[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Previous Issue
Volume 16, December
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Pharmaceutics, Volume 17, Issue 1 (January 2025) – 15 articles

  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
1 pages, 606 KiB  
Correction
Correction: Zhang et al. Interactions of Self-Assembled Bletilla Striata Polysaccharide Nanoparticles with Bovine Serum Albumin and Biodistribution of Its Docetaxel-Loaded Nanoparticles. Pharmaceutics 2019, 11, 43
by Guangyuan Zhang, Jin Qiao, Xin Liu, Yuran Liu, Ji Wu, Long Huang, Danyang Ji and Qingxiang Guan
Pharmaceutics 2025, 17(1), 15; https://doi.org/10.3390/pharmaceutics17010015 (registering DOI) - 25 Dec 2024
Abstract
In the original publication [...] Full article
26 pages, 2866 KiB  
Review
Enhancing Patient-Centric Drug Development: Coupling Hot Melt Extrusion with Fused Deposition Modeling and Pressure-Assisted Microsyringe Additive Manufacturing Platforms with Quality by Design
by Dinesh Nyavanandi, Preethi Mandati, Nithin Vidiyala, Prashanth Parupathi, Praveen Kolimi and Hemanth Kumar Mamidi
Pharmaceutics 2025, 17(1), 14; https://doi.org/10.3390/pharmaceutics17010014 (registering DOI) - 25 Dec 2024
Abstract
In recent years, with the increasing patient population, the need for complex and patient-centric medications has increased enormously. Traditional manufacturing techniques such as direct blending, high shear granulation, and dry granulation can be used to develop simple solid oral medications. However, it is [...] Read more.
In recent years, with the increasing patient population, the need for complex and patient-centric medications has increased enormously. Traditional manufacturing techniques such as direct blending, high shear granulation, and dry granulation can be used to develop simple solid oral medications. However, it is well known that “one size fits all” is not true for pharmaceutical medicines. Depending on the age, sex, and disease state, each patient might need a different dose, combination of medicines, and drug release pattern from the medications. By employing traditional practices, developing patient-centric medications remains challenging and unaddressed. Over the last few years, much research has been conducted exploring various additive manufacturing techniques for developing on-demand, complex, and patient-centric medications. Among all the techniques, nozzle-based additive manufacturing platforms such as pressure-assisted microsyringe (PAM) and fused deposition modeling (FDM) have been investigated thoroughly to develop various medications. Both nozzle-based techniques involve the application of thermal energy. However, PAM can also be operated under ambient conditions to process semi-solid materials. Nozzle-based techniques can also be paired with the hot melt extrusion (HME) process for establishing a continuous manufacturing platform by employing various in-line process analytical technology (PAT) tools for monitoring critical process parameters (CPPs) and critical material attributes (CMAs) for delivering safe, efficacious, and quality medications to the patient population without compromising critical quality attributes (CQAs). This review covers an in-depth discussion of various critical parameters and their influence on product quality, along with a note on the continuous manufacturing process, quality by design, and future perspectives. Full article
(This article belongs to the Special Issue Advances in Hot Melt Extrusion Technology)
Show Figures

Figure 1

Figure 1
<p>Detailed PAM instrumentation with different piston types.</p>
Full article ">Figure 2
<p>Detailed instrumentation of FDM platform.</p>
Full article ">Figure 3
<p>Different types of gear rollers available for FDM instruments and their level of mechanical shear.</p>
Full article ">Figure 4
<p>(<b>a</b>) Continuous manufacturing line for HME and PAM process; (<b>b</b>) pre-filled cartridges for shipping to compounding pharmacies; (<b>c</b>) continuous manufacturing line coupling HME and FDM process; (<b>d</b>) drug-loaded polymeric filaments for shipping to compounding pharmacies.</p>
Full article ">Figure 5
<p>A detailed list of QbD elements critical for the successful development of pharmaceutical medications.</p>
Full article ">Figure 6
<p>SWOT analysis of additive manufacturing for successful development of pharmaceutical medications.</p>
Full article ">
22 pages, 3422 KiB  
Article
Investigation of Novel Aronia Bioactive Fraction-Alginic Acid Nanocomplex on the Enhanced Modulation of Neuroinflammation and Inhibition of Aβ Aggregation
by Bong-Keun Jang, Soo Jung Shin, Hyun Ha Park, Vijay Kumar, Yong Ho Park, Jeom-Yong Kim, Hye-Yeon Kang, Sunyoung Park, Youngsun Kwon, Sang-Eun Shin, Minho Moon and Beom-Jin Lee
Pharmaceutics 2025, 17(1), 13; https://doi.org/10.3390/pharmaceutics17010013 (registering DOI) - 25 Dec 2024
Abstract
Background/Objectives: Aronia extract or its active compounds, especially anthocyanin, have shown potential for Alzheimer’s disease (AD)-related pathologies, including neuroinflammation, fibrillogenesis of amyloid beta (Aβ), and cognitive impairment. However, there was still concern about their structural instability in vivo and in vitro. To solve [...] Read more.
Background/Objectives: Aronia extract or its active compounds, especially anthocyanin, have shown potential for Alzheimer’s disease (AD)-related pathologies, including neuroinflammation, fibrillogenesis of amyloid beta (Aβ), and cognitive impairment. However, there was still concern about their structural instability in vivo and in vitro. To solve the instability of anthocyanins, we combined aronia bioactive factions (ABFs) and alginic acid via electrostatic molecular interactions and created an ABF–alginic acid nanocomplex (AANCP). We evaluated whether it is more stable and effective in cognitive disorder mice and neuroinflammation cell models. Methods: The physicochemical properties of the AANCP, such as nanoparticle size, structural stability, and release rate, were characterized. The AANCP was administered to scopolamine-injected Balb/c mice, and to BV2 microglia treated with lipopolysaccharide (LPS) and amyloid beta (Aβ). Inflammation responses were measured via qPCR and ELISA in vitro, and cognitive functions were measured via behavior tests in vivo. Results: The AANCP readily formed nanoparticles, 209.6 nm in size, with a negatively charged zeta potential. The AANCP exhibited better stability in four plasma samples (human, dog, rat, and mouse) and was slowly released in different pH conditions (pH 2.0, 7.4, and 8.0) compared with non-complexedABF. In vitro studies on microglial cells treated with AANCPs revealed a suppression of inflammatory cytokines (tumor necrosis factor-alpha and interleukin-6) induced by LPS. The AANCP increased microglial Aβ phagocytosis through the activation of triggering receptor expressed on myeloid cell 2 (TREM2)-related microglial polarization. The AANCP inhibited aggregation of Aβ in vitro and alleviated cognitive impairment in a scopolamine-induced in vivo dementia mouse model. Conclusions: Our data indicate that AANCPs are more stable than ABFs and effective for cognitive disorders and neuroinflammation via modulation of M2 microglial polarization. Full article
Show Figures

Figure 1

Figure 1
<p>Physicochemical characterization of the ABF-alginic acid nanocomplex. (<b>A</b>) Schematic representation of chemical bonding in the AANCP. ABFs are represented by (+) and alginic acids by (−). Solid lines express ionic bonds, and dotted lines represent π-π interactions. Zeta potential, PDI, and distribution of zeta average particle size of (<b>B</b>) ABF, (<b>C</b>) alginic acid, and (<b>D</b>) the AANCP using DLS. (<b>E</b>) Anthocyanin release test of ABF and the AANCP according to various pH solutions. (<b>F</b>) SEM image of ABF, alginic acid, and AANCP. Statistical analyses were performed using one-way ANOVA followed by Tukey’s test. Significance levels of * <span class="html-italic">p</span>-value &lt; 0.05, ** <span class="html-italic">p</span>-value &lt; 0.01 and *** <span class="html-italic">p</span>-value &lt; 0.001 indicate differences between the ABF-treated group (white bar), and the AANCP-treated group (black bar).</p>
Full article ">Figure 2
<p>Effect of AANCP on the inhibition of the LPS-induced secretion of pro-inflammatory molecules in BV2 microglial cells. IL-6 and TNF-α levels were determined by ELISA. (<b>A</b>) The quantitative analysis shows that AANCP reduced LPS-induced IL-6 release from BV2 microglial cells. (<b>B</b>) The quantitative graph shows that AANCP decreased the LPS-induced TNF-α secretion from microglia. The mean ± S.E.M. values were calculated. Statistical analyses were performed using one-way ANOVA followed by Tukey’s test. Differences were significant at ### <span class="html-italic">p</span>-value &lt; 0.001 between the control group (black bar) and the vehicle-treated group (white bar). Significance levels of ** <span class="html-italic">p</span>-value &lt; 0.01 and *** <span class="html-italic">p</span>-value &lt; 0.001 indicate differences between the vehicle-treated group, the ABF-treated group (pink bar), and the AANCP-treated group (purple bar).</p>
Full article ">Figure 3
<p>Effect of AANCP on the conversion of the M1 phenotype induced by Aβ to the M2 phenotype in microglial cells. The M1 phenotype was assessed by the expression of cytokines, and the M2 phenotype was examined by the expression of TREM2 in BV2 microglial cells. (<b>A</b>–<b>C</b>) Quantitative analysis shows that AANCP reduced Aβ<sub>42</sub>-mediated upregulation of M1 markers, including TNF-α, IL-6, and IL-1β, in microglia. (<b>D</b>) The quantified graph shows that the AANCP modulated the level of TREM2 mRNA in microglia cells. The mean ± S.E.M. values were calculated. Statistical analyses were performed using one-way ANOVA followed by Tukey’s test. Differences were significant at # <span class="html-italic">p</span>-value &lt; 0.05 and ### <span class="html-italic">p</span>-value &lt; 0.001 between the control group (black bar) and the vehicle-treated group (white bar). Significance levels of * <span class="html-italic">p</span>-value &lt; 0.05, ** <span class="html-italic">p</span>-value &lt; 0.01, and *** <span class="html-italic">p</span>-value &lt; 0.001 indicate differences between the vehicle-treated group and the AANCP-treated group (purple bar).</p>
Full article ">Figure 4
<p>Significant enhancement of AANCP on the microglial phagocytic clearance of Aβ. (<b>A</b>) Representative images show immunoreactivity of ionized calcium-binding adaptor molecule 1 (Iba-1; green) and FAM-labeled Aβ<sub>42</sub> (FAM-Aβ; red) in BV2 microglial cells. DAPI staining was performed to visualize the nucleus (cyan). (<b>B</b>) Phagocytosis rates were expressed as a percentage with a counting number of both Aβ<sub>42</sub>- and Iba-1-positive cells per DAPI-positive cells. The mean ± S.E.M. values were calculated. Statistical analyses were performed using one-way ANOVA followed by Tukey’s test. Significance levels of * <span class="html-italic">p</span>-value &lt; 0.05 and *** <span class="html-italic">p</span>-value &lt; 0.001 indicate differences between the vehicle-treated group and the AANCP-treated group (purple bar).</p>
Full article ">Figure 5
<p>Effect of AANCP on the significant inhibition of Aβ aggregation. (<b>A</b>) An outline of the experimental design for treatment and ThT assay, (<b>B</b>) ThT fluorescence intensity curves from 0 to 48 h, and (<b>C</b>) bar graph at the 12-, 24-, 36-, and 48 h exhibiting the kinetics of Aβ<sub>42</sub> aggregation, both with and without AANCP. Morin serves as a positive control for the inhibitory activity of Aβ<sub>42</sub> aggregation. Values are expressed as the mean ± S.E.M. Statistical analyses were performed by one-way ANOVA, followed by Tukey’s test. *** <span class="html-italic">p</span>-value &lt; 0.001 indicates significant differences between the Aβ<sub>42</sub> + vehicle-treated group (red bar) and the Aβ<sub>42</sub> + AANCP (purple bar) or Aβ<sub>42</sub> + morin-treated group (yellow bar).</p>
Full article ">Figure 6
<p>The effect of AANCP on the significant alleviation of cognitive impairment in scopolamine-treated mice. (<b>A</b>) Schematic design of vivo experiment. Balb/c mice were orally administered AANCP (500 mg/kg) and EGB (50 mg/kg) daily for 2 weeks. EGB was used as a positive control. Scopolamine (2 mg/kg) was administered intraperitoneally 30 min before the behavioral test to impair cognitive function. (<b>B</b>) Changes in body weight by administration of scopolamine, EGB, and AANCP in Balb/c mice. Body weight was measured every 3 days from day 1 to 15. (<b>C</b>) Total arm entry, (<b>D</b>) spontaneous alterations (%), (<b>E</b>) Latency time (s). Values are expressed as the mean ± S.E.M (n = 4 in vehicle-treated Balb/c mice; n = 5 in scopolamine-treated Balb/c mice; n = 4 in scopolamine and EGB-treated Balb/c mice; n = 4 in scopolamine and AANCP-treated Balb/c mice). Statistical analyses were performed by one-way ANOVA, followed by Tukey’s test. ## <span class="html-italic">p</span>-value &lt; 0.01 indicates significant differences compared to the vehicle-treated Balb/c mice (black bar) and scopolamine and vehicle-treated Balb/c mice (white bar) and * <span class="html-italic">p</span>-value &lt; 0.05 indicates significant differences between the scopolamine and vehicle-treated Balb/c mice and or scopolamine and AANCP-treated Balb/c mice (purple bar).</p>
Full article ">Figure 7
<p>Schematic diagram of the effects of AANCP on neuroinflammation, Aβ aggregation, and cognitive impairment in AD and dementia. The aggregation of Aβ and M1 markers, such as TNF-α and IL-6, form a vicious circle that exacerbates the progression of AD. The pro-inflammatory cytokines promote the aggregation of Aβ, which in turn induces the release of inflammatory molecules. During pathological conditions caused by Aβ, resting microglia are activated into pro-inflammatory or anti-inflammatory microglia. However, upon return to physiological conditions via the removal of Aβ, M1- or M2-activated microglia are restored to resting microglia. AANCP promotes the transition of microglia to the anti-inflammatory phenotype and inhibits the aggregation of Aβ. Consequently, AANCP inhibits Aβ aggregation, reduces neuroinflammation, and improves cognitive impairment in AD. Stimulation is indicated by (+) arrows. Inhibition is indicated by (−) arrows.</p>
Full article ">
32 pages, 6631 KiB  
Article
Evaluation of Biologics ACE2/Ang(1–7) Encapsulated in Plant Cells for FDA Approval: Safety and Toxicology Studies
by Henry Daniell, Geetanjali Wakade, Smruti K. Nair, Rahul Singh, Steven A. Emanuel, Barry Brock and Kenneth B. Margulies
Pharmaceutics 2025, 17(1), 12; https://doi.org/10.3390/pharmaceutics17010012 (registering DOI) - 25 Dec 2024
Abstract
Background/Objectives: For several decades, protein drugs (biologics) made in cell cultures have been delivered as sterile injections, decreasing their affordability and patient preference. Angiotensin Converting Enzyme 2 (ACE2) gum is the first engineered human blood protein expressed in plant cells approved by the [...] Read more.
Background/Objectives: For several decades, protein drugs (biologics) made in cell cultures have been delivered as sterile injections, decreasing their affordability and patient preference. Angiotensin Converting Enzyme 2 (ACE2) gum is the first engineered human blood protein expressed in plant cells approved by the FDA without the need for purification and is a cold-chain and noninvasive drug delivery. This biologic is currently being evaluated in human clinical studies to debulk SARS-CoV-2 in the oral cavity to reduce coronavirus infection/transmission (NCT 0543318). Methods: Chemistry, manufacturing, and control (CMC) studies for the ACE2/Ang(1–7) drug substances (DSs) and ACE2 gum drug product (DP) were conducted following USP guidelines. GLP-compliant toxicology studies were conducted on Sprague Dawley rats (n = 120; 15/sex/group) in four groups—placebo, low (1.6/1.0 mg), medium (3.2/2.0 mg), and high (8.3/5.0 mg) doses IP/kg/day. Oral gavage was performed twice daily for 14 days (the dosing phase) followed by the recovery phase (35 days). Plasma samples (n = 216) were analyzed for the product Ang(1–7) by ELISA. Results: The ACE2 protein was stable in the gum for at least up to 78 weeks. The toxicology study revealed the dose-related drug delivery to the plasma and increases in the AUC (56.6%) and Cmax (52.9%) after 28 high-dose gavages (95% C.I.), although this quantitation excludes exogenously delivered membrane-associated ACE2/Ang(1–7). Vital biomarkers and organs were not adversely affected despite the 10-fold higher absorption in the tissues, demonstrating the safety for the first in-human clinical trials of ACE2/Ang(1–7). The NOAEL observed in the rats was 2.5–7.5-fold higher than that of the anticipated efficacious therapeutic dose in humans for the treatment of cardiopulmonary disorders, and it was 314-fold higher than the NOAEL for topical delivery via chewing gum. Conclusions: This report lays the foundation for the regulatory process approval for noninvasive and affordable human biologic drugs bioencapsulated in plant cells. Full article
(This article belongs to the Special Issue Peptide–Drug Conjugates for Targeted Delivery)
Show Figures

Figure 1

Figure 1
<p>FDA-compliant CTB-ACE2/Ang(1–7) drug substances and product eliminates fermentation, purification, cold-chain, and invasive drug delivery methods. Left panel—GLP-complaint toxicology studies performed for a total of 162 animals with toxicokinetic assessment. Middle panel—topical delivery of IND-approved ACE2 gum to debulk SARS-CoV2 and oral delivery of ACE2/Ang(1–7) to clinic. Right panel—chemistry, manufacturing, and control studies of drug product—ACE2 gum. RV: right ventricle; LV: left ventricle; NOAEL: no-observed-adverse-effect level; ACE2: Angiotensin Converting Enzyme 2.</p>
Full article ">Figure 2
<p>Toxicokinetic study design: CTB-ACE2 and CTB-Ang(1–7) proteins expressed in lettuce were orally gavaged twice daily for 14 days to male (<span class="html-italic">n</span> = 21) and female (<span class="html-italic">n</span> = 21) rats in placebo, low-, medium-, and high-dose groups. The plasma samples were collected at 2, 4, 6, 10, and 24 h on day 1 and day 14 and analyzed for the toxicokinetic and pharmacodynamic parameters.</p>
Full article ">Figure 3
<p>Chemistry, manufacturing, and control data of drug substances (DSs) CTB-ACE2 and CTB-Ang(1–7) stored at USP controlled temperature. (<b>A</b>) Stability of DSs assessed at &lt;1 month and after 1 year of storage at USP controlled temperature in FDA-approved black containers protected from light. (<b>B</b>) Effect of grinding time on optimal release of drug protein CTB-ACE2 for oral (3 s) and topical (12 s) delivery. (<b>C</b>) Effect of grinding time on optimal release of drug protein CTB-Ang (1–7) for oral delivery (3 s). (<b>D</b>) Grinding time (3, 6, 9, 12 s)-dependent reduction in particle size.</p>
Full article ">Figure 4
<p>Toxicokinetic analysis of plasma Ang(1–7) concentrations. (<b>A</b>) Comparative plasma Ang-(1–7) concentrations in low-, medium-, and high-dose groups after 2 gavages (0 h and 8 h) and 28 gavages at 0, 2, 4, 10, and 24 h timepoints. Data presented as % change (95% CI) w.r.t pregavage plasma Ang-(1–7). (<b>B</b>) Percentage increase in C<sub>max</sub> after 28 gavages (14 days) vs. 2 gavages (day 1) across placebo, low-, medium-, and high-dose groups. (<b>C</b>) Percentage increase in AUC<sub>last</sub> after 28 gavages (14 days) vs. 2 gavages (day 1) across placebo, low-, medium-, and high-dose groups. Data presented as % increase (95% CI).</p>
Full article ">Figure 5
<p>Hematology Index. Complete blood counts were measured in male (<span class="html-italic">n</span> = 15) and female (<span class="html-italic">n</span> = 15) rats for placebo, low-, medium-, and high-dose groups after 28 gavages at the end of the dosing phase (day 15) followed by the recovery phase (day 35). ANOVA with Dunnett’s method showed that the low, medium, and high doses did not produce more significant differences in the CBC parameters than those of the placebo.</p>
Full article ">Figure 6
<p>Liver function test. Liver enzymes were measured in male (<span class="html-italic">n</span> = 15) and female (<span class="html-italic">n</span> = 15) rats for the placebo, low-, medium-, and high-dose groups after 28 gavages at the end of the dosing phase (day 15) followed by the recovery phase (day 35). ANOVA with Dunnett’s method showed that the low, medium, and high doses did not produce more significant differences in the liver function test parameters than those of the placebo control.</p>
Full article ">Figure 7
<p>Renal function test. Renal enzymes were measured in male (<span class="html-italic">n</span> = 15) and female (<span class="html-italic">n</span> = 15) rats for placebo, low-, medium-, and high-dose groups after 28 gavages at the end of the dosing phase (day 15) followed by the recovery phase (day 35). ANOVA with Dunnett’s method showed that the low, medium, and high doses did not produce more significant differences in the renal function test parameters than those of the placebo control.</p>
Full article ">Figure 8
<p>(<b>A</b>) Urinalysis (Urine volume, specific gravity and pH), (<b>B</b>) coagulation (Prothrombin Time, Act. Partial thromboplastin time, fibrinogen), and (<b>C</b>) metabolic syndrome test (glucose, cholesterol and triglyceride) parameters were measured in male (<span class="html-italic">n</span> = 15) and female (<span class="html-italic">n</span> = 15F) rats for the placebo, low-, medium-, and high-dose groups after 28 gavages at the end of the dosing phase (day 15) followed by the recovery phase (day 35). ANOVA with Dunnett’s method showed that the low, medium, and high doses did not produce more significant differences in the urinalysis, coagulation, and metabolic syndrome test parameters than those of the placebo control.</p>
Full article ">
24 pages, 5118 KiB  
Article
Development of a Novel Co-Amorphous Curcumin and L-Arginine (1:2): Structural Characterization, Biological Activity and Pharmacokinetics
by Jose Antonio Mancillas-Quiroz, Miriam del Carmen Carrasco-Portugal, Karina Mondragón-Vásquez, Juan Carlos Huerta-Cruz, Juan Rodríguez-Silverio, Leyanis Rodríguez-Vera, Juan Gerardo Reyes-García, Francisco Javier Flores-Murrieta, Jorge Guillermo Domínguez-Chávez and Héctor Isaac Rocha-González
Pharmaceutics 2025, 17(1), 11; https://doi.org/10.3390/pharmaceutics17010011 (registering DOI) - 25 Dec 2024
Abstract
Background: Curcumin appears to be well tolerated and effective for managing chronic inflammatory pain, but its poor oral bioavailability has been a hurdle in its use as a therapeutic agent. The current study was performed to characterize a novel co-amorphous compound based on [...] Read more.
Background: Curcumin appears to be well tolerated and effective for managing chronic inflammatory pain, but its poor oral bioavailability has been a hurdle in its use as a therapeutic agent. The current study was performed to characterize a novel co-amorphous compound based on curcumin/L-arginine 1:2 (CAC12). Methods: Stability, solubility and structural characterization of the CAC12 were carried out by spectrometry techniques and in vitro assays, whereas the antinociceptive and anti-inflammatory effects were evaluated by CFA or carrageenan models. The mechanism of action was determined by cytokine quantification, and pharmacokinetic parameters were obtained through UPLC-MS/MS. The co-amorphous compound was prepared by fast solvent evaporation. Powder XRD, 13C-NMR, ATR-FTIR and TGA/DSC thermal analysis showed a 1:2 stoichiometry for the CAC12. Results: CAC12 was 1000 times more soluble than curcumin, and it was stable for 1 month at 40 °C and 75% relative humidity or for 60 min in physiological medium at pH 4.5–6.8. Co-amorphous curcumin/L-arginine, but not curcumin + L-arginine, decreased carrageenan- or CFA-induced inflammation and nociception by decreasing IL-1α, IL-1β, IL-6, TNF-α, MCP-1 and CXCL1 cytokines. The bioavailability of free plasmatic curcumin increased about 22.4 times when it was given as CAC12 relative to a phytosome formulation at the equivalent dose. Conclusions: Results suggest the possible use of CAC12 to treat inflammatory pain disorders in human beings. Full article
(This article belongs to the Section Physical Pharmacy and Formulation)
Show Figures

Figure 1

Figure 1
<p>Powder X-ray diffractograms obtained for CAC12, L-arginine and curcumin.</p>
Full article ">Figure 2
<p>Solid-state carbon-13 nuclear magnetic resonance (<sup>13</sup>C-NMR) spectra of curcumin, L-arginine and CAC12. (<b>A</b>) Proposed chemical structure of CAC12. (<b>B</b>) The main displacements and (<b>C</b>) typical <sup>13</sup>C-NMR spectra of CAC12, L-arginine and curcumin. Abbreviations: ws = wide signal, ss = small shoulder. As expected, the signals in the spectrum corresponding to the co-amorphous compound were observed to be broader and much less defined than those of the raw materials. In the CAC12 spectrum, we observed that the typical L-arginine signal at 54.8 ppm decreases to 49.0 ppm (Δδ = 5.8 ppm); likewise, the signal observed in the curcumin spectrum at 150.2 ppm, which is assigned to the hydroxyl-bonded aromatic carbons (C8 and C8′) shifts to 147.1 ppm (Δδ = 3.1 ppm) in the CAC12 spectrum.</p>
Full article ">Figure 3
<p>Attenuated total reflectance–Fourier transform infrared (ATR-FTIR) spectra for CAC12, L-arginine and curcumin.</p>
Full article ">Figure 4
<p>Differential scanning calorimetry (DSC) and thermal gravimetric analysis (TGA) of CAC12.</p>
Full article ">Figure 5
<p>X-ray powder diffractograms acquired from CAC12 stability studies. (<b>A</b>) Powder X-ray diffractograms of CAC12 obtained after 1 month of storage under several temperature and relative humidity (RH) conditions. (<b>B</b>) Powder X-ray diffractograms of CAC12 obtained after 12 h in distilled water, phosphate buffer (pH = 6.8), acetate buffer (pH = 4.5) or HCl solution (pH = 1.2).</p>
Full article ">Figure 6
<p>Lack of anti-inflammatory effect of oral curcumin and L-arginine in Freund’s complete adjuvant or carrageenan models. Time courses show the effects of increasing doses (100–320 mg/kg) of curcumin and L-arginine on inflammation induced by Freund’s complete adjuvant (<b>A</b>,<b>C</b>) or carrageenan (<b>E</b>,<b>G</b>). The bar graphs show a lack of anti-inflammatory effect of increasing doses of curcumin and L-arginine in Freund’s complete adjuvant (<b>B</b>,<b>D</b>) or carrageenan (<b>F</b>,<b>H</b>) models. Curcumin and L-arginine were orally administered at −1 h with respect to intra-articular injection of Freund’s complete adjuvant (100 µL, 0.1%) or subcutaneous administration of carrageenan (50 µL, 1%). Data are expressed as the mean ± S.E.M. of 6 animals per experimental group. There was no statistical difference based on one-way ANOVA. The area under the curve (AUC) was calculated from the time courses using the trapezoidal rule.</p>
Full article ">Figure 7
<p>Anti-inflammatory effects of oral CAC12 (1:2) in Freund’s complete adjuvant or carrageenan models. Time courses show the effects of increasing doses (10–100 mg/kg) of CAC12 on inflammation induced by Freund’s complete adjuvant (<b>A</b>) or carrageenan (<b>C</b>). The bar graphs show the anti-inflammatory effect produced by increasing doses of CAC12 in Freund’s complete adjuvant (<b>B</b>) or carrageenan (<b>D</b>) models. Curcumin and L-arginine were orally administered at −1 h with respect to intra-articular injection of Freund’s complete adjuvant (100 µL, 0.1%) or subcutaneous administration of carrageenan (50 µL, 1%). Data are expressed as the mean ± S.E.M. of 6 animals per experimental group. * Statistically different from the vehicle based on one-way ANOVA followed by Dunnett’s test with <span class="html-italic">p</span> &lt; 0.05. Statistical differences in time courses were omitted for the sake of clarity. The area under the curve (AUC) was calculated from the time courses using the trapezoidal rule.</p>
Full article ">Figure 8
<p>Effect of oral administration of 100 mg/kg of the crystalline mixture of curcumin (Cur, 51.4 mg) + L-arginine (Arg, 48.6 mg), diclofenac (Dic) and CAC12 (Co-amorph, 1:2) on inflammation induced by Freund’s complete adjuvant or carrageenan. The time courses show the anti-inflammatory effect produced by 100 mg/kg of diclofenac and CAC12 on the inflammation induced by Freund’s complete adjuvant (<b>A</b>) and carrageenan (<b>C</b>). The bar graphs indicate that 100 mg/kg of diclofenac and CAC12, but not the curcumin + L-arginine combination, have anti-inflammatory effects in Freund’s complete adjuvant (<b>B</b>) and carrageenan (<b>D</b>) models. Data are expressed as the mean ± S.E.M. of 6 animals per experimental group. * Statistically different from the vehicle group (Veh) based on one-way ANOVA followed by Tukey’s test with <span class="html-italic">p</span> &lt; 0.05. Statistical differences in time courses were omitted for the sake of clarity. The area under the curve (AUC) was calculated from the time courses using the trapezoidal rule.</p>
Full article ">Figure 9
<p>Effects of oral administration of 100 mg/kg of curcumin (Cur), L-arginine (Arg) and CAC12 (Co-amorph, 1:2) on nociception induced by Freund’s complete adjuvant or carrageenan. The time courses show the antinociceptive effect produced by 100 mg/kg of the CAC12 on the nociception induced by complete Freund’s adjuvant (<b>A</b>) and carrageenan (<b>C</b>). The bar graphs strongly suggest that 100 mg/kg of CAC12 exhibits an antinociceptive effect in Freund’s complete adjuvant (<b>B</b>) and carrageenan (<b>D</b>) models. Data are expressed as the mean ± S.E.M. of 6 animals per experimental group. * Statistically different with respect to the vehicle (Veh) group or ** CAC12 group based on one-way ANOVA followed by Tukey’s test with <span class="html-italic">p</span> &lt; 0.05. Statistical differences in time courses were omitted for the sake of clarity. The area under the curve (AUC) was calculated from the time courses using the trapezoidal rule.</p>
Full article ">Figure 10
<p>Effect of oral administration of CAC12 (1:2) on the levels of interleukin 1 alpha (IL-1α), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein 1 (MCP-1) and CXC motif chemokine ligand 1 (CXCL1), induced by Freund’s complete adjuvant (<b>A</b>) or carrageenan (<b>B</b>). The bar graphs indicate that 100 mg/kg of CAC12 is able to prevent the increase in tissue concentrations of IL-1α, IL-1β, IL-6, TNF-α, MCP-1 and CXCL1 in Freund’s complete adjuvant (<b>A</b>) and carrageenan (<b>B</b>) models of inflammation for 8 h. Data are expressed as the mean ± S.E.M. of 3–4 animals per experimental group. * Statistically different from the vehicle group and ** statistically different from the carrageenan or CFA groups based on Newman–Keuls’ test with <span class="html-italic">p</span> &lt; 0.05. The area under the curve (AUC) was calculated from time courses (from 0 to 8 h) using the trapezoidal rule.</p>
Full article ">Figure 11
<p>Pharmacokinetic profile of oral administration of the reference compound (curcumin phytosome, Mericart<sup>®</sup>) and the test compound (CAC12, CurQsen<sup>®</sup>). The data are expressed as the mean of the plasmatic-free curcumin of 18 healthy subjects.</p>
Full article ">
18 pages, 14945 KiB  
Article
Long-Term Therapeutic Effects of 225Ac-DOTA-E[c(RGDfK)]2 Induced by Radiosensitization via G2/M Arrest in Pancreatic Ductal Adenocarcinoma
by Mitsuyoshi Yoshimoto, Kohshin Washiyama, Kazunobu Ohnuki, Ayano Doi, Miki Inokuchi, Motohiro Kojima, Brian W. Miller, Yukie Yoshii, Anri Inaki and Hirofumi Fujii
Pharmaceutics 2025, 17(1), 9; https://doi.org/10.3390/pharmaceutics17010009 - 24 Dec 2024
Abstract
Background: Alpha radionuclide therapy has emerged as a promising novel strategy for cancer treatment; however, the therapeutic potential of 225Ac-labeled peptides in pancreatic cancer remains uninvestigated. Methods: In the cytotoxicity study, tumor cells were incubated with 225Ac-DOTA-RGD2. [...] Read more.
Background: Alpha radionuclide therapy has emerged as a promising novel strategy for cancer treatment; however, the therapeutic potential of 225Ac-labeled peptides in pancreatic cancer remains uninvestigated. Methods: In the cytotoxicity study, tumor cells were incubated with 225Ac-DOTA-RGD2. DNA damage responses (γH2AX and 53BP1) were detected using flowcytometry or immunohistochemistry analysis. Biodistribution and therapeutic studies were carried out in BxPC-3-bearing mice. Results: 225Ac-DOTA-RGD2 demonstrated potent cytotoxicity against cells expressing αvβ3 or αvβ6 integrins and induced G2/M arrest and γH2AX expression as a marker of double-stranded DNA breaks. 225Ac-DOTA-RGD2 (20, 40, 65, or 90 kBq) showed favorable pharmacokinetics and remarkable tumor growth inhibition without severe side effects in the BxPC-3 mouse model. In vitro studies revealed that 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 swiftly induced G2/M arrest and elevated γH2AX expression. Furthermore, to clarify the mechanism of successful tumor growth inhibition for a long duration in vivo, we investigated whether short-term high radiation exposure enhances radiation sensitivity. Initially, a 4 h induction treatment with 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 enhanced both cytotoxicity and γH2AX expression with 0.5 kBq/mL of 225Ac-DOTA-RGD2 compared to a treatment with only 0.5 kBq/mL of 225Ac-DOTA-RGD2. Meanwhile, the γH2AX expression induced by 5 or 10 kBq/mL of 225Ac-DOTA-RGD2 alone decreased over time. Conclusions: These findings highlight the potential of using 225Ac-DOTA-RGD2 in the treatment of intractable pancreatic cancers, as its ability to induce G2/M cell cycle arrest enhances radiosensitization, resulting in notable growth inhibition. Full article
(This article belongs to the Section Clinical Pharmaceutics)
Show Figures

Figure 1

Figure 1
<p>In vitro cytotoxicity. (<b>a</b>) Cytotoxicity of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in human pancreatic tumor cell lines. (<b>b</b>) Comparison of cytotoxicity between <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> and <sup>225</sup>AcDOTA in BxPC-3. All assays were performed in triplicate. Data are presented as mean ± standard deviation.</p>
Full article ">Figure 2
<p>Induction of γH2AX and 53BP1 foci formation in response to increasing doses of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> at 24 h. (<b>a</b>) Representative images of γH2AX and 53BP1 foci obtained by immunofluorescence microscopy in BxPC-3 cells. Scale bar, 20 μm. (<b>b</b>) The number of γH2AX and 53BP1 foci per cell. Induction of γH2AX and 53BP1 foci in response to increasing doses of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> was monitored at 24 h. The number of γH2AX and 53BP1 foci per cell was counted, and 50–100 cells were analyzed. All assays were performed in triplicate. Data are presented as mean ± standard deviation and analyzed using a one-way analysis of variance with Dunn’s multiple-comparisons test (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>Flow cytometric analysis of BxPC-3 after incubation with <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. (<b>a</b>) Representative fluorescence-activated cell sorting plots for γH2AX. The <span class="html-italic">y</span>-axis indicates γH2AX staining, and the <span class="html-italic">x</span>-axis is the DNA content. (<b>b</b>) Percentage of cells with γH2AX staining. All assays were performed in triplicate. (<b>c</b>) Percentage of cell cycle distribution (G1, S, and G2/M). All assays were performed in triplicate. Data are presented as the mean ± standard deviation (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>Biodistribution of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in BxPC-3-bearing mice. (<b>a</b>) Pharmacokinetics of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. Data are expressed as % ID/g for organs and blood and as % ID for carcass, urine, and feces. Data are shown as the mean ± standard deviation (<span class="html-italic">n</span> = 3–4). (<b>b</b>) Alpha camera imaging of intratumoral distribution and corresponding hematoxylin and eosin images. The scale bars indicate 100 μm. ID, injected dose.</p>
Full article ">Figure 5
<p>Therapeutic efficacy of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in BxPC-3-bearing mice. (<b>a</b>) Individual tumor responses. Each solid color line represents a tumor from a single mouse. (<b>b</b>) Relative tumor growth of the mice groups treated with a single dose of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> compared to the control group (untreated). Data are shown as the mean ± standard deviation. (<b>c</b>) Kaplan–Meier survival curves of the mice treated with <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. Log-rank (Mantel–Cox) test; <span class="html-italic">p</span> = 0.0192, hazard ratio [HR] 2.415, 95% CI 0.7639–7.636 (control vs. 20 kBq); <span class="html-italic">p</span> = 0.0014, HR 3.342, 95% CI 0.9631–11.60 (control vs. 40 kBq); <span class="html-italic">p</span> = 0.0002, HR 3.774, 95% CI 1.042–13.67 (control vs. 65 kBq); <span class="html-italic">p</span> = 0.0009, HR 3.786, 95% CI 1.062–13.49 (control vs. 90 kBq). (<b>d</b>) Change in body weight after administration of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>. Data are shown as the mean ± standard deviation.</p>
Full article ">Figure 6
<p>Cytotoxicity, cell cycle, and γH2AX expression by low-dose <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> after 4 h of treatment with high-dose (5 or 10 kBq/mL) of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub> in BxPC-3 and PANC-1 cells. (<b>a</b>) Cell viability. The white, grey, and blue columns indicate the pretreatment with 0, 5, and 10 kBq/mL of <sup>225</sup>Ac-DOTA-RGD<sub>2</sub>, respectively. (<b>b</b>) Percentage of cell cycle distribution. (<b>c</b>) Time course of γH2AX expression. The significance of γH2AX expression at each time point was compared to 0, 5, or 10 kBq/mL as the control in each graph. Data represent the mean ± standard deviation (<span class="html-italic">n</span> = 2–4). * <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 ">
Article
The Frequency of DPYD c.557A>G in the Dominican Population and Its Association with African Ancestry
by Mariela Guevara, Carla González de la Cruz, Fernanda Rodrigues-Soares, Ernesto Rodríguez, Caíque Manóchio, Eva Peñas-Lledó, Pedro Dorado and Adrián LLerena
Pharmaceutics 2025, 17(1), 8; https://doi.org/10.3390/pharmaceutics17010008 (registering DOI) - 24 Dec 2024
Abstract
Background/Objectives: Genetic polymorphism of the dihydropyrimidine dehydrogenase gene (DPYD) is responsible for the variability found in the metabolism of fluoropyrimidines such as 5-fluorouracil (5-FU), capecitabine, or tegafur. The DPYD genotype is linked to variability in enzyme activity, 5-FU elimination, and toxicity. [...] Read more.
Background/Objectives: Genetic polymorphism of the dihydropyrimidine dehydrogenase gene (DPYD) is responsible for the variability found in the metabolism of fluoropyrimidines such as 5-fluorouracil (5-FU), capecitabine, or tegafur. The DPYD genotype is linked to variability in enzyme activity, 5-FU elimination, and toxicity. Approximately 10–40% of patients treated with fluoropyrimidines develop severe toxicity. The interethnic variability of DPYD gene variants in Afro-Latin Americans is poorly studied, thereby establishing a barrier to the implementation of personalized medicine in these populations. Therefore, the present study aims to analyze the frequency of DPYD variants with clinical relevance in the Dominican population and their association with genomic ancestry components. Methods: For this study, 196 healthy volunteers from the Dominican Republic were genotyped for DPYD variants by qPCR, and individual genomic ancestry analysis was performed in 178 individuals using 90 informative ancestry markers. Data from the 1000 Genomes project were also retrieved for comparison and increased statistical power. Results and Conclusions: The c.557A>G variant (decreased dihydropyrimidine dehydrogenase function) presented a frequency of 2.6% in the Dominican population. Moreover, the frequency of this variant is positively associated with African ancestry (r2 = 0.67, p = 1 × 10−7), which implies that individuals with high levels of African ancestry are more likely to present this variant. HapB3 is completely absent in Dominican, Mexican, Peruvian, Bangladeshi, and all East Asian and African populations, which probably makes its analysis dispensable in these populations. The implementation of pharmacogenetics in oncology, specifically DPYD, in populations of Afro-Latin American ancestry should include c.557A>G, to be able to carry out the safe and effective treatment of patients treated with fluoropyrimidines. Full article
(This article belongs to the Section Pharmacokinetics and Pharmacodynamics)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">DPYD</span> c.557A&gt;G allele frequencies as a function of African ancestry proportions in the 1000 Genomes project populations, plus the Dominican Republic population. East Asian: CDX: Dai Chinese from Xishuangbanna, CHS: Han Chinese from the south, CHB: Han Chinese from Beijing, JPT: Japanese from Tokyo; European: TSI: Italians from Tuscany, IBS: Iberians from Spain, GBR: British from England and Scotland, CEU: European descendants from Utah, FIN: Finns from Finland; South Asian: PJL: Pakistanis, GIH: Gujarati Indians in Houston USA, ITU: Telugu Indians in the UK, STU: Sri Lankan Tamils in the UK, BEB: Bengalis from Bangladesh, and KHV: Kinh Vietnamese from Ho Chi Minh City.</p>
Full article ">Figure 2
<p>World map presenting the frequency of c.557A&gt;G in 1000 Genomes project populations and in the Dominican Republic. EAS: Dai Chinese from Xishuangbanna, Han Chinese from the South, Han Chinese from Beijing, Japanese from Tokyo; EUR: Italians from Tuscany, Iberians from Spain, British from England and Scotland, European descendants from Utah, Finns from Finland; ESN: Esan from Nigeria, LWK: Luhya from Kenya, YRI: Yoruba from Nigeria, MSL: Mande from Sierra Leone, GWD: Mandinka from Gambia, ASW: African-Americans from the southwestern United States, MXL: Mexican descent from Los Angeles, ACB: Afro-Caribbeans from Barbados, CLM: Colombians from Medellín, PEL: Peruvians from Lima, DR: Dominican Republic (present study).</p>
Full article ">
25 pages, 11158 KiB  
Article
Computer-Aided Construction and Evaluation of Poly-L-Lysine/Hyodeoxycholic Acid Nanoparticles for Hemorrhage and Infection Therapy
by Qin Qin, Wenxing Wu, Ling Che, Xing Zhou, Diedie Wu, Xiaohui Li, Yumin Yang and Jie Lou
Pharmaceutics 2025, 17(1), 7; https://doi.org/10.3390/pharmaceutics17010007 (registering DOI) - 24 Dec 2024
Abstract
Background: Traumatic hemorrhage and infection are major causes of mortality in wounds caused by battlefield injuries, hospital procedures, and traffic accidents. Developing a multifunctional nano-drug capable of simultaneously controlling bleeding, preventing infection, and promoting wound healing is critical. This study aimed to design [...] Read more.
Background: Traumatic hemorrhage and infection are major causes of mortality in wounds caused by battlefield injuries, hospital procedures, and traffic accidents. Developing a multifunctional nano-drug capable of simultaneously controlling bleeding, preventing infection, and promoting wound healing is critical. This study aimed to design and evaluate a nanoparticle-based solution to address these challenges effectively. Methods: Using a one-pot assembly approach, we prepared a series of nanoparticles composed of poly-L-lysine and hyodeoxycholic acid (PLL-HDCA NPs). Theoretical simulations and experimental studies were combined to optimize their structure and functionality. In vitro platelet aggregation, antibacterial assays, cytotoxicity tests, and hemolysis evaluations were performed. In vivo efficacy was assessed in various hemorrhage models, a full-thickness skin defect model, and a skin irritation test. Results: PLL-HDCA NPs demonstrated effective induction of platelet aggregation and significantly reduced bleeding time and blood loss in mouse models, including tail vein, femoral vein, artery, and liver bleeding. Antibacterial assays revealed strong activity against E. coli and S. aureus. Wound healing studies showed that PLL-HDCA NPs promoted tissue repair in a full-thickness skin defect model. Cytotoxicity and hemolysis tests indicated minimal impact on human cells and significantly reduced hemolysis rates compared to PLL alone. Skin irritation tests confirmed the safety of PLL-HDCA NPs for external application. Conclusions: PLL-HDCA NPs represent a safe, efficient, and multifunctional nano-drug suitable for topical applications to control bleeding, combat infection, and facilitate wound healing, making them promising candidates for use in battlefield and hospital settings. Full article
(This article belongs to the Special Issue Nanoformulations for Local Treatment of Cancer, Infections and Wounds)
Show Figures

Figure 1

Figure 1
<p>Molecular structures of cholic acid derivatives.</p>
Full article ">Figure 2
<p>The characterization and computer simulation of PLL-cholic acid derivatives NPs. (<b>a</b>) Visual inspection of PLL-cholic acid derivatives complexes. (<b>b</b>) Adsorption energies between PLL and cholic acid derivatives. (<b>c</b>) The hydrophilic and lipophilic surfaces of PLL-cholic acid derivatives complexes after MD simulation. The green area represents hydrophobic, and the purple area represents hydrophilic. (<b>d</b>) The proportion of hydrophobic regions was statistically analyzed on the amphiphilic map of PLL-cholic acid derivative complexes. (<b>e</b>) Count rate distribution. (<b>f</b>) Zeta-potential distribution. (<b>g</b>) The TEM image of PLL-CHE\HDCA\OBE NPs. (<b>h</b>) The particle size distribution. (<b>i</b>) The dynamic mesostructure of PLL-cholic acid derivatives complexes during 20 ns DPD simulation. PLL beads were represented as red colors. HDCA, CHE, and OBE beads were, respectively, represented as blue, purple and yellow colors. Data are mean ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Molecular modeling and experimental characterization of interactions between PLL and HDCA, OBE, CHE. (<b>a</b>) Various binding energies calculated by Autodock 4.2. The binding interaction mainly depends on van der Waals force and electrostatic interaction. (<b>b</b>) 2D images displaying the lowest energy conformation and the interaction of PLL and HDCA, OBE, CHE, respectively. (<b>c</b>) The lowest energy 3D conformation of PLL/HDCA, OBE, CHE complexes. The Blue dotted line (red arrows) indicates H-bonding. (<b>d</b>) Characterization of PLL/HDCA, OBE, CHE forces by FT-IR spectroscopy.</p>
Full article ">Figure 4
<p>The characterization of PLL-HDCA NPs. (<b>a</b>–<b>c</b>) The stability of PLL-HDCA NPs was examined by evaluating the particle size (<b>a</b>), zeta potential (<b>b</b>), and PDI (<b>c</b>). (<b>d</b>) XRD patterns of HDCA, PLL, and PLL-HDCA NPs. The red arrows indicate the peak position. Data are mean ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>Hemostatic properties of PLL-HDCA NPs in vitro. (<b>a</b>) The representative image of coagulation assay. (<b>b</b>) Proportion of blood clotting area of various samples. (<b>c</b>) Statistical analysis of blood clotting time of various samples. (<b>d</b>) Fluorescence microscope images of platelets after being treated with various specimens. The red arrows indicate aggregated platelets. (<b>e</b>) Statistical analysis of platelet aggregation area. Data are mean ± SD (<span class="html-italic">n</span> = 3) of independent experiments; * <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, ns = not significantly different.</p>
Full article ">Figure 6
<p>Hemostatic effects of topical administrations of PLL-HDCA NPs in different bleeding models. (<b>a</b>) Digital photos of hemorrhage at various stages before and after treatment with PLL-HDCA NPs. (<b>b</b>–<b>d</b>) Hemostatic action in tail vein hemorrhage model. (<b>b</b>) Schematic illustration, (<b>c</b>) bleeding time, and (<b>d</b>) blood loss in vein hemorrhage model. (<b>e</b>–<b>g</b>) Hemostatic action in femoral vein hemorrhage model. (<b>e</b>) Schematic illustration, (<b>f</b>) bleeding time, and (<b>g</b>) blood loss in femoral vein hemorrhage model. (<b>h</b>–<b>j</b>) Hemostatic action in femoral artery hemorrhage model. (<b>h</b>) Schematic illustration, (<b>i</b>) bleeding time, and (<b>j</b>) blood loss in femoral artery hemorrhage model. (<b>k</b>–<b>m</b>) Hemostatic action in hepatic hemorrhage model. (<b>k</b>) Schematic illustration, (<b>l</b>) bleeding time, and (<b>m</b>) blood loss in hepatic hemorrhage model. Data are mean ± SD (<span class="html-italic">n</span> = 5) of independent experiments; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001, ns = not significantly different.</p>
Full article ">Figure 7
<p>Antibacterial activity of PLL-HDCA NPs. (<b>a</b>) Plate counting images of <span class="html-italic">E. coli</span> and <span class="html-italic">S. aureus</span> after treatment with HDCA, PLL, PLL-HDCA NPs, using PBS treatment as control. (<b>b</b>) Statistical analysis of <span class="html-italic">E. coli</span> and <span class="html-italic">S. aureus</span> viable count after treatment with HDCA, PLL, PLL-HDCA NPs, using PBS treatment as control. (<b>c</b>) Morphology of <span class="html-italic">E. coli</span> and <span class="html-italic">S. aureus</span> after co-culturing with samples at a concentration corresponding to the PLL (0.375 mg/mL)/(0.1875 mg/mL), respectively. (<b>d</b>) The integrity of <span class="html-italic">E. coli</span> and <span class="html-italic">S. aureus</span> cell membrane was determined by flow cytometry after co-culturing with samples at a concentration corresponding to the PLL (0.375 mg/mL)/(0.1875 mg/mL), respectively. Data are mean ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 8
<p>Wound healing assay to evaluate wound closure ability of PLL-HDCA NPs at different time points. (<b>a</b>) Morphology of wound healing at different time points. (<b>b</b>) Schematic diagram that mimicking wound healing process. (<b>c</b>). Wound healing rate of the samples at the set time point. (<b>d</b>) H&amp;E-stained pictures of wound skin tissue on days 7 after treatment (scale bar = 100 μm). Blue arrows represent newly generated blood vessels. Data are mean ± SD (<span class="html-italic">n</span> = 3) of independent experiments; ns = not significantly different.</p>
Full article ">Figure 9
<p>Biocompatibility test of PLL-HDCA NPs. (<b>a</b>) Cytotoxicity assay of PLL-HDCA NPs. (<b>b</b>) Hemolysis assay of PLL-HDCA NPs. (<b>c</b>) Hemolysis rate statistic of PLL-HDCA NPs. (<b>d</b>) Changes in skin appearance of mice after skin irritation test: the normal saline (zone 1), PLL/HDCA/PLL-HDCA NPs (zone 2). (<b>e</b>) Changes in body weight of mice after skin irritation test. Data are mean ± SD (<span class="html-italic">n</span> = 3) of independent experiments; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
17 pages, 1496 KiB  
Article
Selective Laser Sintering 3D Printing of Carvedilol Tablets: Enhancing Dissolution Through Amorphization
by Nikola Pešić, Branka Ivković, Tanja Barudžija, Branka Grujić, Svetlana Ibrić and Djordje Medarević
Pharmaceutics 2025, 17(1), 6; https://doi.org/10.3390/pharmaceutics17010006 (registering DOI) - 24 Dec 2024
Abstract
Background/Objectives: Selective laser sintering (SLS) is one of the most promising 3D printing techniques for pharmaceutical applications as it offers numerous advantages, such as suitability to work with already approved pharmaceutical excipients, the elimination of solvents, and the ability to produce fast-dissolving, porous [...] Read more.
Background/Objectives: Selective laser sintering (SLS) is one of the most promising 3D printing techniques for pharmaceutical applications as it offers numerous advantages, such as suitability to work with already approved pharmaceutical excipients, the elimination of solvents, and the ability to produce fast-dissolving, porous dosage forms with high drug loading. When the powder mixture is exposed to elevated temperatures during SLS printing, the active ingredients can be converted from the crystalline to the amorphous state, which can be used as a strategy to improve the dissolution rate and bioavailability of poorly soluble drugs. This study investigates the potential application of SLS 3D printing for the fabrication of tablets containing the poorly soluble drug carvedilol with the aim of improving the dissolution rate of the drug by forming an amorphous form through the printing process. Methods: Using SLS 3D printing, eight tablet formulations were produced using two different powder mixtures and four combinations of experimental conditions, followed by physicochemical characterization and dissolution testing. Results: Physicochemical characterization revealed that at least partial amorphization of carvedilol occurred during the printing process. Although variations in process parameters were minimal, higher temperatures in combination with lower laser speeds appeared to facilitate a greater degree of amorphization. Ultimately, the partial conversion to the amorphous form significantly improved the dissolution of carvedilol compared to its pure crystalline form. Conclusions: Obtained results suggest that the SLS 3D printing technique can be effectively used to convert poorly water-soluble drugs to their amorphous state, thereby improving solubility and bioavailability. Full article
(This article belongs to the Special Issue 3D Printing of Drug Delivery Systems)
Show Figures

Figure 1

Figure 1
<p>SLS printed tablets of powder blend 1 (<b>a</b>) and powder blend 2 (<b>b</b>) under varying printing conditions (surface temperature (°C)/chamber temperature (°C)/laser speed (mm/s) from left to right: 80/70/60, 80/70/50, 90/80/60, 90/80/50).</p>
Full article ">Figure 2
<p>Tablets of powder blend 1 (<b>a</b>) and powder blend 2 (<b>b</b>) produced under the following printing conditions: surface temperature (°C)/chamber temperature (°C)/laser speed (mm/s) of 90 °C/80 °C/50 mm/s (right side in (<b>a</b>,<b>b</b>)) and 80 °C/70 °C/60 mm/s (left side in (<b>a</b>,<b>b</b>)).</p>
Full article ">Figure 3
<p>Diffractograms of carvedilol and powdered tablets obtained from powder blend 1 (<b>a</b>) and powder blend 2 (<b>b</b>) (printing parameters are given in the brackets in the following order: surface temperature (°C), chamber temperature (°C), and laser speed (mm/s)).</p>
Full article ">Figure 4
<p>Thermograms of carvedilol and powdered tablets obtained from powder blend 1 (<b>a</b>), and powder blend 2 (<b>b</b>) (printing parameters are given in the brackets in the following order: surface temperature (°C), chamber temperature (°C), and laser speed (mm/s)).</p>
Full article ">Figure 5
<p>FT-IR spectra of carvedilol and powdered tablets obtained from powder blend 1 (<b>a</b>) and powder blend 2 (<b>b</b>) (printing parameters are given in the brackets in the following order: surface temperature (°C), chamber temperature (°C), and laser speed (mm/s)).</p>
Full article ">Figure 6
<p>In vitro release profiles of carvedilol from printed tablets from powder blend 1 (<b>a</b>) and powder blend 2 (<b>b</b>) in comparison with pure active substance (printing parameters are given in the brackets in the following order: surface temperature (°C), chamber temperature (°C), and laser speed (mm/s)).</p>
Full article ">
15 pages, 2802 KiB  
Article
Development and Characterization of Trihexyphenidyl Orodispersible Minitablets: A Challenge to Fill the Therapeutic Gap in Neuropediatrics
by Camila Olivera, Oriana Boscolo, Cecilia Dobrecky, Claudia A. Ortega, Laura S. Favier, Valeria A. Cianchino, Sabrina Flor and Silvia Lucangioli
Pharmaceutics 2025, 17(1), 5; https://doi.org/10.3390/pharmaceutics17010005 - 24 Dec 2024
Abstract
Background: Trihexyphenidyl (THP) has been widely used for over three decades as pediatric pharmacotherapy in patients affected by segmental and generalized dystonia. In order to achieve effective and safe pharmacotherapy for this population, new formulations are needed. Objective: The aim of this work [...] Read more.
Background: Trihexyphenidyl (THP) has been widely used for over three decades as pediatric pharmacotherapy in patients affected by segmental and generalized dystonia. In order to achieve effective and safe pharmacotherapy for this population, new formulations are needed. Objective: The aim of this work is the development of trihexyphenidyl orodispersible minitablets (ODMTs) for pediatric use. Methods: Six different excipients were tested as diluents. The properties of powder mixtures were evaluated before direct compression and pharmacotechnical tests were performed on the final formulation. The determination of the API content, uniformity of dosage, and physicochemical stability studies were analyzed by an HPLC-UV method. Results: The developed ODMTs met pharmacopeia specifications for content, hardness, friability, disintegration, and dissolution tests. The physicochemical stability study performed over 18 months shows that API content remains within 90.0–110.0% at least for this period. Conclusions: These ODMTs will allow efficient, safe, and high-quality pharmacotherapy. Full article
Show Figures

Figure 1

Figure 1
<p>Trihexyphenidyl chemical structure.</p>
Full article ">Figure 2
<p>Color scale according to powder rheological parameter results obtained for each physical mixture (diluent + API).</p>
Full article ">Figure 3
<p>Mixing time optimization.</p>
Full article ">Figure 4
<p>(<b>A</b>) Contrast image API/excipient blend 400×; (<b>B</b>) API/excipient blend elemental mapping; (<b>C</b>) nitrogen mapping; (<b>D</b>) chloride mapping.</p>
Full article ">Figure 5
<p>ATR-FTIR analysis of THP and its respective co-process excipient and powder blend. (<b>A</b>) Star Lac; (<b>B</b>) Cellactose 80; (<b>C</b>) Pharmaburst; (<b>D</b>) Mannogem 1080; (<b>E</b>) Avicel C15; (<b>F</b>) Avicel RC 591.</p>
Full article ">Figure 6
<p>Scanning electron micrographs of (<b>A</b>) Star-Lac, (<b>B</b>) THP, (<b>C</b>) API/excipient blend 200×, and (<b>D</b>) API/excipient blend 1000×.</p>
Full article ">Figure 7
<p>THP ODMT dissolution profile.</p>
Full article ">
29 pages, 6039 KiB  
Article
Innovative Solid Lipid Nanoparticle-Enriched Hydrogels for Enhanced Topical Delivery of L-Glutathione: A Novel Approach to Anti-Ageing
by Mengyang Liu, Manisha Sharma, Guoliang Lu, Zhiwen Zhang, Wenting Song and Jingyuan Wen
Pharmaceutics 2025, 17(1), 4; https://doi.org/10.3390/pharmaceutics17010004 - 24 Dec 2024
Abstract
Background: Skin ageing, driven predominantly by oxidative stress from reactive oxygen species (ROS) induced by environmental factors like ultraviolet A (UVA) radiation, accounts for approximately 80% of extrinsic skin damage. L-glutathione (GSH), a potent antioxidant, holds promise in combating UVA-induced oxidative stress. However, [...] Read more.
Background: Skin ageing, driven predominantly by oxidative stress from reactive oxygen species (ROS) induced by environmental factors like ultraviolet A (UVA) radiation, accounts for approximately 80% of extrinsic skin damage. L-glutathione (GSH), a potent antioxidant, holds promise in combating UVA-induced oxidative stress. However, its instability and limited penetration through the stratum corneum hinder its topical application. This study introduces a novel solid lipid nanoparticle (SLN)-enriched hydrogel designed to enhance GSH stability, skin penetration, and sustained release for anti-ageing applications. Methods: GSH-loaded SLNs were prepared via a double-emulsion technique and optimized using factorial design. These SLNs were incorporated into 1–3% (w/v) Carbopol hydrogels to produce a semi-solid formulation. The hydrogel’s characteristics, including morphology, mechanical and rheological properties, drug release, stability, antioxidant activity, cytotoxicity, and skin penetration, were evaluated. Results: SEM and FTIR confirmed the uniform dispersion of SLNs within the hydrogel. The formulation exhibited desirable properties, including gel strength (5.1 ± 0.5 g), spreadability (33.6 ± 1.9 g·s), pseudoplasticity, and elasticity. In vitro studies revealed a biphasic GSH release profile, with sustained release over 72 h and over 70% cumulative release. The hydrogel significantly improved antioxidant capacity, protecting human fibroblasts from UVA-induced oxidative stress and enhancing cell viability. Stability studies indicated that 4 °C was optimal for storage over three months. Notably, the hydrogel enhanced GSH penetration through the stratum corneum by 3.7-fold. Conclusions: This SLN-enriched hydrogel effectively improves GSH topical delivery and antioxidant efficacy, providing a promising platform for anti-ageing and other bioactive compounds with similar delivery challenges. Full article
(This article belongs to the Special Issue Advances in Delivery of Peptides and Proteins)
Show Figures

Figure 1

Figure 1
<p>The SEM image of dry GSH-SLN-EH (where the black area is the mesh size of dry cross-linked hydrogels).</p>
Full article ">Figure 2
<p>FTIR spectra of GSH, GSH-SLN dry powder, gelling polymers (Carbopol 971), GSH -SLN-EH, and dry GSH-SLN-EH.</p>
Full article ">Figure 3
<p>Gel strength (positive peak), adhesion (negative area), and stickiness (negative peak) of (<b>a</b>) commercial control gel; (<b>b</b>) GSH-SLN-EH with 1% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside; (<b>c</b>) GSH-SLN-EH with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside; and (<b>d</b>) GSH-SLN-EH with 5% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside.</p>
Full article ">Figure 4
<p>Gel spreadability (positive peak area) of (<b>a</b>) commercial control gel; (<b>b</b>) GSH-SLN-EH with 1% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside; (<b>c</b>) GSH-SLN-EH with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside; and (<b>d</b>) GSH-SLN-EH with 5% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside.</p>
Full article ">Figure 5
<p>Rheograms of four tested gels. (<b>a</b>) Merino<sup>®</sup> 97% Pure Aloe Vera Gel (control); (<b>b</b>) GSH-SLN-EH with 1% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971; (<b>c</b>) GSH-SLN-EH with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971; (<b>d</b>) GSH-SLN-EH with 5% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971.</p>
Full article ">Figure 6
<p>Viscograms of four tested gels. (<b>a</b>) Merino<sup>®</sup> 97% Pure Aloe Vera Gel (control); (<b>b</b>) GSH-SLN-EH with 1% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside; (<b>c</b>) GSH-SLN-EH with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside; (<b>d</b>) GSH-SLN-EH with 5% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside.</p>
Full article ">Figure 7
<p>Influence of oscillation rate on both storage and loss modulus in Merino<sup>®</sup> 97% Pure Aloe Vera Gel.</p>
Full article ">Figure 8
<p>Influence of oscillation rate on both storage and loss modulus in GSH-SLN-EH with 1% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside.</p>
Full article ">Figure 9
<p>Influence of oscillation rate on both storage and loss modulus in GSH-SLN-EH with 3% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside.</p>
Full article ">Figure 10
<p>Influence of oscillation rate on both storage and loss modulus in GSH-SLN-EH with 5% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) Carbopol 971 inside.</p>
Full article ">Figure 11
<p>Release profiles of all samples of GSH, namely GSH solution, GSH-loaded hydrogels, GSH-SLN suspension, and GSH-SLN-EH with and without additional GSH in hydrogels (Mean ± S.D. n = 3).</p>
Full article ">Figure 12
<p>Release profiles of (<b>a</b>) GSH hydrogels; (<b>b</b>) GSH-SLN suspension; (<b>c</b>) GSH-SLN-EH without additional GSH in hydrogels; (<b>d</b>) GSH-SLN-EH with additional GSH in hydrogels compared with GSH solution (Mean ± S.D. n = 3).</p>
Full article ">Figure 13
<p>The microscopic images of Fbs cells after 72 h. (<b>a</b>) Fbs cells without any GSH formulations; (<b>b</b>) Fbs cells treated with GSH solution; (<b>c</b>) Fbs cells treated with GSH-SLN suspension; (<b>d</b>) Fbs cells treated with GSH-SLN-EH.</p>
Full article ">Figure 14
<p>Effect of three formulations of GSH on the Fbs cell viability. Cells were incubated for 24, 48, and 72 h; control is cell culture medium only without any treatment of GSH (<span class="html-italic">p</span> value ˂ 0.05) (Mean ± SD, n = 5).</p>
Full article ">Figure 15
<p>The micrographs of two control groups of Fbs cells, treated without (<b>a</b>) and with (<b>b</b>) UVA irradiation after culture for 72 h; the black circles in figure b represent dead Fbs cells.</p>
Full article ">Figure 16
<p>The micrographs of Fbs cells treated with a half hour of UVA irradiation after 72 h. (<b>a</b>) Fbs cells without any GSH formulations; (<b>b</b>) Fbs cells treated with GSH solution; (<b>c</b>) Fbs cells treated with GSH-SLN suspension; (<b>d</b>) Fbs cells treated with GSH-SLN-EH.</p>
Full article ">Figure 17
<p>Cytoprotective effects of GSH formulations at three time intervals (24, 48, and 72 h). The control group (2) was exposed to UVA irradiation without GSH treatment. Cell viability increased significantly for all GSH formulations (<span class="html-italic">p</span> value &lt; 0.05) (Mean ± SD, n = 5).</p>
Full article ">Figure 18
<p>Cumulative permeation data for GSH solution, GSH-SLN, GSH-hydrogel, and GSH-SLN-EH formulations over 48 h (Mean ± S.D., n = 3).</p>
Full article ">Figure 19
<p>Human skin deposition of GSH in SC and epi/dermis layers for five formulations after 48 h: GSH solution, GSH-SLNs, GSH-hydrogels, and GSH-SLN-EH with and without additional GSH in hydrogels, respectively (Mean ± S.D., n = 3).</p>
Full article ">
17 pages, 5873 KiB  
Article
Injectable Tumoricidal Neural Stem Cell-Laden Hydrogel for Treatment of Glioblastoma Multiforme—An In Vivo Safety, Persistence, and Efficacy Study
by Jasmine L. King, Alain Valdivia, Shawn D. Hingtgen and S. Rahima Benhabbour
Pharmaceutics 2025, 17(1), 3; https://doi.org/10.3390/pharmaceutics17010003 - 24 Dec 2024
Abstract
Background/Objectives: Glioblastoma multiforme (GBM) is the most common high-grade primary brain cancer in adults. Despite efforts to advance treatment, GBM remains treatment resistant and inevitably progresses after first-line therapy. Induced neural stem cell (iNSC) therapy is a promising, personalized cell therapy approach that [...] Read more.
Background/Objectives: Glioblastoma multiforme (GBM) is the most common high-grade primary brain cancer in adults. Despite efforts to advance treatment, GBM remains treatment resistant and inevitably progresses after first-line therapy. Induced neural stem cell (iNSC) therapy is a promising, personalized cell therapy approach that has been explored to circumvent challenges associated with the current GBM treatment. Methods: Herein, we developed a chitosan-based (CS) injectable, biodegradable, in situ forming thermo-responsive hydrogel as a cell delivery vehicle for the treatment of GBM. Tumoricidal neural stem cells were encapsulated in the injectable CS hydrogel as stem cell therapy for treatment of post-surgical GBM. In this report, we investigated the safety of the injectable CS hydrogel in an immune-competent mouse model. Furthermore, we evaluated the persistence and efficacy of iNSC-laden CS hydrogels in a post-surgical GBM mouse model. Results: The injectable CS hydrogel was well tolerated in mice with no signs of chronic local inflammation. Induced neural stem cells (iNSCs) persisted in the CS hydrogels for over 196 days in comparison to 21 days for iNSCs (cell injection) only. GBM recurrence was significantly slower in mice treated with iNSC-laden CS hydrogels with a 50% increase in overall median survival in comparison to iNSCs (cell injection) only. Conclusions: Collectively, we demonstrated the ability to encapsulate, retain, and deliver iNSCs in an injectable CS hydrogel that is well tolerated with better survival rates than iNSCs alone. Full article
(This article belongs to the Section Physical Pharmacy and Formulation)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the injectable CS hydrogel under physiological conditions.</p>
Full article ">Figure 2
<p>Schematic illustration depicting the transdifferentiation (TD) process of primary fibroblast to induced neural stem cells for implantation.</p>
Full article ">Figure 3
<p>Surgical procedure for in vivo persistence studies. (<b>1</b>) In step 1, a surgical incision was created to expose the intact skull, and an intracranial window (craniotomy) was created in the right hemisphere of the parietal skull plate using a microsurgical drill. (<b>2</b>) In step 2, using a surgical scope, an aspiration device was used to remove brain tissue to create a mock surgical resection cavity. (<b>3</b>) In step 3, a 5 µL of CS hydrogel solution containing 25,000 iNSCs was implanted into the resection cavity. The iNSC-CS hydrogel was given 1–2 min to settle before closing the wound with Vetbond tissue adhesive (3M 1469SB).</p>
Full article ">Figure 4
<p>In vivo retention and persistence post-implantation of iNSCs via direct injection or seeded in CS hydrogels. (<b>A</b>) In vivo persistence study design. (<b>B</b>) Representative BLI images collected up until signal loss for each group. (<b>C</b>,<b>D</b>) Summary graphs demonstrating the FLuc signal from iNSCs directly injected or encapsulated in CS hydrogel following delivery into the resection cavity. iNSC survival was represented as total FLuc signal from day 1 (* indicates <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA).</p>
Full article ">Figure 5
<p>Histology and safety of mice brain tissue following the resection and the CS hydrogel implantation. Histological images of brain tissue on days 3, 7, 30, 60, and 90 following the resection (n = 3/timepoint) and post-implantation of the CS hydrogel (n = 5/timepoint). (<b>A</b>,<b>B</b>) Resection control histological images at day 3 (<b>A</b>) and (<b>B</b>) a zoomed-in image of (<b>A</b>). (<b>C</b>,<b>D</b>) CS hydrogel histological images at day 3 (<b>C</b>) and (<b>D</b>) a zoomed-in image of (<b>C</b>). (<b>E</b>,<b>F</b>) Resection control histological images at day 7 (<b>E</b>) and (<b>F</b>) a zoomed-in image of (<b>E</b>). (<b>G,H</b>) CS hydrogel histological images at day 7 (<b>G</b>) and (<b>H</b>) a zoomed-in image of (<b>G</b>). (<b>I</b>,<b>J</b>) Resection control histological images at day 30 (<b>I</b>) and (<b>J</b>) a zoomed-in image of (<b>I</b>). (<b>K</b>,<b>L</b>) CS hydrogel histological images at day 30 (<b>K</b>) and (<b>L</b>) a zoomed-in image of (<b>K</b>). (<b>M</b>,<b>N</b>) Resection control histological images at day 60 (<b>M</b>) and (<b>N</b>) a zoomed-in image of (<b>M</b>). (<b>O</b>,<b>P</b>) CS hydrogel histological images at day 60 (<b>O</b>) and (<b>P</b>) a zoomed-in image of (<b>O</b>). (<b>Q</b>,<b>R</b>) Resection control histological images at day 90 (<b>Q</b>) and (<b>R</b>) a zoomed-in image of (<b>Q</b>). (<b>S</b>,<b>T</b>) CS hydrogel histological images at day 90 (<b>S</b>) and (<b>T</b>) a zoomed-in image of (<b>S</b>). R represents resection site, H represents hemorrhage (trauma-related), E represents edema (trauma-related), yellow circles represent swollen axons (trauma-related), and blue triangles represent pigment-laden macrophages. All scale bars represent 500 µm. All scale bars for zoomed-in images represent 50 µm.</p>
Full article ">Figure 6
<p>(<b>A</b>) Schematic illustration of tumor implantation, resection, and cell injection/implantation in vivo. (<b>B</b>) Fluorescent images of U87-MG mCh-FLuc tumors before and after resection and images of resection cavity using an Olympus MVX-10 microscope (1.6× magnification). White arrows represent positive tumor margins following resection.</p>
Full article ">Figure 7
<p>Delivery of tumoricidal iNSCs to inhibit progression of GBM in post-resection model. (<b>A</b>) Representative images of serial BLI showing tumor inhibition and regrowth in iNSC-sTR treated versus control-treated animals. (<b>B</b>) Summary graph depicting the tumor radiance of U87-MG FLuc overtime following post-resection treatment (* indicates <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.005; *** <span class="html-italic">p</span> &lt; 0.001 by ANOVA). (<b>C</b>) Kaplan–Meier survival analysis demonstrating the survival of animals receiving iNSC-sTR therapy in comparison to control-treated animals (*** <span class="html-italic">p</span> &lt; 0.001 by log-rank test).</p>
Full article ">
26 pages, 4557 KiB  
Article
Ethanolic Extract of Averrhoa carambola Leaf Has an Anticancer Activity on Triple-Negative Breast Cancer Cells: An In Vitro Study
by Oscar F. Beas-Guzmán, Ariana Cabrera-Licona, Gustavo A. Hernández-Fuentes, Silvia G. Ceballos-Magaña, José Guzmán-Esquivel, Luis De-León-Zaragoza, Mario Ramírez-Flores, Janet Diaz-Martinez, Idalia Garza-Veloz, Margarita L. Martínez-Fierro, Iram P. Rodríguez-Sanchez, Gabriel Ceja-Espíritu, Carmen Meza-Robles, Víctor H. Cervantes-Kardasch and Iván Delgado-Enciso
Pharmaceutics 2025, 17(1), 2; https://doi.org/10.3390/pharmaceutics17010002 - 24 Dec 2024
Abstract
Background/Objectives: Averrhoa carambola, or star fruit, is a shrub known for its medicinal properties, especially due to bioactive metabolites identified in its roots and fruit with anti-cancer activity. However, the biological effects of its leaves remain unexplored. This study aimed to [...] Read more.
Background/Objectives: Averrhoa carambola, or star fruit, is a shrub known for its medicinal properties, especially due to bioactive metabolites identified in its roots and fruit with anti-cancer activity. However, the biological effects of its leaves remain unexplored. This study aimed to assess the effects of ethanolic extract from A. carambola leaves on triple-negative breast cancer (TNBC), an aggressive subtype lacking specific therapy. Methods: Phytochemical analysis and HPLC profile and additional cell line evaluation employing MDA-MB-231 were carried out. Results: Phytochemical screening revealed that the ethanolic extract was rich in flavonoids, saponins, and steroids, demonstrating an antioxidant capacity of 45%. 1H NMR analysis indicated the presence of flavonoids, terpenes, and glycoside-like compounds. Cell viability assays showed a concentration-dependent decrease in viability, with an IC50 value of 20.89 μg/mL at 48 h. Clonogenic assays indicated significant inhibition of replicative immortality, with only 2.63% survival at 15 μg/mL. Migration, assessed through a wound healing assay, was reduced to 3.06% at 100 μg/mL, with only 16.23% of cells remaining attached. An additive effect was observed when combining lower concentrations of the extract with doxorubicin, indicating potential synergy. Conclusions: These results suggest that the ethanolic extract of A. carambola leaves contains metabolites with anti-cancer activity against TNBC cells, supporting further research into their bioactive compounds and therapeutic potential. Full article
(This article belongs to the Special Issue Pharmaceutical Applications of Plant Extracts, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Chromatograms obtained at 290 nm from HPLC analysis. (<b>A</b>) Chromatogram of standards: gallic acid (GA, Rt 2.385 min), cinnamic acid (CA, Rt 30.795 min), anthrone (ANT, Rt 20.000 min), quercetin (Q, Rt 17.955 min), and 4-methylumbelliferone (4-ML, Rt 10.908 min). (<b>B</b>) Chromatogram of the ethanolic extract of <span class="html-italic">A. carambola</span> (500 ppm). (<b>C</b>) Chromatogram of the hydrolysate of the leaves of <span class="html-italic">A. carambola</span> (500 ppm). S: signal.</p>
Full article ">Figure 2
<p>Viability experiments employing ethanolic extract of <span class="html-italic">A. carambola</span> on MDA-MB-231 cells. (<b>A</b>) No changes in viability were observed in cervical cancer cell line TC-1 exposed to A. carambola extract in increasing concentrations. (<b>B</b>) A concentration-dependent effect was observed on MDA-MB-231 cell line exposed to the extract. (<b>C</b>) The ethanolic extract of <span class="html-italic">A. carambola</span> leaves had an experimental IC<sub>50</sub> of 20.83 μg/mL in triple-negative breast cancer cell line. (<b>D</b>) Morphological changes and detached cells were observed from the concentration of 25 μg/mL of ethanolic extract. Magnification 10×. The <span class="html-italic">p</span>-values correspond to significant differences compared to the control, DMEM-F12 medium with 0.1% DMSO, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Ethanolic extract of <span class="html-italic">A. carambola</span> leaves decreases replicative immortality of MDA-MB-231 cells. (<b>A</b>) Photographs depict the number of colonies formed after the exposition of each treatment. It is observed that a concentration-dependent effect completely inhibits cell survival. (<b>B</b>) The graph shows the percentage of survival treatment. The <span class="html-italic">p</span>-values correspond to significant differences compared to the control, only DMEM medium, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Ethanolic extract of <span class="html-italic">A. carambola</span> leaves interferes with MDA-MB-231 cell migration. (<b>A</b>) Images captured at 48 h of the wound area made in MDA-MB-231 cell monolayers. Magnification 4×. (<b>B</b>) The graph shows the changes in the open area; a concentration-dependent inhibitory effect can be observed at 48 h that was superior to the doxorubicin effect. Comparison to 48 h control, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>The ethanolic extract of <span class="html-italic">A. carambola</span> leaves affects the cell adhesion of MDA-MB-231 cells. (<b>A</b>) The micrographs show the adhesive capacity of cells recovered after exposure to <span class="html-italic">A. carambola</span> extract and reseeded for 24 h. The adhesive capacity decreases as the concentration of the extract increases. Magnification 10×. (<b>B</b>) The graphs show the percentage of cells adhered to the monolayer after being treated with the extract for 48, showing a concentration-dependent decrease in adhesion. (<b>C</b>) The graph shows the percentage of cell death after 48 h of treatment. (<b>D</b>) The graph shows the percentage of adhesion of detached cells after treatment that were recovered and reseeded. The <span class="html-italic">p</span>-values correspond to significant changes compared to the control, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>The combination of a low dose of doxorubicin and intermediate doses of <span class="html-italic">A. carambola</span> extract reduces the cell viability of MDA-MB-231 cells. The graph shows the reduction in cell viability induced by the different combinations after 48 h of treatment. An additive effect was observed between the 1/5 IC<sub>50</sub> dose of doxorubicin (DOX) and the three tested concentrations of the extract. <sup>a</sup> 0.4 μM DOX + 15 μg/mL extract vs. 15 μg/mL of the extract, <sup>b</sup> 0.4 μM DOX + 25 μg/mL vs. 25 μg/mL, <sup>c</sup> 0.4 μM DOX + 50 μg/mL vs. 50 μg/mL, <sup>d</sup> 2 μM DOX + 15 μg/mL vs. 15 μg/mL, <sup>e</sup> 2 μM DOX + 25 μg/mL vs. 25 μg/mL, <sup>f</sup> 2 μM DOX + 50 μg/mL vs. 50 μg/Ml, <sup>a’</sup> 0.4 μM DOX + 15 μg/mL vs. 0.4 μM DOX, <sup>b’</sup> 0.4 μM DOX + 25 μg/mL vs. 0.4 μM DOX <sup>c’</sup> 0.4 μM DOX + 50 μg/mL vs. 0.4 μM DOX, <sup>d’</sup> 2 μM DOX + 15 μg/mL vs. 2 μM DOX, <sup>e’</sup> 2 μM DOX + 25 μg/mL vs. 2 μM DOX, <sup>f’</sup> 2 μM DOX + 50 μg/mL vs. 2 μM DOX, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
17 pages, 5477 KiB  
Article
Development and Optimization of Nano-Hydroxyapatite Encapsulating Tocotrienol-Rich Fraction Formulation Using Response Surface Methodology
by Anis Syauqina Mohd Zaffarin, Shiow-Fern Ng, Min Hwei Ng, Haniza Hassan and Ekram Alias
Pharmaceutics 2025, 17(1), 10; https://doi.org/10.3390/pharmaceutics17010010 (registering DOI) - 24 Dec 2024
Abstract
Background/Objective: The tocotrienol-rich fraction (TRF) is a lipid-soluble vitamin that has good antioxidant and anti-inflammatory properties. The TRF is widely studied as a potential treatment for various diseases, including bone diseases. However, its application is limited due to its poor oral bioavailability [...] Read more.
Background/Objective: The tocotrienol-rich fraction (TRF) is a lipid-soluble vitamin that has good antioxidant and anti-inflammatory properties. The TRF is widely studied as a potential treatment for various diseases, including bone diseases. However, its application is limited due to its poor oral bioavailability profile, warranting an innovative approach to overcome its pharmacokinetic limitations. Recently, the nano-hydroxyapatite (nHA) has been investigated as a drug delivery vehicle for various drugs and active compounds owing to its excellent biocompatibility, biodegradability, and osteogenic properties. The nHA is also a well-known biomaterial which has chemical and structural similarities to bone minerals. Hence, we aim to explore the use of the nHA as a potential nanocarrier for the TRF. Methods: In this study, we develop and optimize the formulation of an nHA-encapsulating TRF (nHA/TRF) by employing the response surface methodology (RSM). Results: RSM outcomes reveal that the mass of the nHA, the concentration of the TRF, and the incubation time have a significant effect on the particle size, zeta potential, and encapsulation efficiency of the nHA/TRF. The outcomes for the optimized formulation are not significantly different from the predicted RSM outcomes. The optimized nHA/TRF formulation is freeze-dried and results in an average particle size of ~270 nm, a negative zeta potential value of ~−20 mV, a polydispersity index of <0.4, and an encapsulation efficiency of ~18.1%. Transmission electron microscopy (TEM) shows that the freeze-dried nHA/TRF has a spherical structure. Conclusions: Taken together, the above findings indicate that the nHA may be established as a nanocarrier for efficient delivery of the TRF, as demonstrated by the promising physical properties. Full article
Show Figures

Figure 1

Figure 1
<p>The experimental set-up for the ultrasonication process during the preparation of nHA/TRF.</p>
Full article ">Figure 2
<p>The normal probability plot of the residuals of (<b>a</b>) particle size, (<b>b</b>) zeta potential, and (<b>c</b>) encapsulation efficiency, indicating the normally distributed experimental values.</p>
Full article ">Figure 3
<p>Response surface plots showing the effect of the interaction between the (<b>a</b>) mass of the nHA and the concentration of the TRF with a fixed incubation time of 14.23 h, (<b>b</b>) mass of the nHA and the incubation time with a fixed TRF concentration of 2.96 mg/mL, and (<b>c</b>) concentration of the TRF and the incubation time with a fixed nHA mass of 200 mg on the particle size of the nHA/TRF.</p>
Full article ">Figure 4
<p>Response surface plots showing the effect of the interaction between the (<b>a</b>) mass of the nHA and the concentration of the TRF with a fixed incubation time of 14.23 h, (<b>b</b>) mass of the nHA and the incubation time with a fixed TRF concentration of 2.96 mg/mL, and (<b>c</b>) concentration of the TRF and the incubation time with a fixed nHA mass of 200 mg on the zeta potential of the nHA/TRF.</p>
Full article ">Figure 5
<p>Response surface plots showing the effect of the interaction between the (<b>a</b>) mass of the nHA and the concentration of the TRF with a fixed incubation time of 14.23 h, (<b>b</b>) mass of the nHA and the incubation time with a fixed TRF concentration of 2.96 mg/mL, and (<b>c</b>) concentration of the TRF and the incubation time with a fixed nHa mass of 200 mg on the encapsulation efficiency of the nHA/TRF.</p>
Full article ">Figure 6
<p>Physical appearance of the freeze-dried nHA/TRF with 10% mannitol (% <span class="html-italic">w</span>/<span class="html-italic">v</span>).</p>
Full article ">Figure 7
<p>Particle size (<b>a</b>), polydispersity index (<b>b</b>), and zeta potential (<b>c</b>) of the unloaded nHA and nHA/TRF. Data expressed as the mean ± SEM (<span class="html-italic">n</span> = 3) (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 8
<p>Transmission electron micrograph of the (<b>a</b>) unloaded nHA and (<b>b</b>) nHA/TRF at 50,000× magnification.</p>
Full article ">
13 pages, 3856 KiB  
Article
Inhibition of Aβ Aggregation by Cholesterol-End-Modified PEG Vesicles and Micelles
by Shota Watanabe, Motoki Ueda and Shoichiro Asayama
Pharmaceutics 2025, 17(1), 1; https://doi.org/10.3390/pharmaceutics17010001 - 24 Dec 2024
Abstract
Background/Objectives: This study aimed to design and evaluate Chol-PEG2000 micelles and Chol-PEG500 vesicles as drug delivery system (DDS) carriers and inhibitors of amyloid-β (Aβ) aggregation, a key factor in Alzheimer’s disease (AD). Methods: The physical properties of Chol-PEG assemblies [...] Read more.
Background/Objectives: This study aimed to design and evaluate Chol-PEG2000 micelles and Chol-PEG500 vesicles as drug delivery system (DDS) carriers and inhibitors of amyloid-β (Aβ) aggregation, a key factor in Alzheimer’s disease (AD). Methods: The physical properties of Chol-PEG assemblies were characterized using dynamic light scattering (DLS), electrophoretic light scattering (ELS), and transmission electron microscopy (TEM). Inhibitory effects on Aβ aggregation were assessed via thioflavin T (ThT) assay, circular dichroism (CD) spectroscopy, and native polyacrylamide gel electrophoresis (native-PAGE). Results: Chol-PEG2000 micelles and Chol-PEG500 vesicles were found to exhibit diameters of 20–30 nm and 70–80 nm, respectively, with neutral surface charges and those physical properties indicated the high affinity for Aβ. At a 10-fold molar ratio, thioflavin T (ThT) assay revealed that Chol-PEG2000 delayed Aβ fibril elongation by 20 hours, while Chol-PEG500 delayed it by 40 hours against Aβ peptide. At a 50-fold molar ratio, both Chol-PEG2000 and Chol-PEG500 significantly inhibited Aβ aggregation, as indicated by minimal fluorescence intensity increases over 48 hours. CD spectroscopy indicated that Aβ maintained its random coil structure in the presence of Chol-PEG assemblies at a 50-fold molar ratio. Native-PAGE analysis demonstrated a retardation in Aβ migration immediately after mixing with Chol-PEG assemblies, suggesting complex formation. However, this retardation disappeared within 5 min, implying rapid dissociation of the complexes. Conclusions: This study demonstrated that Chol-PEG500 vesicles more effectively inhibit Aβ aggregation than Chol-PEG2000 micelles. Chol-PEG assemblies perform as DDS carriers to be capable of inhibiting Aβ aggregation. Chol-PEG assemblies can deliver additional therapeutics targeting other aspects of AD pathology. This dual-function platform shows promise as both a DDS carrier and a therapeutic agent, potentially contributing to a fundamental cure for AD. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

Figure 1
<p>Particle sizes and zeta potentials of Chol-PEGs assemblies at each concentration used in later Aβ experiments. All concentrations of Chol-PEGs assemblies were above the critical aggregation concentration (CAC).</p>
Full article ">Figure 2
<p>TEM images of assembly of (<b>A</b>,<b>C</b>) Chol-PEG<sub>2000</sub> and (<b>B</b>,<b>D</b>) Chol-PEG<sub>500</sub> at the concentration of (<b>A</b>,<b>B</b>) 470 μM above CAC and (<b>C</b>,<b>D</b>) 4.7 × 10<sup>−5</sup> μM below CAC.</p>
Full article ">Figure 3
<p>ThT assay results of Aβ<sub>40</sub> aggregation in the presence of each concentration of Chol-PEGs. (<b>A</b>) Aβ<sub>40</sub> incubated with Chol-PEG<sub>2000</sub>, (<b>B</b>) Aβ<sub>40</sub> incubated with Chol-PEG<sub>500</sub>, (<b>C</b>) Aβ<sub>40</sub> incubated with mPEG<sub>2000</sub>-NH<sub>2</sub> and Chol-PEG<sub>2000</sub> alone (without Aβ<sub>40</sub>), (<b>D</b>) Aβ<sub>40</sub> incubated with mPEG<sub>500</sub>-NH<sub>2</sub> and Chol-PEG<sub>500</sub> alone (without Aβ<sub>40</sub>). The final concentration of Chol-PEGs at Chol-PEG/Aβm = 10 and Chol-PEG/Aβ = 50 is 94 µM and 470 µM, respectively.</p>
Full article ">Figure 4
<p>CD spectra of Aβ<sub>40</sub> and Chol-PEG mixtures. (<b>A</b>,<b>B</b>) CD spectra immediately after mixing (0 h) of Aβ<sub>40</sub> with (<b>A</b>) Chol-PEG<sub>2000</sub> or (<b>B</b>) Chol-PEG<sub>500</sub>, and their respective controls, mPEG<sub>2000</sub>-NH<sub>2</sub> and mPEG<sub>500</sub>-NH<sub>2</sub>. (<b>C</b>,<b>D</b>) CD spectra after 48 h of incubation (48 h) of Aβ<sub>40</sub> with (<b>C</b>) Chol-PEG<sub>2000</sub> or (<b>D</b>) Chol-PEG<sub>500</sub>, and their respective controls.</p>
Full article ">Figure 5
<p>Polyacrylamide gel electrophoresis of Aβ<sub>40</sub> incubated with Chol-PEGs (<b>A</b>) just after mixing Aβ<sub>40</sub> and Chol-PEG assemblies, (<b>B</b>) after incubation for 48 h. (<b>C</b>) Magnified results of repeating the experiment in (<b>A</b>) five times, (<b>D</b>) magnified results of repeating the experiment in (<b>B</b>) five times.</p>
Full article ">Figure 6
<p>Hypothesis on the mechanism by which Chol-PEG inhibits Aβ<sub>40</sub> aggregation.</p>
Full article ">
Previous Issue
Back to TopTop