of different TEB formulations (n = 6).</p>
Full article ">Figure 5
<p>The effect of iontophoresis on the skin microstructure. (<b>A</b>–<b>D</b>) The ultrastructural damage of the epidermis under a scanning electron microscope (SEM) ((<b>A</b>) skin of normal rats (control); (<b>B</b>) the skin of rats with a TEB hydrogel patch; (<b>C</b>) skin of rats with a TEB hydrogel patch under iontophoresis; (<b>D</b>) skin of rats with the TEB cream); (<b>E</b>–<b>H</b>) deeper damage to skin cells under a transmission electronic microscope (TEM) ((<b>E</b>) skin of normal rats (control); (<b>F</b>) skin of rats with a TEB hydrogel patch; (<b>G</b>) skin of rats with a TEB hydrogel patch under iontophoresis; (<b>H</b>) skin of rats with the TEB cream).</p>
Full article ">Figure 6
<p>The preparation process of a TEB hydrogel patch.</p>
Full article ">
Back to TopTop