[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

Search Results (372)

Search Parameters:
Keywords = drug (nano)formulation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 4546 KiB  
Article
Spanlastic Nano-Vesicles: A Novel Approach to Improve the Dissolution, Bioavailability, and Pharmacokinetic Behavior of Famotidine
by Hend I. Almohamady, Yasmin Mortagi, Shadeed Gad, Sawsan Zaitone, Reem Alshaman, Abdullah Alattar, Fawaz E. Alanazi and Pierre A. Hanna
Pharmaceuticals 2024, 17(12), 1614; https://doi.org/10.3390/ph17121614 - 29 Nov 2024
Viewed by 471
Abstract
Background/Objectives: Drugs exhibiting poor aqueous solubility present a challenge to efficient delivery to the site of action. Spanlastics (a nano, surfactant-based drug delivery system) have emerged as a powerful tool to improve solubility, bioavailability, and delivery to the site of action. This [...] Read more.
Background/Objectives: Drugs exhibiting poor aqueous solubility present a challenge to efficient delivery to the site of action. Spanlastics (a nano, surfactant-based drug delivery system) have emerged as a powerful tool to improve solubility, bioavailability, and delivery to the site of action. This study aimed to better understand factors affecting the physicochemical properties of spanlastics, quantify their effects, and use them to enhance the bioavailability of famotidine (FMT), a model histamine H2 receptor antagonist (BCS class IV). Methods: FMT was incorporated into nano-spanlastics drug delivery system. The ethanol injection method, Box–Behnken design, and mathematical modeling were utilized to fabricate famotidine-loaded nano-spanlastics and optimize the formula. Spanlastics were characterized for their particle size, polydispersity index, zeta potential, entrapment efficiency, drug loading, compatibility of the excipients (using DSC), in vitro drug release, and in vivo pharmacokinetics. Results: Span 60 (the non-ionic surfactant) and tween 60 (the edge activator) gave rise to spanlastics with the best characteristics. The optimal spanlastic formulation exhibited small particle size (<200 nm), appropriate polydispersity index (<0.4), and zeta potential (>−30 mV). The entrapment efficiency and drug loading of the optimum formula assured its suitability for hydrophobic drug entrapment as well as practicability for use. DSC assured the compatibility of all formulation components. The drug release manifested a biphasic release pattern, resulting in a fast onset and sustained effect. Spanlastics also showed enhanced Cmax, AUC0–24, and bioavailability. Conclusions: Spanlastics manifested improved FMT dissolution, drug release characteristics, membrane permeation, and pharmacokinetic behavior. Full article
Show Figures

Figure 1

Figure 1
<p>Linear plots of the effects of individual formulation variables on the PS of SNV.</p>
Full article ">Figure 2
<p>Three-dimensional plots of the combined effects of independent variables (<b>a</b>) S 60 and T 60 concentrations on the PS; (<b>b</b>) S 60 concentration and FMT amount on the PS; (<b>c</b>) T 60 concentration and FMT amount on the PS; (<b>d</b>) S 60 and T 60 concentrations on ζ; (<b>e</b>) S 60 and T 60 concentrations on EE; (<b>f</b>) S 60 and T 60 concentrations on DL; (<b>g</b>) S 60 concentration and FMT amount on DL; (<b>h</b>) T 60 concentration and FMT amount on DL.</p>
Full article ">Figure 3
<p>Linear plots of the effects of individual independent variables on the EE of SNV.</p>
Full article ">Figure 4
<p>Linear plots of the effects of individual independent variables on the DL of SNV.</p>
Full article ">Figure 5
<p>(<b>a</b>) Ramp graphs for the optimized variable levels and predicted responses of the optimized SNV (FOSNV); (<b>b</b>) bar graph for the desirability of the optimization process.</p>
Full article ">Figure 6
<p>DSC thermograms of (<b>a</b>) FMT, (<b>b</b>) S60, (<b>c</b>) plain optimized SNV, and (<b>d</b>) FOSNV.</p>
Full article ">Figure 7
<p>TEM of FOSNV.</p>
Full article ">Figure 8
<p>In vitro drug release profiles of FOSNV and FMT suspension.</p>
Full article ">Figure 9
<p>Plasma drug concentration–time profiles of the FOSNV and FMT suspension after the administration of a sublingual liquid single dose.</p>
Full article ">
19 pages, 4172 KiB  
Article
Exploration of the Topical Nanoemulgel Bearing with Ferulic Acid and Essential Oil for Diabetic Wound Healing
by Urati Anuradha, Valamla Bhavana, Padakanti Sandeep Chary, Nitin Pal Kalia and Neelesh Kumar Mehra
Pathophysiology 2024, 31(4), 680-698; https://doi.org/10.3390/pathophysiology31040049 - 25 Nov 2024
Viewed by 634
Abstract
Aim: To investigate the anti-inflammatory, antioxidant, and diabetic wound healing properties of the novel topical formulation [Ferulic acid-loaded nanoemulgel (DLMGO-G)]. Methods: Ferulic acid nanoemulsion developed with lemongrass oil is investigated in diabetic wound healing. Further nanoemulsion is incorporated into 1% carbopol® 934 [...] Read more.
Aim: To investigate the anti-inflammatory, antioxidant, and diabetic wound healing properties of the novel topical formulation [Ferulic acid-loaded nanoemulgel (DLMGO-G)]. Methods: Ferulic acid nanoemulsion developed with lemongrass oil is investigated in diabetic wound healing. Further nanoemulsion is incorporated into 1% carbopol® 934 to obtain the DLMGO-G. Nanoemulsion was characterized for particle size, and polydispersity index (PDI) was obtained by Malvern Zetasizer (Zetasizer Nano ZS, Malvern, AL, USA), and morphology by TEM (JEM 1400, JOEL, Akishima, Japan). Furthermore, in vitro cell line and in vivo studies were carried out. Results: The developed nanoemulsion showed a globule size of 28.04 ± 0.23 nm and PDI of 0.07 ± 0.01. The morphology of nanoformulations by TEM confirmed the spherical and uniform nature. Further, the nanoformulation in in vitro cell line experiments revealed that the IC50 value was increased by 1.52 times compared to the drug solution. The treatment groups have shown that fibroblast morphologies were spindle-shaped, suggesting that nanoformulation was compatible with the cells and developed normally on nanoformulation. It also reduced ROS with improved internalization more than the control group. The in vitro wound healing model also revealed that nanoformulation had better wound healing activity. In the in vivo diabetic wound studies on male SD rats, the levels of inflammatory markers such as TNF-α, IL-6, IL-22, and IL-1β declined significantly when treated with DLMGO-G. IL-10 levels significantly increased compared to the diseased group, and MMP-9 levels were remarkably decreased compared to the diseased group. Furthermore, histopathological studies showed the regeneration and granulation of tissues. Conclusions: Thus, these findings indicate that FA-loaded nanoemulgel greatly accelerates the healing of wounds in diabetic rats. Full article
Show Figures

Figure 1

Figure 1
<p>The schematic representation of the in vivo study design to assess the formulation on STZ-induced SD rats.</p>
Full article ">Figure 2
<p>Characterization of drug-loaded nanoemulsion (DLMGONE) and drug-loaded nanoemulgel (DLMGO-G); (<b>A</b>) Droplet-size distribution analysis of DLMGONE. (<b>B</b>,<b>C</b>) Transmission electron microscopy (TEM) image of DLMGONE and DLMGO-G with scale 100 nm and 200 nm, respectively.</p>
Full article ">Figure 3
<p>(<b>A</b>) Cytotoxicity study by MTT assay of nanoformulation. Cellular uptake study (<b>B1</b>–<b>B4</b>): (<b>B1</b>) Control group; (<b>B2</b>) An amount of 10 UL of dye solution; (<b>B3</b>) IC50 of dye-loaded nanoformulation; and (<b>B4</b>) High dose of dye-loaded nanoformulation. DCFDA staining (<b>C1</b>–<b>C4</b>): (<b>C1</b>) Control group; (<b>C2</b>) LPS-treated group; (<b>C3</b>) IC50 of nanoformulation; and (<b>C4</b>) High dose of nanoformulation.</p>
Full article ">Figure 4
<p>Acridine orange and ethidium bromide (AO/EB) dual staining of nanoformulation.</p>
Full article ">Figure 5
<p>(<b>A</b>–<b>C</b>): (<b>A</b>) In vitro scratch assay of nanoformulation (IC50 and high dose) for wound closure on L929 cell lines. (<b>B</b>) % wound closure was quantified using ImageJ software, version 1.45. * <span class="html-italic">p</span> &lt; 0.05 vs. Control. (<b>C</b>) Morphological assay of nanoformulation.</p>
Full article ">Figure 6
<p>(<b>A</b>–<b>C</b>): (A) Dorsal view phenotypic photos exhibiting the subjects; (<b>B</b>) The % of wound closure of each group on different days; (<b>C</b>) The wound area of each group on different days.</p>
Full article ">Figure 7
<p>(<b>A1</b>–<b>C6</b>): (<b>A1</b>) Glucose levels by glucometer of different groups on different days; (<b>A2</b>) Bodyweight of each group on different days; (<b>A3</b>) God-pod assay of different groups; (<b>B1</b>,<b>B2</b>) Effect of Nitrite and TBARS levels on STZ intoxication and Novel nanoformulation treatment; (<b>C1</b>–<b>C5</b>) Effect of inflammatory and anti-inflammatory cytokines such as TNF-α, IL-1β, IL-22, IL-6, and IL-10 on STZ intoxication and Novel nanoformulation treatment; (<b>C6</b>) MMP-9 levels of different groups. All values were analyzed by one-way ANOVA followed by Tukey’s multiple comparison test and expressed as mean ± S.E.M and <sup>####</sup> <span class="html-italic">p</span> &lt; 0.0001 vs. Control (C), <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. C, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. C, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. C, **** <span class="html-italic">p</span> &lt; 0.0001 vs. disease (D), *** <span class="html-italic">p</span> &lt; 0.001 vs. D, ** <span class="html-italic">p</span> &lt; 0.01 vs. D, * <span class="html-italic">p</span> &lt; 0.05 vs. D.</p>
Full article ">Figure 8
<p>Rat skin stained with hematoxylin and eosin for histopathological analysis (<b>A1</b>–<b>A5</b>): (<b>A1</b>) Healthy group; (<b>A2</b>) Disease group; (<b>A3</b>) Marketed group; (<b>A4</b>) Blank nanoemulgel (BLMGO-G); and (<b>A5</b>) Drug loaded nanoemulgel (DLMGO-G). PSR Red staining of rat skin (<b>B1</b>–<b>B5</b>): (<b>B1</b>) Healthy group; (<b>B2</b>) Disease group; (<b>B3</b>) Marketed group; (<b>B4</b>) BLMGO-G; and (<b>B5</b>) DLMGO-G. PSR staining of rat skin in polarized light images (<b>C1</b>–<b>C5</b>): (<b>C1</b>) Healthy group; (<b>C2</b>) Disease group; (<b>C3</b>) Marketed group; (<b>C4</b>) BLMGO-G; and (<b>C5</b>) DLMGO-G. Each of the histopathology studies has a scale of 100 µm.</p>
Full article ">
15 pages, 1518 KiB  
Article
Two in One: Size Characterization and Accelerated Short-Term Physical Stability of Dual-Drug Suspensions with Two Acidic Compounds (Indomethacin and Naproxen)
by Nadina Zulbeari, Signe Malig Hansen and René Holm
Pharmaceutics 2024, 16(12), 1495; https://doi.org/10.3390/pharmaceutics16121495 - 21 Nov 2024
Viewed by 382
Abstract
Background/Objectives: Co-delivering dual-drug systems have proven to be effective in, for example, anticancer therapy or HIV prophylaxis due to a higher target selectivity and therapeutic efficacy from compound synergism. However, various challenges regarding physical stability can arise during the formulation definition when multiple [...] Read more.
Background/Objectives: Co-delivering dual-drug systems have proven to be effective in, for example, anticancer therapy or HIV prophylaxis due to a higher target selectivity and therapeutic efficacy from compound synergism. However, various challenges regarding physical stability can arise during the formulation definition when multiple drug compounds are included in the same formulation. In this work, the focus was on aqueous suspensions, which could be applied as long-acting injectable formulations to release the drug compounds over weeks to months after administration. Methods: It was possible to gain insights into dual-drug nano- and microsuspensions containing two acidic compounds (indomethacin and naproxen) prepared by milling with dual centrifugation. Information regarding the physical stability of individual suspensions was subtracted and compared to dual-drug suspensions when prepared with the same milling conditions and stored at elevated temperatures of 40 °C. Results: Distinct particle size profiles after milling were obtained dependent on the stabilizer used in both individual and dual-drug suspensions. Most notably, the combination of indomethacin and naproxen in one formulation resulted in smaller sizes of drug particles compared to individual suspensions under the presence of some stabilizers. The obtained particle size profiles further indicated that at least one of the model compounds needed to be sufficiently stabilized from a stabilizer to obtain physically stable dual-drug suspensions over 28 days when stored at 40 °C. Similarly, the particle size distribution was dependent on the individual distribution of the suspensions, which showed a monomodal distribution could be achieved for dual-drug suspensions when at least one of the individual suspensions demonstrated a monomodal distribution in the presence of the stabilizer alone. Over a 28-day period, the smallest particle size was obtained in dual-drug suspensions stabilized with a combination of polysorbate 85 and poloxamer 338 compared to dual-drug suspensions stabilized with only a single stabilizer during preparation, indicating tendencies towards stabilization synergism from a combination of stabilizers as well as the model compounds. Conclusion: Overall, the study showed insights into the preparation and physical stability of dual-drug suspensions containing indomethacin and naproxen. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

Figure 1
<p>Overview of the solubility of indomethacin and naproxen with increasing concentrations of polysorbate 20 dissolved in either 50 mM citric buffer, pH 3.0 or 50 mM acetate buffer, pH 4.0 (n = 3).</p>
Full article ">Figure 2
<p>Overview of obtained sizes of particles (D<sub>50</sub>-values) (µm) from the stabilizer screening (dissolved in 50 mM citric buffer, pH 3.0) of prepared suspensions containing indomethacin, naproxen, or dual-drug suspensions of both compounds analyzed with either Mie’s theory or the Fraunhofer approximation. The span values are presented as symbols with indomethacin as the ellipse symbol, naproxen as the rectangular symbol, dual-drug suspensions analyzed with Mie’s theory as the diamond symbol, and dual-drug suspensions analyzed with the Fraunhofer approximation as the triangular symbol.</p>
Full article ">Figure 3
<p>Particle size distribution of prepared suspensions containing either indomethacin (black), naproxen (red), or both compounds (green) stabilized with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) poloxamer 188 (dissolved in 50 mM citric buffer, pH 3.0). Particle size distributions were analyzed using Mie’s theory.</p>
Full article ">Figure 4
<p>Particle size distribution of prepared suspensions containing either indomethacin (black), naproxen (red), or both compounds (green) stabilized with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) PVP K16-18 (dissolved in 50 mM citric buffer, pH 3.0). Particle size profiles were analyzed using Mie’s theory (<b>left</b>) or the Fraunhofer approximation (<b>right</b>).</p>
Full article ">Figure 5
<p>Overview of the short-term physical stability of dual-drug suspensions presented as particle sizes (d<sub>50</sub>-value, µm) over 28 days of storage at 40 °C. Suspensions were stabilized with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) (dissolved in 50 mM citric buffer) of either polysorbate 20/PVP K16-18 (PS20 + PVP K16-18), polysorbate 85/poloxamer 338 (PS85 + P338), polysorbate 85/vitamin E TPGS (PS85 + Vit E TPGS), poloxamer 188/PVP K30 (P188 + PVP K30), or PVP K16-18/vitamin E TPGS (PVP K16-18 + vit E TPGS). The span values are presented as symbols with day 0 as the ellipse symbol, day 7 as the rectangular symbol, day 14 as the diamond symbol, and day 28 as the triangular symbol.</p>
Full article ">
29 pages, 7806 KiB  
Article
Formulation and Ex Vivo Evaluation of Ivermectin Within Different Nano-Drug Delivery Vehicles for Transdermal Drug Delivery
by Eunice Maureen Steenekamp, Wilna Liebenberg, Hendrik J. R. Lemmer and Minja Gerber
Pharmaceutics 2024, 16(11), 1466; https://doi.org/10.3390/pharmaceutics16111466 - 18 Nov 2024
Viewed by 1173
Abstract
Background/Objectives: Ivermectin gained widespread attention as the “miracle drug” during the coronavirus disease 2019 (COVID-19) pandemic. Its inclusion in the 21st World Health Organization (WHO) List of Essential Medicines is attributed to its targeted anti-helminthic response, high efficacy, cost-effectiveness and favorable safety profile. [...] Read more.
Background/Objectives: Ivermectin gained widespread attention as the “miracle drug” during the coronavirus disease 2019 (COVID-19) pandemic. Its inclusion in the 21st World Health Organization (WHO) List of Essential Medicines is attributed to its targeted anti-helminthic response, high efficacy, cost-effectiveness and favorable safety profile. Since the late 2000s, this bio-inspired active pharmaceutical ingredient (API) gained renewed interest for its diverse therapeutic capabilities. However, producing ivermectin formulations does remain challenging due to its poor water solubility, resulting in low bioavailability after oral administration. Therefore, the transdermal drug delivery of ivermectin was considered to overcome these challenges, which are observed after oral administration. Methods: Ivermectin was incorporated in a nano-emulsion, nano-emulgel and a colloidal suspension as ivermectin-loaded nanoparticles. The nano-drug delivery vehicles were optimized, characterized and evaluated through in vitro membrane release studies, ex vivo skin diffusion studies and tape-stripping to determine whether ivermectin was successfully released from its vehicle and delivered transdermally and/or topically throughout the skin. This study concluded with cytotoxicity tests using the methyl thiazolyl tetrazolium (MTT) and neutral red (NR) assays on both human immortalized epidermal keratinocytes (HaCaT) and human immortalized dermal fibroblasts (BJ-5ta). Results: Ivermectin was successfully released from each vehicle, delivered transdermally and topically throughout the skin and demonstrated little to no cytotoxicity at concentrations that diffused through the skin. Conclusions: The type of nano-drug delivery vehicle used to incorporate ivermectin influences its delivery both topically and transdermally, highlighting the dynamic equilibrium between the vehicle, the API and the skin. Full article
(This article belongs to the Special Issue Transdermal Delivery: Challenges and Opportunities)
Show Figures

Figure 1

Figure 1
<p>SEM micrographs of <b>NPs</b>.</p>
Full article ">Figure 2
<p>XRPD overlay for: (<b>a</b>) ivermectin, (<b>b</b>) ivermectin-loaded <b>NPs</b>, (<b>c</b>) PCL, (<b>d</b>) PVA and (<b>e</b>) sucrose.</p>
Full article ">Figure 3
<p>(<b>a</b>) Average cumulative amount of ivermectin released per area (μg/cm<sup>2</sup>) over 6 h during the in vitro membrane release studies from all the nano-drug delivery vehicles, and (<b>b</b>) boxplot displaying the average (dashed lines) and median (solid lines) flux (μg/cm<sup>2</sup>.h) of ivermectin for the nano-drug delivery vehicles during the in vitro membrane release studies over a period of 6 h.</p>
Full article ">Figure 4
<p>(<b>a</b>) Average cumulative amount of ivermectin diffused per area (μg/cm<sup>2</sup>) over 12 h during the ex vivo skin diffusion studies from all the nano-drug delivery vehicles, and (<b>b</b>) boxplot displaying the average (dashed lines) and median (solid lines) flux (μg/cm<sup>2</sup>.h) of ivermectin for the nano-drug delivery vehicles during the ex vivo skin diffusion studies over a period of 12 h.</p>
Full article ">Figure 5
<p>Boxplot displaying the average (dashed lines) and median (solid lines) concentration (μg/mL) of ivermectin from the different nano-drug delivery vehicles (<b>NE</b> and <b>NEG</b>) that were delivered in the SCE and ED after each 12 h ex vivo skin diffusion study.</p>
Full article ">Figure 6
<p>%Cell viability of the HaCaT cells after treatment with different concentrations of (<b>a</b>) API, <b>NE</b> and <b>PNE</b>, and (<b>b</b>) <b>CS</b> using the MTT assay, while the NR assay was used for (<b>c</b>) API, <b>NE</b> and <b>PNE</b>, and (<b>d</b>) <b>CS</b>.</p>
Full article ">Figure 7
<p>%Cell viability of the BJ-5sta cells after treatment with different concentrations of (<b>a</b>) API, <b>NE</b> and <b>PNE</b>, and (<b>b</b>) <b>CS</b> using the MTT assay, while the NR assay was used for (<b>c</b>) API, <b>NE</b> and <b>PNE</b>, and (<b>d</b>) <b>CS</b>.</p>
Full article ">
17 pages, 4451 KiB  
Article
Design of Nanocrystalline Suspension of Dutasteride for Intramuscular Prolonged Delivery
by Min Young Jeong, Doe Myung Shin, Min Kyeong Kwon, Ye Bin Shin, Jun Soo Park, In Gyu Yang, Jin Hyuk Myung, Dong Geon Lee, Gi Yeong Lee, Chae Won Park, Ji Won Yeo, Myoung Jin Ho, Yong Seok Choi and Myung Joo Kang
Nanomaterials 2024, 14(22), 1781; https://doi.org/10.3390/nano14221781 - 5 Nov 2024
Viewed by 834
Abstract
The aim of the study is to formulate an injectable nanocrystalline suspension (NS) of dutasteride (DTS), a hydrophobic 5α-reductase inhibitor used to treat benign prostatic hyperplasia and scalp hair loss, for parenteral long-acting delivery. A DTS-loaded NS (DTS-NS, 40 mg/mL DTS) was prepared [...] Read more.
The aim of the study is to formulate an injectable nanocrystalline suspension (NS) of dutasteride (DTS), a hydrophobic 5α-reductase inhibitor used to treat benign prostatic hyperplasia and scalp hair loss, for parenteral long-acting delivery. A DTS-loaded NS (DTS-NS, 40 mg/mL DTS) was prepared using a lab-scale bead-milling technique. The optimized DTS-NS prepared using Tween 80 (0.5% w/v) as a nano-suspending agent, was characterized as follows: rod/rectangular shape; particle size of 324 nm; zeta potential of −11 mV; and decreased drug crystallinity compared with intact drug powder. The DTS-NS exhibited a markedly protracted drug concentration-time profile following intramuscular injection, reaching a maximum concentration after 8.40 days, with an elimination half-life of 9.94 days in rats. Histopathological observations revealed a granulomatous inflammatory response at the injection site 7 days after intramuscular administration, which significantly subsided by day 14 and showed minimal inflammation by day 28. These findings suggest that the nanosuspension system is a promising approach for the sustained release parenteral DTS delivery, with a protracted pharmacokinetic profile and tolerable local inflammation. Full article
Show Figures

Figure 1

Figure 1
<p>Illustration of DTS-NS manufacturing process using dual centrifugation-based bead-milling technique.</p>
Full article ">Figure 2
<p>Influence of suspending agents on (<b>a</b>) the appearance and (<b>b</b>) particle size and homogeneity of DTS-NS formulas prepared using a lab-scale bead-milling technology. Notes: (<b>a</b>) All images were captured on glass slides within 10 min after fabrication (scale bar: 5 mm). (<b>b</b>) The concentrations of DTS and suspending agent were fixed at 40 mg/mL and 5 mg/mL, respectively. Bead milling was carried out at 1500 rpm for 1 h under conditions of −10 °C. Data represent means ± SD (n = 3).</p>
Full article ">Figure 3
<p>Effect of the concentration of Tween 80 on the crystal size of DTS-NSs prepared using a lab-scale bead-milling technology. (<b>a</b>) The appearance of drug suspension prepared with different Tween 80 concentrations. (<b>b</b>) Effect of Tween 80 concentration on mean particle size and homogeneity. Notes: (<b>a</b>) All images were captured on glass slides 10 min after preparation (scale bar: 5 mm). (<b>b</b>) Concentrations of DTS and Tween 80 were set at 40 and 0–10 mg/mL, respectively. Bead milling was performed at 1000–2000 rpm for 1 h under conditions of −10 °C. Data are presented as means ± SD (n = 3).</p>
Full article ">Figure 4
<p>Morphological and physical characteristics of DTS-NS. Scanning electron microscopic (SEM) image of (<b>a</b>) raw material and (<b>b</b>) DTS-NS. (<b>c</b>) Particle size distribution and (<b>d</b>) zeta potential of DTS-NS. Representative (<b>e</b>) XRD patterns and (<b>f</b>) DSC curves of the raw material, aqueous vehicle, and DTS-NS.</p>
Full article ">Figure 4 Cont.
<p>Morphological and physical characteristics of DTS-NS. Scanning electron microscopic (SEM) image of (<b>a</b>) raw material and (<b>b</b>) DTS-NS. (<b>c</b>) Particle size distribution and (<b>d</b>) zeta potential of DTS-NS. Representative (<b>e</b>) XRD patterns and (<b>f</b>) DSC curves of the raw material, aqueous vehicle, and DTS-NS.</p>
Full article ">Figure 5
<p>In vitro dissolution profile of DTS raw material and DTS-NS under sink condition. The sink condition was guaranteed by adding 2% <span class="html-italic">w</span>/<span class="html-italic">v</span> Cremophor EL to 10 mM sodium phosphate buffer (pH 7.4). Note: Data represent mean ± SD (n = 3).</p>
Full article ">Figure 6
<p>Multple reaction monitoring chromatograms of double blank rat plasma (<b>a</b>), 50 ng/mL of dutasteride-spiked rat plasma (<b>b</b>), rat plasma taken five minutes after dutasteride intravenous (IV) administration (<b>c</b>), and rat plasma taken eight hours after dutasteride IV administration (<b>d</b>). DTS and IS stand for dutasteride and finasteride (internal standard), respectively.</p>
Full article ">Figure 6 Cont.
<p>Multple reaction monitoring chromatograms of double blank rat plasma (<b>a</b>), 50 ng/mL of dutasteride-spiked rat plasma (<b>b</b>), rat plasma taken five minutes after dutasteride intravenous (IV) administration (<b>c</b>), and rat plasma taken eight hours after dutasteride IV administration (<b>d</b>). DTS and IS stand for dutasteride and finasteride (internal standard), respectively.</p>
Full article ">Figure 7
<p>The plasma concentration–time profiles of DTS following IM injection (5 mg/kg as DTS) of DTS-NS and IV injection (0.2 mg/kg as DTS) of DTS solution in rats. Note: Data represent mean ± SD (n = 5).</p>
Full article ">Figure 8
<p>Histological examination of gastrocnemius muscles stained with hematoxylin and eosin (H&amp;E) following intramuscular (IM) injection of negative control (normal saline) or DTS-NS in rats. Note: The depot formed by injection of DTS-NS is indicated by (*). Red double arrows indicate fibroblastic bands, and (a) denotes angiogenesis.</p>
Full article ">
14 pages, 2118 KiB  
Article
Safety of Zein Nanoparticles on Human Innate Immunity and Inflammation
by Annunziata Corteggio, Tommaso Heinzl, Diana Boraschi, Silvia Voci, Agnese Gagliardi, Donato Cosco and Paola Italiani
Int. J. Mol. Sci. 2024, 25(21), 11630; https://doi.org/10.3390/ijms252111630 - 29 Oct 2024
Viewed by 741
Abstract
In recent years, natural polymers have attracted great interest for the development of release systems for vaccine formulations and drug delivery. Zein, a hydrophobic proline-rich protein mixture obtained from maize, is one of the most widely used polymers, very promising for applications in [...] Read more.
In recent years, natural polymers have attracted great interest for the development of release systems for vaccine formulations and drug delivery. Zein, a hydrophobic proline-rich protein mixture obtained from maize, is one of the most widely used polymers, very promising for applications in tissue engineering and the parenteral delivery of bioactive agents. Still, we have a limited understanding of the interaction between zein particles and the human immune system, in particular innate immunity/inflammation, which is the first line of defense of our body. Assessing the immune safety of nanoparticles is of central importance for ensuring that nano-formulations for medical use do not cause adverse effects on human health. Here, we evaluated the capacity of zein nanoparticles to induce/modulate the innate/inflammatory response, the development of innate memory, and the macrophage polarization by using reliable in vitro systems based on human primary monocytes and monocyte-derived macrophages. We observed that zein nanoparticles do not influence any of these aspects of the innate immune/inflammatory response, suggesting its safety and its potential efficiency as a nanocarrier for drug or antigen delivery. Full article
(This article belongs to the Special Issue Interaction of Nanomaterials with the Immune System: 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Mean sizes, (<b>B</b>) polydispersity index, and zeta potential of surfactant-free and SD-stabilized zein NPs (2 mg protein/mL water). The analysis was performed in MilliQ water (dilution 1:50). The conductivity of samples was ~0.003 mS/cm.</p>
Full article ">Figure 2
<p>Turbiscan stability index (TSI) profile of surfactant-free and SD-stabilized zein NPs (2 mg protein/mL water). (<b>A</b>): 25 °C; (<b>B</b>): 37 °C.</p>
Full article ">Figure 3
<p>Stability of zein NPs in human serum. Zein NPs (2 mg/mL) were incubated in 70% heat-inactivated human AB serum at 37 °C for increasing times. Both naked NPs and surfactant-stabilized NPs were used. Data are the mean ± sd of triplicate evaluations.</p>
Full article ">Figure 4
<p>Primary innate immune response of human monocytes to zein NPs, LPS, or their mixture. Human blood monocytes were exposed in culture to medium alone or containing zein NPs (100 ng/mL), LPS (1 ng/mL), or zein NPs + LPS. The levels of inflammatory (TNFα, IL-6; panels (<b>A</b>,<b>B</b>)) and anti-inflammatory cytokines (IL-10, IL-1Ra; panels (<b>C</b>,<b>D</b>)) were measured in the 24 h supernatants via ELISA. The columns represent the average value + sd from 6 individual donors. The individual values are indicated with different symbols. Statistical significance: *** <span class="html-italic">p</span> &lt; 0.001 and ** <span class="html-italic">p</span> &lt; 0.01 in the comparisons between medium vs. LPS and NPs + LPS. All other comparisons (medium vs. NPs, LPS vs. NPs + LPS) are not significant.</p>
Full article ">Figure 5
<p>Secondary “memory” response of human monocytes primed with LPS, zein NPs, or their mixture. After a primary exposure to medium alone, LPS (1 ng/mL), zein NPs, or NPs admixed with LPS (see <a href="#ijms-25-11630-f002" class="html-fig">Figure 2</a>), cells were washed and rested in culture for 7 days to allow extinction of the primary activation, then challenged for 24 h with medium or with LPS (5 ng/mL). Controls (CTR columns) include cells primed with medium, NPs, LPS, or LPS + NPs, which were all at baseline after the challenge with medium alone, thereby confirming the complete extinction of the primary activation. Inflammatory (TNFα, IL-6; panels (<b>A</b>,<b>B</b>)) and anti-inflammatory cytokines (IL-10, IL-1Ra; panels (<b>C</b>,<b>D</b>)) were measured in the 24 h supernatants via ELISA. The columns represent the average value + sd from 6 individual donors. The individual values are indicated with different symbols. Statistical significance: * <span class="html-italic">p</span> &lt; 0.05 unchallenged controls CTR vs. medium-primed LPS-challenged (medium) for IL-6; ** <span class="html-italic">p</span> &lt; 0.01 medium-primed LPS-challenged (medium) vs. LPS-primed LPS-challenged (LPS) and vs. Zein+LPS-primed LPS-challenged for TNFα; *** <span class="html-italic">p</span> &lt; 0.001 unchallenged controls CTR vs. medium-primed LPS-challenged (medium) for TNFα. Other comparisons (CTR vs. medium for IL-1Ra, medium vs. LPS for IL-10 and IL-1Ra, medium vs. Zein, LPS vs. Zein + LPS) are not significant.</p>
Full article ">Figure 6
<p>In vitro uptake of rhodamine-labeled zein (red) in monocyte-derived macrophages. Cell nuclei are stained blue with Hoechst 33258. Representative images for 0.5, 2, 6, and 24 h time points are shown. Bar 5 μm.</p>
Full article ">Figure 7
<p>Effect of zein NPs on human macrophage polarization. Monocyte-derived macrophages were either left untreated (naïve, 24 h in culture medium alone—MΦ) or polarized toward the M1 or the M2 functional phenotypes (by 24 h exposure to LPS + IFN-γ for M1, and IL-10 for M2) in the absence or in the presence of 100 ng/mL zein NPs. The production of cytokines and other inflammation-related factors was measured via ELISA in the 24 h supernatants: (<b>A</b>), TNFα; (<b>B</b>), IL-6; (<b>C</b>), IL-1β; (<b>D</b>), IL-8; (<b>E</b>), IL-12p40; (<b>F</b>), IL-1Ra; (<b>G</b>), sIL-1R2. Panel (<b>H</b>) reports the calculated free active IL-1β, i.e., the ratio between free IL-1β (the fraction of the cytokine not bound by its inhibitory receptor sIL-1R2) and the receptor antagonist IL-1Ra multiplied by 1000. The columns represent the average value + sd from 3 individual donors. Individual values are shown by different symbols. Statistical significance: ** <span class="html-italic">p</span> &lt; 0.01 MΦ + Zein vs. M1 + Zein for IL-6; *** <span class="html-italic">p</span> &lt; 0.001 MΦ vs. M1 for TNFα and IL-6. MΦ + Zein vs. M1 + Zein for TNFα; **** <span class="html-italic">p</span> &lt; 0.0001 MΦ vs. M1 or MΦ + Zein vs. M1+Zein for IL-8 and IL-1β. Other comparisons (M1 or M2 vs. M1 + NP or M2 + NP, MΦ vs. M2) are not significant.</p>
Full article ">
21 pages, 1094 KiB  
Review
Curcumin Administration Routes in Breast Cancer Treatment
by Bianca Mayo, Silvana Penroz, Keila Torres and Layla Simón
Int. J. Mol. Sci. 2024, 25(21), 11492; https://doi.org/10.3390/ijms252111492 - 26 Oct 2024
Cited by 1 | Viewed by 1820
Abstract
Breast cancer is a public health concern worldwide, characterized by increasing incidence and mortality rates, requiring novel and effective therapeutic strategies. Curcumin is a bioactive compound extracted from turmeric with several pharmacological activities. Curcumin is a multifaceted anticancer agent through mechanisms including the [...] Read more.
Breast cancer is a public health concern worldwide, characterized by increasing incidence and mortality rates, requiring novel and effective therapeutic strategies. Curcumin is a bioactive compound extracted from turmeric with several pharmacological activities. Curcumin is a multifaceted anticancer agent through mechanisms including the modulation of signaling pathways, inhibition of cell proliferation, induction of apoptosis, and production of reactive oxygen species. However, the poor water solubility and bioavailability of curcumin create important barriers in its clinical application. This review elaborates on the therapeutic potential of curcumin in breast cancer treatment, focusing on the efficacy of different administration routes and synergistic effects with other therapeutic agents. The intravenous administration of curcumin-loaded nanoparticles significantly improves bioavailability and therapeutic outcomes compared to oral routes. Innovative formulations, such as nano-emulsifying drug delivery systems, have shown promise in enhancing oral bioavailability. While intravenous delivery ensures higher bioavailability and direct action on tumor cells, it is more invasive and expensive than oral administration. Advancing research on curcumin in breast cancer treatment is essential for improving therapeutic outcomes and enhancing the quality of life of patients. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Curcumin engagement in key biological processes to inhibit breast cancer development.</p>
Full article ">Figure 2
<p>Comparative efficacy of oral, intravenous, transdermal, intraperitoneal, and intratumoral curcumin administration routes in breast cancer therapy.</p>
Full article ">
16 pages, 2988 KiB  
Article
Customizable Self-Microemulsifying Rectal Suppositories by Semisolid Extrusion 3D Printing
by Hye Jin Park and Dong Wuk Kim
Pharmaceutics 2024, 16(11), 1359; https://doi.org/10.3390/pharmaceutics16111359 - 24 Oct 2024
Viewed by 768
Abstract
Objectives: This study aims to create an innovative self-microemulsifying drug delivery system (SMEDDS) suppository for ibuprofen (IBU) using semisolid extrusion (SSE) three-dimensional (3D) printing technology. Methods: Based on solubility studies and the ability to form a transparent microemulsion upon dilution, a [...] Read more.
Objectives: This study aims to create an innovative self-microemulsifying drug delivery system (SMEDDS) suppository for ibuprofen (IBU) using semisolid extrusion (SSE) three-dimensional (3D) printing technology. Methods: Based on solubility studies and the ability to form a transparent microemulsion upon dilution, a selected oil, surfactant, and co-surfactant were utilized to prepare SMEDDS-3DPS containing IBU. The optimal formulation consisted of 10% Triacetin, 80% Gelucire 48/16, and 10% Tetraethylene glycol. SSE 3D printing was employed to create three different-sized suppositories with varying drug contents. These suppositories were assessed for their physicochemical properties, content uniformity, and dissolution profiles. Results: The prepared mixture exhibited suitable physical properties for printing, with nano-sized emulsion droplets providing a large surface area for improved drug absorption in the rectum. Characterization techniques such as differential scanning calorimetry, powder X-ray diffraction, and Fourier transform infrared spectroscopy indicated that IBU was present in the formulation in an amorphous state. Additionally, in vitro dissolution tests demonstrated that SMEDDS-3DPS had a significantly higher initial dissolution rate compared with IBU powder. Conclusions: This research suggests that SMEDDS-3DPS, as a rectal IBU dosage form, can enhance the rectal bioavailability of IBU. It demonstrates the versatility of 3D printing as a novel manufacturing method for lipid-based suppositories and highlights the simplicity and adaptability of SSE 3D printing technology in producing customized suppositories tailored to individual patient needs, surpassing traditional methods. Full article
(This article belongs to the Special Issue 3D Printing of Drug Delivery Systems)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Solubility of IBU in aqueous solution, oils (<b>A</b>), and surfactants (<b>B</b>) (n = 3). * <span class="html-italic">p</span> &lt; 0.05 when compared to all other samples. <sup>#</sup> <span class="html-italic">p</span> &gt; 0.05 when compared to each other.</p>
Full article ">Figure 2
<p>Pseudoternary phase diagram of the oil (Triacetin), surfactant (Gelucire 48/16), and co-surfactant (Tetraethylene glycol).</p>
Full article ">Figure 3
<p>Scanning electron micrograph of IBU (<b>A</b>) and transmission electron micrograph of the SMEDDS formulation (<b>B</b>).</p>
Full article ">Figure 4
<p>DSC (<b>A</b>), PXRD (<b>B</b>), and FTIR (<b>C</b>) images of samples.</p>
Full article ">Figure 5
<p>Picture of IBU-3DPS printed in three different sizes (from left to right: 50 mg, 100 mg, and 200 mg) (<b>A</b>) and different sections of IBU-3DPS (<b>B</b>).</p>
Full article ">Figure 6
<p>Dissolution profile of IBU powder and 3DPS (50 mg, 100 mg, and 200 mg) in pH 7.2 buffer (n = 6).</p>
Full article ">
24 pages, 1540 KiB  
Review
Research Progress on Peptide Drugs for Type 2 Diabetes and the Possibility of Oral Administration
by Xinxin Yang, Ruiting Lin, Changzhuo Feng, Qiyuan Kang, Peng Yu, Yongzhi Deng and Ye Jin
Pharmaceutics 2024, 16(11), 1353; https://doi.org/10.3390/pharmaceutics16111353 - 23 Oct 2024
Viewed by 888
Abstract
Diabetes is a global disease that can lead to a range of complications. Currently, the treatment of type 2 diabetes focuses on oral hypoglycemic drugs and insulin analogues. Studies have shown that drugs such as oral metformin are useful in the treatment of [...] Read more.
Diabetes is a global disease that can lead to a range of complications. Currently, the treatment of type 2 diabetes focuses on oral hypoglycemic drugs and insulin analogues. Studies have shown that drugs such as oral metformin are useful in the treatment of diabetes but can limit the liver’s ability to release sugar. The development of glucose-lowering peptides has provided new options for the treatment of type 2 diabetes. Peptide drugs have low oral utilization due to their easy degradation, short half-life, and difficulty passing through the intestinal mucosa. Therefore, improving the oral utilization of peptide drugs remains an urgent problem. This paper reviews the research progress of peptide drugs in the treatment of diabetes mellitus and proposes that different types of nano-formulation carriers, such as liposomes, self-emulsifying drug delivery systems, and polymer particles, should be combined with peptide drugs for oral administration to improve their absorption in the gastrointestinal tract. Full article
(This article belongs to the Special Issue Advances in Oral Administration)
Show Figures

Figure 1

Figure 1
<p>The mechanism/target of the combination of peptide drugs and nano-preparations on diabetic complications.</p>
Full article ">Figure 2
<p>Glucose stimulates GLP-1 secretion from L-cells: Glucose closes ATP-sensitive K<sup>+</sup> <sub>ATP</sub> channels and opens Ca<sup>2+</sup> channel (VDCC) to induce membrane depolarization, and Ca<sup>2+</sup> inward flow triggers vesicular cytotoxicity and GLP-1 secretion.</p>
Full article ">Figure 3
<p>Schematic representation of the transport mechanisms.</p>
Full article ">
14 pages, 3431 KiB  
Article
Concanavalin a Grafted Nanoemulsions for Nasal Delivery: Preliminary Studies with Fluorescently Labelled Formulations
by Merve Mışraklı, Sebastiano Antonio Rizzo, Valentina Bordano, Annalisa Bozza, Luca Ferraris, Elisabetta Marini, Elisabetta Muntoni, Maria Teresa Capucchio, Anna Scomparin and Luigi Battaglia
Materials 2024, 17(20), 4959; https://doi.org/10.3390/ma17204959 - 11 Oct 2024
Viewed by 2993
Abstract
Nasal delivery is a non-invasive strategy for effective drug delivery. Nevertheless, in order to promote drug uptake by the nasal mucosa, it is fundamental to increase its residence time in the administration site. To this aim, nano-sized drug delivery systems are widely exploited. [...] Read more.
Nasal delivery is a non-invasive strategy for effective drug delivery. Nevertheless, in order to promote drug uptake by the nasal mucosa, it is fundamental to increase its residence time in the administration site. To this aim, nano-sized drug delivery systems are widely exploited. Within this context, the commercially available nanoemulsion for parenteral nutrition is a biocompatible, safe and clinically approved vehicle for drug delivery. Furthermore, the nanodroplet surface can be modified via a well-established protocol to graft Concavalin A, a lectin capable of improving the mucosal adhesion, by binding to the α-mannose and α-glucose residues of the mucosal glycocalyx. The obtained targeted formulation is able to induce haemagglutination, as opposite to non-modified nanoemulsion. Furthermore, the ConA grafting maintains the physicochemical properties of the nanodroplets (size~230 nm, Z < −35 mV) and does not interfere with the loading of the Rose Bengal fluorescent probe. Fluorescently labelled ConA grafted nanodroplets showed enhanced permeation and accumulation in ex vivo bovine nasal mucosa. This study is a proof of concept that Concanavalin A can be used to decorate the surface of nanodroplets, acting as a permeation promoter. Full article
(This article belongs to the Section Biomaterials)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>HPLC of free Concanavalin A (ConA): (<b>A</b>) 280 nm detection; (<b>B</b>) 220 nm detection. Peak retention times: peak 1: 10.6 min; peak 2: 11.6 min; peak 3: 12.2 min.</p>
Full article ">Figure 2
<p>Agglutination assay for free Concanavalin A (ConA). (<b>A</b>) % agglutination in spectrophotometry; (<b>B</b>) visual observation; (<b>C</b>) optical microscopy. Reported scale bar: 2 μm.</p>
Full article ">Figure 3
<p>Optical microscopy of nanoemulsions. (<b>A</b>) Blank IL—normal light; (<b>B</b>) ConA grafted IL—normal light; (<b>C</b>) Rose Bengal labelled ConA grafted IL—normal light; (<b>D</b>) Rose Bengal labelled ConA grafted IL—fluorescence. Reported scale bar: 2 μm. The fluorescence image was captured with the rhodamine filter (λ<sub>exc</sub> = 515; λ<sub>em</sub> = 560, N2.1 filter cube, Leica Microsystems, Wetzlar, Germany). Abbreviations: ConA: Concanavalin A; IL: 20% Lipofundin<sup>®</sup> MCT.</p>
Full article ">Figure 4
<p>Agglutination assay for IL grafted ConA: (<b>A</b>) visual observation; (<b>B</b>) optical microscopy. Condition 1: IL + 2% RBC; condition 2: ConA grafted IL + 2% RBC. Reported scale bar: 2 μm. Abbreviations: ConA: Concanavalin A; IL: 20% Lipofundin<sup>®</sup> MCT; RBC: red blood cells.</p>
Full article ">Figure 5
<p>Nanoemulsion coalescence in the presence of 2% starch mixture by optical microscopy. (<b>A</b>): ConA grafted IL; (<b>B</b>): IL. (<b>C</b>): ConA grafted IL with 2% starch mixture: (<b>D</b>): IL with 2% starch mixture. Reported scale bar: 2 μm. Abbreviations: ConA: Concanavalin A; IL: 20% Lipofundin<sup>®</sup> MCT.</p>
Full article ">Figure 6
<p>Permeation of Rose Bengal (Rose B) as labelled nanoemulsions (<b>a</b>,<b>b</b>,<b>d</b>,<b>e</b>) or as a free drug (<b>c</b>,<b>f</b>) through bovine nasal mucosa and its accumulation (<b>g</b>,<b>h</b>). As the receiving phase it was used either PBS (<b>a</b>,<b>d</b>) or 1% Pullulan (Pul) (<b>b</b>,<b>e</b>); in (<b>c</b>,<b>f</b>) panels PBS and 1% Pullulan are compared. Statistical analysis: ** <span class="html-italic">p</span> &lt; 0.01 Permeation: ConA grafted Rose B IL vs. Rose B IL. # &lt; 0.05 Accumulation: ConA grafted Rose B IL vs. Rose B IL. ### &lt; 0.0005 Accumulation: Pul ConA grafted Rose B IL vs. Pul Rose B IL.</p>
Full article ">
15 pages, 4985 KiB  
Article
Optimization and Synthesis of Nano-Niosomes for Encapsulation of Triacontanol by Box–Behnken Design
by Alfredo Amaury Bautista Solano, Gloria Dávila-Ortiz, María de Jesús Perea-Flores and Alma Leticia Martínez-Ayala
Molecules 2024, 29(18), 4421; https://doi.org/10.3390/molecules29184421 - 18 Sep 2024
Viewed by 1046
Abstract
Triacontanol is a long-chain primary alcohol derived from policosanol, known for its diverse biological activities, including functioning as a plant growth regulator and exhibiting anti-inflammatory and antitumoral effects. However, its application is limited due to its high hydrophobicity, resulting in poor absorption and [...] Read more.
Triacontanol is a long-chain primary alcohol derived from policosanol, known for its diverse biological activities, including functioning as a plant growth regulator and exhibiting anti-inflammatory and antitumoral effects. However, its application is limited due to its high hydrophobicity, resulting in poor absorption and reduced therapeutic effectiveness. A potential solution to this problem is the use of niosomes. Niosomes are carriers composed of non-ionic surfactants, cholesterol, charge-inducing agents, and a hydration medium. They are effective in encapsulating drugs, improving their solubility and bioavailability. The objective of this study was to optimize and synthesize nano-niosomes for the encapsulation of triacontanol. Niosomes were synthesized using a thin-film hydration method combined with ultrasonication, following a Box–Behnken design. Niosomes were characterized using various techniques including dynamic light scattering, Fourier-transform infrared spectroscopy (FTIR), confocal microscopy, high-resolution scanning electron microscopy, and transmission electron microscopy (TEM). Formulation 14 of niosomes achieved the desired size, polydispersity index (0.198 ± 0.008), and zeta potential (−31.28 ± 1.21). FTIR analysis revealed a characteristic signal in the 3400–300 cm−1 range, indicating intermolecular interactions due to a bifurcated hydrogen bond between cholesterol and S60. Confocal microscopy confirmed the presence of triacontanol through Nile Red fluorescence. TEM revealed the spherical structure of niosomes. Full article
Show Figures

Figure 1

Figure 1
<p>Three-dimensional response plots displaying effects on particle size. Dihexadecyl phosphate and relationship of cholesterol and S60 effect on particle size (<b>a</b>), sonication time and dihexadecyl phosphate effect on particle size (<b>b</b>), and sonication time and relationship of cholesterol and S60 effect on the particle size (<b>c</b>).</p>
Full article ">Figure 2
<p>Three-dimensional response plots displaying effects on polydispersity. Dihexadecyl phosphate and relationship of cholesterol and S60 effect on polydispersity (<b>a</b>), sonication time and relationship of cholesterol and S60 effect on polydispersity (<b>b</b>), and sonication time and dihexadecyl phosphate effect on polydispersity (<b>c</b>).</p>
Full article ">Figure 3
<p>Three-dimensional response plots displaying effects on polydispersity. Dihexadecyl phosphate and relationship of cholesterol and S60 effect on zeta potential (<b>a</b>), sonication time and relationship of cholesterol and S60 on zeta potential (<b>b</b>), and sonication time and relationship of cholesterol and S60 effect on zeta potential (<b>c</b>).</p>
Full article ">Figure 4
<p>FTIR spectra of reagents for niosome formulation.</p>
Full article ">Figure 5
<p>FTIR spectra of blank niosomes and triacontanol-loaded niosomes.</p>
Full article ">Figure 6
<p>Micrographs of formulation 14 of niosomes with Nile Red fluorochrome. Blank niosomes (<b>a</b>) and niosomes with encapsulated triacontanol (<b>b</b>).</p>
Full article ">Figure 7
<p>SEM micrograph of formulation 14 of niosomes. Blank niosomes (<b>a</b>) and niosomes with encapsulated triacontanol (<b>b</b>).</p>
Full article ">Figure 8
<p>TEM micrographs of blank niosomes and niosomes with encapsulated triacontanol. Blank niosomes ((<b>a</b>,<b>b</b>); scale bar 0.2 μ) and niosomes with encapsulated triacontanol ((<b>c</b>,<b>d</b>); Scale bar 0.1 μ).</p>
Full article ">Figure 8 Cont.
<p>TEM micrographs of blank niosomes and niosomes with encapsulated triacontanol. Blank niosomes ((<b>a</b>,<b>b</b>); scale bar 0.2 μ) and niosomes with encapsulated triacontanol ((<b>c</b>,<b>d</b>); Scale bar 0.1 μ).</p>
Full article ">
19 pages, 8485 KiB  
Article
Evaluation of the In-Vitro Effects of Albendazole, Mebendazole, and Praziquantel Nanocapsules against Protoscolices of Hydatid Cyst
by Nooshinmehr Soleymani, Soheil Sadr, Cinzia Santucciu, Abbas Rahdar, Giovanna Masala and Hassan Borji
Pathogens 2024, 13(9), 790; https://doi.org/10.3390/pathogens13090790 - 12 Sep 2024
Viewed by 1122
Abstract
Cystic echinococcosis still remains a serious health and economic problem worldwide. The etiologic agent is Echinococcus granulosus sensu lato, giving origin to a fluid-filled cystic lesion. Therapy faces several challenges. Nanodrugs have shown promise as chemotherapeutics against hydatid cysts. The present study [...] Read more.
Cystic echinococcosis still remains a serious health and economic problem worldwide. The etiologic agent is Echinococcus granulosus sensu lato, giving origin to a fluid-filled cystic lesion. Therapy faces several challenges. Nanodrugs have shown promise as chemotherapeutics against hydatid cysts. The present study evaluated a highly safe lipid nano-polymeric capsule for its superior efficacy and ability to overcome drug resistance. Nanocapsule drugs were formulated into six groups: Albendazole, mebendazole, praziquantel, albendazole + mebendazole, albendazole + praziquantel, and praziquantel + mebendazole. The protoscolicidal effects of these six groups were assessed at 10, 60, and 120 min in three concentrations (1, 0.5, and 0.25 mg/mL). Drug formulations were evaluated via zeta potential, droplet size, solubility, particle size analyzer (PSA), and scanning electron microscopy. According to the PSA results, the mean size of the albendazole nanocapsules was 193.01 nm, mebendazole was 170.40 nm, and praziquantel was 180.44 nm. Albendazole + mebendazole showed the greatest protoscolicidal activity at a concentration of 1 mg/mL after 120 min. In contrast, each drug’s 0.25 mg/mL single-dose times showed the least protoscolicidal activity after 120 min. With the right application of nanotechnology, it is possible to produce safe and effective drugs, such as the polymeric combination of albendazole and mebendazole, which has promising implications. Full article
(This article belongs to the Section Parasitic Pathogens)
Show Figures

Figure 1

Figure 1
<p>The schematic of the methodology in the current study.</p>
Full article ">Figure 2
<p>Zeta potential spectra of the nanocapsules: control, loaded with ABZ range of 5 to −55 millivolts (mean: −35.78 mW), MBZ range of 0 to −48 millivolts (mean: −27.38 mW), and PZQ range of 0 to −43 millivolts (mean: −23.34 mW).</p>
Full article ">Figure 3
<p>The mean size of ABZ nanocapsules is 193.01 nm, MBZ is 170.40 nm, and PZQ is 180.44 nm.</p>
Full article ">Figure 4
<p>SEM image of polymeric nanocapsules.</p>
Full article ">Figure 5
<p>The protoscolicidal rate of 0.25 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).</p>
Full article ">Figure 6
<p>The protoscolicidal rate of 0.5 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).</p>
Full article ">Figure 7
<p>Light microscopy images showing the viable protoscolices of <span class="html-italic">Echinococcus granulosus</span> (stained with 0.1% eosin) (<b>A</b>: ×40). Live and red stained dead protoscolices after exposure to nano-ABZ (0.5 mg/mL) after 60 min of exposure (stained with 0.1% eosin) (<b>B</b>: ×10).</p>
Full article ">Figure 8
<p>The protoscolicidal rate of 1 mg/mL concentration nanocapsules in 10, 60, and 120 min (R version 4.3.1 used to generate the graphs).</p>
Full article ">Figure 9
<p>Light microscopy images showing the viable protoscolices of <span class="html-italic">Echinococcus granulosus</span> (<b>A</b>: ×40). Live and red stained dead protoscolices after exposure to nano-ABZ (1 mg/mL) after 10 min of exposure (stained with 0.1% eosin) (<b>B</b>: ×40, <b>C</b>: ×10).</p>
Full article ">
31 pages, 2559 KiB  
Review
Origami of KR-12 Designed Antimicrobial Peptides and Their Potential Applications
by Jayaram Lakshmaiah Narayana, Abraham Fikru Mechesso, Imran Ibni Gani Rather, D. Zarena, Jinghui Luo, Jingwei Xie and Guangshun Wang
Antibiotics 2024, 13(9), 816; https://doi.org/10.3390/antibiotics13090816 - 28 Aug 2024
Cited by 2 | Viewed by 2068
Abstract
This review describes the discovery, structure, activity, engineered constructs, and applications of KR-12, the smallest antibacterial peptide of human cathelicidin LL-37, the production of which can be induced under sunlight or by vitamin D. It is a moonlighting peptide that shows both antimicrobial [...] Read more.
This review describes the discovery, structure, activity, engineered constructs, and applications of KR-12, the smallest antibacterial peptide of human cathelicidin LL-37, the production of which can be induced under sunlight or by vitamin D. It is a moonlighting peptide that shows both antimicrobial and immune-regulatory effects. Compared to LL-37, KR-12 is extremely appealing due to its small size, lack of toxicity, and narrow-spectrum antimicrobial activity. Consequently, various KR-12 peptides have been engineered to tune peptide activity and stability via amino acid substitution, end capping, hybridization, conjugation, sidechain stapling, and backbone macrocyclization. We also mention recently discovered peptides KR-8 and RIK-10 that are shorter than KR-12. Nano-formulation provides an avenue to targeted delivery, controlled release, and increased bioavailability. In addition, KR-12 has been covalently immobilized on biomaterials/medical implants to prevent biofilm formation. These constructs with enhanced potency and stability are demonstrated to eradicate drug-resistant pathogens, disrupt preformed biofilms, neutralize endotoxins, and regulate host immune responses. Also highlighted are the safety and efficacy of these peptides in various topical and systemic animal models. Finaly, we summarize the achievements and discuss future developments of KR-12 peptides as cosmetic preservatives, novel antibiotics, anti-inflammatory peptides, and microbiota-restoring agents. Full article
(This article belongs to the Special Issue Insights into Natural Antimicrobial Peptides)
Show Figures

Figure 1

Figure 1
<p>Properties and functions of human cathelicidin LL-37 discovered in different cells. Depicted in the center is the 3D structure of membrane-bound LL-37 (PDB ID: 2K6O) determined by 3D triple-resonance heteronuclear multidimensional nuclear magnetic resonance (NMR) spectroscopy. When targeting bacterial membranes, the C-terminal tail of LL-37 is not folded and remains highly flexible as confirmed by heteronuclear <sup>15</sup>N backbone dynamics on the ps-ns time scale [<a href="#B32-antibiotics-13-00816" class="html-bibr">32</a>]. The C-terminal tail is disordered in complex with SDS, D8PG, and LPS (abbreviations in the text). Direct interactions of LL-37 with anionic bacterial phosphatidylglycerols (PGs) and LPS as demonstrated by NMR provide basis for antimicrobial and anti-inflammatory effects. NET: neutrophil extracellular traps; PBMC: peripheral blood mononuclear cells; DCs: dendritic cells; MSC: mesenchymal stem cell.</p>
Full article ">Figure 2
<p>Therapeutic strategies based on human cathelicidin LL-37. (<b>A</b>) Humans can use sunlight or vitamin D and its analog to switch on the expression of LL-37 to boost innate defense against infection. Likewise, recombinant DNA technology can be used to express LL-37 to achieve the same production. (<b>B</b>) Human LL-37 can function synergistically with other human AMPs such as defensin or lysozyme to better control pathogens. Similarly, human LL-37 can work synergistically with bacteriocins from commensal bacteria to better control invading pathogens. Using the same strategy, AMPs can be used with existing antibiotics to overcome resistance. (<b>C</b>) LL-37 can be engineered into novel antimicrobial agents based on different fragments (IG-24, GF-17, and KR-12) discovered from (1) peptide library, (2) structure-based design, (3) combined (1) and (2), and (4) feature-based mimicking (reviewed in ref. [<a href="#B26-antibiotics-13-00816" class="html-bibr">26</a>]). This review focuses on a variety of the engineered constructs based on KR-12, the smallest antibacterial fragment of LL-37.</p>
Full article ">Figure 3
<p>The discovery path of KR-12 through structural studies. (<b>A</b>) Amino acid sequences and nomenclature of LL-37 and its fragments. The original peptide names for these fragments are given on the right of the peptide sequences, while the shortened names are provided on the left. While LL-37 has a carboxylic acid at the C-terminus, the C-termini of shorter sequences, including FK-16/GF-17, FK-13, and KR-12, are all amidated to increase the net charge by +1. (<b>B</b>) Backbone structures of LL-37, IG-25, GF-17, FK-13, and KR-12 determined by 2D and 3D NMR spectroscopy [<a href="#B32-antibiotics-13-00816" class="html-bibr">32</a>,<a href="#B39-antibiotics-13-00816" class="html-bibr">39</a>,<a href="#B60-antibiotics-13-00816" class="html-bibr">60</a>]. Except for LL-37 and IG-25, GF-17, FK-13, and KR-12 are C-terminally amidated. (<b>C</b>,<b>D</b>) Horizontal and vertical views of the NMR structure of KR-12 in complex with anionic D8PG [<a href="#B32-antibiotics-13-00816" class="html-bibr">32</a>].</p>
Full article ">Figure 4
<p>Helical wheel plots for LL-37 (<b>A</b>), KR-12 (<b>B</b>) and its selective derivatives (<b>C</b>–<b>L</b>). The helical wheel was generated using the NetWheel program (<a href="http://lbqp.unb.br/NetWheels/" target="_blank">http://lbqp.unb.br/NetWheels/</a>, accessed on 31 July 2024). In this program, amino acids are classified into four groups: (1) polar/basic (red square): RHK, (2) polar/acidic (blue triangle): DE, (3) polar/uncharged (green diamond): STNQC, and (4) nonpolar (yellow circle): AGVILMFYWP. Although included in the plot, it is evident that the HIV TAT sequence is not amphipathic (<b>J</b>). However, the amphipathic helical structure can still be seen in the presence of additional sequence from the Trp cage (<b>K</b>). Finally, some symmetry can be seen in the helical wheel plot of cyclic KR-12 dimer (<b>L</b>).</p>
Full article ">Figure 5
<p>Various design strategies that transform human LL-37-derived KR-12 to new constructs: (1) amino acid changes, terminal capping, peptide hybridization, and peptide conjugates (<b>top</b>), (2) sidechain stapling and backbone cyclization (<b>right</b>), (3) surface immobilization (<b>left</b>), and (4) peptide formulation (<b>bottom</b>). See the text for further details. These strategies can be applied to other linear antimicrobial peptides as well.</p>
Full article ">Figure 6
<p>2D array of the lengths of the KR-12 peptide with 4 to 12 amino acids (aa4 to aa12) and fatty acids (c6 to C14) for antibacterial activity (<b>A</b>) and hemolytic toxicity (<b>B</b>) uncovered a zone for designing selective lipopeptides. On the left, the closer the curves to the green plane, the more potent the peptides are. In contrast, the farther away from the red plane on the right, the less hemolytic the peptides are.</p>
Full article ">Figure 7
<p>Like LL-37, KR-12 peptides also possess numerous desired properties such as antimicrobial, antibiofilm, and LPS neutralization. KR-12, as well as LL-37, has been covalently immobilized onto titanium (Ti) implants [<a href="#B123-antibiotics-13-00816" class="html-bibr">123</a>,<a href="#B124-antibiotics-13-00816" class="html-bibr">124</a>].</p>
Full article ">Figure 8
<p>In vivo safety and efficacy of KR-12 constructs. (<b>A</b>) Systemic toxicity of nanobiotics Myr-KR-12N and Myr-KR-12C via intravenous administration in mice. (<b>B</b>) Ototoxicity of a KR-12-a2 by applying the solution topically into the middle ears of guinea pigs. (<b>C</b>) Identification of the non-toxic dose of C10-KR8d via the intraperitoneal route in mice. (<b>D</b>) Efficacy of Ti-C10-KR8d implant on catheter-associated MRSA biofilm in mice. (<b>E</b>) LPS neutralization and bone restoration efficacy of KR-12-a2 in mice. (<b>F</b>) Cryogel-HA/TA/KR12 topical application in a mouse wound model. (<b>G</b>) PEEK-PDA-KR-12 coating on implants shows both antibacterial and osteointegration potential in mice. (<b>H</b>) KR-12 has anti-colitis ability against chemical induced colitis in mice. (<b>I</b>) Myr-KR-12N and Myr-KR-12C protection from LPS sepsis in mice. (<b>J</b>) C10-KR8d showcased anti-MRSA efficacy in a neutropenic murine infection model.</p>
Full article ">
18 pages, 3978 KiB  
Article
Liposomal Drug Delivery against Helicobacter pylori Using Furazolidone and N-Acetyl Cysteine in Augmented Therapy
by Muhammad Irfan Alam, Timothy Paget, Najla Yussuf Moosa, Husein Alghurairy and Amal Ali Elkordy
Pharmaceutics 2024, 16(9), 1123; https://doi.org/10.3390/pharmaceutics16091123 - 26 Aug 2024
Viewed by 1288
Abstract
Helicobacter pylori (H. pylori) infection is a significant global health concern, affecting approximately 50% of the world’s population and leading to gastric ulcers, gastritis, and gastric cancer. The increase in antibiotic resistance has compromised the efficacy of existing therapeutic regimens, necessitating [...] Read more.
Helicobacter pylori (H. pylori) infection is a significant global health concern, affecting approximately 50% of the world’s population and leading to gastric ulcers, gastritis, and gastric cancer. The increase in antibiotic resistance has compromised the efficacy of existing therapeutic regimens, necessitating novel approaches for effective eradication. This study aimed to develop a targeted liposomal drug delivery system incorporating furazolidone and N-acetylcysteine (NAC) to enhance mucopenetration and improve Helicobacter pylori eradication. Liposomes were formulated with furazolidone, NAC, and Pluronic F-127 using a modified reverse-phase evaporation technique. The formulations were categorized based on charge as neutral, negative, and positive and tested for mucopenetration using a modified silicon tube method with coumarin-6 as a fluorescent marker. The encapsulation efficiency and particle size were analyzed using HPLC and an Izon q-nano particle size analyzer. The results indicated that charged liposomes showed a higher encapsulation efficiency than neutral liposomes with Pluronic F-127. Notably, combining furazolidone with 1% NAC achieved complete eradication of H. pylori in 2.5 h, compared to six hours without NAC. The findings of this study suggest that incorporating NAC and Pluronic F-127 into liposomal formulations significantly enhances mucopenetration and antimicrobial efficacy. Full article
(This article belongs to the Topic Personalized Drug Formulations)
Show Figures

Figure 1

Figure 1
<p>Spot plating technique for different dilutions from (<b>A</b>). 10<sup>−1</sup> through 10<sup>−3</sup> and (<b>B</b>). from 10<sup>−1</sup> to 10<sup>−6</sup>.</p>
Full article ">Figure 2
<p>Transmission electron microscopic, TEM, images of mucopenetrative liposomes (<b>A</b>). MP3, cationic liposomes with Pluronic F-127 (<b>B</b>). MP1, neutral liposomes with Pluronic F-127. For formulations’ composition, refer to <a href="#pharmaceutics-16-01123-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 3
<p>Diffusion of mucopenetrative liposomal particles from MP1 to MP6 through 1 mm thick sigma mucin type I in a silicon tube maintained at pH 6.0 at 37 °C at 1, 2, and 3 hours’ time (n = 3, mean ± SD shown). Abbreviations: MP1 (neutral with Pluronic F-127), MP4 (neutral without Pluronic F-127), MP2 (negative with Pluronic F-127), MP5 (negative without Pluronic F-127), MP3 (positive with Pluronic F-127), MP6 (positive without Pluronic F-127). For formulations’ composition, refer to <a href="#pharmaceutics-16-01123-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 4
<p>(<b>A</b>). In vitro drug release of furazolidone from the mucopenetrative formulation MP1 and MP4 up to a 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP1 (neutral with Pluronic F-127), MP4 (neutral without Pluronic F-127). (<b>B</b>). In vitro drug release of furazolidone from the mucopenetrative formulation MP3 and MP6 up to a 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP3 (positive with Pluronic F-127), MP6 (positive without Pluronic F-127). (<b>C</b>). In vitro drug release of furazolidone from the mucopenetrative formulation MP2 and MP5 for up to a 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP2 (negative with Pluronic F-127), MP5 (negative without Pluronic F-127).</p>
Full article ">Figure 4 Cont.
<p>(<b>A</b>). In vitro drug release of furazolidone from the mucopenetrative formulation MP1 and MP4 up to a 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP1 (neutral with Pluronic F-127), MP4 (neutral without Pluronic F-127). (<b>B</b>). In vitro drug release of furazolidone from the mucopenetrative formulation MP3 and MP6 up to a 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP3 (positive with Pluronic F-127), MP6 (positive without Pluronic F-127). (<b>C</b>). In vitro drug release of furazolidone from the mucopenetrative formulation MP2 and MP5 for up to a 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP2 (negative with Pluronic F-127), MP5 (negative without Pluronic F-127).</p>
Full article ">Figure 5
<p>(<b>A</b>). In vitro drug release of NAC from the mucopenetrative formulation from MP1 and MP4 for up to 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP1 (neutral with Pluronic F-127), MP4 (neutral without Pluronic F-127). (<b>B</b>). In vitro drug release of NAC from the mucopenetrative formulation from MP2 and MP5 for up to 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP2 (negative with Pluronic F-127), MP5 (negative without Pluronic F-127). (<b>C</b>). In vitro drug release of NAC from the mucopenetrative formulation from MP3 and MP6 for up to 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP3 (positive with Pluronic F-127), MP6 (positive without Pluronic F-127).</p>
Full article ">Figure 5 Cont.
<p>(<b>A</b>). In vitro drug release of NAC from the mucopenetrative formulation from MP1 and MP4 for up to 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP1 (neutral with Pluronic F-127), MP4 (neutral without Pluronic F-127). (<b>B</b>). In vitro drug release of NAC from the mucopenetrative formulation from MP2 and MP5 for up to 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP2 (negative with Pluronic F-127), MP5 (negative without Pluronic F-127). (<b>C</b>). In vitro drug release of NAC from the mucopenetrative formulation from MP3 and MP6 for up to 4 h time at pH 6.0 (n = 3, mean ± SD shown). Abbreviations: MP3 (positive with Pluronic F-127), MP6 (positive without Pluronic F-127).</p>
Full article ">Figure 6
<p>A time–kill curve experiment of furazolidone augmented with NAC at a concentration of 1% of its minimum inhibitory concentration (MIC) against <span class="html-italic">H. pylori</span> inoculated on a blood agar plate and incubated at 37 °C for 24 h in an anaerobic jar with CampyGen gas packs before counting the colonies. The mean value of the log number of cfu per milliliter was plotted against time (n = 6, mean ± SD).</p>
Full article ">Scheme 1
<p>The coding for mucopenetrative liposomal formulation compositions (MP1-MP6).</p>
Full article ">
30 pages, 9634 KiB  
Review
Nano-Formulations of Natural Antioxidants for the Treatment of Liver Cancer
by Mariateresa Cristani, Andrea Citarella, Federica Carnamucio and Nicola Micale
Biomolecules 2024, 14(8), 1031; https://doi.org/10.3390/biom14081031 - 19 Aug 2024
Viewed by 1590
Abstract
Oxidative stress is a key factor in the pathological processes that trigger various chronic liver diseases, and significantly contributes to the development of hepatocarcinogenesis. Natural antioxidants reduce oxidative stress by neutralizing free radicals and play a crucial role in the treatment of free-radical-induced [...] Read more.
Oxidative stress is a key factor in the pathological processes that trigger various chronic liver diseases, and significantly contributes to the development of hepatocarcinogenesis. Natural antioxidants reduce oxidative stress by neutralizing free radicals and play a crucial role in the treatment of free-radical-induced liver diseases. However, their efficacy is often limited by poor bioavailability and metabolic stability. To address these limitations, recent advances have focused on developing nano-drug delivery systems that protect them from degradation and enhance their therapeutic potential. Among the several critical benefits, they showed to be able to improve bioavailability and targeted delivery, thereby reducing off-target effects by specifically directing the antioxidant to the liver tumor site. Moreover, these nanosystems led to sustained release, prolonging the therapeutic effect over time. Some of them also exhibited synergistic effects when combined with other therapeutic agents, allowing for improved overall efficacy. This review aims to discuss recent scientific advances in nano-formulations containing natural antioxidant molecules, highlighting their potential as promising therapeutic approaches for the treatment of liver cancer. The novelty of this review lies in its comprehensive focus on the latest developments in nano-formulations of natural antioxidants for the treatment of liver cancer. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the molecular mechanisms and cascade pathways involved in liver damage and related to oxidative stress (OS). Intercellular communication that occurs during liver injury includes sinusoidal endothelial cells (LSECs) capillarization, Kupffer cells (KCs) activation, and hepatic stellate cells (HSCs) activation. These activated cells release warning signals, including pro-inflammatory cytokines and growth factors, which contribute to the progression and persistence of liver damage. HBV: hepatitis B virus; HCV: hepatitis C virus; NASH: nonalcoholic steatohepatitis; HCC: hepatocellular carcinoma; ECM: extracellular matrix; ET-1: endothelin-1; EVs: extracellular vesicles; ILs: interleukins; PDGF: platelet-derived growth factor; TGFβ: transforming growth factor β; TNFα: tumor necrosis factor α; TXA: thromboxane.</p>
Full article ">Figure 2
<p>Schematic representation of the targeted drug delivery system developed by Lian B. et al.: (<b>A</b>) Chemical structure of RES; (<b>B</b>) Synthesis of the FA-HSA conjugate; (<b>C</b>) Representation of the FA-HSA-RES-NPs obtained by the encapsulation of RES within FA-HSA using a high-pressure fluid nano-homogeneous emulsification method; (<b>D</b>) Proposed mechanism of active tumor targeting.</p>
Full article ">Figure 3
<p>Schematic representation of the synthesis of galactosylated chitosan (GCS) and its application in curcumin-loaded nanoparticles (CUR-GCS@NPs) developed by Huang M. et al. These GCS-coated NPs were designed to target the asialoglycoprotein receptors (ASGPRs) overexpressed on hepatocellular carcinoma cells, offering enhanced biocompatibility and potential therapeutic efficacy.</p>
Full article ">Figure 4
<p>Chemical structure of apocynin (APO).</p>
Full article ">Figure 5
<p>Schematic representation of the tumor-targeting and IR-sensitive nanosystem (FA-CA@AgNPs) designed and developed by Abdelwahab T. et al. Caffeic acid (CA) was used both to chemically conjugate the targeting agent (folic acid; FA) and as a reducing agent to form AgNPs.</p>
Full article ">Figure 6
<p>Chemical structure of quercetin (Q).</p>
Full article ">Figure 7
<p>Chemical structure of rutin (RU).</p>
Full article ">Figure 8
<p>Chemical structure of hesperetin (HP).</p>
Full article ">Figure 9
<p>Chemical structure of honokiol (HK).</p>
Full article ">Figure 10
<p>Chemical structure of epigallocatechin gallate (EGCG).</p>
Full article ">Figure 11
<p>Chemical structure of umbelliferone β-<span class="html-small-caps">d</span>-galactopyranoside (UFG).</p>
Full article ">Figure 12
<p>Chemical structure of andrographolide (AG).</p>
Full article ">Figure 13
<p>Chemical structure of oleanolic acid (OA).</p>
Full article ">Figure 14
<p>Chemical structure of parthenolide (PLT).</p>
Full article ">Figure 15
<p>Chemical structure of vitamin C (Vit. C) and vitamin E (Vit. E).</p>
Full article ">Figure 16
<p>Chemical structure of α-tocotrienol.</p>
Full article ">Figure 17
<p>Chemical structure of coenzyme Q10 (CoQ10).</p>
Full article ">Figure 18
<p>Chemical structure of docosahexaenoic acid (DHA).</p>
Full article ">
Back to TopTop